Skip to main content

Full text of "كتب في علم الميكروبيولوجي"

See other formats




5/1/06 9:41:41 AM 





rolnieduna 

MICROBIOLOGY 



edited by 

Arthur C Ouwehand 

Danisco Innovation 
Kantvik, Finland 

Elaine E. Vaughan 

Unilever R&D, Vlaardingen and 
Wageningen University, Wageningen, 

The Netherlands 




Taylor & Francis 

Taylor & Francis Group 

New York London 

Taylor St Francis is an imprint of the 

Taylor & Francis Group, an informa business 



DK2999 FM.indd 2 



4/25/06 12:54:06 PM 





DK2999_Discl.fm Page 1 Thursday, February 9, 2006 1 1:36 AM 




+ 







Published in 2006 by 
Taylor & Francis Group 
270 Madison Avenue 
New York, NY 10016 

© 2006 by Taylor & Francis Group, LLC 

No claim to original U.S. Government works 

Printed in the United States of America on acid-free paper 

10 987654321 

International Standard Book Number- 10: 0-8247-2641-3 (Hardcover) 
International Standard Book Number- 13: 978-0-8247-2641-6 (Hardcover) 

This book contains information obtained from authentic and highly regarded sources. Reprinted material is quoted with 
permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts have been made to publish 
reliable data and information, but the author and the publisher cannot assume responsibility for the validity of all materials 
or for the consequences of their use. 

No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic, mechanical, or 
other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any information 
storage or retrieval system, without written permission from the publishers. 

For permission to photocopy or use material electronically from this work, please access www.copyright.com 
(http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood Drive, Danvers, MA 
01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and registration for a variety of users. For 
organizations that have been granted a photocopy license by the CCC, a separate system of payment has been arranged. 

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for 
identification and explanation without intent to infringe. 




Library of Congress Cataloging-in-Publication Data 



Catalog record is available from the Library of Congress 



informa 

Taylor & Francis Group 
is the Academic Division of Informa pic. 



Visit the Taylor & Francis Web site at 
http : //www. taylor andfr ancis. com 









We dedicate this book to Willem M. de Vos and Seppo Salminen, who contribute 
significantly to insight in gut microbe-host interactions, and moreover, 

as our colleagues and mentors in this field. 



in 



Preface 



The human gastrointestinal tract microorganisms, termed the "microbiota," have been 
investigated since the beginning of microbiological studies, when Antonie van 
Leeuwenhoek, the father of microbiology, investigated the microorganisms in his own 
stools. The human microbiota comprises trillions of microbes distributed in various niches 
throughout the intestinal tract and is one of the most complex microbial ecosystems on 
earth. The host and its microbiota have co-evolved together, and considering the 
staggering numbers and diversity, it is therefore not surprising that the microbiota exert a 
major influence on the host. The original term for the microbiota upon discovery was the 
"flora" or "microflora," literally translated as "small plants," which has a botanical 
connotation. These terms are still used widely today and internationally recognized. 
Nevertheless, it is considered more appropriate to use the term microbiota, i.e., "small 
life" taking into account that the human microbiota is comprised of bacteria, archaea, 
bacteriophage, a smaller number of yeasts, and some protozoa; hence, this term is mainly 
used throughout this book. With this book, we have made an attempt to cover all issues 
associated with the gastrointestinal microbiota, from health to disease and from sampling 
to identification. Although various books have addressed the intestinal microbiota, this 
has mainly been from the perspective of disease or nutrition, while the microbiota itself 
has rarely been the focus. This current book aims to fill this gap and provide the reader 
with a comprehensive overview of all aspects related to the gastrointestinal microbiota. 
There have been major scientific advances especially in human intestinal microbiology in 
the recent past, which are also covered by the contributions. 

Early studies were limited to description of the culturable microbes, which as we 
now realize, made up only a minority of the gastrointestinal tract microbiota. Due to the 
development of molecular biological techniques over the last decade, microbes can now 
be detected and studied to a large extent, without the need for culturing. In the first chapter, 
Kaouther Ben Amor and Elaine Vaughan review the major achievements of recent times in 
determining the diversity of the microbiota using modern molecular techniques, based on 
16S ribosomal RNA, as well as methods to evaluate their activity within the various 
niches. Research of the gastrointestinal tract microbiota, especially in the case of humans, 
is often restricted to fecal material. In fact, a range of other sampling techniques are 
available, which are presented by Angele Kerckhoffs and colleagues, to access the small 
intestine, as well as noninvasive sampling methods that are routinely used in medical 
practice. This is an important issue since feces represent only the luminal material of the 
terminal colon and will provide insufficient information about other locations of the gut. 
Anne McCartney and Glenn Gibson describe the succession of the microbiota in infants, as 
well as the earlier culturing studies, and the methodology to characterize the microbiota 



vi Preface 

down to subspecies level. It has long been recognized that the intestinal microbiota plays 
an important role in maintaining health in infants. Currently, much attention is also 
focused on the intestinal microbiota of the elderly, as is discussed in the chapter by Fang 
He. In western nations, the elderly are becoming a more numerous segment of the 
population, and it is becoming increasingly established that intestinal health has a major 
role in their quality of life. 

While establishing the microbiota diversity and their activity (live versus dead) is a 
major challenge, it is essential to know and understand their effects on the host. The 
intestinal microbiota has a major influence on the development and maintenance of our 
immune system as described by Marie-Christiane Moreau. Because of their direct contact 
with the host, the activity and interaction of the microbiota with the intestinal mucosa may 
be more important than the activity of microbes in the lumen, as described by Wai Ling 
Chow and Yuan-Kun Lee. The human microbiota also play a major role in our nutrition. 
Barry Goldin reviews the myriad of metabolic possibilities of the human microbiota 
concerning the metabolism of food ingredients and drugs we consume, as well as host- 
derived substrates. Max Bingham focuses on the metabolism by the microbiota of 
polyphenols, which are considered to be key active constituents of fruits and vegetables 
and responsible for many of the health protective effects of diets rich in these foods. 
Today, functional genomics technologies are developing and will facilitate our ability to 
detect the microbes and determine the molecular mechanisms of their impact on the host. 
Through the sequencing of an ever-increasing number of microbiota genomes, and elegant 
molecular studies, a further understanding is being obtained into the molecular functioning 
of the host-microbiota interactions, a dynamic area that is discussed by Peter Bron, Willem 
de Vos, and Michiel Kleerebezem. 

The gastrointestinal tract microbiota is receiving more attention than ever in 
particular in relation to disease. Fergus Shanahan, Barbara Sheil, and coworkers review 
the relationship between the intestinal microbiota and inflammatory bowel diseases, as 
well as give an overview of the probiotic clinical trials and the potential mechanisms of 
probiotics for ameliorating these intestinal diseases. Through its metabolism, the intestinal 
microbiota is thought to play an important role in both the etiology and prevention of 
colorectal cancer, as discussed by Patricia Heavey, Ian Rowland, and Joseph Rafter. In 
addition to diseases of the gastrointestinal tract, Pirkka Kirjavainen and Gregor Reid also 
discuss that diseases such as allergy are being recognized to have an "intestinal 
component," again mediated through the interaction between the microbiota and the 
intestinal immune system. 

In order to gain a better understanding of the composition and functioning of the 
intestinal microbiota and how this can be influenced, intestinal models have been 
developed; this allows for a simplification of the complex intestinal ecosystem as presented 
by Harri Makivuokko and Paivi Nurminen. Experimental animals, as described by 
Anders Henriksson, have also been highly valuable for this purpose, especially with the 
availability of various knockout animal models for disease. Also the use of animals with a 
"human" microbiota provide valuable models to investigate the influence of substances 
on the microbiota and host physiology. The best animal models to show the importance of 
the intestinal microbiota are germ-free animals. Their physiological differences compared 
to conventional animals are striking and show clearer than any other model the role 
intestinal microbes play, as discussed by Elisabeth Norin and Tore Midtvedt. Because of its 
influence on the health and well being of the host, strategies have been devised to alter the 
composition and/or activity of the intestinal microbiota. Antibiotics have long been known 
to alter the composition of the intestinal microbiota, as discussed by Asa Sullivan and Carl 
Erik Nord, which may lead to various side effects, depending on the activity spectrum of the 



Preface vii 

antibiotic. Methods to improve the activity and composition of the intestinal microbiota 
include probiotics, microbes ingested orally that provide beneficial effects, and prebiotics 
substrates that are selectively metabolized by the beneficial native gastrointestinal tract 
microbes, as discussed in the chapters by Chandraprakash Khedkar and Arthur Ouwehand, 
and Ross Crittenden and Martin Playne, respectively. 

The major part of the book deals with the microbiota of humans, and when animals 
are studied, it is often as a model for humans. Minna Rinkinen describes the microbiota of 
companion animals, an area that has received very little attention to date, although the 
well being of pets can contribute significantly to the well being of the owner. In the case of 
farm animals, discussed by Alojz Bomba and colleagues, there is an important economic 
drive where the role of the microbiota on performance is a major focus. This will only 
become more important from 2006 onward as antimicrobial growth promoters will be 
prohibited in the European Union. 

Gastrointestinal Microbiology is a vibrant field of research that is benefiting from 
many interdisciplinary interactions between different research groups in the world, that are 
using, developing, and applying novel technologies. Exciting initiatives are emerging with 
high through put technologies such as sequence analysis of the human microbiome 
(collective genomes of the gut microbiota) and metabolomics applied to microbiota and 
nutritional research. There is occasionally some overlap in information scattered 
throughout the book that is valuable since the reader will get an appreciation for the 
different opinions and perspectives that reflect the current state of research findings in the 
literature for this subject. It remains a highly complex task to understand the mutual 
relationship between members of the microbial community in the gut and their interaction 
with the host. 

Finally, we hope that all readers will share our excitement for this dynamic subject 
that impacts on all our lives. 

Arthur C. Ouwehand 
Elaine E. Vaughan 



Acknowledgments 



We are most grateful to the contributing authors who have been willing to share their 
knowledge and experience in their field of intestinal microbiology. They are all busy 
researchers and yet they committed themselves to writing these chapters. It has been a 
pleasure to cooperate with these experts for the production of this book. Together their 
excellent contributions provide the state-of-the-art research on the human intestinal 
microbiology as well as informative chapters about the animal microbiota for comparative 
purposes. Elaine Vaughan acknowledges the staff and colleagues in Unilever Research and 
Development, and in the Laboratory of Microbiology, especially the Molecular Ecology 
Group, Wageningen University, for inspiring discussions on intestinal microbiology and 
critical support in this field. She further acknowledges the enjoyable and stimulating 
collaborations with the Wageningen Center of Food Sciences. Arthur Ouwehand similarly 
acknowledges the support and inspiration from the colleagues at Danisco Innovation and 
the Functional Foods Forum, University of Turku. We thank the Egerton Group Ltd. for 
their excellent support during production of this book. Importantly, our families, 
especially our spouses (Dr. Patrick Wouters and Anna-Maija Ouwehand), who have 
learned to live with Gut Microbiology, we give our heartfelt thanks. 



IX 



Contents 



Preface .... v 
Contributors .... xvii 



1. Molecular Ecology of the Human Intestinal Microbiota 

Kaouther Ben Amor and Elaine E. Vaughan 

Introduction .... 1 

GI-Tract Microbiota as Identified by 16S rRNA 

Gene Analysis .... 2 

Fingerprinting Reveals Characteristics of 

the Microbiota .... 5 
16S rRNA-Targeted Probes Quantify the GI-Tract 

Microbiota .... 7 
New Molecular Diversity Approaches .... 12 
Assessment of Microbiota Vitality and Metabolic 

Activity .... 13 
Perspectives .... 15 
References .... 16 



2. Sampling Microbiota in the Human Gastrointestinal Tract 25 

Angele P. M. Kerckhoffs, Melvin Samsom, Gerard P. van Berge Henegouwen, 
Louis M. A. Akkermans, Vincent B. Nieuwenhuijs, and Maarten R. Visser 
Introduction .... 25 
Esophagus: Microbiota and Sampling 

Techniques .... 28 
Stomach: Microbiota and Sampling Techniques .... 29 
Small Intestine: Microbiota and 

Sampling Techniques .... 32 
Large Intestine: Microbiota and Sampling 

Techniques .... 42 
Conclusion .... 45 
References .... 46 

xi 



xii Contents 

3. The Normal Microbiota of the Human Gastrointestinal Tract: 
History of Analysis, Succession, and Dietary Influences 51 

Anne L. McCartney and Glenn R. Gibson 

Introduction .... 51 

Role of the Gastrointestinal Microbiota 

in Humans .... 52 
Acquisition of the Gut Microbiota .... 54 
Composition of the Adult Fecal Microbiota Assessed 

by Culturing .... 55 
Composition of the Adult Fecal Microbiota Assessed 

by Molecular Techniques .... 58 
Investigations at the Subspecies Level .... 68 
Conclusion .... 69 
References .... 70 

4. The Intestinal Microbiota of the Elderly 75 

Fang He 

Introduction .... 75 

Colonization and Succession of Human Intestinal 

Microbiota with Age .... 75 
Bifidobacteria in the Elderly .... 79 
Conclusion .... 88 
References .... 88 

5. Immune Modulation by the Intestinal Microbiota 93 

Marie-Christiane Moreau 

Introduction .... 93 

Brief Review of the Intestinal Microbiota .... 94 

Brief Review of Immune Responses .... 95 

The Intestinal Immune System .... 97 

Relationships Between the Intestinal Immune System and 

Intestinal Microbiota .... 103 
Relationships Between the Peripheral Immune System and 

Intestinal Microbiota .... 112 
Conclusion .... 116 
References .... 117 

6. Mucosal Interactions and Gastrointestinal Microbiota 123 

Wai Ling Chow and Yuan-Kun Lee 

Introduction .... 123 

Features of the Gastrointestinal Tract .... 124 

Microbiota and Gastrointestinal System .... 127 

Development of GI Tract Normal Microbiota 

in Humans .... 129 
Cross-Talk Between Bacteria and Intestinal 

Epithelial Cells .... 131 
Conclusion .... 132 
References .... 133 



Contents xiii 

7. The Metabolism of Nutrients and Drugs by the Intestinal 
Microbiota 137 

Barry R. Goldin 

Introduction .... 137 

General Metabolism and Function 

of the Microbiota .... 137 
Nutrients and Dietary Plant Compounds .... 138 
Intestinal Bacterial Metabolism of Host Endogenously 

Synthesized Compounds .... 142 
Other Bacterial Reactions .... 143 
Bacterial Intestinal Formation of Mutagens .... 144 
Bacterial Intestinal Drug Metabolism .... 147 
Conclusion .... 150 
References .... 150 

8. The Metabolism of Polyphenols by the Human 

Gut Microbiota 155 

Max Bingham 

Summary .... 155 

Introduction .... 155 

Types of Polyphenols and Microbial 

Metabolism .... 156 
Phenolic Acids — Hydroxycinnamates and 

Hydroxybenzoates .... 157 
Flavonoids .... 159 
Anthocyanidins .... 162 
Proanthocyanidins .... 163 
Perspectives .... 163 
References .... 164 

9. Molecular Analysis of Host-Microbe Interactions in the 
Gastrointestinal Tract 169 

Peter A. Bron, Willem M. de Vos, and Michiel Kleerebezem 

Introduction .... 169 

Bacterial Responses to the Host .... 170 

Insights from Genomics .... 176 

In Situ Profiling of Transcription in the 

GI Tract 180 

Concluding Remarks and Future 

Perspectives .... 182 
References .... 183 



10. The Infant Intestinal Microbiota in Allergy 189 

Pirkka V. Kirjavainen and Gregor Reid 
Introduction .... 189 



xiv Contents 

Allergies — An Overview .... 190 
Allergy-Associated Compositional Characteristics of 

Infant Gut Microbiota .... 191 
Interpreting the Gut Microbiota 

Characteristics .... 194 
Conclusion .... 198 
References .... 199 

11. Probiotics: A Role in Therapy for Inflammatory 

Bowel Disease 207 

Barbara Sheil, Jane McCarthy, Liam O'Mahony, 
Malik M. Anwar, and Fergus Shanahan 
Introduction .... 207 
The Role of the Enteric Microbiota in the 

Normal Gut .... 208 
The Importance of the Enteric Microbiota in 

Inflammatory Bowel Disease .... 209 
Probiotics .... 211 
Efficacy of Probiotics in Inflammatory 

Bowel Disease .... 213 
Discussion .... 218 
References .... 219 

12. The Gastrointestinal Microbiota in Cancer 225 

Patricia M. Heavey, Ian R. Rowland, and Joseph J. Rafter 
Introduction .... 225 
The Stomach .... 226 
The Large Intestine .... 228 
Surrogate Markers for Diet-Related 
Colon Cancer Studies .... 230 
Conclusion .... 233 
References .... 234 

13. In Vitro Methods to Model the Gastrointestinal Tract 237 

Harri Mdkivuokko and Pdivi Nurminen 

Introduction .... 237 

Types of Intestinal Simulator Models .... 237 

Simulating the Rumen .... 244 

Conclusion .... 250 

References .... 250 

14. Animal Models for the Human Gastrointestinal Tract 253 

Anders Henriksson 
Introduction .... 253 



Contents xv 

Animal Models Used for Studies on the Human 

GI Microbiota .... 256 
Conclusion .... 265 
References .... 265 

15. Born Germ-Free — Microbial Dependent 273 

Elisabeth Norin and Tore Midtvedt 
Introduction .... 273 
Terminology .... 274 
Germ-Free Animals and Dietary 

Requirements .... 274 
Intestinal Microbiota, Gross Anatomy, Histology, 

and Motility .... 275 
Biochemical Functions and the Gastrointestinal 

Microbiota .... 276 
Immunology and Germ-Free Life .... 279 
Conclusion .... 281 
References .... 281 

16. Modifying the Human Intestinal Microbiota with Prebiotics .... 285 

Ross Crittenden and Martin J. Playne 
Introduction .... 285 

Why Modify the Intestinal Microbiota? .... 285 
The Prebiotic Strategy to Modifying the Intestinal 

Microbiota .... 286 
A Brief History of the Development of Bifidus 

Factors and Prebiotics .... 287 
Currently Available Prebiotic Carbohydrates .... 289 
Modifying the Intestinal Bifidobacterium 

Population .... 290 
Synbiotics .... 294 

Mechanisms of the Bifidogenic Effect .... 295 
Advantages of the Prebiotic Strategy .... 295 
Disadvantages of the Prebiotic Approach .... 300 
Safe Dosage Levels .... 300 
Conclusion and Future Directions .... 300 
References .... 302 

17. Modifying the Gastrointestinal Microbiota with Probiotics 315 

Chandraprakash D. Khedkar and Arthur C. Ouwehand 

Introduction .... 315 

Probiotics .... 316 

Proposed Health Benefits of Probiotics .... 317 

Modifying Intestinal Microbiota Composition Through 

Intake of Probiotics .... 322 
Modifying the Microbial Metabolic Activity .... 324 



xvi Contents 

Conclusion .... 327 
References .... 327 

18. Modifying the Intestinal Microbiota with Antibiotics 335 

Asa Sullivan and Carl Erik Nord 

Introduction .... 335 

Antimicrobial Agents That Inhibit the Synthesis 

of the Bacterial Cell Wall — (3-Lactam 

Antibiotics .... 336 
Other Agents with Inhibitory Effect on the Synthesis 

of the Cell Wall— Glycopeptides .... 348 
Antimicrobial Agents Interfering with the Synthesis 

of Proteins .... 349 
Agents Blocking the Metabolism of 

Folic Acid .... 355 
Antimicrobial Agents That Interfere with the Synthesis 

ofDNA 355 

Conclusion .... 362 
References .... 362 

19. The Intestinal Microbiota of Pets: Dogs and Cats 371 

Minna Rinkinen 

Introduction .... 371 

Development of Intestinal Microbiota in 

Dogs and Cats .... 371 
Canine and Feline Gastrointestinal 

Microbiota .... 372 
Modifying the Intestinal Microbiota: Pre- and 

Probiotics .... 375 
Conclusion .... 376 
References .... 376 

20. The Gastrointestinal Microbiota of Farm Animals 381 

Alojz Bomba, Zuzana Jonecovd, Sofia Gancarcikovd, and 

Radomira Nemcovd 

Introduction .... 381 

Microbiota of the Gastrointestinal Tract 

in Farm Animals .... 382 
Influencing the Ecosystem of the Digestive Tract in 

Farm Animals .... 387 
The Use of Gnotobiotic Animals in Studies of the 

Gastrointestinal Microbiota in Farm Animals .... 393 
Conclusion .... 395 
References .... 395 



Index .... 401 



Contributors 



Louis M. A. Akkermans Department of Surgery, Utrecht University Medical Center, 
Utrecht, The Netherlands 

Kaouther Ben Amor Laboratory of Microbiology, Wageningen University, 
Wageningen, The Netherlands 

Malik M. Anwar Alimentary Pharmabiotic Centre, Departments of Medicine 
and Surgery, Microbiology, National Food Biotechnology Centre, National University 
of Ireland, Cork, Ireland 

Max Bingham Unilever Research and Development, Vlaardingen, The Netherlands 

Alojz Bomba Institute of Gnotobiology and Prevention of Diseases in Young, 
University of Veterinary Medicine, Kosice, Slovak Republic 

Peter A. Bron Wageningen Centre for Food Sciences and NIZO Food Research, 
BA Ede, Wageningen, The Netherlands 

Wai Ling Chow National University of Singapore, Department of Microbiology, 
Faculty of Medicine, Singapore 

Ross Crittenden The Preventative Health Flagship, Food Science Australia, 
Werribee, Victoria, Australia 

Willem M. de Vos Wageningen Centre for Food Sciences and Laboratory of 
Microbiology, Wageningen University, Wageningen, The Netherlands 

Sofia Gancarcikova Institute of Gnotobiology and Prevention of Diseases in Young, 
University of Veterinary Medicine, Kosice, Slovak Republic 

Glenn R. Gibson Food Microbial Sciences Unit, School of Food Biosciences, 
Whiteknights, University of Reading, Reading, U.K. 

Barry R. Goldin Department of Public Health and Family Medicine, Tufts University 
School of Medicine, Boston, Massachusetts, U.S.A. 

Fang He Technical Research Laboratory, Takanashi Milk Products Co., Ltd, 
Yokohama, Kanagawa, Japan 

Patricia M. Heavey School of Life Sciences, Kingston University, Kingston-upon- 
Thames, U.K. 

xvii 



xviii Contributors 

Gerard P. van Berge Henegouwen Department of Gastroenterology, Utrecht 
University Medical Center, Utrecht, The Netherlands 

Anders Henriksson DSM Food Specialties, Sydney, Australia 

Zuzana Jonecova Institute of Gnotobiology and Prevention of Diseases in Young, 
University of Veterinary Medicine, Kosice, Slovak Republic 

Angele P. M. Kerckhoffs Department of Gastroenterology, Utrecht University 
Medical Center, Utrecht, The Netherlands 

Chandraprakash D. Khedkar Department of Dairy Microbiology and Biotechnol- 
ogy (Maharashtra Animal and Fishery Sciences University, Nagpur), College of Dairy 
Technology, Warud (Pusad), India 

Pirkka V. Kirjavainen Canadian Research and Development Center for Probiotics, 
The Lawson Health Research Institute, London, Ontario, Canada 

Michiel Kleerebezem Wageningen Centre for Food Sciences and NIZO Food 
Research, BA Ede, Wageningen, The Netherlands 

Yuan-Kun Lee National University of Singapore, Department of Microbiology, 
Faculty of Medicine, Singapore 

Harri Makivuokko Danisco Innovation, Kantvik, Finland 

Jane McCarthy Alimentary Pharmabiotic Centre, Departments of Medicine 

and Surgery, Microbiology, National Food Biotechnology Centre, National University 

of Ireland, Cork, Ireland 

Anne L. McCartney Food Microbial Sciences Unit, School of Food Biosciences, 
Whiteknights, University of Reading, Reading, U.K. 

Tore Midtvedt Microbiology and Tumor Biology Center, Karolinska Institutet, 
Stockholm, Sweden 

Marie-Christiane Moreau French Institute of Agronomical Research (INRA), 
Nancy, France 

Radomira Nemcova Institute of Gnotobiology and Prevention of Diseases in Young, 
University of Veterinary Medicine, Kosice, Slovak Republic 

Vincent B. Nieuwenhuijs Department of Surgery, Utrecht University Medical Center, 
Utrecht, The Netherlands 

Carl Erik Nord Department of Laboratory Medicine, Karolinska University Hospital, 
Karolinska Institutet, Stockholm, Sweden 

Elisabeth Norin Microbiology and Tumor Biology Center, Karolinska Institutet, 
Stockholm, Sweden 

Paivi Nurminen Danisco Innovation, Kantvik, Finland 



Contributors xix 

Liam O'Mahony Alimentary Pharmabiotic Centre, Departments of Medicine and 
Surgery, Microbiology, National Food Biotechnology Centre, National University of 
Ireland, Cork, Ireland 

Arthur C. Ouwehand Danisco Innovation, Kantvik, and Functional Foods Forum, 
University of Turku, Turku, Finland 

Martin J. Playne Melbourne Biotechnology, Hampton, Victoria, Australia 

Joseph J. Rafter Department of Medical Nutrition, Novum, Huddinge University 
Hospital, Karolinska Institutet, Stockholm, Sweden 

Gregor Reid Canadian Research and Development Center for Probiotics, The Lawson 
Health Research Institute, London, Ontario, Canada 

Minna Rinkinen Department of Clinical Veterinary Sciences, Faculty of Veterinary 
Medicine, University of Helsinki, Helsinki, Finland 

Ian R. Rowland Northern Ireland Center for Food and Health, University of Ulster, 
Coleraine, Northern Ireland, U.K. 

Melvin Samsom Department of Gastroenterology, Utrecht University Medical 
Center, Utrecht, The Netherlands 

Fergus Shanahan Alimentary Pharmabiotic Centre, Departments of Medicine and 
Surgery, National University of Ireland, Cork, Ireland 

Barbara Sheil Alimentary Pharmabiotic Centre, Departments of Medicine and 
Surgery, Microbiology, National Food Biotechnology Centre, National University 
of Ireland, Cork, Ireland 

o 

Asa Sullivan Department of Laboratory Medicine, Karolinska University Hospital, 
Karolinska Institutet, Stockholm, Sweden 

Elaine E. Vaughan Unilever Research and Development, Vlaardingen, and 
Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands 

Maarten R. Visser Department of Microbiology, Utrecht University Medical Center, 
Utrecht, The Netherlands 



1 



Molecular Ecology of the Human 
Intestinal Microbiota 



Kaouther Ben Amor 

Laboratory of Microbiology, Wageningen University Wageningen, The Netherlands 

Elaine E. Vaughan 

Unilever Research and Development, Vlaardingen, and Laboratory of Microbiology, 
Wageningen University, Wageningen, The Netherlands 



INTRODUCTION 

The human gastrointestinal (GI) tract is the home of a huge microbial assemblage, the 
microbiota, the vast extent of which is only now being revealed. The number of micro 
organisms within the intestine greatly exceeds human cells, resulting in one of the most 
diverse and dynamic microbial ecosystems. Relationships among the microbes, and 
between the microbiota and the host, have a profound influence on all concerned (1,2). The 
Gl-tract offers various niches with nutrients, those ingested and generated by the host, and 
a relatively non-hostile environment to the microbes. The microbiota play essential roles 
in a wide variety of nutritional, developmental, and immunological processes and 
therefore significantly contribute to the well being of the host (3-6). During the last 
decade, specific bacterial isolates, termed "probiotics," have been extensively used in an 
attempt to modulate the intestinal microbiota to benefit the host. Today, there is persuasive 
evidence for probiotics in prevention or treatment of a number of intestinal disorders in 
humans, especially for reducing bouts of diarrhea and providing relief for lactose 
intolerant individuals (7,8). In order to rationally use probiotics, prebiotics or other 
functional foods as therapeutic agents, in-depth knowledge of the structure, dynamics, and 
function of the bacterial populations of the Gl-tract microbiota is crucial. 

Studying the microbial ecology in the intestine involves determining the abundance 
and diversity of the microorganisms present, their activity within this niche, and their 
interactions with each other and their host (symbiosis, commensalism and pathogenicity). 
Although the human intestinal microbiota have been extensively investigated by culture- 
based methods more than any other natural ecosystem (9-11), our knowledge about the 
culturable fraction of this community is limited. This is essentially due to the challenges of 
obtaining pure cultures of intestinal inhabitants, which are hindered by the largely 
anaerobic nature of this community, and the paucity of suitable enrichment strategies to 
simulate intestinal conditions. The advent of molecular techniques based on 16S 

1 



2 Amor and Vaughan 

ribosomal RNA (rRNA) gene analysis is now allowing a more complete assessment of this 
complex microbial ecosystem by unraveling the extent of the diversity, abundance and 
population dynamics of this community (12,13). These techniques have extended our view 
of those microorganisms that have proven difficult to culture and which play an important 
role in gut physiology. This huge intestinal microbial reservoir is estimated to contain 
1000 bacterial species and as much as 10 14 cells (1,14). Besides studying the diversity, it is 
essential to identify these microbes based upon their eco-physiological traits, i.e., those 
that are functionally active versus those that are effectively redundant and play little or no 
role at a particular time or at a given site of the intestinal tract. The latter requires the 
development of approaches that monitor the activity of these microorganisms at the single 
cell level in their natural habitat. This chapter initially reviews molecular techniques to 
study the diversity of the microbiota, and subsequently highlights newly developed 
molecular methods to study the eco-physiology of the Gl-tract. 



GI-TRACT MICROBIOTA AS IDENTIFIED BY 16S rRNA 
GENE ANALYSIS 

The human Gl-tract microbiota comprise bacteria, archaea and eukarya. It is by far the 
bacteria that dominate and reach the highest cell density documented for any microbial 
ecosystem (1). The comparative analysis of environmentally retrieved nucleic acid 
sequences, most notably of rRNA molecules and the genes encoding them, has become 
the standard over the last decade for cultivation-independent assessment of bacterial 
diversity in environmental samples (Fig. 1) (15,16). The 16S rRNA gene comprises 
highly variable to highly conserved regions, and the differences in sequence are used to 
distinguish bacteria at different levels from species to domain and determine phylogenetic 
relationships. rRNA gene fragments are today routinely retrieved without prior 
cultivation of the microbes by constructing 16S ribosomal DNA (rDNA) libraries. The 
procedure is based upon polymerase chain reaction (PCR)-mediated amplification of 16S 
rRNA genes or gene fragments, isolated from the environmental sample, followed by 
segregation of individual gene copies by cloning into Escherichia coli. In this way a 
library of community 16S rRNA genes is generated, the composition of which can be 
estimated by screening clones, full or partial sequence analysis, and comparing them with 
adequate appropriate reference sequences in databases to infer their phylogenetic 
affiliation. Large databases of 16S rRNA gene sequence information (> 200,000 
sequences) for described as well as uncultured microorganisms are available, which 
provide a high-resolution platform for the assignment of those new sequences obtained in 
16S rDNA libraries. Databases harboring 16S rRNA sequences include the ARB software 
package (17), the Ribosomal Database project (http://rdp.cme.msu.edu/index.jsp) (18) 
and EMBL (www.embl-heidelberg.de/). 

Sequencing of 16S rDNA clone libraries generated from various sites of the Gl-tract 
including terminal ileum, colon, mucosa and feces have confirmed that relevant fractions 
of gut bacteria were derived from new, as yet undescribed bacterial phylotypes (19-23). 
Clearly the biases of culturing studies in the 1960s and 1970s such as incomplete 
knowledge of culture conditions and selectivity had prejudiced the outcome. The new 
molecular studies revealed that the vast majority of rDNA amplicons generated directly 
from fecal or biopsy samples of adults, originated from the phyla of the Firmicutes 
(including the large class of Clostridia and the lactic acid bacteria), Bacteroidetes, 
Actinobacteria (including Atopobium and Bifidobacterium spp.) and Proteobacteria 



Molecular Ecology of the Human Intestinal Microbiota 

DNA/rRNA of /& 

intestinal ecosystem// 



Comparative analysis 
by PCR-DGGE 



PCRon 16SrDNA 

or rRNA —^ 



Quantitative real time 
PCR to monitor 



GC clamp on 

PCR primer & PCR <■ 

:■ ii n i i :■ 



u 



it 



16SrRNA 
PCR products 



16srDNA 

clone libraries 

and sequence analysis 



Probe and primer 
design 



4 4 



G 



A 




<^=C> 



Identification 
and phylogeny 



Band extraction 
and identification 



fl 



Figure 1 PCR-based approaches to monitor the Gl-tract microbiota. The 16S rDNA or rRNA 
isolated from a Gl-tract sample may be amplified by (reverse transcriptase-) PCR using primers that 
target all or some bacteria. The amplicons may be cloned and sequenced in order to identify the 
bacteria present in the sample. The 16S rRNA gene comprises highly variable to highly conserved 
regions, and the differences in sequence are used to determine phylogenetic relationships and 
distinguish bacteria at different levels from species to domain. The DGGE technique is based on 
16S rRNA sequence-specific melting behavior of the PCR products, generated with primers one of 
which contains a 40-bp GC clamp. Statistical software enables the calculation of similarity indices 
and cluster analysis to compare the samples. The 16S rRNA sequences may also be used to design 
new primers specific for bacterial groups or species in order to quantify them in samples by real 
time PCR. Abbreviations: DGGE, denaturing gradient gel electrophoresis; DNA, deoxyribonucleic 
acid; GC, guanine cytosin; PCR, polymerase chain reaction; rDNA, ribosomal deoxyribonucleic 
acid; rRNA, ribosomal ribonucleic acid. 



(including Escherichia coli). The large class of Clostridia comprises the Clostridium 
coccoides-Eubacterium rectale group, and the Clostridium leptum group consists of 
Ruminococcus species and Faecalibacterium prausnitzii. These analyses indicated that 
the adult intestinal microbiota constitutes a majority of low and high G + C content 
Gram-positive bacteria. The latter has been indirectly confirmed by analysis of the 
metagenome of bacterial viruses recovered from fecal samples that revealed 
predominantly viral sequences with similarity to genomes of bacteriophages specific for 
Gram-positive bacteria (24). In fact, this bacterial diversity at the division level relative to 
other microbial ecosystems is quite low, mainly deriving from the divisions Firmicutes 
and the Cytophaga-Flavobacterium-Bacteroides (9,19). 

Interestingly, molecular inventories based on 16S rDNA clone libraries of microbial 
communities in inflammatory bowel disease (IBD) patients differed from healthy 



4 Amor and Vaughan 

subjects (25). In several Crohn's disease (CD) patients numerous clones were isolated 
belonging to phylogenetic groups that are commonly not dominant in adult fecal 
microbiota of healthy persons, while Bacteroides vulgatus was the only molecular species 
shared by all patients, and E. coli clones were also detected unlike in healthy persons (25). 
In another study, 16S rDNA libraries generated from mucosa-associated microbiota of 
patients with IBD revealed a reduction in diversity due to a loss of normal anaerobic 
bacteria, especially those belonging to the Bacteroides, Eubacterium and Lactobacillus 
species. Most of the sequenced clones retrieved (70%) were assigned to known intestinal 
bacteria, but a significant number of the cloned sequences were affiliated to normal 
residents of the oral mucosa such as Streptococcus species (26). It was suggested that 
alteration of the microbiota in mucosal inflammation reflects a metabolic imbalance of the 
complex microbial ecosystem with severe consequences for the mucosal barrier rather 
than disrupted defense to single microorganisms (26). 

Even though sequencing of cloned 16S rDNA amplicons provides relevant 
information about the identity of uncultured bacteria, the data are not quantitative. 
Moreover, PCR and cloning steps are not without bias (27): a recent comparative 
analysis of clone libraries from a fecal sample pointed out that the number of PCR 
cycles may affect the diversity of the amplified 16S rDNAs and thus should be 
minimized (28). More rapid culture-independent options to the cloning procedures 
include exploring of the complex microbial populations using a variety of fingerprinting 
methods. See Table 1 for an overview of some current methods used to investigate the 
intestinal microbiota. 



Table 1 Potential and Limitations of Various Methods for Investigating the Diversity of the 
Human Intestinal Microbiota 



Method 



Application 



Comments 



Culturing 

16S rRNA gene 
libraries and 
sequencing 

Dot-blot 

hybridization 

FISH 



PCR-DGGE/TGGE 



T-RFLP 

Quantitative real 
time PCR 



Isolation of pure cul- 
tures, enumeration 

Identification and 
phylogeny 

Detection, quantifi- 
cation and activity 

Single cell detection 
and enumeration 

Rapid profiling of total 
microbiota 

Rapid profiling of total 
microbiota 

Detection and quanti- 
fication 



Not representative for microbiota; insufficient 
selective media; time consuming 

Large scale cloning is laborious; primer bias 
can be an issue 

Gives information about activity of microbiota; 

of rRNA; comprehensive set of probes 

published 
High throughput with image analysis software 

and flow cytometry; requires probe design; 

comprehensive set of probes published 
Detection of specific groups possible; semi- 
quantitative identification by band extraction 

and sequencing 
Identification by cloning and sequencing; bank 

of T-RF under construction 
Requires probe/primers design; very high 

throughput 



Abbreviations: DGGE, denaturing gradient gel electrophoresis; FISH, fluorescent in situ hybridization; PCR, 
polymerase chain reaction; rRNA, ribosomal ribonucleic acid; TGGE, temperature gradient gel electrophoresis; 
T-RF, terminal restriction fragment; T-RFLP, terminal restriction fragment length polymorphism. 



Molecular Ecology of the Human Intestinal Microbiota 5 

FINGERPRINTING REVEALS CHARACTERISTICS 
OF THE MICROBIOTA 

PCR-Denaturing Gradient Gel Electrophoresis 

The most commonly applied fingerprinting methods used to study the Gl-tract 
microbiota are denaturing and temperature gradient gel electrophoresis (DGGE and 
TGGE, respectively) of PCR-amplified genes coding for 16S rRNA (Fig. 1) (12,23). 
Other techniques such as terminal restriction fragment length polymorphism (T-RFLP) 
and single strand conformation polymorphism (SSCP) analysis are being applied but 
less frequently (26,29). The common principle of these methods is based on the 
separation of PCR-amplified segments of 16S rRNA genes of the same length, but 
with different sequence to visualize the diversity within the PCR amplicons by a 
banding pattern. One of the PCR primers has a 40-bp GC clamp to hold the DNA 
strands of the PCR product or amplicon together. With DGGE/TGGE, separation is 
based on the decreased electrophoretic mobility of partially melted double-stranded 
DNA molecules in polyacrylamide gels containing a linear gradient of DNA 
denaturants (a mixture of formamide and urea) or a linear temperature gradient, 
respectively. As a result mixed amplified PCR products will form a banding pattern 
after staining that reflects the different melting behaviors of the various sequences 
(30,31). Subsequent identification of specific bacterial groups or species present in the 
sample can be achieved either by cloning and sequencing of the excised bands or by 
hybridization of the profile using phylogenetic probes (30). Furthermore, comple- 
mentation of the fingerprinting results with statistical analysis provides additional 
information of the observed diversity by highlighting some putative correlation 
between different sets of variables (32). 

Since its application to study the intestinal microbiota, PCR-DGGE/-TGGE 
fingerprinting has advanced our knowledge of the intestinal microbiota by unraveling 
the complexity of this ecosystem and providing insight in the establishment and 
succession of the bacterial community within the host (23,33). The succession of the 
microbiota in the feces of infants over the first year of life has been visualized using DGGE 
profiles of the total microbial community, which showed the relatively simple and unstable 
infant fecal ecosystem (31). In healthy adults, the predominant fecal microbiota was 
shown to be complex, host-specific and remarkably stable in time (23,34,35). DGGE 
profiles for monozygotic twins were significantly more similar than for unrelated 
individuals, while marital partners showed less similar profiles than twins, indicating the 
influence of genotype over dietary or environmental factors (35). DGGE profiles also 
revealed that the predominant bacterial species associated with the colonic mucosa are 
uniformly distributed along the colon, but significantly different from the predominant 
fecal community (36,37). 

Under certain environmental circumstances and/or in genetically susceptible 
individuals, there is clear evidence that the Gl-tract microbiota may play a role in the 
pathogenesis and etiology of a number of inflammatory diseases such as ulcerative colitis 
(UC), and CD (30,38,39). Using DGGE, TGGE and SSCP fingerprinting analyses, it was 
demonstrated that fecal and mucosal-associated microbiota of patients with UC and CD is 
altered, less complex, and also unstable over time as compared to matched healthy people 
(26,40,41). In subjects with irritable bowel syndrome (IBS), higher temporal instability 
was also seen in comparison to healthy persons, but this was likely influenced by 
antibiotics used during the study (42). 



Amor and Vaughan 



Group-Specific PCR-DGGE 



Bands originating from lactobacilli in fecal samples could not be detected on the DGGE 
profiles since they represent less than 1% of the community, which is approximately the 
detection limit of this method (43,44). The dominant fecal microbiota of adults as assessed 
by DGGE was not significantly altered following consumption of certain probiotic strains 
(34,43). Although DGGE or TGGE were initially developed for total ecosystem 
communities, the sensitivity of the method for detecting specific groups that are present 
in lower numbers in the Gl-tract such as bifidobacteria and especially lactobacilli has been 
considerably enhanced by using group- or genus-specific primers (34,45-47). 
Consequently, it was possible to monitor the effect of the administration of prebiotics 
and/or probiotics on the composition of indigenous bifidobacterial species, and to track the 
probiotic strain itself (46). In the latter case, DGGE profiles showed that the simultaneous 
administration of the prebiotic and probiotic (synbiotic approach) did not improve the 
colonization of the probiotic strain in the gut of the tested individuals. In another study, 
the DGGE profiles generated from fecal samples of healthy individuals fed a probiotic 
strain Lactobacillus paracasei F19, allowed the tracing of the probiotic and supported its 
presence as autochthonous within the intestinal community of a number of indivi- 
duals (45). A nested PCR-DGGE approach has been developed to determine the diversity 
of sulfate-reducing bacteria (SRB) in complex microbial communities (48). SRB have 
been implicated in the pathogenesis of IBD, and consequently are an interesting 
population to investigate. 

Recently an approach combining GC fractionation with DGGE (GC-DGGE) 
effectively reduced the complexity of the community DNA mixture being analyzed such 
that the total diversity within each fraction could be more effectively assessed (49). Thus, 
initially the total DNA of the complex community was fractionated using buoyant density 
gradient centrifugation based on the % G + C content, using bisbenzimidazole which 
preferentially binds to A + T rich regions (50). This fractionation based on G + C content 
effectively reduced the complexity of the community DNA mixture being analyzed and 
the total diversity within each fraction could be more effectively assessed by the 
subsequent DGGE. 



Terminal-Restriction Fragment Length Polymorphism 

Another community fingerprinting technique which is gaining in popularity is T-RFLP (51). 
The basis is a PCR reaction for the 16S rRNA gene in the complex community followed by 
restriction enzyme digestion that generates the terminal restriction fragments (T-RFs). The 
latter are separated by electrophoresis or by using a capillary electrophoresis sequencer, 
which is more high throughput and reproducible (52), to produce a fingerprint. The 
technique has been used in several studies, including characterizing the human fecal 
bifidobacteria, as well as the tracking of probiotic Lactobacillus strains, and monitoring 
antibiotic-induced alterations in intestinal samples (53,54). Further improvements in this 
technique include the application of new primer-enzyme combinations for specifically 
bacterial populations in human feces (29). Furthermore, a novel phylogenetic assignment 
database for the specific T-RFLP analysis of human fecal microbiota (PAD-HCM) has been 
designed, which enables a high-level prediction of the terminal-restriction fragments at the 
species level (55). This will facilitate the use of this technique in studies on the microbiota. 
While the application of 16S rDNA-based fingerprinting methods are particularly 
well suited for examining time series and population dynamics, a more quantitative 



Molecular Ecology of the Human Intestinal Microbiota 7 

approach is useful to complement our knowledge about the composition and structure of 
this complex intestinal ecosystem. 



16S rRNA-TARGETED PROBES QUANTIFY 
THE GI-TRACT MICROBIOTA 

Hybridization with rRNA-targeted oligonucleotide probes has become the method of choice 
for the direct cultivation-independent identification of individual bacterial cells in natural 
samples. During the last decade, this technique has extended our view of bacterial 
assemblages and the population dynamics of complex microbial communities (15,56,57). The 
most commonly used biomarker for hybridization techniques, whether dot-blot or fluorescent 
in situ hybridization (FISH), is the 16S rRNA molecule because of its genetic stability, domain 
structure with conserved and variable regions, and high copy number. Highly conserved 
stretches may thus be used to design domain- specific probes such as EUB338/EUBII/EUBIII 
which collectively target most of the bacteria, whereas specific probes for each taxonomic 
level, between bacterial and archaeal, down to genus-specific and species-specific, can be 
designed according to the highly variable regions of the 16S rRNA (15,58-60). The increasing 
availability of 16S rRNA sequences has contributed significantly to the development of the 
hybridization methods and their application in different microbial ecosystems. Unquestion- 
ably, the success of the implementation of 16S rRNA hybridization strategies depends on 
different factors, among them rational design and validation of newly designed rRNA- 
targeted probes. 



Probe Design and Validation 

There is an online resource for oligonucleotide probes, called probeBase (142), which 
contains published FISH rRNA-targeted probes as well as recommended conditions of use, 
and many probes for dominant or interesting microbiota groups are described here (61). 
When designing new probes, one must consider specificity, sensitivity and accessibility to 
the target sequence. Nucleic acid probes can be designed to specifically target taxonomic 
groups at different levels of specificity (from species to domain) by virtue of variable 
evolutionary conservation of the rRNA molecules. The probes are typically 15-25 
nucleotides in length. Appropriate software such as the ARB software package (17) and 
availability of large databases (http://rdp.cme.msu.edu/html/) are useful tools for rapid 
probe design and in silico specificity profiling. Additional experimental evaluation of the 
probes with target and non-target microorganisms is necessary to ensure the specificity and 
the sensitivity of the newly designed probe. It is important to notice that the validation of a 
newly designed probe requires different procedures for the dot blot (62) and FISH format 
(60). Moreover, the hybridization and washing conditions (temperature, salt concentration 
and detergent) are also crucial for obtaining a detectable probe signal (63). The 
accessibility of the probe to its target site is another factor to be considered when designing 
new probes. The accessibility of probe target sites on the 16S and 23 S rRNA of Escherichia 
coli has been mapped systematically by flow cytometry (FCM) and FISH, and it was shown 
that probe-conferred signal intensities vary greatly among different targets sites (64,65). 
More recently, it was demonstrated that accessibility patterns of 16S rRNA's are more 
similar for phylogenetically related organisms; these findings may be the first description 
of consensus probe accessibility maps for prokaryotes (66). 



8 



Amor and Vaughan 



Hybridization Techniques 

Nucleic acid probing of complex communities comprises two major techniques: dot blot 
hybridization and FISH. In the dot blot format, total DNA or RNA is extracted from the 
sample and is immobilized on a membrane together with a series of RNA from reference 
strains. Subsequently, the membrane is hybridized with a radioactively labeled probe and 



Specific 

fluorescent 

oligonucleotides 




rRNA in 
ribosomes 




Antibodies 



□ 




Enzyme 
activity 




Membrane 
integrity 



Fluorescent microscopy 
and Image analysis 

(A) 



. .f 



Flow cytometry 



• . .* i 







Fluorescent activated 
cell sorting 



( 



\ 



Culturing 



Molecular Analysis 
-e.g., DGGE 



Figure 2 FISH involves whole cell hybridization with fluorescent oligonucleotide probes targeted 
against specific bacterial groups and species {left-hand scheme). The fluorescent probe hybridized 
cells may be visualized and/or counted using fluorescent microscopy and image analysis. The right- 
hand scheme illustrates how the viability of the cells may be assessed using functional probes that 
can also be visualized by fluorescent microscopy (A). FISH-labeled or functional probe-labeled cells 
may also be detected and enumerated using the flow cytometer (FCM). (B) shows a dot blot of fecal 
cells that were hybridized with a Bifidobacterium-specific probe. Following FCM the cells can be 
sorted according to the functional properties based on the probe stains, and subjected to further 
analysis. Abbreviation: DGGE, denaturing gradient gel electrophoresis. 



Molecular Ecology of the Human Intestinal Microbiota 9 

after a stringent washing step the amount of target rRNA is quantified. The membrane can 
be rehybridized with a general bacterial probe, and the amount of population-specific 
rRNA detected with the specific probe is expressed as a fraction of the total bacterial RNA. 
Quantification of the absolute and relative (as compared to total rRNA) amounts of a 
specific rRNA reflects the abundance of the target population. Consequently this technique 
does not represent a direct measure of cell number since cellular rRNA content varies with 
the current environmental conditions and the physiological activity of the cells at the time 
of sampling (67). Dot-blot hybridization has been successfully used to quantify rRNA 
from human fecal and cecal samples (68,69). It was found that strict anaerobic bacterial 
populations represented by the Bacteroides, Clostridium leptum and Clostridium 
coccoides groups were significantly lower in the cecum (right colon) than in the feces, 
while the Lactobacillus group was significantly higher in the feces than in the cecum (68). 
In contrast to dot-blot hybridization, FISH is applied to morphologically intact cells 
and thus provides a quantitative measure of the target organism without the limitation of 
culture-dependent methods (Fig. 2) (15,70). Following fixation, bacteria from any given 
sample can be hybridized with an appropriate probe or set of probes. The fixation allows 
permeabilization of the cell membrane and thus facilitates the accessibility of the 
fluorescent probes to the target sequence. For some Gram-positive bacteria, especially 
lactobacilli, additional pre-treatments including the use of cell wall lytic enzymes e.g., 
lysozyme, mutanolysin, protease K or a mixture is needed (71-73). Prior to hybridization, 
the cells can be either immobilized on gelatine-treated glass slides or simply kept in 
suspension when analyzed by FCM. The oligonucleotide probe is labeled covalently at the 
5' end with a fluorescent dye, such as fluorescein iso(thio)cyanate, while any necessary 
competitor probes are unlabeled. The stringency, i.e., conditions of hybridization that 
increase the specificity of binding between the probe and its target sequence, can be adjusted 
by varying either the hybridization temperature or formamide concentration. Under highly 
stringent conditions oligonucleotide probes can discriminate closely related target sites. 
Post-hybridization stringency can be achieved by lowering the salt concentration in the 
washing buffer in order to remove unbound probe and avoid unspecific binding. 

Quantification of FISH Signals 

Over the past years, significant methodological improvements of the probe fluorescent- 
conferred signal have been reported. These include the use of brighter fluorochromes 
including Cy3 and Cy5 (74,75), and unlabeled helper oligonucleotide probes (76) that bind 
adjacent to and increase the accessibility of the selected target site. Horseradish peroxidase 
labeled probes and tyramide signal amplification (also termed CARD-FISH) can be used 
to significantly enhance the signal intensity of hybridized cells (77). However, the latter 
requires effective permeabilization for the large enzyme-probe complex to enter the cell 
with the risk of damaging and lysing fixed cells. A further possibility is the use of peptide 
nucleic acid (PNA) probes which can confer very bright signals to the cell (78,79). 
However, currently PNA probes are rather expensive and previously published 
oligonucleotide probes cannot be simply translated into PNA probes. 

Epifluorescence microscopy is the standard method by which fluorescent-stained 
cells are enumerated; however, the method is time consuming and subjective (56,57). This 
technique has been improved by development of automated image acquisition and analysis 
software allowing accurate microscopic enumeration of fecal bacterial cells (73). 
Alternatively, FCM offers a potential platform for high-resolution, high throughput 
identification and enumeration of microorganisms using fluorescent rRNA-targeted 
oligonucleotides with the possibility of cell sorting (40,80-84). 



10 Amor and Vaughan 

An FCM method for direct detection of the anaerobic bacteria in human feces was first 
described over a decade ago (85). A membrane-impermeable nucleic acid dye propidium 
iodide (PI) was used in combination with the intrinsic scatter parameters of the cells to 
discriminate fecal cells from large particles. Coupling FCM results and image analysis, the 
authors showed that most of the particles detected with a large forward scatter value 
corresponded to aggregates most likely representing mucus fragments and undigested 
dietary compounds. They confirmed by means of cell sorting that the Pi-stained cells (fecal 
cells) corresponded to a 2-D surface area of <1.5 urn while the unstained particles 
(aggregates) were around 5.0 um (85). The work highlighted the potential of FCM to study 
anaerobic fecal bacteria without culturing. Despite this valuable work and to quote from 
Shapiro "the subject matter may stink, but the method is superb" (86), the application of 
FCM to study the intestinal microbiota is still in development. 

FISH-FCM was applied to detect and accurately quantify both fecal and mucosa- 
associated bacteria, and statistical analysis showed a high correlation between the FCM 
counts and microscopic counts (Fig. 2) (37,44,84). Using FCM, several thousands of cells 
can be counted accurately in a few seconds. Following the hybridization step, fecal 
cells are stained with a nucleic acid dye, for example PI, SYTO BC, and TOTO-1, to 
detect the total cells and subsequently spiked with standard beads of known size and 
concentration. The beads are thus used as an internal standard to calibrate the measured 
volume and to determine the absolute count of the probe-detected cells (40,87). In 
addition to the determination of the absolute cell counts, the fluorescence intensity signal 
can also be quantified using fluorescent beads with known fluorescent intensities (86). 
This is of major importance for determining optimal hybridization conditions for newly 
designed probes (37,82,88). FCM is becoming a popular method for high-resolution, high 
throughput identification of microorganisms using fluorescent rRNA-targeted 
oligonucleotides. 

Application of FISH to Study the Gl-Tract Ecosystem 

During the last five years, hybridization studies with rRNA-targeted probes have provided 
significant knowledge about the composition and structure of the gut microbiota. A large 
panel of oligonucleotide probes specific for various genera predominant in the GI tract 
have been designed and validated (Table 2), and have been used intensively in 
these studies. 

The uniqueness and complexity of the human gut microbiota revealed by finger- 
printing techniques were supported by results of analysis using nucleic-acid probe-based 
methods. These studies revealed that the majority of fecal bacteria belong to the 
Clostridium coccoides-Eubacterium rectale group and the Clostridium leptum group 
( ~ 20-30% each), Bacteroides ( ~ 10%), Atopobium and bifidobacteria groups in that order 
of abundance (81,89,91,96,97). The Clostridium coccoides-Eubacterium rectale probe 
(Erec482) (Table 2) covers Eubacterium hallii, Lachnospira and Ruminococcus members, 
while the Clostridium leptum group comprises members of Ruminococcus species and 
Faecalibacterium prausnitzii (89,98). In particular members of C. coccoides-E. rectale, 
C leptum, and the Bacteroides groups constituted more than half of the fecal microbiota. 
Atopobium and bifidobacteria groups comprised typically 4-5% each. The Lactobacillus - 
Enterococcus group, Enterobacteriaceae, Phascolarctobacterium and relatives, and 
Veillonella were less dominant (0.1 to a few percent) (90,91). However, differences in 
the occurrence of these bacterial groups have been reported by different research groups. 
These deviations may be due to the different methods or probes used, but it is also likely 
that the observed variance is due to the differences in the genetic background, lifestyle, 



Molecular Ecology of the Human Intestinal Microbiota 



11 



Table 2 FISH Probes Used to Study the Gastrointestinal Microbiota 



Probe 


Probe sequence (5"— 3") 


Target organism 


% Formamide 


Reference 


Eub338 


GCTGCCTCCCGTAGGAGT 


Most bacteria 


0-80 


(58) 


Eubll 


GCAGCCACCCGTAGGTGT 


Planctomycetes 


0-60 


(60) 


Eublll 


GCTGCCACCCGTAGGTGT 


Verrucomicrobia 


0-60 


(60) 


Bac303 


CCAATGTGGGGGACCTT 


Bacteroides/ 
Prevotella 





(59) 


Bdis656 


CCGCCTGCCTCAAACATA 


Bacteroides 
distasonis 





(89) 


Bfra602 


GAGCCGCAAACTTTCACAA 


Bacteroides fragilis 


30 


(89) 


Bvulgl017 


AGATGCCTTGCGGCT- 
TACGGC 


Bacteroides vulgatus 


30 


(82) 


Bfrag998 


GTTTCCACATCATTCCACTG 


Bacteroides fragilis 


30 


(83) 


Bdistl025 


CGCAAACGGCTATTGGTAG 


Bacteroides 
distasonis 


30 


(68) 


Erec482 


GCTTCTTAGTCAR a GTACCG 


Clostridium 

coccoides group 





(89) 


Clep866 


GGTGGATWACTTATTGTG 


Clostridium leptum 
group 


30 


(90) 


Rfla729 


AAAGCCCAGTAAGCCGCC 


Ruminococcus 
flavefaciens 


20 


(91) 


Rbro730 


TAAAGCCCAGY a AGGCCGC 


Ruminococcus bromii 




(91) 


Rcal733 


CAGTAAAGGCCCAG- 
TAAGCC 


Ruminococcus 
callidus 


30 


(90) 


ElgcOl 


GGGACGTTGTTTCTGAGT 


Clostridium leptum 
subgroup 





(89) 


Fprau645 


CCTCTGCACTACTCAA- 
GAAAA 


Faecalibacterium 
prausnitzii 


15 


(92) 


Bifl64 


CATCCGGCATTACCACCC 


Bifidobacteria 





(93) 


Ato291 


GGTCGGTCTCTCAACCC 


Atopobium group 





(94) 


Veil223 


AGACGCAATCCCCTCCTT 


Veillonella 





(91) 


Ecyl387 


CGCGGCATTGCTGCTTCA 


Eubacterium 
cylindroides 


20 


(91) 


Cvirl414 


GGGTGTTCCCGRCTCTCA 


Clostridium viride 


30 


(90) 


Edes635 


AGACCARCAGTTTGAAA 


Eubacterium 
desmolans 


30 


(90) 


Lach571 


GCCACCTACACTCCCTTT 


Lachnospira group 


40 


(91) 


Ehall469 


CCAGTTACCGGCTCCACC 


Eubacterium halii 
group 


20 


(91) 


Phasco741 


TCAGCGTCAGACACAGTC 


Phascolarctobacte 
Hum group 





(91) 


Enterl432 


CTTTTGCAACCCACT 


Enteric group 


30 


(68,69) 


Strc498 


GTTAGCCGTCCCTTTCTGG 


Lactococcus lactis 
ssp. lactis 


30 


(89,90) 


Labl58 


GGTATTAGCAY a CTGT 
TTCCA 


Lactobacillus! 
Enterococcus 





(95) 


Urobe63 


AATAAAGTAATTCCCGTTCG 


Uncultured Rumino- 
coccus obeum-like 
bacteria 


20 


(84) 


Urobeb 


AAARAARTATTTCCCGTTCG 








Non338 


ACATCCTACGGGAGGC 


Negative control 




(83) 



N, R, W, and Y are the International Union of Pure and Applied Chemistry codes for ambiguous bases. 



12 Amor and Vaughan 

and diet in the human populations studied. Two large studies, where an extensive array of 
oligonucleotide probes that targeted the major bacterial groups in the GI- tract of northern 
European adults was used, showed that 62-75% of the fecal bacteria could be detected and 
identified. The remainder (~ 30%) could either belong to members of the Archaea, 
Eukarya or most likely to yet unknown bacteria (90,91). These types of studies provide a 
valuable basis in order to eventually determine factors that change the microbiota such as 
lifestyle, diet or illness. Interestingly, FISH-FCM analysis of fecal microbiota of 
patients with UC revealed substantial temporal variations in the major bacterial groups 
studied (i.e., Bacteroides, C. coccoides-E. rectale, Atopobium, bifidobacteria and 
lactobacilli), which was further was supported by PCR-DGGE profiles (40). 



NEW MOLECULAR DIVERSITY APPROACHES 

Real Time PCR 

Real time quantitative PCR (qPCR) of the 16S rRNA gene is being developed the last 
few years for the detection and quantification of human intestinal microbiota, which has 

Q 

the advantages of being high throughput and measuring from 1 to up to 10 CFU (99). 
Both SYBR Green I and TaqMan chemistries have been used to target Bacteroides 
frag His, Bifidobacterium species, E. coli, L. acidophilus and Ruminococcus productus, 
and the method was demonstrated to be easier and faster than dot-blot hybridization 
methodology (100). Real-time qPCR (5 f nuclease PCR assay) has been used to study the 
microbiota that adhere to the colonic mucosa (101). The primer-probe combinations 
were applied to DNA for the detection of E. coli and Bacteroides vulgatus from pure 
cultures and colonic biopsy specimens. The assay was very sensitive detecting as little 
as 1 and 9 CFU of E. coli and B. vulgatus, respectively. Many of the qPCR assays being 
developed target the lactobacilli and Bifidobacterium species that may be incorporated 
in functional foods (102,103). Besides real time PCR of the 16S rRNA gene, the option 
to use the transaldolase gene of Bifidobacterium species has also been investigated and 
appeared to be superior to the former in quantifying bifidobacterial populations in 
infants (104). The qRT-PCR assays have been used for various applications such as 
comparison of healthy persons versus patients suffering from IBS (105), and in patients 
with active IBD (26). Recently, a TaqMan real-time PCR-based method for the 
quantification of 20 dominant bacterial species and groups of the microbiota was 
developed (106). This method involved a pair of conserved primers, as well as universal 
and specific quantification probes, for species, group or genus in question, in a single 
reaction, and allowed relative and absolute quantification of bacteria in human biopsy 
and fecal samples. Further developments in real-time qPCR will facilitate our insight 
into the dynamics of the microbiota. 

Diagnostic DNA Microarrays 

The development of DNA oligonucleotide microarrays offer a fast, high throughput option 
for detection and estimation of the diversity of microbes in a complex ecosystem (107). 
Alternative terms for the microarrays are phylochips, microbial diagnostic microarrays and 
identification arrays. Their principle is based on the dot-blot hybridization described above. 
Typically microarrays contain hundreds of oligonucleotide probes, usually based on the 16S 
rRNA gene, specific for different strains or species or genera of microorganisms that are 
detected in a single assay. Total DNA or RNA is isolated from the sample, fragmented, 
and amplified by PCR with the simultaneous incorporation of labeled nucleotides, or 



Molecular Ecology of the Human Intestinal Microbiota 13 

directly chemically labeled. The labeled fragments are hybridized to the probes 
immobilized on a surface, and following washing hybridized fragments are detected by a 
fluorescence scanner. There are many different forms of arrays to which the probes can be 
attached including macroarrays, and glass microarrays that are low to medium density, and 
very high density Affymetric microarrays (> 10 4 probes typically 25 mer per chip) (108). 
Three-dimensional form microarrays such as the Pamgene system and gel-pads allow the 
option for quantitative detection (109). Studies are underway to apply microarray 
technology to the human intestinal microbiota (16). A macroarray membrane-based method 
with 60 40-mer oligonucleotide probes specific for the dominant microbiota demonstrated 
the feasibility of arrays for detection (110). The high throughput potential of arrays will 
undoubtedly encourage further efforts in this area in the coming years. 



ASSESSMENT OF MICROBIOTA VITALITY AND METABOLIC ACTIVITY 

The aforementioned molecular techniques have greatly contributed to our fundamental 
understanding of the biodiversity, establishment, succession and structure of the intestinal 
microbiota; yet little is known about the in situ association between the microbial diversity 
and the metabolic activity of a phylogenetic affiliated group. A further challenge is to 
determine the physiological activity of the detected cells. This includes those cells that are 
naturally present within the ecosystem as well as the ingested members from fermented or 
functional foods. Moreover, the use of specific food-grade lactic acid bacteria as vectors 
for therapeutic delivery of molecules with targeted activity in the host is being investigated 
(111,112). These bacteria appear capable of surviving and of being physiologically active 
at the mucosal surfaces in animal models. Biological containment systems are being 
developed for these genetically modified lactic acid bacteria to limit their activity to the 
host and allow their use in human healthcare (113). 

In Situ Activity 

Quantitative hybridization with fluorescent rRNA probes (as in FISH) is a useful indicator 
of activity as there is a correlation between the growth rate, which is coupled to efficient 
protein synthesis, and the number of ribosomes. The FISH technique has been used to 
estimate growth rates of Escherichia coli cells colonizing the intestinal tract of mice (1 14). 
In situ activity of pure cultures of the human commensal Lactobacillus plantarum strain 
has been measured by correlating the rRNA, as determined by fluorescence intensity, with 
the cell growth rate (72). However, at very high cell densities, a typical property of 
L. plantarum at late stages of growth, changes in the cell envelope appeared to prevent 
effective entry of the probe into the cells. Permeabilization issues may confound 
application of this technique to certain microbes in complex environments like the 
intestine. Furthermore, recent data suggest that cellular ribosome content is not always an 
indicator of physiological activity. Apparently some bacterial cells might be highly active 
but possess a low ribosome content (115), while other bacterial types possess high RNA 
even after extended starvation periods (116). 

During the last years several innovative methods have been developed to resolve the 
linkage between taxonomic identity, activity and function in microbial communities. One 
of these techniques involves microautoradiography (MAR), which when combined with 
FISH (MAR-FISH), determines the uptake of specific radiochemicals by individual cells 
(1 17,1 18). MAR-FISH allows monitoring of the radio-labeled substrate uptake patterns of 
the probe-identified organisms under different environmental conditions (117,119). This 



14 Amor and Vaughan 

method has been applied with high throughput DNA microarray analysis to study the 
complex activated sludge ecosystem (120). 



Linking Taxomony to Function 

Another recently developed molecular technique coupled with substrate labeling is stable 
isotope probing (SIP) (121,122). In SIP, either lipid biomarkers (123), DNA (121) or RNA 
(124) are extracted from microbial communities incubated with C-labeled substrates. If 
cells grow on the added compounds, their pool of macromolecules will be isotopically 
enriched (heavy) compared to those of inactive organisms. For DNA- or RNA-SIP, 
identification of the metabolically active organisms (heavy) is achieved by separation of 
community DNA/RNA according to their buoyant density by means of equilibrium 
density-gradient centrifugation, followed by PCR-amplification of 16S rRNA genes in the 
isotopically heavy DNA/RNA pool, cloning and sequencing. The use of RNA was 
proposed as a more responsive biomarker as its turnover is much higher than that of 
DNA (124). Phospholipid fatty acids are also used as biomarker for C enrichments, but 
their resolution for diversity analysis is less powerful than for sequence analysis. 



Reporters to Monitor Gene Expression 

Molecular reporter systems may also be used to monitor activity of specific genes of a 
microbe of the complex intestinal ecosystem. Generally this involves fusing the reporter 
gene to the promoter of the bacterial gene of interest, such as stress- and starvation- 
induced genes and other growth physiology-related genes. It is noteworthy that this 
approach involves a genetically modified microbe, and consequently, its application is 
limited to animal studies. The adaptation of ingested lactic acid bacteria has received 
particular attention in terms of how they adapt their metabolism in order to survive and 
colonize within the gastrointestinal niches. 

The fusion of bacterial promoters from Lactococcus lactis with genes of the reporter 
protein luciferase (luxA-luxB genes of Vibrio harveyi) was developed to investigate gene 
expression of this food-grade bacteria in the mouse intestinal tract (125). L. lactis strains 
marked with reporter genes for luciferase and the green fluorescent protein (GFP; from 
Aequorea victoria) were studied for their metabolic activity and survival by assessment of 
lysis, respectively, which revealed differential expression depending on the intestinal 
conditions and mode of administration (126). Following consumption by rats and analysis 
of the strains in the different regions of the intestinal tract, the lactococci were 
demonstrated to survive gastric transit quite well but the majority lost activity and 
underwent lysis in the duodenum. The luciferase gene reporter system has also been 
applied to a probiotic Lactobacillus casei strain that is added to fermented dairy products. 
The luciferase-harboring L. casei derivative was consumed in milk by mice harboring 
human microbiota. Luciferase activity was undetectable in the stomach to jejunum, but 
detected when the cells reached the ileum, and the activity remained at a maximum level in 
the cecum, confirming reinitiation of protein synthesis in the ileal and cecal compartments 
(127,128). 

Several variants of the GFP have been developed such as GFPs with alternative 
emission wavelengths, or with reduced stability to monitor shifts in gene expression 
(129,130). 



Molecular Ecology of the Human Intestinal Microbiota 15 

Flow Cytometry-Based Approaches 

FCM in combination with a variety of fluorescent physiological probes and cell sorting 
analysis is invaluable for measuring viability of cells in environmental samples 
(80,87,131,132). Ability to grow in medium is the current standard to assess viability, but 
it is recognized that some cells enter a non-culturable state although still exhibit metabolic 
activity. The criteria by which viability is evaluated by the FCM include membrane 
permeability or integrity, enzyme activity, and/or maintenance of a membrane-potential 
(Fig. 2). One of the most widely used dyes for assessment of viability is carboxy-fluorescein 
diacetate, a non-fluorescent precursor that diffuses across the cell membrane, but is retained 
only by viable cells with intact membranes which convert it into a membrane-impermeant 
fluorescent dye by non-specific esterases of active cells. Another probe is PI, a nucleic acid 
dye, which is excluded by viable cells with intact membranes, but enters cells with damaged 
membranes and binds to their DNA or RNA. Simultaneous staining of fecal Bifidobacterium 
species with these two probes was used to assess their viability during bile salt stress (133). 
Subsequent detection with the FCM and cell sorting revealed three populations representing 
viable, injured and dead cells, whereby a significant portion (40%) of the injured cells could 
be cultured. This approach highlights the importance of multi-parametric FCM as a 
powerful technique to monitor physiological heterogeneity within stressed populations at 
the single cell level. 

FCM also allows monitoring of bacterial heterogeneity at the single cell level and 
provides a mean to sort sub-populations of interest for further molecular analysis (15). 
Recently, the viability of fecal microbiota in fecal samples was assessed by combining a 
viability assay with flow sorting, and subsequent analysis by PCR-DGGE and 
identification by cloning and 16S rRNA sequencing (80). The fecal cells of four adults 
were initially discriminated with physiological probes PI and SYTO BC into viable, 
injured and dead cells. This revealed that only approximately half of the microbial 
community in fecal samples is viable, while the remainder was injured or dead (about a 
quarter each of the total community). This is in agreement with a previous analysis of 
proportions of dead bacteria in 10 persons which ranged from 17% to 34%, as assessed by 
PI only (134). The 16S rRNA analysis indicated which bacterial groups comprised 
live, dead or injured populations, for example many butyrate-producers were in the live 
fractions, while many clones from Bacteroides were found in the dead fractions (80). 
Specific PCR-DGGE and 16S rRNA analysis of the bifidobacterial and lactobacilli 
populations showed sequences with low similarity to the characterized species suggested 
the potential of as yet uncultured novel species in humans (80,135). This interesting 
combination of technologies provided ecological information on the in situ diversity and 
activity of the fecal microbes. 



PERSPECTIVES 

This chapter has highlighted the extraordinary advances in the molecular technologies that 
have substantially contributed to our knowledge and understanding of the human intestinal 
microbiota. The application of these molecular tools has greatly facilitated our analysis 
of the composition of the human microbiota. A picture of the "typical" microbiota for at 
least the northern European population of infants and adults is emerging, as are differences 
in individuals with intestinal diseases. The diversity is far greater than previously 
predicted from the initial culturing studies in the 1960s. Consequently, further 
technological improvements to perform the techniques at higher throughput, and for 



16 Amor and Vaughan 

measurement of more subtle changes in the diversity of the microbiota due to, for example, 
specific dietary components, require further development. Microarray technology is 
amenable to both these requirements, and currently DNA microarrays are being 
constructed for the human microbiota using 16S rRNA sequences of microbiota (136); 
[Mirjana Rajilic and Willem M. de Vos, personal communication]. FCM with its unique 
capacity for quantitative and high throughput analysis is resulting in the development of an 
alternative type of array using beads with oligonucleotide probes on the surface that can be 
applied in hybridization assays in suspension (137-139). 

The substantial impact of this highly diverse microbiota on the health of the human 
host is now well recognized, such as processing of undigested food, contributing to the host 
defense and regulating fat storage amongst others (6, 140, 141). It is a particular challenge to 
develop methods that allow monitoring of microorganisms according to their eco- 
physiological traits in situ. The application of cytometric protocols using fluorescent 
probes in combination with molecular techniques opens the potential for examining key 
microbial processes and community function in complex microbial ecosystems. Further 
efforts to determine the molecular foundations of the host-microbiota interactions will 
require multi-disciplinary approaches. The rewards of this research in terms of promoting 
host health via our microbiota and diet can be substantial, as well as novel approaches for 
treating intestinal diseases and infections caused by pathogens. 



REFERENCES 

1. Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the 
human intestine. Science 2005; 5717:1915-1920. 

2. Rawls JF, Samuel BS, Gordon JI. Gnotobiotic zebrafish reveal evolutionarily conserved 
responses to the gut microbiota. Proc Natl Acad Sci USA 2004; 101:4596-4601. 

3. Falk PG, Hooper LV, Midtvedt T, Gordon JI. Creating and maintaining the gastrointestinal 
ecosystem: what we know and need to know from gnotobiology. Microbiol Mol Biol Rev 
1998;62:1157-1170. 

4. Stappenbeck TS, Hooper LV, Gordon JI. Developmental regulation of intestinal 
vasculogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci USA 2002; 
99:15451-15455. 

5. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of 
commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004; 
118:229-241. 

6. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science 2001; 
292:1115-1118. 

7. Ouwehand AC, Salminen S, Isolauri E. Probiotics: an overview of beneficial effects. Antonie 
Van Leeuwenhoek 2002; 82:279-289. 

8. Rachmilewitz D, Katakura K, Karmeli F, et al. Toll-like receptor 9 signaling mediates the 
anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 
2004; 126:520-528. 

9. Finegold SM, Sutter VL, Mathisen GE. Normal indigenous flora. In: Hentges DJ, ed. Human 
Intestinal Microflora in Health and Disease. New York: Academic Press, 1983:3-31. 

10. Holdeman LV, Good IJ, Moore WE. Human fecal flora: variation in bacterial composition 
within individuals and a possible effect of emotional stress. Appl Environ Microbiol 1976; 
31:359-375. 

11. Moore WE, Holdeman LV. Human fecal flora: the normal flora of, 20 Japanese-Hawaiians. 
Appl Microbiol 1974; 27:961-979. 

12. Vaughan EE, Schut F, Heilig HGHJ, Zoetendal EG, de Vos WM, Akkermans ADL. A 
molecular view of the intestinal ecosystem. Curr Issues Intest Microbiol 2000; 1:1—12. 



Molecular Ecology of the Human Intestinal Microbiota 17 

13. Zoetendal EG, Cheng B, Koike S, Mackie RI. Molecular microbial ecology of the 
gastrointestinal tract: from phylogeny to function. Curr Issues Intest Microbiol 2004; 5:31-47. 

14. Whitfield J. Features-science and health: microbial soup of life is sieved for treasure. 
Financial Times, 16 April 2004 http://news.ft.com. 

15. Amann R, Ludwig W, Schleifer K. Phylogenetic identification and in situ detection of 
individual microbial cells without cultivation. Microbiol Rev 1995; 59:143-169. 

16. Namsolleck P, Thiel R, Lawson P, et al. Molecular methods for the analysis of gut microbiota. 
Microb Ecol Health Dis 2004; 16:71-85. 

17. Ludwig W, Strunk O, Westram R, et al. ARB: a software environment for sequence data. 
Nucleic Acids Res 2004; 32:1363-1371. 

18. Cole JR, Chai B, Marsh TL, et al. The Ribosomal database project (RDP-II): previewing a 
new autoaligner that allows regular updates and the new prokaryotic taxonomy. Nucleic Acids 
Res 2003; 31:442-443. 

19. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. 
Sci Express 2005; 14:1-4. 

20. Hayashi H, Sakamoto Y, Benno M. Phylogenetic analysis of the human gut microbiota using 
16S rDNA clone libraries and strictly anaerobic culture-based methods. Microbiol Immunol 
2002; 46:535-548. 

21. Hold GL, Pryde SE, Russell VJ, Furrie E, Flint HJ. Assessment of microbial diversity in 
human colonic samples by 16S rDNA sequence analysis. FEMS Microbiol Ecol 2002; 
39:33-39. 

22. Suau A, Bonnet R, Sutren M, et al. Direct analysis of genes encoding 16S rRNA from 
complex communities reveals many novel molecular species within the human gut. Appl 
Environ Microbiol 1999; 65:4799-4807. 

23. Zoetendal EG, Akkermans ADL, De Vos WM. Temperature gradient gel electrophoresis 
analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities 
of active bacteria. Appl Environ Microbiol 1998; 64:3854-3859. 

24. Mangin I, Bonnet R, Seksik P, et al. Molecular inventory of fecal microflora in patients with 
Crohn's disease. FEMS Micro Ecol 2004; 50:25-36. 

25. Breitbart M, Hewson I, Felts B, et al. Metagenomic analyses of an uncultured viral 
community from human feces. J Bacteriol 2003; 185:6220-6223. 

26. Ott SJ, Musfeldt M, Wenderoth DF, et al. Reduction in diversity of the colonic mucosa 
associated bacterial microflora in patients with active inflammatory bowel disease. Gut 2004; 
53:685-693. 

27. Von Wintzingrode F, Gobel UB, Stackebrandt E. Determination of microbial diversity in 
environmental samples: pitfalls of PCR-based rRNA analysis. FEMS Microbiol Rev 1997; 
21:213-229. 

28. Bonnet R, Suau A, Dore J, Gibson GR, Collins MD. Differences in rDNA libraries of 
fecal bacteria derived from 10- and 25-cycle PCRs. Int J Syst Evol Microbiol 2002; 
52:757-763. 

29. Nagashima K, Hisada T, Sato M, Mochizuki J. Application of new primer-enzyme 
combinations to terminal restriction fragment length polymorphism profiling of bacterial 
populations in human feces. Appl Environ Microbiol 2003; 69:1251-1262. 

30. Muyzer G, de Waal E, Uitterlinden A. Profiling of complex microbial populations by 
denaturing gradient gel electrophoresis analysis of polymerase chain reaction-amplified genes 
coding for 16S rRNA. Appl Environ Microbiol 1993; 59:695-700. 

31. Rosenbaum V, Riesner D. Temperature-gradient gel electrophoresis. Thermodynamic 
analysis of nucleic acids and proteins in purified form and in cellular extracts. Biophys 
Chem 1987; 26:235-246. 

32. Formin N, Hamelin S, Tarnawski S, et al. Statistical analysis of denaturing gel electrophoresis 
(DGE) fingerprinting patterns. Environ Microbiol 2002; 4:634-643. 

33. Favier CF, Vaughan EE, De Vos WM, Akkermans ADL. Molecular monitoring of succession 
of bacterial communities in human neonates. Appl Environ Microbiol 2002; 68:219-226. 



18 Amor and Vaughan 

34. Tannock GW, Munro K, Harmsen HJM, Welling GW, Smart J, Gopal PK. Analysis of the 
fecal microflora of human subjects consuming a probiotic product containing Lactobacillus 
rhamnosus DR20. Appl Environ Microbiol 2000; 66:2578-2588. 

35. Zoetendal EG, Akkermans ADL, Akkermans-van Vliet WM, de Visser JAGM, de Vos WM. 
The host genotype affects the bacterial community in the human gastrointestinal tract. 
Microbiol Ecol Health Dis 2001; 13:129-134. 

36. Nielsen D, Moller P, Rosenfeldt V, Paerregaard A, Michaelsen K, Jakobsen M. Case study of 
the distribution of mucosa-associated Bifidobacterium species, and Lactobacillus species, 
and other lactic acid bacteria in the human colon. Appl Environ Microbiol 2003; 
69:7545-7548. 

37. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben- Amor K, Akkermans ADL, de 
Vos WM. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly 
distributed along the colon and differ from the community recovered from feces. Appl 
Environ Microbiol 2002; 68:3401-3407. 

38. Campieri M, Gionchetti P. Bacteria as the cause of ulcerative colitis. Gut 2001; 48:132-135. 

39. Shanahan F. Probiotics: a perspective on problems and pitfalls. Scand J Gastroenterol Suppl 
2003;(237):34-46. 

40. Ben- Amor K, Heikamp-de jong I, Verhaegh S, de Vos WM, Vaughan EE. Population 
dynamics and diversity of fecal microbiota of patients with ulcerative colitis participating in a 
probiotic trial. In: Ben- Amor K, eds. Microbial ecophysiology of the human intestinal tract: A 
flow cytometric approach. Netherland: PhD Thesis Wageningen University, 2004:83-121. 

41 . Seksik P, Rigottier-Gois L, Gramet G, et al. Alterations of the dominant fecal bacterial groups 
in patients with Crohn's disease of the colon. Gut 2003; 52:237-242. 

42. Matto J, Maunuksela L, Kajander K, et al. Composition and temporal stability of 
gastrointestinal microbiota in irritable bowel syndrome-a longitudinal study in IBS and 
control subjects. FEMS Immunol Med Microbiol 2005; 43:213-222. 

43. Vaughan EE, Heilig HGHJ, Zoetendal EG, et al. Molecular approaches to study probiotic 
bacteria. Trends Food Sci Technol 1999; 10:400-404. 

44. Zoetendal EG, Ben Amor K, Akkermans AD, Abee T, de Vos W. DNA isolation protocols 
affect the detection limit of PCR approaches of bacteria in samples from the human 
gastrointestinal tract. Syst Appl Microbiol 2001; 24:405-410. 

45. Heilig HGHJ, Zoetendal EG, Vaughan EE, Marteau P, Akkermans ADL, de Vos WM. 
Molecular diversity of Lactobacillus spp. and other lactic acid bacteria in the human intestine 
as determined by specific amplification of, 16S ribosomal DNA. Appl Environ Microbiol 
2002;68:114-123. 

46. Satokari RM, Vaughan EE, Akkermans AD, Saarela M, De Vos WM. Polymerase chain 
reaction and denaturing gradient gel electrophoresis monitoring of fecal Bifidobacterium 
populations in a prebiotic and probiotic feeding trial. Syst Appl Microbiol 2001; 
24:227-231. 

47. Walter J, Hertel C, Tannock GW, Lis CM, Munro K, Hammes WP. Detection of 
Lactobacillus, Pediococcus, Leuconostoc, and Weissella species in human feces by using 
group- specific PCR primers and denaturing gradient gel electrophoresis. Appl Environ 
Microbiol 2001; 67:2578-2585. 

48. Dar SA, Kuenen JG, Muyzer G. Nested PCR-denaturing gradient gel electrophoresis approach 
to determine the diversity of sulfate-reducing bacteria in complex microbial communities. 
Appl Environ Microbiol 2005; 71:2325-2330. 

49. Holben WE, Feris KP, Kettunen A, Apajalahti JH. GC fractionation enhances microbial 
community diversity assessment and detection of minority populations of bacteria by 
denaturing gradient gel electrophoresis. Appl Environ Microbiol 2004; 70:2263-2270. 

50. Holben WE, Harris D. DNA-based monitoring of total bacterial community structure in 
environmental samples. Mol Ecol 1995; 4:627-6331. 

5 1 . Kitts CL. Terminal restriction fragment patterns: a toll for comparing microbial communities 
and assessing community dynamics. Curr Issues Intest Microbiol 2001; 2:17-25. 



Molecular Ecology of the Human Intestinal Microbiota 19 

52. Osborn AM, Moore ER, Timmis KN. An evaluation of terminal-restriction fragment length 
polymorphism (T-RFLP) analysis for the study of microbial community structure and 
dynamics. Environ Microbiol 2000; 2:39-50. 

53. Jernberg C, Sullivan A, Edlund C, Jansson JK. Monitoring of antibiotic-induced alterations in 
the human intestinal microflora and detection of probiotic strains by use of terminal restriction 
fragment length polymorphism. Appl Environ Microbiol 2005; 71:501-506. 

54. Sakamoto M, Hayashi H, Benno Y. Terminal restriction fragment length polymorphism 
analysis for human fecal microbiota and its application for analysis of complex bifidobacteria! 
communities. Microbiol Immunol 2003; 47:133-142. 

55. Matsumoto M, Sakamoto M, Hayashi H, Benno Y. Novel phylogenetic assignment database 
for terminal-restriction fragment length polymorphism analysis of human colonic microbiota. 
J Microbiol Methods 2005; 61:305-319. 

56. Lipski A, Friedrich U, Altendorf K. Application of rRNA-targeted oligonucleotide probes in 
biotechnology. Appl Microbiol Biotechnol 2001; 56:40-57. 

57. Moter A, Gobel UB. Fluorescence in situ hybridization (FISH) for direct visualization of 
microorganisms. J Microbiol Methods 2000; 41:85-112. 

58. Amann RI, Binder B, Olson R, Chisholm SW, Devereux R, Stahl D. Combination of 16S 
rRNA-targeted oligonucleotide probes with flow cytometry for analyzing mixed microbial 
populations. Appl Environ Microbiol 1990; 56:1919-1925. 

59. Manz W, Amann R, Ludwig W, Vancanneyt M, Schleifer K. Application of a suite of 16S 
rRNA-specific oligonucleotide probes designed to investigate bacteria of the phylum 
cytophaga-flavobacter-bacteroides in the natural environment. Microbiology 1996; 
142:1097-1106. 

60. Daims H, Bruhl A, Amann R, Schleifer KH, Wagner M. The domain-specific probe EUB338 
is insufficient for the detection of all Bacteria: development and evaluation of a more 
comprehensive probe set. Syst Appl Microbiol 1999; 22:434-444. 

61. Loy A, Horn M, Wagner M. Probebase: an online resource for rRNA-targeted oligonucleotide 
probes. Nucleic Acids Res 2003; 31:514-516. 

62. de los Reyes FL, Ritter W, Raskin L. Group-specific small-subunit rRNA hybridization 
probes to characterize filamentous foaming in activated sludge systems. Appl Environ 
Microbiol 1997; 63:1107-1117. 

63. Stahl DA, Amann R. Development and application of nucleic acid probes. In: 
Stackerbrandt E, Goodfellow M, eds. Nucleic Acid Techniques in Bacterial Systematics. 
Chichester: John Wiley & Sons Ltd, 1991:205-248. 

64. Fuchs BM, Syutsubo K, Ludwig W, Amann R. In situ accessibility of Escherichia coli, 23S 
rRNA to fluorescently labeled oligonucleotide probes. Appl Environ Microbiol 2001; 
67:961-968. 

65. Fuchs BM, Wallner G, Beisker W, Schwippl I, Ludwig W, Amann R. Flow cytometric 
analysis of the in situ accessibility of Escherichia coli 16S rRNA for fluorescently labeled 
oligonucleotide probes. Appl Environ Microbiol 1998; 64:4973-4982. 

66. Behrens S, Fuchs BM, Mueller F, Amann R. Is the in situ accessibility of the 16S rRNA 
of Escherichia coli for Cy3-labeled oligonucleotide probes predicted by a three- 
dimensional structure model of the 30S ribosomal subunit? Appl Environ Microbiol 2003; 
69:4935-4941. 

67. Molin S, Givskov M. Application of molecular tools for in situ monitoring of bacterial growth 
activity. Environ Microbiol 1999; 1:383-391. 

68. Marteau P, Pochart P, Dore J, Bera-Maillet C, Bernalier A, Corthier G. Comparative study of 
bacterial groups within the human cecal and fecal microbiota. Appl Environ Microbiol 2001; 
67:4939-4942. 

69. Sghir A, Gramet G, Suau A, Rochet V, Pochart P, Dore J. Quantification of bacterial groups 
within human fecal flora by oligonucleotide probe hybridization. Appl Environ Microbiol 
2000; 66:2263-2266. 

70. Amann R, Fuchs BM, Behrens S. The identification of microorganisms by fluorescence in situ 
hybridization. Curr Opin Biotechnol 2001; 12:231-236. 



20 Amor and Vaughan 

71. Beimfohr C, Krause A, Amann R, Ludwig W, Schleifer KH. In situ identification of 
lactococci, enterococci and streptococci. Syst Appl Microbiol 1993; 16:450-456. 

72. de Vries MC, Vaughan EE, Kleerebezem M, De Vos WM. Optimising single cell activity 
assessment of Lactobacillus plantarum by fluorescent in situ hybridization as affected by 
growth. J Microbiol Methods 2004; 59:109-115. 

73. Jansen GJ, Wildeboer-Veloo ACM, Tonk RHJ, Franks AH, Welling GW. Development and 
validation of an automated, microscopy-based method for enumeration of groups of intestinal 
bacteria. J Microbiol Methods 1999; 37:215-221. 

74. Glockner FO, Amann R, Alfreider A, et al. An in situ hybridization protocol for detection and 
identification of planktonic bacteria. Syst Appl Microbiol 1996; 19:403-406. 

75. Southwick PL, Ernst LA, Tauriello EW, et al. Cyanine dye labeling reagents — 
carboxymethylindocyanine succinimidyl esters. Cytometry 1990; 11:418-430. 

76. Fuchs BM, Glockner FO, Wulf J, Amann R. Unlabeled helper oligonucleotides increase the 
in situ accessibility to 16S rRNA of fluorescently labeled oligonucleotide probes. Appl 
Environ Microbiol 2000; 66:3603-3607. 

77. Pernthaler A, Pernthaler J, Amann R. Fluorescence in situ hybridization and catalyzed 
reporter deposition for the identification of marine bacteria. Appl Environ Microbiol 2002; 
68:3094-3101. 

78. Oliveira K, Brecher SM, Durbin A, et al. Direct identification of Staphylococcus aureus from 
positive blood culture bottles. J Clin Microbiol 2003; 41:889-891. 

79. Perry-O'Keefe H, Rigby S, Oliveira K, et al. Identification of indicator microorganisms using 
a standardized PNA FISH method. J Microbiol Methods 2001; 47:281-292. 

80. Ben-Amor K, Heilig HG, Smidt H, Vaughan EE, Abee T, de Vos WM. Genetic diversity of 
viable, injured and dead fecal bacteria assessed by fluorescence activated cell sorting and, 
16S rRNA gene analysis. Appl Environ Microbiol 2005; 71:4679-4689. 

81. Rigottier-Gois L, Le Bourhis A-G, Gramet G, Rochet V, Dore J. Fluorescent hybridization 
combined with flow cytometry and hybridization of total RNA to analyse the composition of 
microbial communities in human faeces using 16S rRNA probes. FEMS Microbiol Ecol 2003; 
43:237-245. 

82. Rigottier-Gois L, Rochet V, Garrec N, Suau A, Dore J. Enumeration of Bacteroides species in 
human faeces by fluorescent in situ hybridization combined with flow cytometry using 16S 
rRNA probes. Syst Appl Microbiol 2003; 26:110-118. 

83. Wallner G, Amann R, Beisker W. Optimizing fluorescent in situ hybridization with rRNA- 
targeted oligonucleotide probes for flow cytometric identification of microorganisms. 
Cytometry 1993; 14:136-143. 

84. Zoetendal EG, Ben-Amor K, Harmsen HJM, Schut F, Akkermans ADL, de Vos WM. 
Quantification of uncultured Ruminococcus obeum-\ike bacteria in human fecal samples by 
fluorescent in situ hybridization and flow cytometry using 16S rRNA-targeted probes. Appl 
Environ Microbiol 2002; 68:4225-4232. 

85. van der Waaij LA, Mesander G, Limburg PC, Wan der Waaij D. Direct flow cytometry of 
anaerobic bacteria in human feces. Cytometry 1994; 16:270-279. 

86. Shapiro HM. Practical Flow Cytometry. New York: Wiley-Liss Inc., 1995. 

87. Ben- Amor K, Abee T, De Vos WM. Flow cytometric analysis of microorganisms. In: Ben- 
Amor K, eds. Microbial Eco-physiology of the Human Intestinal Tract: A flow cytometric 
approach. Netherland: PhD thesis Wageningen University, 2004:21-47. 

88. Derrien M, Ben- Amor K, Vaughan EE, de Vos WM. Validation of 16S rRNA probe specific 
for the novel intestinal mucin-degrader Akkermansia muciniphila. In: Ahonen R, Saarela M, 
Mattila-Sandholm T, eds. The FOOD, Gl-tract Functionality and Human Heatlh Cluster. 
PROEUHEALTH. Vol. VTT Biotechnolgy. Sitges, Spain, 2004. http://www.vtt.fi/inf/pdf/. 

89. Franks AH, Harmsen HJM, Raangs GC, Jansen GJ, Schut F, Welling GW. Variations of 
bacterial populations in human feces measured by fluorescent in situ hybridization with 
group- specific 16S rRNA-targeted oligonucleotide Probes. Appl Environ Microbiol 1998; 
64:3336-3345. 



Molecular Ecology of the Human Intestinal Microbiota 21 

90. Lay C, Rigottier-Gois L, Holmstrum K, et al. Colonic microbiota signatures across five 
northern European countries. Appl Environ Microbiol 2005; 717:4153-4155. 

9 1 . Harmsen HJM, Raangs GC, He T, Degener JE, Welling GW. Extensive set of 1 6S rRNA-based 
probes for detection of bacteria in human feces. Appl Environ Microbiol 2002; 68:2982-2990. 

92. Suau A, Rochet V, Sghir A, et al. Fusobacterium prausnitzii and related species represent a 
dominant group within the human fecal flora. Syst Appl Microbiol 2001; 24:139-145. 

93. Langendijk P, Schut F, Jansen G, et al. Quantitative fluorescence in situ hybridization of 
Bifidobacterium spp. with genus-specific 16S rRNA-targeted probes and its application in 
fecal samples. Appl Environ Microbiol 1995; 61:3069-3075. 

94. Harmsen HJM, Wildeboer-Veloo ACM, Grijpstra J, Knol J, Degener JE, Welling GW. 
Development of 16S rRNA-based probes for the Coriobacterium group and the Atopobium 
cluster and their application for enumeration of Coriobacteriaceae in human feces from 
volunteers of different age groups. Appl Environ Microbiol 2000; 66:4523-4527. 

95. Harmsen JHM, Elfferich P, Schut F, Welling GW. A 16S rRNA-targeted probe for detection 
of lactobacilli and enterococci in fecal samples by fluorescent in situ hybridization. Micro 
Ecol Health Dis 1999; 11:3-12. 

96. Lay C, Sutren M, Rochet V, Saunier K, Dore J, Rigottier-Gois L. Design and validation of 16S 
rRNA probes to enumerate members of the Clostridium leptum subgroup in human fecal 
microbiota. Environ Microbiol 2005; 7:933-946. 

97. Rochet V, Rigottier-Gois L, Rabot S, Dore J. Validation of fluorescent in situ hybridization 
combined with flow cytometry for assessing interindividual variation in the composition of 
human fecal microflora during long-term storage of samples. J Microbiol Methods 2004; 
59:263-270. 

98. Collins MD, Lawson PA, Willems A, et al. The phylogeny of the genus Clostridium: proposal 
of five new genera and eleven new species combinations. Int J Syst Bacteriol 1994; 
144:812-826. 

99. Nadkarni MA, Martin FE, Jacques NA, Hunter N. Determination of bacterial load by real- 
time PCR using a broad-range (universal) probe and primers set. Microbiology 2002; 
148:257-266. 

100. Malinen E, Kassinen A, Rinttila T, Palva A. Comparison of real-time PCR with SYBR 
Green I or 5 ' -nuclease assays and dot-blot hybridization with rDNA-targeted 
oligonucleotide probes in quantification of selected fecal bacteria. Microbiology 2003; 
149:269-277. 

101. Huijsdens XW, Linskens RK, Mak M, Meuwissen SG, Vandenbroucke-Grauls CM, 
Savelkoul PH. Quantification of bacteria adherent to gastrointestinal mucosa by real-time 
PCR. J Clin Microbiol 2002; 40:4423-4427. 

102. Bartosch S, Fite A, Macfarlane GT, McMurdo ME. Characterization of bacterial communities 
in feces from healthy elderly volunteers, and hospitalized elderly patients by using real-time 
PCR and effects of antibiotic treatment on the fecal microbiota. Appl Enivr Microbiol 2004; 
70:3575-3581. 

103. Matsuki T, Watanabe K, Fujimoto J, Takada T, Tanaka R. Use of 16S rRNA gene-targeted 
group-specific primers for real-time PCR analysis of predominant bacteria in human feces. 
Appl Environ Microbiol 2004; 71:7220-7228. 

104. Requena T, Burton J, Matsuki T, et al. Identification, detection, and enumeration of human 
Bifidobacterium species by PCR targeting the transaldolase gene. Appl Environ Microbiol 
2002; 68:2420-2427. 

105. Malinen E, Rinttila T, Kajander K, et al. Analysis of the fecal microbiota of irritable bowel 
syndrome patients and healthy controls with real-time PCR. Am J Gastroenterol 2005; 
100:373-382. 

106. Ott SJ, Musfeldt M, Ullmann U, Hampe J, Schreiber S. Quantification of intestinal bacterial 
populations by real-time PCR with a universal primer set and minor groove binder probes: a 
global approach to the enteric flora. J Clin Microbiol 2004; 42:2566-2572. 

107. Bodrossy L, Sessitsch A. Oligonucleotide microarrays in microbial diagnostics. Curr Opin 
Microbiol 2004; 7:245-254. 



22 Amor and Vaughan 

108. Wilson KH, Wilson WJ, Radosevich JL, et al. High-density microarray of small- subunit 
ribosomal DNA probes. Appl Environ Microbiol 2002; 68:2535-2541. 

109. Guschin DY, Mobarry BK, Proudnikov D, Stahl DA, Rittmann BE, Mirzabekov AD. 
Oligonucleotide microchips as genosensors for determinative and environmental studies in 
microbiology. Appl Environ Microbiol 1997; 63:2397-2402. 

110. Wang RF, Kim SJ, Robertson LH, Cerniglia CE. Development of a membrane-array method 
for the detection of human intestinal bacteria in fecal samples. Mol Cell Probes 2002; 
16:341-350. 

111. Geoffroy MC, Guyard C, Quatannens B, Pavan S, Lange M, Mercenier A. Use of green 
fluorescent protein to tag lactic acid bacterium strains under development as live vaccine 
vectors. Appl Environ Microbiol 2000; 66:383-391. 

1 12. Hanniffy S, Wiedermann U, Repa A, et al. Potential and opportunities for use of recombinant 
lactic acid bacteria in human health. Adv Appl Microbiol 2004; 56:1-64. 

113. Steidler L. Live genetically modified bacteria as drug delivery tools: at the doorstep of a new 
pharmacology? Expert Opin Biol Ther 2004; 4:439-441. 

1 14. Rang CU, Licht TR, Midtvedt T, et al. Estimation of growth rates of Escherichia coli BJ4 in 
streptomycin-treated and previously germfree mice by in situ rRNA hybridization. Clin Diagn 
Lab Immunol 1999; 6:434-436. 

115. Pernthaler A, Pernthaler J, Schattenhofer M, Amann R. Identification of DNA- 
synthesizing bacterial cells in coastal North Sea plankton. Appl Environ Microbiol 
2002; 68:5728-5736. 

116. Morgenroth E, Obermayer A, Arnold E, Bruhl A, Wagner M, Wilderer PA. Effect of long- 
term idle periods on the performance of sequencing batch reactors. Water Sci Technol 2000; 
41:105-113. 

117. Lee N, Nielsen PH, Andreasen KH, et al. Combination of fluorescent in situ hybridization and 
microautoradiography-a new tool for structure-function analyses in microbial ecology. Appl 
Environ Microbiol 1999; 65:1289-1297. 

118. Nielsen JL, Christensen D, Kloppenborg M, Nielsen PH. Quantification of cell-specific 
substrate uptake by probe-defined bacteria under in situ conditions by microautoradiography 
and fluorescence in situ hybridization. Environ Microbiol 2003; 5:202-211. 

119. Daims H, Nielsen JL, Nielsen PH, Schleifer KH, Wagner M. In situ characterization of 
Nitrospira-like nitrite-oxidizing bacteria active in wastewater treatment plants. Appl Environ 
Microbiol 2001; 67:5273-5284. 

120. Adamczyk J, Hesselsoe M, Iversen N, et al. The isotope array, a new tool that employs 
substrate-mediated labeling of rRNA for determination of microbial community structure and 
function. Appl Environ Microbiol 2003; 69:6875-6887. 

121. Radajewski S, Ineson P, Parekh NR, Murrell JC. Stable-isotope probing as a tool in microbial 
ecology. Nature 2000; 403:646-649. 

122. Radajewski S, McDonald IR, Murrell JC. Stable-isotope probing of nucleic acids: a window 
to the function of uncultured microorganisms. Curr Opin Biotechnol 2003; 14:296-302. 

123. Boschker HTS, Middelburg JJ. Stable isotope and biomarkers in microbial ecology. FEMS 
Microbiol Ecol 2002; 40:85-95. 

124. Manefield M, Whiteley AS, Griffiths RI, Bailey MJ. RNA stable isotope probing, a novel 
means of linking microbial community function to phylogeny. Appl Environ Microbiol 2002; 
68:5367-5373. 

125. Corthier G, Delorme C, Ehrlich SD, Renault P. Use of luciferase genes as biosensors to study 
bacterial physiology in the digestive tract. Appl Environ Microbiol 1998; 64:2721-2722. 

126. Drouault S, Corthier G, Ehrlich SD, Renault P. Survival, physiology, and lysis of Lactococcus 
lactis in the digestive tract. Appl Environ Microbiol 1999; 65:4881-4886. 

127. Oozeer R, Mater DD, Goupil-Feuillerat N, Corthier G. Initiation of protein synthesis by a 
labeled derivative of the Lactobacillus casei DN-114 001 strain during transit from the 
stomach to the cecum in mice harboring human microbiota. Appl Environ Microbiol 2004; 
70:6992-6997. 



Molecular Ecology of the Human Intestinal Microbiota 23 

128. Oozeer R, Furet JP, Goupil-Feuillerat N, Anba J, Mengaud J, Corthier G. Differential 
activities of four Lactobacillus casei promoters during bacterial transit through the 
gastrointestinal tracts of human-microbiota-associated mice. Appl Environ Microbiol 2005; 
71:1356-1363. 

129. Cormack BP, Valdivia RH, Falkow S. FACS-optimized mutants of the green fluorescent 
(GFP). Gene 1996; 173:33-38. 

130. Andersen JB, Sternberg C, Poulsen LK, Bjorn SP, Givskov M, Molin S. New unstable variants 
of green fluorescent protein for studies of transient gene expression in bacteria. Appl Environ 
Microbiol 1998; 64:2240-2246. 

131. Bernard L, Courties C, Duperray C, Schafer H, Muyzer G, Lebaron P. A new approach to 
determine the genetic diversity of viable and active bacteria in aquatic ecosystems. Cytometry 
2001;43:314-321. 

132. Whiteley AS, Griffiths RI, Bailey MJ. Analysis of the microbial functional diversity within 
water- stressed soil communities by flow cytometric analysis and CTC + cell sorting. 
J Microbiol Methods 2003; 54:257-267. 

133. Ben-Amor K, Breeuwer P, Verbaarschot P, et al. Multiparametric flow cytometry and cell 
sorting for the assessment of viable, injured, and dead Bifidobacterium cells during bile salt 
stress. Appl Environ Microbiol 2002; 68:5209-5216. 

134. Apajalahti JHA, Kettunen A, Nurminen PH, Jatila H, Holben WE. Selective plating 
underestimates abundance and shows differential recovery of bifidobacterial species from 
human feces. Appl Environ Microbiol 2003; 69:5731-5735. 

135. Vaughan EE, Heilig H, Ben Amor K, De Vos WM. Diversity, vitality and activities of 
intestinal lactic acid bacteria and bifidobacteria assessed by molecular approaches. FEMS 
Microbiol Rev 2005; 29:477-490. 

136. Palmer C, Bik EM, Eckburg MB, Eisen MB, Relman DA, Brown PO. Microarray-based 
characterisation of the human colonic flora: a direct comparison to 16S rDNA sequencing — 
21C abstract. p37. In Beneficial Microbes ASM conferences, April 2005, Nevada U.S.A. 

137. Kohara Y, Noda H, Okano K, Kambara H. DNA probes on beads arrayed in a capillary, 
'Bead-array,' exhibited high hybridization performance. Nucl Acids Res 2002; 30:e87. 

138. Nolan JP, Sklar LA. Suspension array technology: evolution of the flat- array paradigm. 
Trends Biotechnol 2002; 20:9-12. 

139. Spiro A, Lowe M. Quantitation of DNA sequences in environmental PCR products by a 
multiplexed, bead-based method. Appl Envir Microbiol 2002; 68:1010-1013. 

140. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Ann Rev Nutr 2002; 22:283-307. 

141. Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that 
regulates fat storage. Proc Natl Acad Sci USA 2004; 101:15718-15723. 

142. www.microbial-ecology.net/probebase/. 



2 



Sampling Microbiota in the Human 
Gastrointestinal Tract 



Angele P. M. Kerckhoffs, Melvin Samsom, and Gerard P. van Berge Henegouwen 

Department of Gastroenterology, Utrecht University Medical Center, Utrecht, 
The Netherlands 

Louis M. A. Akkermans and Vincent B. Nieuwenhuijs 

Department of Surgery Utrecht University Medical Center, Utrecht, The Netherlands 

Maarten R. Visser 

Department of Microbiology, Utrecht University Medical Center, Utrecht, 
The Netherlands 



INTRODUCTION 

General Introduction 

Antonie van Leeuwenhoek (1632-1723) was the first to describe numerous micro- 
organisms from the gastrointestinal tract, which he described as "animalcules," having 
designed the first glass lenses for the microscope that were powerful enough to observe 
bacteria. His curiosity brought him to investigate samples taken from his own mouth 
and other people who never brushed their teeth, and he compared these findings with 
people who brushed their teeth daily and used large amounts of alcohol. He even 
investigated his own fecal samples in a period of diarrhea, compared these findings 
with fecal samples of animals, and reported these observations to the Royal Society in 
London (1). 

We now know that the mucosal surface of the human gastrointestinal tract is about 
300 m 2 and is colonized by 10 13 — 10 14 bacteria consisting of hundreds of different species. 
The prevalence of bacteria in different parts of the gastrointestinal tract depends on pH, 
peristalsis, oxidation-reduction potential within the tissue, bacterial adhesion, bacterial 
cooperation, mucin secretion containing immunoglobulins (Ig), nutrient availability, diet, 
and bacterial antagonism. The composition of the Gram-negative, Gram-positive, aerobic, 
and anaerobic microbiota has been extensively studied by culturing methods, and shown to 
change at the various sites of the gastrointestinal tract (Fig. 1). 

The stomach and proximal small bowel normally contain relatively small numbers 
of bacteria because of peristalsis, and the antimicrobial effects of gastric acidity. An intact 
ileocecal valve is likely to be an important barrier to backflow of colonic bacteria into the 

25 



26 



Kerckhoffs et al. 




w 


_c 


13 


o 


a> 


03 


CO 


E 


1— 


o 


O 


CO 


CO 










E 

=3 
C 
CD 
O 
=5 
T3 



10i 



E 

=3 




C 

_o 

o 

o 







Q Gram-negative bacteria 

■ Gram-positive bacteria 

E3 Anaerobes 

g Aerobes/facultative 
anaerobes 



Figure 1 Numbers ( lu log) of gram-negative bacteria, gram-positive bacteria, anaerobes and 
aerobes and facultative/anaerobes per gram of intestinal material in the human intestinal tract. 
Source: From Refs. 2-8. 



ileum. The intestinal microbiota play a prominent role in gastrointestinal physiology and 
pathology. A bacterial population is essential for the development of the gastrointestinal 
mucosal immune system, for the maintenance of a normal physiological environment, and 
for providing essential nutrients (9). Culturing techniques suggested that dietary changes 
had a negligible effect on the intestinal microbiota composition (2,10). More recently 
molecular techniques indicated that diet can alter the microbiota composition, but the 
predominant groups are generally not substantially altered (11,12). In contrast, antibiotics 
can dramatically alter the composition of the intestinal microbiota. 



Physiology of Microbiota Host Interaction in Humans 

Normal gastrointestinal tract microbiota is essential for the physiology of its host. The 
microbiota in the gastrointestinal tract have important effects on nutrient processing, 
immune function, and a broad range of other host activities some of which are briefly 
described below (13). Pasteur (1822-1895) suggested that the intestinal microbiota might 
play an essential role in the digestion of food. We now know that bacteria harbor unique 
metabolic capabilities which enable otherwise poorly utilizable nutrients to be 
metabolized (14). The intestinal microbiota possess enzymes that can convert endogenous 
substrates, and dietary components, such as fibers, to provide short-chain fatty acids, and 
other essential nutrients, which are absorbed by the host (10). This interaction of host 
and bacteria, when one or both members derive specific benefits from metabolic 
capabilities, is defined as mutualism. Bacteria also produce a number of vitamins that the 
host can utilize, especially those of the B-complex (15). 

The microbiota affords resistance to colonization by potential pathogens that cannot 
compete with entrenched residents of the microbial community for nutrients (13). 
Autochthonous or native microorganisms colonize specific intestinal habitats, whereas 
allochthonous or transient bacteria can only colonize particular habitats under abnormal 
conditions. The normal microbiota prevent colonization of allochthonous species or 
potential pathogens by releasing metabolic waste products as well as bacteriocins, and 
colicins which have antibacterial activity. A pathogenic relationship results in damage to 
the host. Most pathogens are allochthonous microorganisms. However, some pathogens 



Sampling Microbiota in the Human Gastrointestinal Tract 27 

can be autochthonous to the ecosystem, and live in harmony with the host unless the 
system is disturbed. Antibiotic therapy can drastically reduce the normal microbiota, and 
the host may then be overrun by introduced pathogens or by overgrowth of commensal 
microbial members normally present in small numbers. One notable example is following 
treatment with clindamycin, overgrowth by Clostridium difficile that survives the 
antibiotic treatment can give rise to pseudomembranous colitis (10,16). 

Microbial factors are known to influence host postnatal development. Commensals 
acquired during the early postnatal life are essential for the development of tolerance, not 
only to themselves but also to other luminal antigens. Development of B- and T-cell 
responses depend on the microbiota. The natural antibodies that arise in response to the 
antigens of the normal gut microbiota are of great importance in immunity to a number of 
pathogenic species. Somatic hypermutation of Ig genes in intestinal lymphoid follicles 
plays a key role in regulating the composition of the microbial community (14). 

The microbiota participate in bile acid metabolism. In the colon, bacterial enzymes 
convert cholic acid and chenodeoxycholic acid into the secondary bile acids deoxycholic 
acid and lithocholic acid, respectively, which in general are poorly reabsorbed; most of 
these are then eliminated in the stool. In patients with small bowel bacterial overgrowth 
(SBBO), bile acids are deconjugated and metabolized more proximally in the small bowel, 
and removed from further participation in the normal enterohepatic circulation, resulting 
in bile acid malabsorption and steatorrhea. Steatorrhea is defined as excessive loss of fat in 
the stool, i.e., greater than 7 g or 9% of intake for 24 hours (3). 

The effects of having a normal intestinal microbiota has been determined by 
comparing the characteristics of germ-free and conventionally reared animals. In the small 
bowel of germ-free animals there are dramatic reductions in leukocytic infiltration of 
the lamina propria, and both the size and number of Peyer's patches. Moreover, the 
intraluminal pH is more alkaline, and the reduction potential more positive. Colonization 
of the intestinal tract of germ-free animals with even a single strain of bacteria is followed 
by the rapid development of physiologic inflammation of the mucosa resembling that of 
conventional animals. The migrating motor complex (MMC) is a cyclic pattern of motility 
that occurs during fasting, and is an important mechanism in controlling bacterial 
overgrowth in the upper small bowel. Gut transit is slow in the absence of the 
intestinal microbiota. The effect of selected microbial species in germ-free rats on 
small intestinal myoelectric activity is promotion or suppression of the initiation and 
migration of the MMC depending on the species involved. Anaerobes, which have a 
fermentative metabolism, emerge as important promoters of regular spike burst activity in 
the small intestine. Introduction of the fermentative species Clostridium tabificum, 
Lactobacillus acidophilus, and Bifidobacterium bifidum into the gastrointestinal tract of 
germ-free rats significantly reduces the MMC period, and accelerates small intestinal 
transit. In contrast introduction of bacteria with respiratory potential such as Micrococcus 
luteus and Escherichia coli in the germ-free rats prolongs the MMC period. Intestinal 
microbiota accelerate transit through the small intestine in the fasting state compared to 
the unchanged intestinal myoelectric response to food. Overall, the promoting influence 
of the conventional intestinal microbiota on MMC reflects the net effect of bacterial 
species with partly opposite effects (17-19). 

In conclusion, the bacterial microbiota has a range of specific functions including 
intestinal transit, absorption of nutrients, and in the modulation of the immune system of the 
gastrointestinal tract. The introduction of pathogen bacteria can disturb the normal 
physiological functions of the gastrointestinal tract to a great extent. A number of functional 
tests for the detection of intestinal pathogenic bacteria have been developed, and are 
described below. 



28 Kerckhoffs et al. 

Importance of Sampling the Gastrointestinal Tract 

The current knowledge of the human intestinal microbiota is mostly based on culture 
techniques but also more recently on molecular biology techniques that are applied to feces 
and gastrointestinal fluids or biopsies. Sampling of the gastrointestinal tract is clinically 
necessary for the diagnosis of Helicobacter pylori, and the etiology of diarrhea. The 
gastrointestinal tract is also sampled for research questions on SBBO or for the investigation 
of host-bacterial relationships in the gut. There are various methods of obtaining material to 
study the microbiota. Research or diagnosis of bacteria anywhere in the gastrointestinal 
tract can be performed using invasive or noninvasive methods. The various methods of 
investigating microbiota in the gastrointestinal tract will be specified for different 
compartments of the gastrointestinal tract, and the advantages and disadvantages of the 
sampling methodologies will be described below. 



ESOPHAGUS: MICROBIOTA AND SAMPLING TECHNIQUES 

Normal Microbiota 

The mouth and the oropharynx predominantly harbor Gram-positive organisms (20). The 
most numerous species comprise the streptococci, Neisseria, and Veillonella, but 
Fusobacteria, Bacteroides, lactobacilli, staphylococci, yeasts, and Enter ob act eria are 
also present in smaller amounts (4). The esophagus is covered with a stratified squamous 
epithelium layer, which is a mechanical barrier coated with saliva and mucus, that has high 
peristalsis and Ig containing mucus secretion, all of which contribute to prevention of 
infection. Because of the lack of absolute anatomic or known physiological barriers, 
bacteria can be introduced into the esophagus by the swallowing of food, by resident oral 
microbiota or by reflux from a colonized stomach (21). The esophagus, with its large 
mucosal surface located just downstream of the bacterial species-rich oropharynx, 
provides a potential environment for bacterial colonization, but so far limited research has 
been performed. A recent molecular analysis of the distal esophagus indicated members of 
6 phyla, of which Streptococcus (39%), Prevotella (17%), and Veillonella (14%) were the 
most prevalent, and also demonstrated that most esophageal bacteria are similar or 
identical to residents of the upstream oral microbiota (21). Quantitative cultivation-based 
studies indicated that aerobic organisms were present in all, and obligate anaerobes in 80% 
of the subjects investigated. No differences in frequencies of isolation or composition of 
the microbiota were found between different subjects (5,22). 



Disease-Causing Microbiota 

A pathogen is a microorganism which by direct contact with or infection of another 
organism causes disease in that organism. Thus a microbe which produces a toxin that 
causes disease in the absence of the microbe itself would not be regarded a pathogen. 
Members of the commensal microbiota may become pathogenic and cause disease if the 
host defense mechanisms are compromised, or if they are introduced into normally sterile 
body sites. The esophagus of individuals with deficient immune systems (HIV or post- 
transplantation patients) may become infected with Candida albicans, cytomegalovirus, 
herpes simplex virus, Histoplasma capsulatum, Mycobacterium avium, and Cryptospor- 
idium. These microorganisms are usually not seen in immunocompetent persons. With the 
exception of Mycobacterium species, bacterial etiologies for inflammation involving 
the distal esophagus have not been explored (23). Mycobacterial involvement of the 



Sampling Microbiota in the Human Gastrointestinal Tract 29 

esophagus is rare (incidence 0.14%) in both immunocompromised and immunocompetent 
hosts with advanced pulmonary tuberculosis (23). 



Luminal Washes 

Luminal washes to sample esophageal bacteria give poor yields. The washes may contain 
a few transient bacteria of oropharyngeal origin, or even no microbes at all, or an average 
of 16 colony forming units per ml (CFU/ml) with no common species found (24,25). 
Either intestinal contents are passed through the alimentary canal with high peristalsis, and 
prevent bacteria from residing in the esophagus, or the bacteria present in the washes are 
not culturable. Another possibility is that the bacteria are very closely associated with the 
esophageal mucosa, and cannot be removed by simple washes. This technique is not 
commonly used for research questions, and is clinically irrelevant. 



Biopsy 

Esophageal mucosal biopsy specimens from the distal esophagus can be obtained during 
upper endoscopy. The endoscope passes orally into the esophagus, and the biopsy forceps 
can be shielded from the oral microbiota. The forceps consists of a pair of sharpened cups. 
Forceps with a central spike make it easier to take specimens from lesions which have to 
be approached tangentially (such as in the esophagus). The maximum diameter of the cups 
is limited by the size of the operating channel. The length of the cups is limited by the 
radius of curvature through which they must pass in the instrument tip (26). Patients are 
instructed not to eat or drink for at least 4-6 hours before endoscopy (small sips of water 
are permissible for comfort) (27). The channel of the endoscope can also harbor bacteria if 
secretions have inadvertently been suctioned while advancing the endoscope. 
Oropharyngeal and gastric bacteria can contaminate the biopsy. Chlorhexidine or 
acidified sodium chlorite mouth rinse has been used to decontaminate the oropharynx. To 
compare biopsy samples of two individuals or to compare the reproducibility in one 
subject the biopsies have to be taken at the same level (28). 



STOMACH: MICROBIOTA AND SAMPLING TECHNIQUES 

Normal Microbiota 

The human stomach is lined with columnar secreting epithelium. Normally most of the 
bacteria in the stomach are killed because of the low pH levels, and the typical numbers 
detected are less than 10 CFU/ml (2,6,26). Lactic acid bacteria are commonly isolated 
from the human gastric acid contents, especially when good anaerobic techniques are 
used. Candida and some other yeast species are also detected. Bacteria isolated from 
gastric contents are considered transient members. These bacteria have been passed down 
from habitats above the stomach or have been present in ingested materials (29). The 
normal resident microbiota of the stomach consists mainly of Gram-positive aerobic 
bacteria, such as streptococci, staphylococci, and lactobacilli (2,6,26,30). The microbiota 
isolated from gastric contents are presented in Table 1. In healthy fasting patients large 
numbers of Enterococcus, Pseudomonas, Streptococcus, Staphylococcus, and Rothia 
(Stomatococcus) may be isolated in culture when acidity is physiologically reduced, as 
occurs at night, and during phase I (motor quiescence) of the MMC (32-34). 



30 Kerckhoffs et al. 

Table 1 Microorganisms Isolated from the Stomach by Culturing 

Microbial type 

Lactobacilli 

Streptococci 

Bifidobacteria 

Clostridia 

Veillonella 

Coliforms 

Peptostreptococcus, Bacteroides 

Staphylococcus, Actinobacillus 

Candida albicans 

Torulopsis 

Unidentified yeasts 

Neisseria 

Micrococcus 

Note: The most prevalent bacterial types are italicized. 
Source: From Refs. 2, 15, 22, 31. 



Disease-Causing Microbiota 

Bacteria closely associated, and attached to the epithelium like Helicobacter pylori, may 
be sampled from gastric contents with difficulty (29). H. pylori is a Gram-negative 
bacterium that resides below the mucous layer next to the gastric epithelium. H. pylori is 
rarely found before age 10 but increases to 10% in those between 18 and 30 years of age, 
and to 50% in those older than age 60 (35). In developing nations the majority of children 
are infected before age 10, and adult prevalence peaks at more than 80% before age 50. 
Thus H. pylori infection ranges depend on age and socioeconomic differences (36). 
H. pylori produces urease, an enzyme that breaks down urea into ammonium and 
bicarbonate. Ammonium provides an alkaline environment, which helps the bacterium 
protect itself from gastric acid injury. Most infected subjects do not have symptoms of 
H. pylori infection. However, H. pylori may induce acute gastritis with symptoms such as 
epigastric pain, bloating, nausea and vomiting, and/or chronic gastritis. Furthermore, it 
may also be associated with ulcer disease and gastric carcinomas. 

Other gastric bacteria besides Helicobacter species only become apparent in patients 
with reduced acidity (achlorhydria). Achlorhydria may occur in elderly persons (37). 
Colonization of the gastric lumen may occur in patients on anti- secretory medication 
meant to reduce gastric acid secretion. Many subjects regularly use these anti- secretory 
drugs. Acid suppression may allow bacteria to survive in the stomach which results in 
gastric bacterial overgrowth with the degree of overgrowth depending upon the elevation 
of the pH (20). Infectious gastritis is more rarely caused by Mycobacterium tuberculosis, 
Mycobacterium avium, Actinomyces israellii, and Treponema pallidum (3). 



Biopsy 

To investigate the gastric microbiota, tissue is generally obtained by an endoscopic biopsy. 
Slightly less invasive methods are available to obtain a specimen such as the use of a small 
bowel biopsy tube or capsule, or biopsy forceps that can be passed through a modified 
nasogastric tube positioned either in the gastric body or antrum. A biopsy is clinically 
unnecessary to diagnose H. pylori via microbiological methods unless one wishes to 



Sampling Microbiota in the Human Gastrointestinal Tract 31 

isolate the organism for antibiotic susceptibility testing. Recommendations to maximize 
the diagnostic yield of endoscopic biopsies include the use of large-cup biopsy forceps, 
obtaining at least two samples from the lesser curvature and the greater curvature (the 
prepyloric antrum and the body), and proper mounting and preparation of the samples. 
Special stains (H&E, Giemsa, and Warthin- Starry staining) are often used to help detect 
the presence of H. pylori (38). 

The rapid urease test (by agar gel slide tests) involves placing a biopsy specimen from 
the antrum of the stomach on a test medium that contains urea (39). The biopsy specimens 
for the rapid urease test have to be removed from the sterilized biopsy forceps with a sterile 
toothpick, and have to be placed immediately into a tube. The urea is hydrolyzed by urease 
enzymes of H. pylori, and the ammonium formed increases the pH. A phenol indicator that 
changes the color from yellow at pH 6.8 to magenta at pH 8.4 can detect the pH alteration. 
The color change read off 1 hour after and 24 hours after the introduction of the gastric 
biopsy is an indication for the presence of H. pylori. Recommendations to maximize the 
rapidity and sensitivity of rapid urease tests are to warm the slide, and to use two regular or 
one jumbo biopsy specimen(s) (40). Increasing the number of biopsies to more than two 
biopsies from the antrum may increase the sensitivity, given that this probably increases 
the H. pylori load, and therefore the amount of urease. However, this will prolong the 
endoscopy time and add to the discomfort of the patient. The agar gel test may take up to 
24 hours to turn positive, particularly in the presence of a low bacterial density. Recent use 
of antibiotics, bismuth, or proton pump inhibitors may render rapid urease tests falsely 
negative. Compared with histology as the gold standard in the diagnosis of H. pylori 
infection, the sensitivity of the rapid urease test is 70-99%, and the specificity is 92-100% in 
untreated patients (40). Mucosal biopsies can be fixed in neutral buffered formaldehyde, and 
if the rapid urease test is negative the biopsy can sent in the next day for histologic 
assessment. The presence or absence of//, pylori can be established by examining three sets 
of tissue levels within 12 consecutive sections. On microscopic examination of the tissue 
obtained by biopsy, the bacteria may be seen lining the surface epithelium. The sensitivity 
for histologic examination is 70-90%. Giemsa staining is required for H. pylori diagnosis. 
Culture for//, pylori is insensitive. Biopsies should be plated within 2 hours (or transported 
in a special medium) on nonselective media enriched with blood or serum, and incubated in 
a moist and microaerobic atmosphere. The identity of any colonies grown can be confirmed 
using Gram's stain and biochemical tests. 

Aspiration 

In order to sample gastric fluid a Shiner tube may be used. This is a polyvinyl tube with a 
stainless steel sampling capsule at the end with which the specimens are obtained by 
suction. This tube can be sterilized in the autoclave or by boiling (6). Sampling the luminal 
content of the stomach may lead to underestimation of the size or even misinterpretation 
of the composition of gastric microbial communities (29). Estimates per unit weight of 
material of the population levels of microbes attached to an epithelium surface made from 
samples of the mucosa itself have been found to be higher than estimates made from the 
luminal content in the region (29). This technique is not clinically relevant, and is hardly 
ever used in research models. 

Urea Breath Test 

The urea breath test is a noninvasive test that detects radio-labeled carbon dioxide excreted 
in the breath of persons with H. pylori infection; orally administered urea is hydrolyzed to 



32 Kerckhoffs et al. 

carbon dioxide and ammonium in the presence of the enzyme urease, which is present in 
H. pylori. In non-infected subjects, urea leaves the stomach unchanged, unless there is 
urease activity from bacteria in the oral cavity or in situations of gastric bacterial 
overgrowth. The urea breath test is a highly sensitive (93.3%) and specific (98.1%) 
method (41). The two breath tests available are the 14 C urea (radioactive), and 13 C urea 
(stable isotope) breath tests. The C urea breath test avoids radioactivity, and is the test of 
choice for children and pregnant women. The major limitation is the need for a gas isotope 
mass spectrometer to analyze the breath samples and calculate the ratio of C to C. A 
4-hour fast is generally recommended before the urea breath test, and a test meal is given 
before the solution of labeled urea. This test meal delays gastric emptying, and increases 
contact time with the bacterial urease. It is relatively inexpensive compared to the "gold 
standard" of endoscopy with biopsy, and histological examination described above. The 
urea breath test avoids sampling errors that can occur with random biopsy of the antrum. 
False positive results can occur if gastric bacterial overgrowth with urease-producing 
bacteria other than H. pylori are present. False positive results can also occur if the 
measurements are taken too soon after the urea ingestion because the action of the oral 
microbiota on the urea may be measured. False negative results can be obtained if the 
patients were recently treated with antibiotics, bismuth preparations or acid suppression 
therapy, because the test is dependent on the numbers of//, pylori (42). Performance of the 
urea breath test has been associated with several disadvantages especially in infants, 
toddlers or handicapped children because one needs active collaboration. False positive 
results in infants affect the accuracy of the test, but correction for the carbon dioxide 
production of the tested individual will improve the specificity (43,44). 

Other tests that do not require a mucosal biopsy include serologic tests and stool 
antigen tests. Chronic H. pylori infection elicits a circulating IgG antibody response that 
can be quantitatively measured by enzyme-linked immunosorbent assay (ELISA tests). 
The ELISA is based on a specific anti-//. pylori immune response, and this serologic test is 
as sensitive (95.6%) and specific (92.6%) as biopsy-based methods (41). The presence of 
IgG does not indicate an active infection. IgG antibody titers may decrease over time 
(6-12 months) in patients who have been successfully treated. ELISA or immuno- 
chromatographic methods can be performed on the fecal samples to detect H. pylori 
antigen. The limit of sensitivity of the test is 10 5 H. pylori cells per g of feces (45). 
Sensitivities and specificities of 88-97% and 76-100% have been reported (41,44-47). 
The stool antigen test is not used for follow-up evaluation of the H. pylori eradication as it 
gives false positive results. In conclusion, the noninvasive tests are sufficiently accurate 
for the diagnosis of H. pylori infection. 



SMALL INTESTINE: MICROBIOTA AND SAMPLING TECHNIQUES 

Normal Microbiota 

The small intestine comprises the proximal, mid, and distal areas, which are designated 
the duodenum, jejunum, and ileum. The velocity of the intraluminal content of the small 
intestine decreases from the duodenum to the ileum. The microbes isolated from the 
small intestine include those descending from habitats above the small intestine such as the 
mouth, and ingested food. The microbes pass through the intestine with the chyme, and in the 
fasting state by the MMC. The MMC interdigestive motility prevents colonic microbiota from 
entering the proximal small intestine which would cause SBBO. The microbial species 
isolated from the small intestine are listed in Table 2. The density of microbiota increases 
towards the distal small intestine. The upper two thirds of the small intestine (duodenum and 



Sampling Microbiota in the Human Gastrointestinal Tract 33 

jejunum) contain only low numbers of roughly the same microorganisms, which range 
from 10 to 10 bacteria/ml (2). Culturing studies indicated that acid- and aero-tolerant 
Gram-positive species such as lactobacilli and streptococci dominate in the proximal part, 
while distally anaerobic, and more Gram-negative bacteria increasingly dominate. Whipple's 
disease is a rare multisy stemic bacterial infection caused by Tropheryma whipplei. T. whipplei 
could not be cultured from the small intestine for decades, and was diagnosed by 
histopathology. Nowadays T. whipplei can be detected using polymerase chain reaction 
(PCR) or ribosomal RNA techniques on duodenal biopsies or fecal samples (48). The rich 
microbiota of the initial section of the large intestine (cecum) find their way through the 
ileocecal valve back into the ileum. The microbiota of the ileum begins to resemble that of the 
colon with around 10 to 10 bacteria/ml of the intestinal contents. With decreased 
intraluminal transit, decreased acidity, and lower oxidation-reduction potentials, the ileum 
maintains a more diverse and numerous microbial community (29). Factors that compromise 
the oxidation-reduction potential within the tissues are obstruction and stasis, tissue 
anoxia, trauma to tissues, vascular insufficiency, and foreign bodies (49). Decreased 
oxidation-reduction potential specifically predisposes to infection with anaerobes (50). 



Disease-Causing Microbiota 

Pathogenic bacteria of the small intestine, which cause severe diarrhea, are enterotoxic 
Escherichia coli (ETEC) and Vibrio cholerae. V. cholerae is diagnosed when it is present in 
fecal material. ETEC produces enterotoxins that cause intestinal secretion and diarrhea, and is 
a common cause of traveler's diarrhea. In SBBO, the proximal small intestine is populated by 
a substantially higher number of microorganisms than usual. These are frequently anaerobic 
bacteria that are normally not present in large numbers in the duodenum and the proximal 
jejunum. A total count of microorganisms exceeding 10 5 colony forming units/ml in a 
duodenal or jejunal aspirate is generally accepted as SBBO (51). Some gastroenterologists 



Table 2 Microorganisms Isolated from the Small Intestine by Culturing 

Most prevalent microbes in duodenum Most prevalent microbes in 

Microbial types and proximal jejunum distal jejunum and ileum 

Lactobacilli Lactobacilli 

Streptococci Streptococci 

Bifidobacteria Bifidobacteria 

Clostridia Clostridia 

Coliforms 

Bacteroides Bacteroides 

Veillonellae Veillonellae 

Gram positive 

nonsporing 

anaerobes 
Staphylococci Staphylococci 

Actinobacilli Actinobacilli 

Yeasts Yeasts 

Candida albicans 
Haemophilus 

Fusobacterium 

Note: The most prevalent bacterial types are italicized. 
Source: From Refs. 2, 15, 22. 



34 



Kerckhoffs et al. 



also accept a concentration of colonic microorganisms above 10 CFU/ml as positive for 
SBBO. A profound suppression of gastric acid may facilitate the colonization of the upper 
small intestine (20). To diagnose SBBO, the quantitative culture of a small intestine is used, 
and considered to be the gold standard. Fluid aspirated from the descending part of the 
duodenum may be cultured in order to detect bacterial overgrowth in diffuse small 
bowel disorders. 



Biopsy 

To obtain biopsy samples from the small intestine upper endoscopy has to be performed. 
Upper endoscopy is performed after an overnight fast of at least 10 hours. An endoscope 
has a length of approximately 1 meter, and has a biopsy channel. During endoscopy the 
esophagus, stomach, and duodenal wall can be systematically inspected. To allow a good 
view air insufflation is required; the patient may complain of bloating during the 
endoscopy. When the endoscope reaches the site of interest, the biopsy from the small 
intestinal mucosa is rapidly taken by standard biopsy forceps. Figure 2 shows the size 
(in centimeters) of the tip of an endoscope, and a biopsy forceps. The distal part of the 
jejunum and the ileum cannot be reached using a standard endoscope, and therefore is not 
sampled. Endoscopic biopsies are an adequate substitute for jejunal suction biopsies. The 
advantage over capsule biopsy is that the site of interest can be inspected before the biopsy 
is taken (52-54). Adequacy of mucosal biopsies is a function of size and numbers of 
biopsies obtained (54). Alligator-type forceps obtain larger specimen pieces than oval- 
shaped forceps (55). Forceps with a needle, or the multibite forceps, allow more biopsies 
to be taken per passage, and improve the quality of tissue obtained (55). Biopsy forceps 
without a needle can be used to obtain two samples per passage through the endoscope that 
are quantitatively as good as when only one sample is collected. This approach can save 
time, and causes no significant damage to the biopsy specimens. Because air insufflation 
may distort the intraluminal anaerobic environment, nitrogen could be used as a substitute 
if the intention is to culture anaerobic bacteria. There is also the risk of contamination with 
microbiota from more proximal habitats that were passed along via the endoscope. 




Figure 2 Tip of a standard endoscope and biopsy forceps with needle (tape measure in 
centimeters). 



Sampling Microbiota in the Human Gastrointestinal Tract 35 

The biopsies have to be taken at a certain distance from the endoscope to prevent sampling 
contaminated parts of the intestine. 

Intestinal biopsies taken from living persons may not yield satisfactory results 
because the biopsies are only a minimal part of the total intestinal wall (56). The number 
of persons sampled must be large to generate reliable results. The best source of 
information on microbiota in the small intestine so far has been achieved with sampling 
from autopsy studies of accident victims. As slow cooling of the gastrointestinal tract can 
cause alterations in bacterial localization the samples have to be taken immediately after 
death (57), and the number of individuals sampled must still be quite large. 

Full Thickness Biopsy 

Full thickness biopsy is a peroperative or laparoscopic biopsy (muscularis-containing 
biopsy) used to diagnose motility disturbances. One incision is situated below the 
umbilicus, and one in the left fossa. The bowel loop is identified laparoscopically, and will 
then be exteriorized through the incision below the umbilicus. The full thickness biopsy of 
at least 10X10 mm will then be taken with a surgical knife. The bowel loop is closed with 
absorbable sutures, and repositioned into the abdomen (56). Drawbacks of biopsies taken 
at surgery are the manipulation of the patients' diet (fasting), and the bowel preparation or 
preoperative treatment with antibiotics (29,58). Biopsies taken at surgery have the 
advantage of larger sample size than endoscopic biopsies, and various analyses may be 
applied such as molecular typing of bacteria in intestinal tissue of Crohn's patients (59). 

Mucosal Brushings 

Mucosal brushings may be used to sample bacteria from the intestinal mucosa. The 
cytology brush, protected by a sheath, is passed through the instrument channel of 
the endoscope. After the endoscope is placed at the location of interest, the brush is 
advanced from its sleeve within sight of the mucosal surface, and rubbed and rolled across 
the surface. Thereafter, the brush is pulled back into the sleeve. Normally, cytology 
brushes are only covered with a plastic sleeve to protect the specimen during withdrawal. 
This sleeve, however, does not protect against contamination; the use of suction of saliva 
and gastric fluid during endoscopy contaminates the suction channel of the endoscope, and 
the subsequent passage of the brush without a sheath through the suction channel causes 
loss of sterility of the brush (27). These brushes cannot be used for sampling bacteria in the 
lumen of the gastrointestinal tract. Avoidance of any suction during endoscopy is 
extremely difficult. To obtain small bowel samples without contamination one could 
utilize a catheter with a specimen brush plugged with sterile Vaseline. Brushes cannot be 
protected from contact with air, so it is not useful for the isolation of anaerobes for culture. 
To determine the concentration of bacteria obtained by the brush present per milliliter, one 
has to standardize the loading capacity of the brush used. Brushing is a highly reproducible 
technique (92%) (60). 

Peroperative Needle Aspiration 

Peroperative needle aspiration is useful for relatively inaccessible locations within the 
intestinal tract. The technique is only applicable for patients with an underlying disease who 
will undergo laparotomy or laparoscopy. The microbiota may be influenced by pre-operative 
fasting, antibiotic prophylaxis, and anesthesia. Until 1959 the peroperative needling technique 
was regularly performed at operation (61,62) but is currently no longer performed routinely. 



36 Kerckhoffs et al. 

The advantages of this technique are asepsis and the lack of contamination from other regions 
of the gastrointestinal tract. 



Self-Opening Capsule 

The Crosby capsule, first applied in 1957, was used to obtain biopsies from the small 
intestine before the introduction of the endoscope. This self-opening capsule is a metallic 
capsule of 19 to 1 1 mm with a round opening of 4 mm (53). A long tiny tube is attached to 
the capsule, and this is muscle loaded through an endoscope which is passed into the 
second part of the duodenum. Intestinal mucosa is sucked into the tube by suction and 
excised. Every part of the stomach and the small intestine can be reached (63). Sizes of the 
biopsies are 5-8 mm, with stomach biopsies usually being smaller. Failure of obtaining 
biopsies is 6%. The mucous membrane is very mobile with respect to the muscular layer so 
only mucosa is sucked into the capsule, and the risk of perforation is very small. 
Muscularis propria is never cut. The risk of bleeding (0.14%) and intestinal perforation is 
very small (64). 

Capsules that can be opened electronically are also available. They have the 
disadvantage of a long interval between sample collection and culturing. During this 
interval, bacteria inside the capsule can replicate, and influence growth of other bacteria in 
the capsule. It is a very imprecise method. The advantage of this technique is that, like the 
Crosby capsule, every part of the small intestine can be examined. The disadvantage of the 
suction biopsy capsule used to provide specimens from the proximal jejunum is the need 
for radiological screening for the location of the capsule. This makes it unsuitable for 
repeated use in young children, and women who are or might be pregnant. There may be 
some discomfort when the procedure is prolonged. The technique fails in up to 10% of the 
cases. To overcome the problem of determining the sampling location with the capsule 
biopsy, it is better to take specimens with endoscopic forceps. Capsule biopsies are not 
common in current clinical gastroenterology practice (52). 



Aspirate 

Small bowel aspiration for quantitative and qualitative culture specimens is still regarded 
as the gold standard for diagnosis of SBBO. The sample should be properly harvested with 
respect to sterile technique and accurate location. The exact composition of the microbiota 
is not important for the diagnosis of SBBO if one uses the definition that more than 10 5 
colony forming units/ml small intestinal fluid represents SBBO, but it is of use when 
antibiotic therapy is being considered. It should be realized that cultures of randomly 
harvested samples can produce false-negative results if the sample is not taken from the 
actual site of bacterial overgrowth. 

Culturing is not necessary if one uses gas chromatographic detection and analysis of 
volatile fatty acids in the aspirates. The volatile fatty acids are produced by the metabolism 
of microorganisms such as Bacteroides and Clostridia. This is essentially a rapid test for 
the presence of anaerobic bacteria. When gas chromatography of volatile fatty acids is 
compared with cultures of jejunal aspirates, it shows a sensitivity of 56% and specificity of 
100% (51). When the tests for volatile fatty acids in jejunal aspirates are positive, this 
always indicates the presence of bacterial overgrowth. This procedure avoids the more 
complicated, time-consuming, and expensive bacteriological analysis of jejunal samples 
(51,65,66). The numbers of bacteria per milliliter of intestinal fluid taken at two different 
levels of the proximal jejunum show highly significant correlations (rs = 0.90, p< 0.001); 



Sampling Microbiota in the Human Gastrointestinal Tract 37 

thus one does not have to obtain the aspirate from the exact same location in the proximal 
jejunum (51). 

Aspirate can be acquired by intestinal intubation with sterile or nonsterile tubes, the 
capsule method, direct needle aspiration of the gut contents, peroral intubation, and by 
the string test as described below. 

Intubation with Sterile or Nonsterile Tubes 

This endoscopic method for collection of proximal gastrointestinal fluid for culture is 
simple and can be performed during routine endoscopy. When the endoscope reaches the 
descending part of the duodenum, the polyethylene tube will pass through the biopsy 
channel into the intestinal lumen. Intestinal intubation seems to be the most suitable and 
reliable method for studying small intestinal microbiota, because of the short sample 
collection time and minimal disturbance of physiological conditions. Care must be taken to 
prevent contamination with upper respiratory tract microbiota during the passage of the 
tube, and to maintain oxygen-free conditions for anaerobic culturing. A closed 
polyethylene tube filled with water through the suction channel of the endoscope is 
therefore recommended, as it is not necessary to keep the suction channel sterile. The water 
has to have been boiled for sterilization and the removal of dissolved oxygen. The distal end 
is closed with a plug of agar. Because the innertube remains sterile even after the 
passage through the nonsterile suction channel of the endoscope, the use of an overtube 
eliminates the possibility of contamination. The proximal end can be attached to a double 
way stopcock connected to a syringe containing boiled water. In the duodenum the agar plug 
can be expelled from the tube by injection of the water in the syringe. After several minutes 
the expelled water has gone through and the duodenal contents can be aspirated into the 
tube, after which the tube is removed from the endoscope. Precision of the sample site and 
proven absence of contamination are the main advantages. Since fresh aspirate is known to 
tolerate oxygen fairly well for an exposure time of at least 8 hours, it is a good method for 
obtaining aerobic and anaerobic samples (60,62,67). 

Highly significant correlations (rs = 0.84, p< 0.001) were found between the 
numbers of bacteria/ml of jejunal aspirate obtained from the closed and open tubes, 
confirming that the intubation method is highly reproducible (51). The use of suction 
during endoscopy contaminates the suction channel of the endoscope. The first milliliter of 
aspirate can be discarded to avoid this, although this is very difficult in the duodenum, 
where at best only a few milliliters of aspirate will be found (67). Using an open tube for 
collection of small bowel fluid can theoretically lead to contamination, but according to 
reported studies this does not seem to be the case (68,69). 

Duodenal String Test (Enterotest) 

The duodenal string test capsule is a cheap and simple device used for sampling the 
contents of the upper gastrointestinal tract. It has been used for the diagnosis of typhoid 
fever, whereby sampling duodenal contents by a "string" test yields a positive culture in 
70% of patients (70). The weighted gelatin capsule contains a silicone rubber bag and a 
140 cm highly absorbent nylon string. After a 10-hour fast the device is administered. The 
first 10 cm of the nylon line is pulled out from the capsule by the protruding loop. 
The capsule is then swallowed with water while the loop is held outside the mouth. The 
loop is then taped to the face to secure the line. After approximately 3.5 hours the thread 
has moved into the duodenum. The volume of the duodenal fluid absorbed by the distal 
end of the thread is calculated by subtracting the dry weight of the segment. The distal end 
is squeezed out between sterile gloved fingers in order to collect the intestinal contents. 



38 Kerckhoffs et al. 

Its major applications in pediatrics are the diagnosis of enteric parasitic infestations, and 
the diagnosis of Salmonella infection, Giardia lamblia, and assessment of duodenal bile 
salts in the diagnosis of neonatal cholestasis in duodenal contents. A drawback of the 
Enterotest is that when the string is pulled out of the gastrointestinal tract, the intestinal 
contents adhering to it are exposed first to the sterilizing effect of gastric acid, and 
afterwards to contamination with microbiota present in the esophagus and pharynx. The 
Enterotest is not useful for the isolation of anaerobes because samples cannot be protected 
from contact with air. The clinical value of the string test compared with a sterile 
endoscopic method for sampling small bowel secretions is limited by poor sensitivity, 
specificity, and positive predictive value. Thus the string test is not an adequate substitute 
for oro-duodenal intubation for the detection of SBBO (60,71). 

Peroral Intubation 

Peroral intubation and aspiration of luminal contents can be achieved using Miller- Abbott 
or Levin tubes. These tubes were modified to suit the special needs for culture studies. The 
headpiece of a Miller- Abbott tube comprises a capsule, which may be opened and closed 
by hydraulic pressure. The capsule has an advantage of large size (44.5 X 12 mm), but it 
has been proven possible for bacteria to gain access into the closed capsule in vitro. A 
Levin tube is clinically used as a gastroduodenal feeding tube with a length of 
125 centimeters. A long radio-opaque tube is used, marked for accurate placement, either 
single- or double-lumened, with or without balloons, and perforated by one or more holes 
at its distal end. These perforations were either left free or were protected by means of a 
collodion membrane, a thin rubber sheath, or by plugs, which could be either dissolved or 
dispelled by positive pressure at the moment of taking samples for culture. Contamination 
of the tubes depends on the degree of contamination of the surrounding fluid, the exposure 
time, and the static environment. The small intestine contains only a very small quantity of 
fluid in contrast to gastric juice, which may be aspirated in large quantities. A disadvantage 
of peroral intubation is the lack of certainty that the specimen obtained from the desired 
level of the intestine has not been contaminated by bacteria from a higher position during 
its passage. 



Noninvasive Methods 

Because small intestinal intubation for quantitative culture is inconvenient, expensive, and 
not widely available, a variety of surrogate tests for bacterial overgrowth in the small 
intestine have been devised based on the metabolic actions of enteric bacteria rather than 
on increases in the number of bacteria. Several indirect methods have been developed to 
overcome the problem of location-dependence of aspirates for culturing. A comparison 
between the small intestinal noninvasive tests versus invasive methods with culture of 
material obtained for diagnosis of SBBO is presented in Table 3. Most of these indirect 
tests lack sensitivity for reliable detection of SBBO. The main reason for this is the great 
variability of the microbiota and its metabolic profile. The tests are based on a specific 
bacterial metabolic activity. Thus, if this particular activity is not present in the microbiota 
of a SBBO patient, the test will yield a false-negative result. For this reason urinary 
excretion tests (e.g., indican excretion, D-xylose, conjugated para-aminobenzoic acid), 
and analysis of intestinal aspirates for bacterial metabolic products (e.g., deconjugated bile 
acids in serum) lack the required reliability for detection of SBBO, and have become 
obsolete (71-75). These tests will not be described further. 



Sampling Microbiota in the Human Gastrointestinal Tract 



39 



Table 3 Small Intestinal Noninvasive Tests Compared to Jejunal Culture (Gold Standard) 



Test 



Sensitivity (%) Specificity (%) 



Simplicity 



14C-D-xylose BT 


42-100 


85-100 


Excellent 


Lactulose H 2 BT 


68 


44 


Excellent 


Glucose H 2 BT 


62-93 


78-83 


Excellent 


13C and 14C- glycocholate BT 


20-70 


76-90 





Abbreviations: BT, breath test; H2, hydrogen. 
Source: From Refs. 42, 51, 78, 80, 84, 101-105. 



To diagnose bacterial overgrowth, various breath tests may be used including the 
14 C-glycocholate, 14 C-D-xylose, lactulose-H 2 , and glucose-H 2 tests. The rationale for 
the breath test is the production of volatile metabolites i.e., carbon dioxide (C0 2 ), 
hydrogen (H 2 ) or methane (CH 4 ), by intraluminal bacteria from the administered 
substrates, which can be measured in the exhaled air. The most successful and popular 
methods analyze either expired isotope-labeled C0 2 after timed oral administration of 
14 C- or 13 C-enriched substrates, or breath hydrogen following feeding of a non-labeled 
fermentable carbohydrate substrate. 

The 14 C- and 13 C-breath tests measure the pulmonal excretion of labeled C0 2 
produced by the fermentation of labeled substrates, using either a radioactive or a 
stable isotope. The increasing availability of methods for analyzing stable isotopes 
has raised interest in replacing the radioactive 14 C by non-radioactive 13 C. The use of 
radioactive isotopes is not recommended for study of children or women who are or 
might be pregnant. C0 2 can be measured by mass spectrometry. Because of concerns 
about diagnostic accuracy, costs of the substrates and equipment, and limited availability, 
these tests have not gained widespread acceptance. 

The first breath test to diagnose SBBO was the hydrogen breath test described by 
Levitt in 1969 (76). Hydrogen is a constituent of human breath derived exclusively 
from bacterial fermentation reactions in the intestinal lumen. Detection of hydrogen in 
expired breath is considered a measure of the metabolic activity of the hydrogen- 
producing bacteria. Bacteria produce hydrogen from carbohydrate substrates, and human 
tissue does not generate hydrogen. The colon is considered to be the only place in the 
human body where hydrogen is produced, because of the high amount of hydrogen- 
producing bacteria. In cases of SBBO, hydrogen is also produced in the small intestine. 
Part of the produced hydrogen is reabsorbed from the intestine into the blood, and is 
exhaled. Measurement of breath hydrogen could circumvent the administration of a 
radioactive isotope in testing for bacterial overgrowth. This test assumes the presence of 
a hydrogen-producing microbiota, but in 15-20% of humans the microbiota of the subject 
does not meet this condition. Hydrogen breath analysis is therefore not sufficiently reliable 
as a diagnostic tool in SBBO. 



14 C-Glycocholate Breath Test 

l4 C-glycocholate breath test or bile acid test is based on the bile salt deconjugating 
capacity of bacteria in the proximal small bowel. Conjugated bile acids are excreted 
through the bile in the duodenum, and they are reabsorbed in the terminal ileum. 
Conjugated bile acids are in the enterohepatic circulation. Physiologically, less than 5% of 
the conjugated bile acids reach the colon. After excretion in the duodenum, bile acids 
stimulate micellization of dietary lipids. After oral administration of glycocholic bile 



40 Kerckhoffs et al. 

acid (a normal component of bile) this is normally reabsorbed in the terminal ileum. In 
cases of SBBO some bacteria split off glycine on the amide bond of cholylglycine. Glycine 
is absorbed, and fermented in the liver to C0 2 , H 2 0, and ammonia (NH 4 ); the C0 2 
produced is exhaled. When using 14 C glycocholate, the 14 C0 2 in the exhaled air can 
be measured. 

The sensitivity is too low (20-70%) to allow SBBO to be demonstrated without 
additional intestinal culturing. A rise in labeled C0 2 does not differentiate bile salt wastage 
from bacterial overgrowth. This is a disadvantage given that a significant number of SBBO 
patients may have had ileal resection. Ruling out bile salt malabsorption as an explanation 
for a positive breath test can be done with stool collection (42,77). 

The false negative rate for the 14 C-glycocholate breath test is 30-40%. There are 
three reasons for false negative outcomes. Firstly, one needs anaerobic organisms to 
deconjugate bile salts. Secondly, not all cases of bacterial overgrowth involve bile salt 
deconjugation. Lastly, the fatty meal (usually a polymeric supplement) given with the 
cholylglycine may, in theory, affect the ratio of labeled and unlabeled carbon dioxide 
absorbed, diluting the labeled carbon dioxide with that produced from the metabolism of 
the meal. False positive results are possible in case of ileal pathology, ileal resection, and 
increased intestinal transit. In those cases bile acids are deconjugated by the (anaerobic) 
colonic microbiota. The disadvantage of using radioactivity in 14 C-substrate breath tests 
can be overcome by using the stable C-isotope, which is measured by mass spectrometry 
in breath samples. However, the use of C-isotope does not improve the sensitivity. 



14 



C-D-xylose Breath Test 

The 14 C-D-xylose breath test was considered to be the only breath test for the detection of 
bacterial overgrowth with high sensitivity (95-100%) and 100% specificity, but these 
promising results have not been sustained (42). Compared with cultures of the duodenal 
aspirates, the sensitivity and specificity are 60% and 40%, respectively (78). 

This test is based on the assumption that the overgrown aerobic Gram-negative 
microbiota ferment D-xylose. The 14 C0 2 produced, and unmetabolized xylose are 
absorbed by the proximal small bowel, which thus avoids confusion of results caused by 
metabolism of substrate by colonic bacteria. Subjects must fast at least 8 hours before the 
test, and no smoking or exercise is permitted for 12 hours before the breath test. Following 
a 1 g oral dose of 14 C-D-xylose in water, elevated 14 C0 2 levels are detected in the breath 
within 60 minutes in 85% of patients with SBBO. 

False negative rates for the 14 C-D-xylose breath test are 35-78%. False negative 
results cannot be entirely attributed to the absence of D-xylose fermentation of the 
microbiota (overgrown bacteria in 81.8% of SBBO patients are capable of D-xylose 
fermentation); body weight is correlated to endogenous C0 2 production, and should 
therefore also be taken into account (79). Disturbed gastric emptying and small intestinal 
motility can also contribute to a false-negative result of the 14 C-D-xylose breath test 
because of delayed delivery of the labeled substrate to the metabolizing microbiota. 
Refinement of the 14 C-D-xylose breath test to include a transit marker for intestinal 
motility increases its specificity. With the transit marker one can determine whether the 
site of metabolism is in the small intestine or the colon (80). 



Lactulose Hydrogen Breath Test 

Lactulose is an easily fermented disaccharide, and is used for the detection of bacterial 
overgrowth, and for determination of the orocecal transit time. The lactulose hydrogen 



Sampling Microbiota in the Human Gastrointestinal Tract 41 

breath test is a simple, inexpensive, and noninvasive technique to diagnose SBBO. The 
lactulose breath test is performed after 12 hours fasting previous to the test. Hydrogen 
breath samples are taken at baseline, and subsequently every 10-30 minutes after the test 
meal that contains 10-12 g of lactulose. The hydrogen breath samples are analyzed gas 
chromatographically (81). Baseline samples average 7.1+5 parts per million (ppm) of H 2 
and 0-7 ppm for CH 4 (82). Values of the baseline sample over 20 ppm H 2 are suspect for 
bacterial overgrowth. Values between 10 and 20 suggest incomplete fasting before the test 
or ingestion of slowly digested foods the day before the test, the colon being the source of 
the elevated levels (82). Slowly digested foods like beans, bread, pasta, and fiber must not 
be consumed the night before the test because these foods produce prolonged hydrogen 
excretion (82). The patient is not allowed to eat during the complete test. Antibiotics and 
laxatives must be avoided for weeks prior to breath hydrogen testing. Cigarette smoking, 
sleeping, and exercise must be avoided at least a half hour before and during the test 
because these may induce hyperventilation (42). Chlorhexidine mouthwash must be used 
before the test to eliminate oral bacteria, which might otherwise contribute to an early 
hydrogen peak after the substrate is given. Lactulose, which reaches the colon, shows 
peaks usually more than 20 ppm above baseline after 2-3 hours of testing. Lactulose is not 
absorbed in the small intestine so every patient should have a colonic peak, assuming the 
colonic microbiota has not been altered. Peaks associated with SBBO occur within 1 hour, 
and are less prominent. Some laboratories measure H 2 and CH 4 simultaneously whereas 
others test CH 4 selectively after flat lactulose tests (42). Figure 3 shows lactulose breath 
test results in a patient with small bowel bacterial overgrowth. 

The lactulose hydrogen breath test is positive for small intestinal bacterial overgrowth 
if there is an increase in breath hydrogen of > 10 parts per million above basal that occurs at 
least 15 minutes before the cecal peak. Strict interpretative criteria, such as requiring two 
consecutive breath hydrogen values more than 10 ppm above the baseline reading, and 
recording a clear distinction of the small bowel peak from the subsequent colonic peak 
(double peak criterion), are recommended. Application of the double peak criterion alone 
for interpretation of the lactulose hydrogen breath test is inadequately sensitive, even with 
scintigraphy, to diagnose bacterial overgrowth. Twenty-seven percent of normal subjects 
have no peak due to organic acid reduction or dilution from voluminous diarrhea (42). 

The disadvantage of this test is that it is not always easy to distinguish breath 
hydrogen arising from small bowel colonization from that resulting from cecal 
fermentation in patients with an exceptionally rapid orocecal transit time. A comparison 
with the jejunal culture sensitivity of 68% and specificity of 44% has been described (51). 
A sensitivity of 16% for SBBO has been described (83). 

Despite the attractive aspects of ease of performance and avoidance of a radioactive 
tracer, breath hydrogen tests are not sufficiently sensitive or specific to justify their 
substitution for the 14 C-D-xylose breath test for noninvasive detection of intestinal 
bacterial overgrowth. 

Glucose Hydrogen Breath Test 

Glucose hydrogen breath tests can also be used to detect SBBO. Glucose is completely 
absorbed before reaching the colon even in patients with previous gastric surgery, who 
have faster than normal transit. Patients receive a solution containing 50-80 g of glucose 
dissolved in 250 ml water after fasting for 12 hours. Breath hydrogen concentrations are 
analyzed with an H 2 monitor after direct expiration through a Y-piece that prevents air 
from mixing with the exhaled hydrogen (84). Hydrogen concentration is determined 
every 10-15 minutes for two hours. Results of the hydrogen breath test are considered 



42 



Kerckhoffs et al. 



Lactulose breath test 



E 

Q. 
Q. 

C/) 

as 
CD 



200 -i 

180- 

160- 

140- 

120- 

100- 

80- 

60- 

40- 

20- 

0- 

-20- 




-♦ — Hydrogen (H 2 ) 
«— Methane (CH 4 ) 



o 



9? tf> 



# # x# JV° >° 



V 



\" NT 



Time (minutes) 



Figure 3 Production of hydrogen (H 2 ) and methane (CH 4 ) in a patient with bacterial overgrowth 
of the small bowel (SB BO). Fasting H 2 and CH 4 production at- 10 and minutes; 10 grams of 
lactulose was administered at minutes. 



positive when the hydrogen concentration increases by 14-20 ppm (85). Smoking and 
exercise are not allowed during the test, and the day previous to the test (86). The 
hydrogen breath test shows stable intra-individual results in healthy people. However, in 
patients with high values there is a large day-to-day variation (87). The coefficient of 
variation is 5-10% (84,88). Sensitivity of 93% and specificity of 78% have been 
described (85). The glucose hydrogen breath test has a sensitivity of 62% and a 
specificity of 83% compared with jejunal culture (51). Poor sensitivity due to rapid 
absorption of glucose substrate in the proximal small bowel, which inhibits hydrogen 
generation, can be explained by a washout effect of concomitant diarrhea, loss of 
bacterial microbiota because of recent antibiotic therapy, or an acidic bowel lumen. 



LARGE INTESTINE: MICROBIOTA AND SAMPLING TECHNIQUES 
Normal Microbiota 

The large intestine including the cecum, colon, and the rectum harbors over 500 species of 

11 10 

bacteria, mainly obligate anaerobes (99.9%) with 10 -10 CFU/g (2,10). Microorganisms 
isolated from large intestine and fecal samples are listed in Table 4. Bacteroides, Bifido- 
bacteria, Eubacteria, Clostridia, and Enterobacteriaceae can predominantly be found in 
the colon. Novel molecular methods are aiding better understanding of the microbiota, 
which is challenging to culture due to the anaerobic nature of most of the microbiota, and 
insufficient knowledge of the culturing conditions (90,91). Knowledge about the mucosa- 
associated bacterial communities in different parts of the colon is limited as most attention 
has been focused on bacteria present in feces. Enormous microbial populations can develop 
in the lumen of the large bowel, and especially in that of the cecum because these areas have 
a relative stagnation in the flowing stream (up to 60 hours) and very low oxidation- 
reduction potentials. The transit time of the lumenal content exceeds the doubling times of 
bacteria. Whether the microbiota is transient or truly autochthonous to habitats in the region 
remains a main concern. Bacteria in food are known to pass into human feces at high 
population levels. Bacteria from habitats above the large bowel pass down into the lumen of 
that region. The population levels of transients probably do not contribute significantly to 



Sampling Microbiota in the Human Gastrointestinal Tract 



43 



Table 4 Microbiota Isolated from the Large Intestine and Feces by Culturing 



Microbial types in large intestine 



Microbial types in feces 



Lactobacilli 

Streptococci 

Bifidobacteria 

Clostridia 

Propionibacterium 

Eubacterium 

Bacteroides 

Fusobacterium 

Veillonella 

Staphylococcus 

Coliforms 

Bacillus sp 

Yeasts 

Spiral shaped microbes 

Actinobacillus 

Enterobacteriaceae 

Enterococci 



Lactobacilli 

Streptococci 

Bifidobacteria 

Clostridia 

Propionibacterium 

Eubacterium 

Bacteroides 

Fusobacterium 

Veillonella 

Staphylococcus 

Coliforms 

Bacillus sp 

Yeasts 

Spiral shaped bacteria 

Peptococcus 

Ruminococcus 

Coprococcus 

Acidaminococcus, Succinivibrio, Butyrivibrio, 

Megasphaera, Gemminger 
Catenabacterium 
Peptostreptococcus 



Note: The most prevalent bacterial types are italicized. 
Source: From Refs. 2, 10, 15, 22, 89. 



the level in the region. Bacteria in the colon are important in processing maldigested 
carbohydrates (92). 



Disease-Causing Microbiota 

Yersinia enterocolitica, Salmonella, Shigella, Campylobacter, Clostridium difficile, 
enterohemorragic Escherichia coli (EHEC), and enteropathogenic Escherichia coli 
(EPEC) are the most common pathogenic bacteria in the colon that cause diarrhea. 
Diarrhea can also occur after oral antibiotic treatment. Poorly absorbed antibiotics change 
the normal composition of the microbiota in the colon (93). Suppression of the normal 
microbiota may lead to reduced colonization resistance with subsequent overgrowth of 
resistant microbiota, yeasts, and Clostridium difficile. This organism produces a protein 
toxin which causes necrosis and ulceration of the colonic mucosa, called antibiotic- 
associated hemorrhagic colitis. 



Biopsy 

A standard colonoscope has a length of 1 .30 to 1 .60 m, so that the colon and the distal ileum 
can be evaluated. Long colonoscopes (165-180 cm) are able to reach the cecum even in 
overly long and tortuous colons (27). Biopsy specimens can be collected with a flexible 
colonoscope and flexible biopsy forceps. Patients are given a laxative solution to drink the 
day before the examination. The object of full preparation is to cleanse the entire colon of 
fecal material, especially the proximal parts, to allow a clear view (27). So it is very likely 



44 Kerckhoffs et al. 

that the bacteria in the biopsy sample are mucosa associated as the luminal bacteria will have 
been washed away (94). Typically biopsy samples contain 10 5 -10 6 bacteria, and the 
predominant mucosa-associated bacterial community is host specific and uniformly 
distributed along the colon but differs significantly from the fecal community (95). Biopsy 
samples are very small in size, and therefore more easily exposed to oxygen during 
sampling; therefore, the number of viable strict anaerobes might be reduced easily. 
Relatively high levels of facultative anaerobes are reported to be present in intestinal biopsy 
samples. To minimize contamination during sampling, the colonoscope jaws will have to be 
washed in tap water after each biopsy is performed. 

Pyxigraphy 

Pyxigraphy is a technique which makes use of a capsule that can be swallowed, and by 
which contents of the gastrointestinal tract can be sampled under remote control. 
Pyxigraphy is a simple and safe sampling method that allows the microbial population of 
the proximal colon to be studied (96). 



Fecal Samples 

Feces are a complex microbial habitat, with many niches occupied by bacteria. It is 
estimated that bacteria account for about 30% of the fecal mass, and 40-55% of fecal 
solids. All of the bacteria in feces are exposed to the influences of dehydrating and 
concentrating mechanisms of the colon and rectum, and intense biochemical activity of the 
organisms living in the material. When the samples consist of only feces, the composition 
and localization of communities anywhere in the tract cannot be revealed. Bacteroides 
accounts for nearly 20% of the species that can be cultivated from feces (10). The 
Bacteroides and Prevotella group (gram-negative anaerobes), and Eubacterium rectale 
and Clostridium coccoides species (gram-positive anaerobes) are predominantly present in 
the fecal samples (90,92). The predominant bacterial community in feces is stable in time, 
host specific, affected by ageing, and not significantly altered after consumption of 
probiotic strains (97). 

Fecal samples have to be collected in sterile bags, and kept at low temperature 
( — 80°C to +4°C) before processing (88). Stool specimens or rectal swabs can be used 
for the diagnosis of cholera. Dipsticks in rectal swabs are used for the rapid diagnosis 
of cholera caused by Vibrio cholerae. Dipstick analysis uses colloidal gold particles, 
and is based on a one-step immunochomatography principle. The sensitivity and the 
specificity of the dipsticks is greater than 92% and 91% respectively. This rapid test 
(diagnosis within 10 minutes) requires minimal technical skills (98,99). 

Most knowledge of the gastrointestinal microbiota stems from colon or feces 
bacteriology. A major limitation in studying the proximal human colonic microbiota is the 
lack of suitable sampling methods. Studies in which only feces are sampled can never 
reveal the composition and localization of epithelial and cryptal communities anywhere in 
the tract. Such studies reveal little about the composition of lumenal communities in any 
area except perhaps the large bowel (29). 

Low fecal pH is caused by ingestion of poorly absorbed carbohydrates or 
carbohydrate malabsorption in the small intestine, and consequently, the bacteria in the 
colon ferment the carbohydrate. Fecal pH of less than six is highly suggestive of 
carbohydrate malabsorption. A breath hydrogen test with lactose can confirm 
carbohydrate malabsorption. In this test a fasting patient is given 25 g of lactose dissolved 
in water, and exhaled breath is assayed for hydrogen content at baseline, and at intervals 



Sampling Microbiota in the Human Gastrointestinal Tract 



45 




Lactase deficiency 



Figure 4 Principle of the hydrogen breath test with lactose to determine carbohydrate 
malabsorption in the small intestine. 



for several hours as described in Figure 4. As explained above, because hydrogen is not a 
normal product of human metabolism, any increase in breath hydrogen concentration 
represents bacterial fermentation, and indicates that unabsorbed lactose has reached 
the colon. 



CONCLUSION 



The different methods of investigating the intestinal microbiota in humans all have their 
advantages, and their drawbacks as described above. If one desires information about the 
gastrointestinal tract one should also weigh the benefits of the (research) question, and 
their financial consequences. Sampling of the gastrointestinal tract in humans is far more 
difficult than in animal models. The sampled area is relatively small in comparison with 
the total area. In animal models the animal can be sacrificed so that the complete intestinal 
tract can be sampled and investigated. Unfortunately, individuals who are killed in 
accidents are the best source of complete information about microbiota in the 
gastrointestinal tract (29). 

In general, the patient prefers the noninvasive method. Noninvasive methods are of 
particular importance for very young pediatric patients, pregnant women, and the elderly, 
as well as for research purposes. The difficulties of sampling the entire gastrointestinal 
tract are reduced by the noninvasive tests. However, noninvasive methods are often less 
sensitive and less specific. Invasive methods, such as endoscopy, are extremely unpleasant 
but are highly sensitive and specific, and have the advantage of sampling at the accurate 
location. The conditions that have to be satisfied in obtaining an uncontaminated specimen 



46 Kerckhoffs et al. 

from anywhere in the gastrointestinal tract have to include: (1) strict asepsis of method, 
which necessitates that the instrument used must be suitable for sterilization by heat or gas; 
(2) prevention of contamination of the internal channels in which the culture specimen is 
to be lodged, until the site of sampling is reached, and protection against further 
contamination on withdrawal of the instrument; and (3) verification of the location from 
which cultures have been obtained. 

As the development of molecular biology techniques increases the current sampling 
techniques can be revised. The condition of anaerobic sampling is becoming less important. 
Possible improvement of the current sampling methods only seems possible in small details. 
Nanotechnology is one of the promising techniques for possible improvement of sampling 
and analysis of bacteria in the human gastrointestinal tract. 



REFERENCES 

1. Leeuwenhoek AV, Dobell C. Antony van Leeuwenhoek and His "Little Animals": Being 
Some Account of the Father of Protozoology and Bacteriology and His Multifarious 
Discoveries in these Disciplines. London: Staples Press, 1932. 

2. Simon GL, Gorbach SL. The human intestinal microbiota. Dig Dis Sci 1986; 3L147S-162S. 

3. Feldman M, Friedman LS, Sleisenger MH, Fortran JS. Sleisenger & Fortran's Gastrointestinal 
and Liver Disease: Pathophysiology, Diagnosis, Management. 7th ed. Philadelphia: W.B. 
Saunders, 2002. 

4. Galatola G, Grosso M, Barlotta A, et al. Diagnosis of bacterial contamination of the small 
intestine using the 1 g [14C] xylose breath test in various gastrointestinal diseases. Minerva 
Gastroenterol Dietol 1991; 37:169-175. 

5. Mannell A, Plant M, Frolich J. The microbiota of the oesophagus. Ann R Coll Surg Engl 1983; 
65:152-154. 

6. Drasar BS, Shiner M, McLeod GM. Studies on the intestinal flora. I. The bacterial flora of the 
gastrointestinal tract in healthy and achlorhydric persons. Gastroenterology 1969; 56:71-79. 

7. King TS, Elia M, Hunter JO. Abnormal colonic fermentation in irritable bowel syndrome. 
Lancet 1998; 352:1187-1189. 

8. Mallory A, Savage D, Kern F, Jr., Smith JG. Patterns of bile acids and microbiota in the 
human small intestine. II. Microflora. Gastroenterology 1973; 64:34-42. 

9. Cunningham-Rundles S, Ahrn S, Abuav-Nussbaum R, Dnistrian A. Development of 
immunocompetence: role of micronutrients and microorganisms. Nutr Rev 2002; 
60:S68-S72. 

10. Carman RJ, Van Tassell RL, Wilkins TD. The normal intestinal microbiota: ecology, 
variability and stability. Vet Hum Toxicol 1993; 35:11-14. 

11. Blaut M, Collins MD, Welling GW, Dore J, van Loo J, de Vos W. Molecular biological 
methods for studying the gut microbiota: the EU human gut flora project. Br J Nutr 2002; 
87:S203-S211. 

12. Blaut M. Relationship of prebiotics and food to intestinal microbiota. Eur J Nutr 2002; 
41:111-116. 

13. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science 2001; 
292:1115-1118. 

14. Gordon JI, Stappenbeck TS, Hooper LV, et al. Response from Jeffrey I. Gordon. Commensal 
bacteria make a difference. Trends Microbiol 2003; 11:150-151. 

15. Zinsser H. Zinsser Microbiology. 20th ed. Norwalk, CT: Appleton & Lange, 1992. 

16. Hopkins MJ, Macfarlane GT. Changes in predominant bacterial populations in human feces 
with age and with Clostridium difficile infection. J Med Microbiol 2002; 51:448-454. 

17. Husebye E, Hellstrom PM, Sundler F, Chen J, Midtvedt T. Influence of microbial species on 
small intestinal myoelectric activity and transit in germ-free rats. Am J Physiol Gastrointest 
Liver Physiol 2001; 280:G368-G380. 



Sampling Microbiota in the Human Gastrointestinal Tract 47 

18. Husebye E, Hellstrom PM, Midtvedt T. Intestinal microbiota stimulates myoelectric activity 
of rat small intestine by promoting cyclic initiation and aboral propagation of migrating 
myoelectric complex. Dig Dis Sci 1994; 39:946-956. 

19. Nieuwenhuijs VB, Verheem A, Duijvenbode-Beumer H, et al. The role of interdigestive small 
bowel motility in the regulation of gut microbiota, bacterial overgrowth, and bacterial 
translocation in rats. Ann Surg 1998; 228:188-193. 

20. Williams C. Occurrence and significance of gastric colonization during acid-inhibitory 
therapy. Best Pract Res Clin Gastroenterol 2001; 15:511-521. 

21. Pei Z, Bini EJ, Yang L, Zhou M, Francois F, Blaser MJ. Bacterial biota in the human distal 
esophagus. Proc Natl Acad Sci USA 2004; 101:4250-4255. 

22. Sjostedt S. The upper gastrointestinal microbiota in relation to gastric diseases and gastric 
surgery. Acta Chir Scand Suppl 1989; 551:1-57. 

23. Jain SK, Jain S, Jain M, Yaduvanshi A. Esophageal tuberculosis: is it so rare? Report of 12 
cases and review of the literature Am J Gastroenterol 2002; 97:287-291. 

24. Gagliardi D, Makihara S, Corsi PR, et al. Microbial flora of the normal esophagus. Dis 
Esophagus 1998; 11:248-250. 

25. Pajecki D, Zilberstein B, dos Santos MA, et al. Megaesophagus microbiota: a qualitative and 
quantitative analysis. J Gastrointest Surg 2002; 6:723-729. 

26. Franklin MA, Skoryna SC. Studies of the natural gastric flora: I Bacterial flora of fasting 
human subjects. Can Med Assoc J 1966; 95:1349-1355. 

27. Cotton Peter B, Williams Christopher B. 4th ed. Practical Gastrointestinal Endoscopy. 
London: Blackwell Scientific Publications, 1996. 

28. Osias GL, Bromer MQ, Thomas RM, et al. Esophageal bacteria and Barrett's esophagus: 
a preliminary report. Dig Dis Sci 2004; 49:228-236. 

29. Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 1977; 
31:107-133. 

30. Simon GL, Gorbach SL. Intestinal flora in health and disease. Gastroenterology 1984; 
86:174-193. 

31. Franklin MA, Skoryna SC. Studies on natural gastric flora: survival of bacteria in fasting 
human subjects. Can Med Assoc J 1971; 105:380-386. 

32. Monstein HJ, Tiveljung A, Kraft CH, Borch K, Jonasson J. Profiling of bacterial flora in 
gastric biopsies from patients with Helicobacter py/on-associated gastritis and histologically 
normal control individuals by temperature gradient gel electrophoresis and 16S rDNA 
sequence analysis. J Med Microbiol 2000; 49:817-822. 

33. Verhagen MA, Roelofs JM, Edelbroek MA, Smout AJ, Akkermans LM. The effect of 
cisapride on duodenal acid exposure in the proximal duodenum in healthy subjects. Aliment 
Pharmacol Ther 1999; 13:621-630. 

34. Pasricha PJ. Effect of sleep on gastroesophageal physiology and airway protective 
mechanisms. Am J Med 2003; 1 15:1 14S-1 18S. 

35. Pounder RE, Ng D. The prevalence of Helicobacter pylori infection in different countries. 
Aliment Pharmacol Ther 1995; 9:33-39. 

36. Graham DY, Adam E, Reddy GT, et al. Seroepidemiology of Helicobacter pylori infection in 
India. Comparison of developing and developed countries. Dig Dis Sci 1991; 36:1084-1088. 

37. Guerre J, Vedel G, Gaudric M, Paul G, Cornuau J. Bacterial flora in gastric juice taken at 
endoscopy in 93 normal subjects. Pathol Biol (Paris) 1986; 34:57-60. 

38. Wong BC, Wong WM, Wang WH, et al. An evaluation of invasive and non-invasive tests for 
the diagnosis of Helicobacter pylori infection in Chinese. Aliment Pharmacol Ther 2001; 
15:505-511. 

39. Cotton, Williams. Practical gastrointestinal endoscopy. 4th ed. London: Blackwell Scientific 
Publications, 1996. 

40. Lim LL, Ho KY, Ho B, Salto-Tellez M. Effect of biopsies on sensitivity and specificity of 
ultra-rapid urease test for detection of Helicobacter pylori infection: A prospective 
evaluation. World J Gastroenterol 2004; 10:1907-1910. 



48 Kerckhoffs et al. 

41. Monteiro L, de Mascarel A, Sarrasqueta AM, et al. Diagnosis of Helicobacter pylori 
infection: noninvasive methods compared to invasive methods and evaluation of two new 
tests. Am J Gastroenterol 2001; 96:353-358. 

42. Romagnuolo J, Schiller D, Bailey RJ. Using breath tests wisely in a gastroenterology practice: 
an evidence-based review of indications and pitfalls in interpretation. Am J Gastroenterol 
2002;97:1113-1126. 

43. Koletzko S, Feydt-Schmidt A. Infants differ from teenagers: use of non-invasive tests for 
detection of Helicobacter pylori infection in children. Eur J Gastroenterol Hepatol 2001; 
13:1047-1052. 

44. Makristathis A, Pasching E, Schutze K, Wimmer M, Rotter ML, Hirschl AM. Detection of 
Helicobacter pylori in stool specimens by PCR and antigen enzyme immunoassay. J Clin 
Microbiol 1998; 36:2772-2774. 

45. MacKay WG, Williams CL, McMillan M, Ndip RN, Shepherd AJ, Weaver LT. Evaluation of 
protocol using gene capture and PCR for detection of Helicobacter pylori DNA in feces. 
J Clin Microbiol 2003; 41:4589-4593. 

46. Basset C, Holton J, Ricci C, et al. Review article: diagnosis and treatment of Helicobacter: a 
2002 updated review. Aliment Pharmacol Ther 2003; 17:89-97. 

47. Vakil N, Rhew D, Soil A, Ofman JJ. The cost-effectiveness of diagnostic testing strategies for 
Helicobacter pylori. Am J Gastroenterol 2000; 95:1691-1698. 

48. Maibach RC, Dutly F, Altwegg M. Detection of Tropheryma whipplei DNA in feces by PCR 
using a target capture method. J Clin Microbiol 2002; 40:2466-2471. 

49. Edmiston CE, Jr., Krepel CJ, Seabrook GR, Jochimsen WG. Anaerobic infections in the 
surgical patient: microbial etiology and therapy. Clin Infect Dis 2002; 35:S1 12— SI 18. 

50. Finegold SM. Host factors predisposing to anaerobic infections. FEMS Immunol Med 
Microbiol 1993; 6:159-163. 

51. Corazza GR, Menozzi MG, Strocchi A, et al. The diagnosis of small bowel bacterial 
overgrowth. Reliability of jejunal culture and inadequacy of breath hydrogen testing. 
Gastroenterology 1990; 98:302-309. 

52. Mee AS, Burke M, Vallon AG, Newman J, Cotton PB. Small bowel biopsy for malabsorption: 
comparison of the diagnostic adequacy of endoscopic forceps and capsule biopsy specimens. 
Br Med J (Clin Res Ed) 1985; 291:769-772. 

53. Crosby WH, Kugler HW. Intraluminal biopsy of the small intestine; the intestinal biopsy 
capsule. Am J Dig Dis 1957; 2:236-241. 

54. Flick AL, Quionton WE, Rubin CE. A peroral hydraulic biopsy tube for multiple sampling at 
any level of the gastro-intestinal tract. Gastroenterology 1961; 40:120-126. 

55. Chu KM, Yuen ST, Wong WM, et al. A prospective comparison of performance of biopsy 
forceps used in single passage with multiple bites during upper endoscopy. Endoscopy 2003; 
35:338-342. 

56. Tornblom H, Lindberg G, Nyberg B, Veress B. Full-thickness biopsy of the jejunum reveals 
inflammation and enteric neuropathy in irritable bowel syndrome. Gastroenterology 2002; 
123:1972-1979. 

57. Davis CP. Postmortem alterations of bacterial localization. Scan Electron Microsc 1980; 523- 
6:542. 

58. Bengmark S, Jeppsson B. Gastrointestinal surface protection and mucosa reconditioning. 
JPEN J Parenter Enteral Nutr 1995; 19:410-415. 

59. Tiveljung A, Soderholm JD, Olaison G, Jonasson J, Monstein HJ. Presence of eubacteria in 
biopsies from Crohn's disease inflammatory lesions as determined by 16S rRNA gene-based 
PCR. J Med Microbiol 1999; 48:263-268. 

60. Leon-Barua R, Gilman RH, Rodriguez C, et al. Comparison of three methods to obtain upper 
small bowel contents for culture. Am J Gastroenterol 1993; 88:925-928. 

61. Shiner M, Waters TE, Gray JD. Culture studies of the gastrointestinal tract with a newly 
devised capsule. Results of tests in vitro and in vivo. Gastroenterology 1963; 45:625-632. 

62. Tally FP, Stewart PR, Sutter VL, Rosenblatt JE. Oxygen tolerance of fresh clinical anaerobic 
bacteria. J Clin Microbiol 1975; 1:161-164. 



Sampling Microbiota in the Human Gastrointestinal Tract 49 

63. Nakshabendi IM, McKee R, Downie S, Russell RI, Rennie MJ. Rates of small intestinal 
mucosal protein synthesis in human jejunum and ileum. Am J Physiol 1999; 
277:E1028-E1031. 

64. Sheehy TW. Intestinal biopsy. Lancet 1964; 41:959-962. 

65. Mayhew JW, Gorbach SL. Internal standards for gas chromatographic analysis of metabolic 
end products from anaerobic bacteria. Appl Environ Microbiol 1977; 33:1002-1003. 

66. Gorbach SL, Mayhew JW, Bartlett JG, Thadepalli H, Onderdonk AB. Rapid diagnosis of 
anaerobic infections by direct gas-liquid chromatography of clinical speciments. J Clin Invest 
1976; 57:478-484. 

67. Bardhan PK, Gyr K, Beglinger C, Vogtlin J, Frey R, Vischer W. Diagnosis of bacterial 
overgrowth after culturing proximal small-bowel aspirate obtained during routine upper 
gastrointestinal endoscopy. Scand J Gastroenterol 1992; 27:253-256. 

68. Tabaqchali S. The pathophysiological role of small intestinal bacterial flora. Scand 
J Gastroenterol Suppl 1970; 6:139-163. 

69. Norby RS, Haagen NO, Justesen T, Jacobsen IE, Lave J, Magid E. Comparison of an open and 
a closed tube system for collection of jejunal juice. Scand J Gastroenterol 1983; 18:353-357. 

70. Muraca M, Vilei MT, Miconi L, Petrin P, Antoniutti M, Pedrazzoli S. A simple method for the 
determination of lipid composition of human bile. J Lipid Res 1991; 32:371-374. 

71. Riordan SM, Mclver CJ, Duncombe VM, Bolin TD. An appraisal of a 'string test' for the 
detection of small bowel bacterial overgrowth. J Trop Med Hyg 1995; 98:117-120. 

72. Bardhan PK, Feger A, Kogon M, et al. Urinary choloyl-PABA excretion in diagnosing small 

intestinal bacterial overgrowth: evaluation of a new noninvasive method. Dig Dis Sci 2000; 
45:474-479. 

73. Mayer PJ, Beeken WL. The role of urinary indican as a predictor of bacterial colonization in 
the human jejunum. Am J Dig Dis 1975; 20:1003-1009. 

74. Krawitt EL, Beeken WL. Limitations of the usefulness of the D-xylose absorption test. Am 
J Clin Pathol 1975; 63:261-263. 

75. Einarsson K, Bergstrom M, Eklof R, Nord CE, Bjorkhem I. Comparison of the proportion of 
unconjugated to total serum cholic acid and the [ 14 C] -xylose breath test in patients with 
suspected small intestinal bacterial overgrowth. Scand J Clin Lab Invest 1992; 52:425-430. 

76. Levitt MD. Production and excretion of hydrogen gas in man. N Engl J Med 1969; 
281:122-127. 

77. Hepner GW. Increased sensitivity of the cholylglycine breath test for detecting ileal 
dysfunction. Gastroenterology 1975; 68:8-16. 

78. Valdovinos MA, Camilleri M, Thomforde GM, Frie C. Reduced accuracy of 14 C-D-xylose 
breath test for detecting bacterial overgrowth in gastrointestinal motility disorders. Scand 
J Gastroenterol 1993; 28:963-968. 

79. Riordan SM, Mclver CJ, Duncombe VM, Bolin TD, Thomas MC. Factors influencing the 1-g 
14 C-D-xylose breath test for bacterial overgrowth. Am J Gastroenterol 1995; 90:1455-1460. 

80. Lewis SJ, Young G, Mann M, Franco S, O'Keefe SJ. Improvement in specificity of [ 14 C]D- 
xylose breath test for bacterial overgrowth. Dig Dis Sci 1997; 42:1587-1592. 

81. Metz G, Gassull MA, Drasar BS, Jenkins DJ, Blendis LM. Breath-hydrogen test for small- 
intestinal bacterial colonisation. Lancet 1976; 1:668-669. 

82. Perman J A, Modler S, Barr RG, Rosenthal P. Fasting breath hydrogen concentration: normal 
values and clinical application. Gastroenterology 1984; 87:1358-1363. 

83. Riordan SM, Mclver CJ, Walker BM, Duncombe VM, Bolin TD, Thomas MC. The lactulose 
breath hydrogen test and small intestinal bacterial overgrowth. Am J Gastroenterol 1996; 
91:1795-1803. 

84. Brummer RJ, Armbrecht U, Bosaeus I, Dotevall G, Stockbruegger RW. The hydrogen (H 2 ) 
breath test. Sampling methods and the influence of dietary fibre on fasting level. Scand 
J Gastroenterol 1985; 20:1007-1013. 

85. Kerlin P, Wong L. Breath hydrogen testing in bacterial overgrowth of the small intestine. 
Gastroenterology 1988; 95:982-988. 



50 Kerckhoffs et al. 

86. Thompson DG, O'Brien JD, Hardie JM. Influence of the oropharyngeal microbiota on the 
measurement of exhaled breath hydrogen. Gastroenterology 1986; 91:853-860. 

87. Riordan SM, Mclver CJ, Bolin TD, Duncombe VM. Fasting breath hydrogen concentrations 
in gastric and small-intestinal bacterial overgrowth. Scand J Gastroenterol 1995; 30:252-257. 

88. Rumessen JJ, Kokholm G, Gudmand-Hoyer E. Methodological aspects of breath hydrogen 
(H 2 ) analysis. Evaluation of a H 2 monitor and interpretation of the breath H 2 test. Scand J Clin 
Lab Invest 1987; 47:555-560. 

89. Moore WE, Holdeman LV. Special problems associated with the isolation and identification 
of intestinal bacteria in fecal flora studies. Am J Clin Nutr 1974; 27:1450-1455. 

90. Mai V, Morris JG, Jr. Colonic bacterial flora: changing understandings in the molecular age. 
J Nutr 2004; 134:459^164. 

91. Tlaskalova-Hogenova H, Stepankova R, Hudcovic T, et al. Commensal bacteria (normal 
microbiota), mucosal immunity and chronic inflammatory and autoimmune diseases. 
Immunol Lett 2004; 93:97-108. 

92. Zhong Y, Priebe MG, Vonk RJ, et al. The role of colonic microbiota in lactose intolerance. 
Dig Dis Sci 2004; 49:78-83. 

93. Edlund C, Nord CE. Effect on the human normal microbiota of oral antibiotics for treatment 
of urinary tract infections. J Antimicrob Chemother 2000; 46:41-48 discussion 63-65: 41-48. 

94. Huijsdens XW, Linskens RK, Mak M, Meuwissen SG, Vandenbroucke-Grauls CM, 
Savelkoul PH. Quantification of bacteria adherent to gastrointestinal mucosa by real-time 
PCR. J Clin Microbiol 2002; 40:4423-4427. 

95. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben Amor K, Akkermans AD, de 
Vos WM. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly 
distributed along the colon and differ from the community recovered from feces. Appl 
Environ Microbiol 2002; 68:3401-3407. 

96. Pochart P, Lemann F, Flourie B, Pellier P, Goderel I, Rambaud JC. Pyxigraphic sampling to 
enumerate methanogens and anaerobes in the right colon of healthy humans. Gastroenter- 
ology 1993; 105:1281-1285. 

97. Zoetendal EG, Cheng B, Koike S, Mackie RI. Molecular microbial ecology of the 
gastrointestinal tract: from phylogeny to function. Curr Issues Intest Microbiol 2004; 5:31-47. 

98. Bhuiyan NA, Qadri F, Faruque AS, et al. Use of dipsticks for rapid diagnosis of cholera 
caused by Vibrio cholerae 01 and 0139 from rectal swabs. J Clin Microbiol 2003; 
41:3939-3941. 

99. Nato F, Boutonnier A, Rajerison M, et al. One-step immunochromatographic dipstick tests for 
rapid detection of Vibrio cholerae 01 and 0139 in stool samples. Clin Diagn Lab Immunol 
2003; 10:476^78. 

100. Sayler GS, Simpson ML, Cox CD. Emerging foundations: nano-engineering and bio- 
microelectronics for environmental biotechnology. Curr Opin Microbiol 2004; 7:267-273. 

101. Stotzer PO, Kilander AF. Comparison of the 1-gram ( 14 C)-D-xylose breath test and the 50- 
gram hydrogen glucose breath test for diagnosis of small intestinal bacterial overgrowth. 
Digestion 2000; 61:165-171. 

102. King CE, Toskes PP, Guilarte TR, Lorenz E, Welkos SL. Comparison of the one-gram d- 
[ 14 C]xylose breath test to the [ 14 C]bile acid breath test in patients with small-intestine 
bacterial overgrowth. Dig Dis Sci 1980; 25:53-58. 

103. Sherr HP, Sasaki Y, Newman A, Banwell JG, Wagner HN, Jr., Hendrix TR. Detection of 
bacterial deconjugation of bile salts by a convenient breath-analysis technic. N Engl J Med 
1971; 285:656-661. 

104. Metz G, Drasar BS, Gassull MA, Jenkins DJA, Blendis LM. Breath-hydrogen test for small- 
intestinal bacterial overgrowth. Lancet 1976; 1:668-669. 

105. Rumessen JJ, Gudmand-Hoyer E, Bachmann E, Justesen T. Diagnosis of bacterial overgrowth 
of the small intestine. Comparison of the 14 C-D-xylose breath test and jejunal cultures in 60 
patients. Scan J Gastroenterol 1985; 20:1267-1275. 



3 

The Normal Microbiota of the Human 
Gastrointestinal Tract: History of Analysis, 
Succession, and Dietary Influences 



Anne L. McCartney and Glenn R. Gibson 

Food Microbial Sciences Unit, School of Food Biosciences, Whiteknights, 
University of Reading, Reading, U.K. 



INTRODUCTION 

The human body is a wonderland for the microbial world, with harsh uninhabitable lands 
in some regions and lush fertile metropolis in others. The normal microbiota of humans is 
an extensive and diverse microbial community, which is composed primarily of bacteria 
from numerous phylogenetic clusters (1-5). The largest proportion of the human 
microbiota is found in the gastrointestinal (GI) tract, or more specifically the colon. Other 
regions of the body harboring indigenous bacterial populations include the skin, oral 
cavity, upper respiratory tract, and urogenital tract (3). This chapter aims to discuss the 
normal microbiota of the human GI tract and our current understanding of its composition 
and role in human health. Discussion of the interactions between the gut microbiota and 
the host will also abridge the impact of extrinsic factors, such as diet and environment. 

The GI tract of humans can be divided into three anatomical regions, namely, the 
stomach, small intestine (comprising duodenum, jejunum, and ileum) and large intestine 
or colon. Distinctive physicochemical environments are found within the different regions 
and the microbial populations harbored reflect this, both quantitatively and qualitatively 
(3,6,7). Thus, the normal microbiota of the human GI tract is often subdivided into three 
distinct bacterial communities: that of the upper GI tract, the ileum and the colon. 

The rapid transit time and acidic conditions of the stomach restrict the levels of 
microbial colonization of this region (6,8). Gastric juices and small-intestine secretions 
(bile and pancreatic fluids) amplify the hostile nature of the upper GI tract to microbial 
establishment. However, some aciduric Gram-positive bacteria (lactobacilli and 
streptococci) can be detected in this region (~10 2 -10 4 bacterial cells per milliliter of 
contents). In addition, some micro-organisms, such as Helicobacter pylori (the possible 
etiological agent in peptic ulcers and Type B gastritis), are able to survive, evade or 
combat the harsh conditions of the stomach (9-12). Helicobacter spp. use their flagellae to 

51 



52 McCartney and Gibson 

avoid peristaltic movement and burrow into the mucosal lining of the stomach, where they 
are partially protected from the acidic conditions by producing NH 3 from urea to 
neutralize the acid (11,12). 

The flow of digesta (intestinal motility) is somewhat slower in the ileum, 
compared with the upper GI tract, and conditions are thus more favorable for microbial 
colonization. Available data indicate increasing bacterial population levels (10 -10 
bacterial cells per milliliter of contents) and a higher diversity of micro-organisms, with 
the presence of Gram-negative facultatively anaerobic bacteria (such as members of the 
family Enterobacteriaceae) and obligate anaerobes (including Bacteroides, Veillonella, 
Fusobacterium and Clostridium species) in conjunction with lactobacilli and entero- 
cocci (1,3,6). 

The typical GI transit time is between 55 and 70 hours (13,14). Taken together with 
a more neutral pH and relative abundance of nutrients (including non-digestible 
carbohydrates and food components which have escaped digestion in the upper GI tract, 
sloughed off epithelial cells and microbial cell debris), this region of the human GI tract is 
an oasis for microbial growth, attaining levels of 10 10 — 10 12 bacterial cells per gram of 
contents (3,6,8). The composition of the colonic microbiota is extremely complex, 
generally estimated to comprise greater than 500 bacterial species, although it is thought 
that 30-40 predominate. The majority of members of the colonic microbiota are obligate 
anaerobic genera, including Bacteroides, Bifidobacterium, Clostridium, Enterococcus, 
Eubacterium, Fusobacterium, Peptococcus, Peptostreptococcus and Ruminococcus 
(2,3,15). Our understanding of the composition of the normal colonic microbiota has 
largely resulted from studies of the fecal microbiota. Questions regarding the accuracy of 
fecal samples to represent the colonic microbiota have been initially addressed by 
bacteriological analysis of the intestinal contents of sudden-death victims (14,16). This 
work demonstrated that cultivation studies of the fecal microbiota accurately reflected the 
culturable component of the distal colon. However, with recent advances in molecular 
technology (and indeed in cultivation assays), as well as sampling methods (including 
medical advances affording biopsy samples), analysis of the microbiota in different 
regions of the GI tract is now feasible as discussed below and in previous chapters. Future 
studies will, no doubt, begin to unravel the impact of impairment or disease on the mucosal 
microbiota, as well as the interaction between the luminal microbiota, the mucosally 
associated microbiota and the host. 



ROLE OF THE GASTROINTESTINAL MICROBIOTA IN HUMANS 

Traditionally, the colon has been considered to largely be the human sewage system 
which, as well as storing and removing waste material from the GI tract, was capable 
of recycling water (i.e., absorption). However, we now recognize that the GI tract is one of 
the most metabolically and immunologically active organs of the human body. Indeed, the 
primary function of the microbiota is generally considered to be salvage of energy 
via fermentation of carbohydrates, such as indigestible dietary residues (plant cell walls, 
non-digestible fibers and oligosaccharides), mucin side-chains and sloughed-off epithelial 
cells (5,6,8,13,17). It has been estimated that between 20 and 60 grams of carbohydrate are 
available in the colon of healthy human adults per day, as well as 5-20 grams of protein. In 
addition to salvaging energy, principally through production of short-chain fatty acids 
(SCFAs) and their subsequent absorption and use by the host, microbial fermentation 
produces gases (principally hydrogen, carbon dioxide and methane) and increases 
biomass. These all impact upon gut physiology. Components of the gut microbiota also 



The Normal Microbiota of the Human Gastrointestinal Tract 53 

synthesize certain B and K vitamins, metabolize xenobiotics, contribute to amino acid 
homeostasis, may impact drug efficacy and are an integral part of the host defense (both 
through host-microbe and microbe-microbe interactions; including colonization resist- 
ance) (6,17,18). Recent observations, using molecular techniques and germ-free/ 
gnotobiotic animals, have also identified that intestinal bacteria can influence gene 
expression of epithelial cells (5,19). Taken together, the activity of the microbiota, or 
certain components thereof, may be more important to the homeostasis of the ecosystem 
than specific numerics. Although the combination of all these factors, as well as host and 
environmental factors, will ultimately determine the equilibrium of the colon. 

Three main SCFAs are produced by microbial fermentation in the human colon: 
acetate, butyrate, and propionate (the approximate molar ratio for which is 70:10:20 — 
although diet and microbiota composition influence the exact ratio) (5). SCFAs supply 
energy to cells (acetate, muscle; butyrate, colonocytes; propionate, liver), affect colonic 
metabolism, control epithelial cell proliferation and differentiation, and impact upon 
bowel motility and circulation (including water absorption and the hepatic regulation of 
lipids and sugars) (5,8,13). 

Uptake and utilization of acetate is the primary method of the host salvaging energy 
from non-digestible dietary carbohydrates. Acetate may also play a role in lipogenesis by 
adipocytes and, together with propionate, may be involved in modulation of glucose 
metabolism (via the glycaemic index). Butyrate is estimated to provide between 40 and 
70% of the required energy of the colonic mucosa (5,6). In vitro studies have demonstrated 
inhibition of proliferation of neoplastic cell lines by butyrate, suggesting a possible 
beneficial role of butyrate against the progression of colorectal carcinoma. Such work has 
also shown that butyrate stimulates cell differentiation, promoting reversion to non- 
neoplastic phenotypes. 

In addition to carbohydrate fermentation, bacterial metabolism of amino acids may 
generate branched-chain fatty acids (such as isobutyrate, isovalerate, and 2-methyl 
butyrate), whilst microbial degradation of peptides and proteins forms potentially toxic 
compounds (including ammonia, amines, phenols, and indoles) (8,17). 

The colonic microbiota impacts upon amino acid homeostasis, with 1-20% of 
circulating plasma lysine being derived from the activity of gut bacteria (18). In addition, 
microbial hydrolysis of urea to ammonia by the gut microbiota is important in the 
recycling of nitrogen in the intestine. 

The protective effect of the gut microbiota against pathogenic microorganisms falls 
under two umbrellas: 1, colonization resistance and, 2, stimulation of immune function. In 
the healthy state, the resident microbiota effectively inhibits the establishment and/or 
overgrowth of harmful bacteria. A number of mechanisms appear to be responsible, 
including competition for adhesion sites, competition for nutrients, production of 
environmental conditions restrictive to pathogenic growth (pH, redox potential), 
production of anti-microbial compounds (either toxic metabolites or bacteriocins) 
and/or generation of signals which interact with gene expression of exogenous organisms 
(3,8,13). In addition, certain members of the intestinal microbiota are known to stimulate 
immune function (both locally and systemically) (17,20,21). Interactions between the 
mucosal barrier, the indigenous microbiota and the gut-associated lymphoid tissue 
(GALT) are paramount to the host defense against pathogenic invasion and infection. This 
three-component system is integral to the equilibrium of the GI tract ecosystem and 
defines the balance between oral tolerance and mounting an immune response. 

Bacterial-host cell communications can also impact upon expression by host cells. 
One example of this is the ability of Bad. thetaiotaomicron to influence fucosylated 
glycoconjugate production by intestinal cells in relation to the availability of fucose 



54 McCartney and Gibson 

(a substrate for the organism) (5,19). In this manner, the bacteria can essentially order 
nutrients from the epithelial cells as necessary. Such microbial induced signals may also 
act in cell-cell communications between different bacterial species and play an important 
role in homeostasis of their environmental niche. 



ACQUISITION OF THE GUT MICROBIOTA 

Acquisition of the normal microbiota is a biological succession which commences during 
or immediately following birth (depending on the mode of delivery). During natural birth, 
the neonate is exposed to the maternal microbiota, both vaginal and fecal (22-24). 
However, colonization is delayed in infants born via Caesarian section and the major 
source of inoculation is thought to be from the environment (including nosocomially from 
within the maternity ward) (23). Caesarean section delivery has been correlated with an 
increased clostridial component in the infant microbiota. Indeed, recent studies have 
demonstrated that higher clostridial counts in children delivered by Caesarean section 
relative to children delivered vaginally persist even after 7 years of age (25). 

During the initial phase of acquisition, facultative anaerobes predominate 
(enterobacteria and streptococci) and effectively reduce the redox potential of the gut 
environment enabling colonization by obligate anaerobes (including bacteroides, 
bifidobacteria, Clostridia, and eubacteria). Factors such as diet and host genetics play 
important roles in the development of the microbiota (with some bacterial populations 
eliminated and others maintained) (3,24). The classical studies by Tissier almost a century 
ago first highlighted the significant difference of the fecal microbiota harbored by breast- 
fed and formula-fed infants. Indeed, Tissier described three phases of microbial 
acquisition in infants: 1, initial hours of life when the fecal bacterial content was nil; 
2, beginning between the tenth and twentieth hour of life, comprising a heterogeneous 
microbiota; 3, after passage of maternal milk through the intestinal tract, the microbiota 
being predominated by bifidobacteria (an obligately anaerobic Gram-positive bacillus 
which often exhibits bifurcating morphology, formerly named Bacillus bifidus by Tissier) 
(3,26,27). A fourth phase, following introduction of solid foods (weaning), was later 
described and is characterized by modulation of the breast-fed microbiota towards an 
adult-type microbiota (climax community) harboring a more complex and diverse 
bacterial community (13,28,29). It is worth noting that Tissier also speculated that 
subdominant populations (including facultative anaerobes) were harbored during phase 
three of acquisition and that complete bacteriological examination was necessary to 
determine this. No doubt some such populations are then re-established as predominant 
members within the heterogeneous climax community through the introduction of 
complex carbohydrates into the diet. 

Bottle-fed infants did not demonstrate the same succession of micro-organisms as 
seen in their breast-fed counterparts. Indeed, Tissier observed that formula- fed infants 
maintained a heterogeneous fecal microbiota beyond day 4. Much work has been compiled 
over the last 30 years comparing the fecal microbiota of exclusively milk-fed infants. Until 
recently, such studies were performed using traditional cultivation techniques. A range of 
data has accumulated and while notable differences may still be observed between breast- 
fed and formula-fed infants, they are not as startling as those shown by Tissier. In general, 
the bifidobacterial microbiota, both carriage (percentage of infants harboring 
bifidobacteria) and population level, of exclusively milk-fed infants was not significantly 
different (30-33). However, levels of other organisms, notably Bacteroides, Clostridia and 
enterobacteria, were significantly higher in formula-fed infants. Thus, breast-fed infants 



The Normal Microbiota of the Human Gastrointestinal Tract 55 

harbored a bifidobacterially predominant fecal microbiota, whereas formula-fed 
infants harbored a larger bacterial load comprising greater heterogeneity with higher 
levels of Bacteroides, enterobacteria and Clostridia. Studies investigating the fecal 
microbiota of infants fed different formulae (for example, following fortification with iron 
and/or oligosaccharides) have shown that the constituents of the infant formulae impact 
upon the microbial composition (24,34). Recent studies employing molecular biological 
methods have further clarified the situation, demonstrating an initial diverse microbiota 
during the first 4-6 days of life (phase 2) followed by establishment of a bifidobacterially 
predominant microbiota in breast-fed infants (phase 3) which is not as obvious in formula- 
fed infants. Namely, bifidobacteria formed 60-91% of the bacterial composition of 
breast-fed infants (n = 6) and between 28 and 75% of the total microbial load of formula- 
fed infants (n = 6) after day six (35). Inter-individual differences were noted in both 
feeding groups, with respect to the relative proportions of the bacterial groups studied. 
Molecular characterization studies of the predominant isolates from concurrent cultivation 
work further highlighted the distinction between the microbiota harbored by infants [both 
between feeding groups and inter-individually (35)]. 



COMPOSITION OF THE ADULT FECAL MICROBIOTA 
ASSESSED BY CULTURING 

Much of the early information on the composition of the human colonic microbiota was 
elucidated using traditional cultivation techniques. The majority of such work was driven 
by the quest to determine the relationship between diet and colonic cancer (16,36-38). 
Epidemiological studies had identified that risk of colon cancer correlated with dietary 
habit, with higher colorectal cancer incidence in populations consuming a high-fat, low- 
fiber diet. In 1969, Aries and coworkers (39) postulated that this correlation between diet 
and cancer should be reflected in the composition of the colonic microbiota. Thus, interest 
in the effect of diet on the GI microbiota began in earnest. The majority of these early 
studies compared the fecal microbiota of individuals from different populations which had 
significantly different incidences of colon cancer. For example, Aries and coworkers (39) 
compared the fecal microbiota of English subjects (relatively high incidence) to that of 
Ugandans (low incidence). Significantly higher numbers of Bacteroides and bifidobacteria 
were enumerated from English individuals (Table 1), whilst enterococci, lactobacilli, 
streptococci, and yeasts were present at higher numbers in the fecal microbiota of 
Ugandan subjects. Subsequent studies compared the microbial compositions of multiple 
populations with either a high or a low incidence of colon cancer (38). Again, higher yields 
of bacteroides were seen for the high-risk populations (Table 1). However, an even more 
striking observation was the higher anaerobe-to-aerobe ratio in fecal samples from the 
high-incidence populations. Moore and colleagues (40) similarly showed higher levels of 
Bacteroides and bifidobacteria in subjects from high-risk populations (North Americans), 
when compared to low-risk populations (Africans). However, these observations were not 
consistent for a second low-risk population (Japanese) for whom the greatest percentage of 
isolates was Bacteroides (Table 2). More detailed characterization of these isolates 
identified that Bacteroides vulgatus, Bacteroides distasonis and Peptostreptococcus 
productus (reclassified as Ruminococcus productus) were the more predominant members 
of the fecal microbiota of high-risk populations (40). In addition, a notably higher 
percentage of isolates in the low-risk populations belonged to the species Bacteroides 
fragilis, Eubacterium aerofaciens (reclassified as Collinsella aerofaciens) and Escherichia 
coli (Table 2). Such detailed analyses of the microbial community have highlighted the 



56 



McCartney and Gibson 



O 
C 
03 

u 

c 
o 

o 
U 



O 

a 

• i-H 

o 

c 

HH 

o 

-J 

o 

X3 

00 

•I-H 

J3 



0) 

• -H 

c 

o 
U 

B 

o 

GO 

Is 

73 

• i-H 

> 

• i-H 

C 

I— I 

e 

o 

73 

■4-J 

o 
-^ 
'o 
U 

or 
<D 

S 

03 

13 
o 

Ph 



c 
o 

• i-H 

■4-J 

• i-H 

o 

s 

o 
U 

• T-H 

Sh 

<U 

■*— » 

o 

03 

Oh- 
io 

si 



60 

a 

o 

. i-H 

o3 
00 

• i-H 

> 



n 

CO 



r3 



Sh 



CD 

O 
s- 



< 



c3 

S-H 



o 

o3 pfi 

C O 

b 

c3 



< 



83 2s 1 SP •* 

c « o « 



-v — 1 

X> 

O 



60 



o 
H 



03 

-*— ' 

O 

H 



o 

c3 



60 

X) 

o 

Sh 

O 

c3 
C 
c3 



Sh O 
<D O 

O 

o 

o 






O .-H 

o ° 
ST ° 

£ ° 

c/3 



c3 

. — 

o 



O 

& 

60 

■4— » 

60 






O o3 



<D 
> 



o 





o3 




• i-H 




73 




•c 




■4— ' 




60 




o 




t 4 




u 


! 


c3 


o 


•a 


73 


<D 


eg 

s 


■4—4 

O 


0J 

•4— < 

o 


60 
73 




• i-H 

o 

Sh 


M-H 

o 


60 


. 


"a. 


o 


£ 


Z 


c3 
60 



o\car^esvo<Nvo 

£; Z; <n -h o o -h en 
~* en ir> u~> 



QQ-Hini^i^^Hm>n 






Q Q «-h <N I (N I <N CT | on t> 

^^OOOOOnOnGn 

<h ^-< r- H i- H i — I i-H 



r--ooqcooNOO^co 
irihiniriinint^odh 



qooHooqHoo«no\ 



inooN^q^trnocoos 



-mZZZZ^ZZ 






^■iri^rnrnioiri^in 



^^■iriiriiriiriioirii^ 



ONcnoooNi-jONcnr^vq 



t^ cn I 00 oq r^ oq cN ^t ; fN 

0\CCO\0\0\0\COO\0\ 



OoooocncNCNoot^^H 

^t^j-^0(N(N-H'N|-^Hin 



Gd Cd 

.y o 



b 



S-H 



o 

s: 

• i-H 

00 

c 



o 

60 



j g 
< < 



-° s 

60 73 

OD ^ 

W D W (^ ^ ffl P 



3 ^ 
•t= o 



o 

C 

03 

73 

C 

00 



u 



o 

60 

a) 

c 

03 

Oh 73 

,03 a 



c 

c3 



43 

11 

O 

o 



00 

a 

3 



3 



cd 



O- bH 



O ON 
CO 



c 
- 

O 



o 



E 
o 

X) .. 






00 

m 

«+H 

<u 

P«H 



c 

tin 



73 

o 

£ 

■— 

o 
o 

73 

■*— • 

o 

c 

Q 

Z 

is 



— a) 

si a ^ 

u < Co 



•»-H 

CO g 






^ 



The Normal Microbiota of the Human Gastrointestinal Tract 57 

Table 2 Incidence of Bacterial Populations in Fecal Samples of Individuals from Countries with 
High or Low Risk of Colon Cancer 





High incidence 




Low incidence 






North American and 












polyp patients 










Bacterial population 


(rc = 40-160) a 


Japanese (n 


= 10) 


Africans (n = 4) 


Bacteroides spp. 


29.2 b 


34.4 






23.1 


Bacteroides vulgatus 


12.5 


7.7 






2.6 


Bacteroides distasonis 


4.0 


1.7 






0.9 


Bacteroides thetaiotaomicron/ 


5.2 


7.0 






1.7 


uniformis group 












Bacteroides fragilis 


2.3 


3.2 






8.0 


Bifidobacterium spp. 


7.7 


7.8 






1.8 


Bifidobacterium adolescentis 


4.3 


6.1 






1.2 


Peptostreptococcus productus I 


3.0 


2.1 






1.3 


Peptostreptococcus productus II 


5.7 


2.2 






1.9 


Eubacterium aerofaciens II 


0.8 


2.7 






9.2 


Escherichia coli 


0.5 


1.0 






4.6 


Fusobacterium prausnitzii 


5.6 


3.4 






3.5 



a Incidence of colon cancer per 100,000. 
b Percentage of isolates. 
Source: From Ref. 40. 

importance of investigating population dynamics and not merely population levels. For 
more information on the influence of the intestinal microbiota and diet on the risk for colon 
cancer, see the chapter by Rafter and Rowland in this book. 

At this time (mid-1970s), researchers became concerned with the inherent variation 
between the different populations and the possible impact this may have on interpretation 
of the data (e.g., geographical, and genetic differences between the study groups). 
Subsequent investigations concentrated on comparing dietary changes within cultural 
populations. Initial work included comparison of two generations of Japanese living in Los 
Angeles, one maintaining the traditional Japanese (low-risk) diet and the other having 
adopted a high-risk Western diet (41). Interestingly, no statistically significant differences 
were seen in the predominant genera of the fecal microbiota of the two groups. In addition, 
though significant differences in the prevalence of certain species were observed between 
the dietary groups, the average age of the two groups was also significantly different 
(Table 3). So commenced the era of longitudinal studies, using individual subjects as their 
own controls. One of the first such studies investigated the fecal microbiota of three North 
Americans over several months and different dietary regimens (42). Greater inter- 
individual variation (between different subjects) in species composition was seen than 
intra-individual variation (between multiple samples from the same subject). Drasar and 
coworkers (36) monitored volunteers' fecal habits and composition over a six- week period 
(3 weeks on a conventional diet, followed by 3 weeks on a high-fiber diet). The only 
significant changes corresponded to stool weight and transit times. Hentges and colleagues 
(43) followed 10 subjects during baseline (1 month on a typical American diet; control), a 
meatless diet (1 month), a high-beef diet (1 month) and control diet again (1 month). Three 
stool samples were collected from each subject during the fourth week of each dietary 
period. Bacteroides spp. counts were significantly higher during the high-beef diet than the 
meatless diet (P<0.01). Similar statistically significant observations were, however, seen 



58 McCartney and Gibson 

Table 3 Summary of the Statistically Significant Differences Between Japanese Subjects 
Consuming Different Diets 





Japanese diet 


Western diet 


P value 3 


Age (years) 


60.30 


41.3 


0.013 b 


Streptococcus faecalis var faecalis 


9.83 c 


8.46 


0.038 b 


Other facultative or aerobic organisms 


7.20 


4.75 


<0.01 


Eubacterium contortum 


9.58 


ND 


0.033 


Eubacterieum Intum 


10.20 


10.07 


0.015 


Bifidobacterium infantis other 


ND 


10.29 


0.009 


Peptostreptococcus sp. 1 


10.53 





0.033 


Peptostreptococcus sp. 1-25 


8.29 


4.64 


0.00 l b 



a Based on contingency table analysis (Fisher's exact probability statistic). 

Confirmed by Student's r-test. 
c Mean logio counts per gram feces (dry weight). 
Abbreviation: ND, none detected. 
Source: From Ref. 37. 



between the Bacteroides spp. counts of the two control diet periods. Perhaps a better study 
strategy would have incorporated a control diet between the cross-over from meatless to 
high-beef diets. Indeed, this work demonstrated that short-term cross-over design dietary 
investigations may hinder identification of the effects of the different diets on the 
microbiota. Studies incorporating either prolonged diet regimens (allowing the microbiota 
to stabilize) or interspersed with control diet (enabling a return to baseline) may better 
demonstrate the microbial impact of each diet. 

Another important aspect in studies monitoring the microbial composition over time 
and between subjects is the analytical method employed. For example, in the study by 
Hentges and colleagues (43), the data were essentially averaged twice (first by subject, 
then by dietary period). Such analysis is flawed due to the inter-individual variation which 
negates significance observed intra-individually. Indeed, this was discussed by Cummings 
(44), who concluded that overall changes in a group would be obscured due to inter- 
individual variations using such analytical methods. 

Overall, data from early cultivation studies have indicated that the major bacterial 
populations harbored by individuals within a given society (i.e., Japanese, British, 
American) are reasonably stable to species level (3,45). Intra-individual, as well as inter- 
individual (even within a given society), subspecies variation has been documented in a 
number of studies following stability of the E. coli biotypes in humans (46-50). As will be 
discussed below, molecular fingerprinting techniques have demonstrated the complexity 
and dynamics of the bifidobacterial and lactobacilli populations of healthy New 
Zealanders (51,52). Such studies have highlighted the complex nature of the bacterial 
community residing in the distal regions of the human GI tract, with variation observed 
both in stability and in composition. 



COMPOSITION OF THE ADULT FECAL MICROBIOTA 
ASSESSED BY MOLECULAR TECHNIQUES 

With the advent of molecular-based techniques, bacterial characterization has become 
much more accurate, since it no longer relies upon phenotypic traits (which often vary 



The Normal Microbiota of the Human Gastrointestinal Tract 59 

due to the elastic nature of bacterial growth). In addition, more direct comparisons can be 
made between laboratories and across different studies. Initial work employed molecular 
methods to identify and/or discriminate different bacterial isolates from cultivation 
studies. One such study demonstrated that the majority of bacterial isolates from six 
healthy humans belonged to either the Bad. fragilis group or the Clostridium coccoides 
group (53). Bifidobacterium, the Clostridium leptum subgroup, Collinsella and 
Prevotella were also shown to be common phylogenetic lineages represented in healthy 
humans. Recent developments in molecular biology afford not only accurate and 
reproducible identification techniques for microbial isolates, but also strategies for direct 
community analysis at a number of genetic levels. Improved understanding of microbial 
taxonomy has generated a wealth of probing and polymerase chain reaction (PCR)-based 
strategies for quantification and/or qualification studies. Community profiling assays, 
including denaturing gradient gel electrophoresis (DGGE) and sequencing of clonal 
libraries from GI samples, have revolutionized our knowledge of the microbial 
composition of the GI tract. 

The development and application of PCR-based methods and probing strategies, 
which have circumvented cultivation, highlighted the "tip-of-the-iceberg" scenario that 
our knowledge of the GI tract microbiota amounted to. The coverage that cultivation 
studies afforded has been calculated to be as low as 10%, although others suggest it may be 
as high as 40-58% (15,54-56). Modern cultivation media and incubation conditions 
enable greater diversity, and therefore coverage, to be recognized. However, many 
components of the human gut microbiota remain elusive to cultivation in vitro. Molecular 
strategies also have their limitations, including detection limits and inherent biasing. As 
such, the overall objective of the study generally determines which assay is most 
appropriate. In the case of investigations to elucidate the diversity and dynamics of the 
human gut microbiota, a polyphasic approach is best, allowing thorough analysis at 
multiple taxonomic levels. 

Microbiota Assessed by Clone Libraries and Community 
Profiling Techniques 

Two PCR-based profiling strategies have been used to obtain an overall profile of complex 
bacterial communities — clone libraries and PCR-DGGE [or alternatively PCR-TGGE 
(temperature gradient gel electrophoresis)]. Both utilize universal PCR primers to amplify 
the 16S rRNA genes from total DNA isolated from samples. 

Suau and colleagues (15) prepared a detailed phylogenetic inventory of the fecal 
microbiota of a healthy 40-year-old male subject using PCR-cloning. A total of 520 clones 
were obtained from two transformations of the same ligation product from the 10-cycle 
PCR amplification (120 from the first and 400 from the second). The 282 clones that were 
sequenced were classified as belonging to 82 molecular species, 20 of which corresponded 
to bacteria previously cultivated from human stool samples (i.e., 24% corresponded to 
sequences available in public databases). Three major monophyletic groups contained 270 
(95.7%) of the 282 clones; the Clos. coccoides group (125 clones), the Baderoides group 
(88 clones) and the Clos. leptum group (57 clones). The remainder of the clones were 
distributed among a variety of phylogenetic clusters; two belonged to recognized 
molecular species {Streptococcus salivarius and Streptococcus parasanguinis), whilst the 
remainder were potentially novel molecular species. Most interesting was a lack of 
bifidobacterial sequences amongst the clones analyzed (even though rRNA dot-blot 
hybridizations indicated the carriage of bifidobacteria). Two possibilities could explain 
this: (1) lack of amplification of bifidobacterial rRNA genes, due to DNA extraction 



60 McCartney and Gibson 

protocol, denaturation conditions during PCR, or amplification efficiency; and (2) 
coverage of the biodiversity provided by the 282 clones was insufficient (coverage was 
calculated to be 85%; thus, the probability that the 283rd clone was a different molecular 
species from the 82 already observed was 15%). An investigation of the 25-cycle PCR 
clone library was performed in parallel to this work, using the same subject (57). 
Comparison of the 10- and 25-cycle approaches demonstrated that PCR cycle number 
influences the diversity of the resulting phylogenetic profile. The clonal library obtained 
from the 25-cycle PCR was less diversified than that from the 10-cycle PCR. However, 
differences in diversity were seen between the two methods. That is, molecular species or 
operational taxonomic units (OTUs) were present in the 25-cycle PCR clone library that 
were not represented in the 10-cycle PCR clone library. 

Previous work by Wilson and Blitchington (58) demonstrated somewhat similar 
results, with 25 of 50 clones (50%) classified as Clos. leptum subgroup, 34% as 
Bacteroides group and 10% as Clos. coccoides group. The disparity in the clostridial 
representation of the different clone libraries most probably reflects either inter-individual 
variations or disparity of the protocols. However, bifidobacteria were again absent from 
the clone library. In addition, Eubacterium rectale was not covered in the clone library in 
this earlier study, although Eub. rectale isolates were cultured from the same sample (58). 
These data highlight the difficulty to approach full coverage of the complex microbiota 
and further demonstrate that a polyphasic approach is pertinent. However, such work has 
enabled identification of previously unknown components of the fecal microbiota, and the 
sequence data can be used to develop new probing strategies to accurately quantify 
such bacteria. 

Work carried out as part of the European Union (EU) human gut microbiota project 
using PCR clone libraries demonstrated that microbial diversity increased with age (57). In 
addition, the percentage of OTUs corresponding to known molecular species was highest 
in infants and lowest in the elderly subjects. Thus, not only was the microbial diversity 
greater in the elderly subjects, but also 92% of OTUs were undescribed (potentially 
novel) species. 

The alternative to sequencing and subsequent phylogenetic analysis of clone 
libraries is to employ TGGE or DGGE to separate the 16S rRNA gene clones. Such 
techniques essentially provide a fingerprint representation of the numerically dominant 
members of the microbial community and allow rapid profiling of the microbial diversity 
of different samples (59). In addition, the TGGE/DGGE patterns can be used to selectively 
identify 16S rRNA amplicons of interest for characterization (which is achieved by 
sequencing and phylogenetic analysis). Recent years have seen an explosion in the 
development and application of TGGE and DGGE in human gut microbiology (Table 4). 
Zoetendal and coworkers (56) demonstrated the use of TGGE for monitoring the bacterial 
composition of human fecal samples. They compared the PCR-TGGE profiles of 16 
healthy adults and identified host-specific patterns reflecting inter-individual variation in 
the predominant microbiota of stool samples. Some bands were seen in samples from 
multiple subjects, suggesting that certain members of the predominant human fecal 
microbiota were common across the volunteers (56). In addition, the study encompassed 
longer-term surveillance of the microbial community of two subjects. The PCR-TGGE 
profiles of each individual did not differ greatly with time, demonstrating that 
the predominant bacterial species were relatively stable. Phylogenetic analysis of the 
predominant bacteria was performed via cloning and sequencing. PCR-TGGE of each 
clone enabled mobility comparisons and showed 45 of the 78 clones had similar mobility 
to one of the 15 prominent bands of the fecal PCR-TGGE profile. This work demonstrated 
that the majority of predominant bacterial species represented in the fingerprint did not 



The Normal Microbiota of the Human Gastrointestinal Tract 



61 



bo 

o 



-O 
o 

cj 



d 

a 

c 

§ 



a 
a 

a 
a 

H 



c 
o 

• i-H 

4— ' 

cd 
CJ 

• H 



.Q 

(0 



co 

■4— » 

CO 

O 

S-H 



cd 

> 

o 



c 
o 

. -H 
4—1 

cd 

00 

. — 
■*-' 
co 
CJ 

> 

C 



co 

4—1 
O 

C/3 



c 
o 

• i-H 

4-J 

Oh 

O 
Oh 

4— ' 

CD 

00 

Sh 

H 



CJ 

d 

Sh 
C4-H 

<D 

c4 



.d 

o 

cd 
CJ 

73 

CD 

4-J 

Cd 
J-H 
4— I 

a 
o 



00 

d 

•iH 

4—1 

cd 
^-< o 

3-9 

> 



73 
C 



d 
CD 

<u 

CO 



o 

pq cd 

o §* 

a 3 

- i 

0) cd 

"Eh 



I 

53 



cd 
73 

CD 

cd 
O 

Oh 

00 

c 

• i-H 

d 

X) 
P-) 

a 
a 

H 



•— 

.9 £ 

^^ CO 

cd 73 

£> -o 

o 

Jh 



cd 
CO 



cd 



CO 

CD 

4— > 
'— 

o 



CJ 



•s e 



d 

c 



1 s 



<D 

. — 
c 

d 



O 
CJ 

a) 



c 

CO 

O- 

S-H D 

« to 



53 
> 
c 



£ -a 



cd CO 

73 J3 

CD 00 

fa 3 

ll 

Cd —i 

-d cd 



1.2 



Jh 

o 

4— ' 

o 

Cd 
X) 



CD 
CJ 
d 

.CD 



cd 

d 

73 



d 
C 



? a 



o 

73 



> cd CD 



73 
d 



cd 
> 



S-H 

cd 

> 



Jh 

CD 

> 

O 



cd 
73 X> 



CN 

Jh 

.O 



> 

• i-H 

73 

C 



cd 

4—1 

CO 
C 

o 



CO 

cd 

co d 

~~ Jh 

O 



d 
c 



S h 



si 



Q&o 
73 Hi 

« s 



2 a 



^o 



c3 

CO 

• i-H 

C 
C 



CO 

' 15 



co 



<u 



3 _ 

h_, 73 
** cd G\ 



4-J 
• i-H 

c 

i 

a 

o 

CJ 

4— > 

o 
H 






cd 

GJ 

'cd 

73 

c 

4-4 
CJ 

O 

N 



CO 

4— ' 

CJ 
CD 

's? 

D 
CO 

^O 

CJ 

-C 

4— » 

tw 

O 

d 

•I-H 

c 

cj 
cj 

CO 

CJ 

s- 

CJ 

CO 
CJ 
Oh 



CO 

d 
c 

CJ 

>> 

00 



>% o 



•a s .2 



^3 

co 53 



CO 

g i 

Oh O 

W o 
O d 

X CD 

U CD 
i CO 

PSl CO 

ft. g> 

4-J 

CJ 

cd 

o 

73 

S § 



CJ 

73 
C 
'— 
Oh 

0^ 

u 

Cm 

o 

d 



t>3 



-d 

00 



CJ 

4— I 

CJ 



£ 

CJ 

4— < 
4— < 

cd 

Oh 



73 

CJ 

4— < 

CO 

'— 

CO 

> 

CJ 



Cj 

Q 

4— 
Cj— i CO 

w cj 

O T -1 
-4= 7-t 
O 

cj 



CD C-t 
73 P 



53 
> 

o 

d 
o 

• i-H 
4—4 

cd 

•c 

cd 
> 

C4-H 'rt 

O 3 

00 3 



73 

cj 



Uh 

u 



IT) 



c -d 

CD 

co 



cd 

d 

73 

• i-H 

> 

• i-H 

73 



CJ 

cd 

O 
73 



d ^ 
9 P 

CO Qh 



CO 

4— » 

CJ 
d 'SP 

° d 

d co 



cd cd 

CO 

Id >, 

d ^ 

73 cd 

•r" s-i 

> CD 



cd 

*C 

cj 

4—1 

CJ 

cd 
£> 

O 
73 

cd 



cd O 
d ^ 

A 



> 

• i-H 

73 
d 

• i-H 

I 

cd 



CD 



IT, 



d S 



73 

d 



cd 

ij Oh 



o -7 

5-H 

0J 



d 
CD 
00 

cj 

5-H 

CJ 



.5 ^ 



o 

CD 

.« o 

•S fa 

d o 

CA «H-H 



"H o in 

O cd u co 

•r- -d -^ 

>> « d <D 

• - B CD CD 

5 cd O > 

4-- 



v: 



d 






73 




CO 


cd 


CO 


o 




CD 


1 < 


-d 




cd 


CO 


cd 


a 

CD 


•IH 

d 


a 


d 

•H 




C+H 


m 




Ph 


w 


C< ) 


^O 







cd 

• iH 

CD 

4— < 

CJ 

cd 
-O 

O 
73 

cd 

•H 

CQ 



o 



cd 

*-• 
cj 

•H 
^d 

o 

4— < 

cd 



& td 

U w 

^ g 

3 a 

O co 

cd CD 

I CD 

^ H Oh 

CJ CO 

cd 



d 

o 

• -H 
4-J 

cd 

° -d 

cd 
> 



cd 
d 

73 

• H 

> 

-d co 

c d 

•t 1 d 

Jh O 

4^ £ 



73 

d , 

cd «g 

d c 

CD o 

.2 - 
>1 

CD ^ 



J-, 

CD 

Id 

B 

4— > CD 

"d Id 
73 d 

«» s 

N ^ 



cd 

■fi 

4-< 



cd oq 

.a < 

CJ w 

cd 



S-H 

cd co .CD 



CJ 



Oh 
CO 



Oh d 
Oh 7 Cd 

2 y § 

oo Cm -d 



^o 



CJ 

53 



B 
O 

CO 

cd 

-o 
S 









CJ 

cd 

-o 5 
d 

■-* 

a 

d 

o 



a 



d 

cd 

.3 a 

d *h 

o g c3 

'4 Ha 1 

o- d -^ 

o o| 

CD 73 •T' 



^ .s 



cd 
d 



o 5 - 1 

5 ^ d 

Oh On-d 



CN 



d oo 

o d 

CJ 1^ *-' 

• -H 73 O 

% 8 .& 

a ^ ■§ 

cd 00 55 

W •§ 

O d 

Q g 



Jh 

O 

+-H 

o 



-o 

Cc< CD 3 

p7 co co 

Cm 



•9 1 oo 

CD ^ 'JB 

oo oo -g 

d c CD 

° 73 S 

o CQ ^ ^h 

■^ 3 1 -a 

O MH "G d 



I 

o 

•— 
Oh 

d 
o 

co cd 
d- 'd 

d ° 

73 CJ 

cd • jh 
N .2 

CN 



73 

d 

* CD 

co O 

co c 

CD CD 

73^ 

cd 73 



CJ 
Jh 

CJ 



d 
cd 
cj 

3 £ 

d C 

00 *-h 
c^ 

4-J 

co o 

■§ ?. 

d ^ 

CD <£ 

8 y= 
r P 

•*-» Vh 
CD CL 

^ w 

•d O 

S Q 
o T 

^3 u 

CD Cm 
Jh ^ 

Jh 7T( 

Cd O 

S 4^ 

•-H .j-H 
^H (h 

CD 3 

> *d 

•d a 

•-H . — . 
CO c/5 

o 

Cm 



Oh 

a 

c 

CJ 

d 

cd 



'— 

43 H o 

73 ^ 

CD -d O 
Ch^OO w 

d d 73 

O CD 

H >-» fa 

00-^ ^ 

co d 

d cd 

° Sh 

co cd 

5h ih 

cd d 

Oh 00 _ 
73 'co -d 

co co 
id c 

d cd 

d a 

d d 

CD r- 
M-) 00 

o d ^3 
cd cd 

co U 4-» 

cd C cd 
o cd 75 

73 oo d 

c c § 

-H -lH 3 

d w 

d U CD 

_cd O ^ 

^^ Vh •*- J 

a « 2 

d 73 -o 
M d >. 

- CD 41^ 

g oo 'd 

cd ^h cd 

> CD .73 

oj .d .a 

pO CD co 



o 

oo 

cd 

73 

d 

cd 

■si 
O 
CJ 

• H 

73 

d 

• H 

>> 

4-J 

• H 

5h 

cd 



13 

d 
d 



CO 
5- 

cd 

CJ 



co VsO 



d 

CJ 

o 



CN 



CJ CD 

o 00 

t: d 

Oh cd 

•i-H S-H 

si w 

CO 

d 

o 



'CO 

(30 

d 



CO 

C4- 

o 



4h 

13 

Jh 

o 
d 
... «> 

CO CJ 

cd d 

d « 
2 fa 

Jig 

5h '^h 
Oh 73 



CJ 

-d 



o 

. -H 
4-J 

CJ 

d 

cj 



cd 



d 

c 

co 

CO 



oo 



00 CD 

O ^ 
CD 

4— > — ' 

cj cd 



c?+ a 
> 



n 


X5 


o 

-H 


U 
Jh 


4— ' 


( > 


-H 


•H 


o 


a 


Oh 


^^ 


a 


cd 
n 


u 


(i) 


CJ 


C+H 



CO feH 

c^ Oh 
«D ^ 



C+H 

° S d 

d 

73 
^ Ih 73 

o 



cd 






CN 



cd 

4—1 

CJ 

Id 

73 

d 

cj 
O 

N 



73 
S 

4>a 
S 

a 



62 



McCartney and Gibson 



ft 

s 

r 

■»* 

*~ 

r 

>^ 

tofi 

o 



o 

o 



G 

o 

C 

cd 

e 



a 
a 

Q 

w 
a 
a 

H 



c 
o 

• i-H 
4-J 

cd 

O 

• i-n 

"a. 



a> 

.q 

re 



CO 

4— > 

CO 
»H 



cd 

CD 

> 
O 



c 

c 

• l-H 
-4— > 

cd 
OX) 

• l-H 
4— » 

co 
cd 
> 
c 



CO 

4—1 

o 
a) 

IE 5 

00 



d 
c 

• l-H 

4— » 

ex 

o 
o- 

4— ' 

<D 

W) 

s-. 

cd 
H 



a) 

o 

d 
cd 
S-H 

«8 



3 



a) 

Sh Sh 

CD -O 



cd 



g 

<D 

d -G 

<D £ 

CD "^ 

CO g 

• !-H 1) 

?-H CO 

CD 

4=5 dj 

O C 

cd CD 

O ^ 

73 co 

IS « 

• •" '— 

^ Oh 

CO Q 

2 Q 

* ri-H 

CO Qj 

CD C 

P4 CD 

W 9 

a a 

^ co 

Q & 

03 

.2 "° 



c 

• ^H 

G 

cd 
CD 

£ 

C 
« d 

■2 .a 

a) -G 



cA 



<4-H 
I 

CD 
X 



T3 

CD 

4— ' 

g 

CD 
CD 



Oh co 

G CD 

CO « 

cd 



O 

a 

c 

CD 
CD 

Cd 

d 

o 



G 

CO 



gp.S 

d H 
• d CD 



CD 



CD 



CD g 



a , c 
° k - 

^^ 03 cO 

CO 

CD 
CD 
CD 
3 

CO 



O g 

•^ d 



or 
CD 



'CO 

cd 

CD 

Ph 



a 3 

O CD 



i 73 
43 X 



CO 

4— » ^^ 

9 r 

1 £ 



CN 



4-J 

• I-H 

c 

=5 



C 
CD 

4— I 

o 
H 



en 



03 

4— ' 

CD 

U 
CD 

•l-H 
> 

03 

Ph 



T3 

d 

c3 

d 
o 

•I-H 
4— < 

03 

•!-H 

o9 
> 

d 

T3 

•I-H 

> 

•I-H 

c 

•I-H 

I 

Jh 
CD 



PJ 

a 
a 

Q 



73 

d 

c3 

d 
o 

• i-H 

4— < 

c3 



CD 

B 

o 

CO 



-H CD 



> -3 



CD 



^^ CO 

"■* r- 

§ 8 

■S •- 

a 

^3 



cd 
73 



cd 



>^ 

03 
73 



W) d 

d cd 



> 



d 03 



CD 



CO 



>. 



O 

•— 

o 



>^ 



a * 

a 



CO 



I -l-H 



u 5 

• S ,s 

d *g 

II 

s a 

O 03 

03 



c3 

d 

u > 
^ '-3 

d.a 



c3 
O 

•I-H 

T3 
^^ d 
1o 3 

5-H 

CD lO 

o >,, 

_h 03 ■*-• 

.a -o -a 
o o .5 

.-H ■*-» ■— l 

£ b d 
c3 o o 
d C 'Z3 

>-. Ph O 

73 d B 

CD CD CD 

CD T3 

CO 



d 

O 
-G 

d 

o 

4— > 

CD 

- 4—1 
»- CO 



o 

Oh 



CD 

d 

c3 

jd 
o 

Vh 

c3 

4— < 

CD 

•l-H 

T3 



O 



3 .H 03 
CD co 



d -^ 



CD 

4d 

4^ 

c 






JO 
03 

•c 

03 



CD 

(D 



O 

»H 

D- CD 



CD 



O 

H 



d ^j > co 



U 



^ 



d 

c3 



5<. 



2 ^ 

top c3 

.a ^ 

73 O 

d CO 
CX "d 

co d 

CD O 

6 a 

o m 

d 
o 



d 

J2 CO 

• - a 

■IE 

3 *"-4 

3 ^ 

C3 

Q 



d 

.03 



O 

d 

o 



d 1) 

03 *-* 

bo.a 
.a cd 

73 c>C 



CD 

.-H Ch 



03 



03 
CO 



c3 



co CD 

CD O 

o d 

O 73 

d o 

CO £ 

13 .a 

•i-H 

y 2 

03 .O 



>^ 



i 

Vh 
CD 

> "rd 

03 

4— 1 

CO 



T3 
d 



E>3 



73 

d 



CO 

jd 

4— < 

d 

O 



^O g 



_, o _ 

d .§ s 

8 a .^ o o 

d O Jo w CN 

^3 



»H 

CD O 

> T3 

73 g 

>j O 

=2 d 

O 



Cj 

Q 
"ft 

ft 

O co 



2 
'o 

03 
73 

CD 

d 

CD 



S-.9 



CO 

-d 

4— > 

d 

o 



m 



CO 
■*-» 

03 
(N 



5- 

■a 

-o 



Oh 


1 


5 I 




d 


ft 


O 







Co 


O 








ft 




c^ 


^ 


"ft 






OX) 

d 

•l-H 

T3 

d 

13 
d 

•l-H 






• I*«4 

-ft 

a 


O 

a 
ft 


Oh 
CO 

a 


'o 


R 


ft 



3 



^1- 



03 

4— > 

CD 

W) 

•l-H 
•l-H 

CD 



O 
CD 

3 



13 

1 B? 

ft G 

** id 

ft ^3 

ft X) 

kJ 03 

^ 4— > 

H-H CO 

O ^H 

a d 

.2 "o 

3 '!H 

c3 > 

i-H 73 

^ d 



d 

c^ 

o ^ 

co 

13 ^ 

.2 ^ 

CD rH 

Oh.S 



d 

CD 
CD 

CO , , 

03 c3 
U d 

d *-3 

Jh CD 
Oh d 

MP 

U 

Oh 



^h 03 

Ch • " 

CD -jj 

S .1 
O co 

!h 

Cm ^ 

>,-d 

co tJ) 

Oh "d 

o -d 



4—> 

d 

03 
CD 

cd 

• l-H 

d 

• I-H 

CO 

<u 
'— 
(D 



03 



c^ 



cn 



o 



O X) 



CD 

CO 

03 



d 

o 

. pH 

4— ' 

03 

^H 

d X5 



o 
d 



'CO 

CD 

'cl 



s e 



CD 
> 

•I-H 

T3 
3 



ft 

cs 

-ft 

ft 

4— 

ft 

C3 



CO 

4— > 

I 

'-4— 

d 






73 2 

d cd 

1 b 

-- cd 

« > 



cd cd 

•E Ch 

^5 d 

in > 

'-d CD 

P ° 

b cd 

Oh U 



PJ 

a 
a 
a 

& 
u -^ 

Ch d 

>^^ 

o 

CD 



CD 

2 -° 

cd |3 
co 



cd 

'CO 

>^ 

CO 

O- 
O 



d 
d 



o 

CD 

13 

4— ' 

o 
H 



03 

CD 

.CD 



d 
O 

. -H 
4-J 

cd 

• pH 

cd 
> 



CD 

cd 



O 

CO 

"o- 



CD 

'CO 

•— 

CD 
> 

'CO 

d 

cd 
'— 



CO 

r-CD 

c 
•— 
o- 

03 



O 

•— 
CD 



cd 

'CO 

C 
CD 

d 

a 

T3 

d 

cd 

cd 



C/j 

CD 


»-H 

• i-H 


d 




B 


CD 


"o- 


CD 


£ 


• * 


CO 


cd 

CO 


"d 

o 


CD 


CO 


CD 

• i-H 


'Ph 

£ 

cd 


O- 


'Eh 


O 

•l-H 


a 


CO 


-O 


cd 


>. 


d 


4—1 

d 


CO 

O- 


CD 


O 


cd 


d 

• i-H 


•l-H 


13 

CD 
.(D 

M-H 


a 

o 

>H 

Oh 


4— < 

d 

CD 
U 


Ph 


<D 


fc 


a 


*W) 73 


a 


d 

• i-H 


«+H 


Q 


C^ 


o 



li -d co 

y ^ cd 



o 

•— 

Oh 



cd 03 



d 
73 

• i-H 

> 

• i-H 

73 



«* -d 



co 

Oh 

O 

.3 S 



X5 

o 

Jh 



d 

CD 
CD 



.a to 

£ 3 



Ch 

^C co 

Oh •>-' 

O- ^ - 

Lo .« .a 

«o JO co 

S d CD 

=2 co O 

*r* r~i d 

g S g 8 

ft .5 -a £ 



CA 



a 



00 

d 

►*-] CD 

toO^ 

d 



d 



ft 

C3 



15 s 

CD It! 

co X3 cd 

M d 



d 

o 



o .a 



i 
o 

O 



cd .d 



73 ^ 

ft 

• - s 

CO ft. 



w 03 

co O 

d .cd 

o ^ 



03 



13 O o Sh c3 

o o 5b^ o 

O g CD d •-" 

3 X) u. cd -O 



d 

o 

£ 

00 



2 
'o 



4— ' 

d 

^ ^^ 

• i-H •^~ s 

■M r. CO 

03 co CD 

^^ 13 

±i w d 

73 C +H 



T3 Oh 

d d 

cd O 

. Jh 



d 
d 



CO 

d 



II 

d 5 

o 
H 



io 



cd 

4— I 

CD 

13 
d 

CD 
O 

N 



'co 

u 

c 

D. 

C 
•— 
— - 

o 
0) 

CD 

13 

— < 

c 
t3 

cd 
i- 

Sb 

CD 
•— 

3 

— i 

u 

CD 



0) 

— < 

pq 

a 
a 

H 

c 
_o 

"4—' 
O 

cd 

0) 

■— 

d 

•a 

o 
0) 

Cfl 

cd 
u 

cu 

a 

p. 

of 

U 

Ch 
CO 

0) 

u 

c 

-C 
Q. 

O 

•— 
— < 

CD 

0) 

13 

— 1 

C 

CD 

73 

cd 
•— 

too 

too 

c 

a 

cd 

C 

CD 

73 

w 

a 
a 

Q 
s 



C3 



The Normal Microbiota of the Human Gastrointestinal Tract 63 

correspond to known species. However, the 15 prominent bands were identified as 
belonging within different Clostridium clusters. In addition, the common biotypes found in 
virtually all subjects' TGGE patterns were identified as Ruminococcus obeum, 
Eubacterium hallii and Fusobacterium prausnitzii (reclassified as Faecalibacterium 
prausnitzii) (56). 

More recent studies by this group have shown a positive linear relationship between 
host genetic relatedness and the similarity index of PCR-DGGE profiles (Table 4) (62). 
Higher similarity was seen between profiles obtained for monozygotic twins living apart 
than that seen for married couples. In addition, similarity was highest between 
monozygotic twin individuals than between pairs of twins. No correlation was shown 
between similarity index and gender or living arrangements of unrelated individuals, 
suggesting these factors did not significantly impact upon the bacterial composition. 
Inclusion of samples collected from four different primates (chimpanzee, gorilla, 
macaque, and orangutan) and subsequent analysis demonstrated that PCR-DGGE profiles 
of unrelated humans showed significantly greater similarity than that between humans and 
other primates. This work has indicated that host genotype factors have an important 
impact upon the bacterial composition of the gut microbiota (62). 

A number of studies have also evaluated the application of PCR-DGGE to monitor 
the composition and dynamics of particular components of the human gut microbiota 
(60,61,64). To date, such research has concentrated on the lactic acid bacteria (LAB), as 
well as bifidobacteria. Each of these studies has displayed evidence of the ability to use 
PCR-DGGE for group- or genus-specific investigations. Overall, these studies 
demonstrated inter-individual variation within specific bacterial populations (Table 4). 
Differences were seen regarding the dynamics of different bacterial groups over time: 
fluctuations were seen in the LAB of two New Zealand adults over 6 months (61); the 
bifidobacterial population of five Finnish adults remained relatively stable over 4 weeks 
(60); Lactobacillus spp. PCR-DGGE of several healthy adults displayed varying stability 
over 20 months (stable for certain individuals and more dynamic for others) (64). The 
study by Heilig and colleagues (64) also monitored the lactobacilli diversity in one baby 
boy, from birth to 5 months of age. No Lactobacillus spp. PCR product was obtained for 
the first 55 days (suggesting this population was either absent or below the detection limit). 
Subsequently, two prominent amplicons were seen to persist throughout the study period 
(64). These were identified as belonging to the species Lactobacillus rhamnosus and 
Lactobacillus casei. In addition, this work displayed bacterial succession of the 
lactobacilli corresponding to the introduction of solid foods (~3 months of age), from 
which time a third prominent amplicon was observed (Lactobacillus salivarius). Two of 
these studies further investigated the usefulness of this technique in probiotic feeding trials 
(61,64). Both groups demonstrated the ability to identify probiotic- specific amplicons 
within the group- specific bacterial profiles. 

Favier and colleagues (63) performed a pilot study with two infants investigating the 
feasibility of DGGE profiling to monitor bacterial succession during the first 
10-12 months of life. One infant was exclusively breast-fed prior to weaning, whilst the 
other was breast-fed for a fortnight and then mixed-fed (both formula- and breast-milk) 
until weaning. The results demonstrated a simple fecal microbiota initially, which 
progressively diversified with time. Bifidobacterial amplicons were predominant in the 
fecal microbiota of both infants during the first 6 months. Alterations in diet, such as the 
supplementation of breast-feeding with formula-milk and introduction of solid foods 
(weaning), was associated with changes in the bacterial profiles. The shift in bacterial 
profiles seen following weaning, was more pronounced in the exclusively breast-fed infant 
(compared to the mixed-fed infant) — although this may be a reflection of the relative 



64 McCartney and Gibson 

simplicity of the pre- weaning profile of this infant (compared to the more complex pre- 
weaning profile of the mixed-fed infant, comprised of multiple dominant amplicons) (63). 

PCR-DGGE has also been used to compare the microbial component of biopsy 
samples taken from different regions of the colon, both with each other and the fecal 
microbiota (65). Inter-individual variation was shown for both fecal and biopsy samples. 
Interestingly, the biopsy samples taken from three distinct regions of the colon (ascending, 
transverse, and descending colon) of the same individual provided extremely similar 
DGGE profiles (total community). Significant differences were evident in the total 
community PCR-DGGE of fecal and biopsy samples. This is by no means alarming, as one 
can readily appreciate the distinction of the two ecological niches (i.e., the luminal 
microbiota and mucosally associated community), and the numbers within the species are 
likely to differ and result in different profiles. However, Lactobacillus spp. PCR-DGGE 
patterns from fecal and biopsy samples were very similar in 6/10 subjects. Minor 
differences were seen in the Lactobacillus spp. PCR-DGGE profiles of the different biopsy 
samples from three of the 10 individuals. Overall, no differences were noted in the 
mucosally associated lactobacilli of different individuals based on host health (i.e., healthy 
versus diseased tissues). 

In summary, molecular methods enabling community analysis of the human fecal 
microbiota have demonstrated that a large proportion of the predominant microbial 
component are novel or unknown species — which have not yet been cultivated. Inter- 
individual variation and intra-individual stability are consistent features of studies of the 
prominent members of the total community. However, investigations of specific bacterial 
groups or genera indicate varying levels of stability, with fluctuations seen in some cases. 
Host genetic factors appear to play an important role in the microbial composition of 
healthy human adults, though it is as yet undetermined what impact bacterial acquisition 
and succession during childhood plays. 

Directed PCR Analysis 

In addition to PCR-cloning and PCR-DGGE profiling techniques, PCR strategies have 
been employed in gut microbiology for many years to investigate the presence/absence or 
activity of bacterial groups, genera, and even species. Such methods were initially 
developed for identification purposes but have subsequently been utilized for detection, 
essentially allowing qualitative analysis of the microbial component of samples. Modern 
developments in PCR technology now afford quantitative PCR assays (e.g., real-time 
PCR), though the major application of such methods to date has been clinical diagnostics. 
Wang and coworkers (66) developed 12 species-specific PCR primer sets to 
monitor the predominant gut microbiota of humans {Bact. distasonis, Bacteroides 
thetaiotaomicron, Bact. vulgatus, Bifidobacterium adolescentis, Bifidobacterium longum, 
Clostridium clostridioforme, E. coli, Eubacterium biforme, Eubacterium limosum, Fuso. 
prausnitzii, Lactobacillus acidophilus and Pep. productus). During validation of the 
species-specific PCR assays, the sensitivity of each primer set was examined with DNA 
extracts from pure cultures. Interestingly, such work demonstrated that PCR sensitivities 
varied markedly. Following validation of the PCR assays, Wang and coworkers (66) 
examined the presence of the bacterial species in fecal samples from humans (seven 
adults and two infants), two BALB/c mice, two Fischer rats, two cats, one dog, one 
rhesus monkey and one rabbit. High titers of Clos. clostridioforme, Fuso. prausnitzii and 
Pep. productus were detected in all samples examined. High titers of Bact. 
thetaiotaomicron, Bact. vulgatus and Eub. limosum were also detected in all adult 
human samples, whereas the Bacteroides spp. specific assays gave either weak or no 



The Normal Microbiota of the Human Gastrointestinal Tract 65 

PCR products for infant samples. Bifidobacteria! levels were higher in human infants 
compared to adults and other animals. 

Similar research by Matsuki and colleagues (53) developed four group-specific 
primer sets to monitor the predominant bacteria in human feces. These 16S rRNA gene- 
targeted primer sets included group-specific primers for the Bact. fragilis group and the 
Clos. coccoides group, and genus-specific primers for Bifidobacterium and Prevotella. 
DNA extracts were prepared from fecal samples collected from six healthy adults (five 
males and one female) and used for the group-specific PCR detection assays. The Bact. 
fragilis group, Bifidobacterium and Clos. coccoides group were detected in all six subjects, 
whilst PCR detected Prevotella in only two of the six subjects (53). 

PCR techniques have also been developed for identification and detection of 
bacterial isolates or components at species level. One bacterial group that has enjoyed 
particular interest in this regard is bifidobacteria (67-69). Investigation of the distribution 
of the nine bifidobacterial species known to be harbored by humans was performed by 
Matsuki and coworkers (68), using fecal samples from 48 healthy adults and 27 breast-fed 
infants. No Bifidobacterium gallicum amplification products were obtained from any 
sample. In addition, no Bifidobacterium infantis products were seen from the adult 
samples. The bifidobacterial species that were most consistently detected in adult samples 
were Bifidobacterium catenulatum (44/48), Bif. longum (31/48), Bif. adolescentis (29/48) 
and Bifidobacterium bifidum (18/48). Overall, 29 of the 48 adult samples contained three 
or four different bifidobacterial species, with 17 of the remaining 18 samples comprising 
less than three species. The majority of breast-fed infants harbored Bifidobacterium breve 
(19/27), with a smaller proportion of samples containing Bif. infantis (11/27) and 
Bif. longum (10/27; six of which were positive for Bif. infantis). Interestingly, three breast- 
fed infant samples were negative with all nine bifidobacterial species-specific primers. In 
general, breast-fed infant fecal samples were positive for three or less bifidobacterial 
species (23/27). 

Germond and coworkers (67) designed and validated species-specific primers for 
human bifidobacterial species and then developed PCR primer mixtures which enabled 
detection of multiple species concurrently (i.e., in a single reaction). PCR mix one 
comprised species-specific primers for seven bifidobacterial species: namely, 
Bif. adolescentis, Bif. angulatum, Bif. bifidum, Bif. breve, Bif. catenulatum! pseudocate- 
nulatum, Bif. infantis and Bif. longum. Application of this PCR primer mixture with fecal 
DNA from two healthy human adults demonstrated both subjects harbored Bif. longum 
and Bif. adolescentis, whilst a weak PCR amplification product was also seen for 
Bif. angulatum for one subject. Confirmation assays performed with the individual 
species-specific primer sets indicated that Bif. bifidum was under-represented during 
concurrent PCR analysis as amplification was positive for both subjects when single 
species PCR was used but negative using PCR mix one. 

Requena and colleagues (69) investigated the use of the transaldolase gene in 
identification and detection of nine bifidobacterial species {Bif. adolescentis, 
Bif. angulatum, Bif. bifidum, Bif. breve, Bif. catenulatum, Bif. infantis, Bifidobacterium 
lactis, Bif. longum and Bif. pseudo catenulatum). These workers examined its application 
for both PCR-DGGE and real-time PCR. Seven of the nine bifidobacterial species could be 
differentiated by transaldolase gene PCR-DGGE; Bif. angulatum and Bif. catenulatum 
displayed the same mobility characteristics. Examination of the bifidobacterial species 
diversity in fecal samples using this method showed 6/10 healthy adults contained two 
amplicons, one being Bif. adolescentis. In four of the six profiles the second amplicon was 
Bif. longum, the fifth profile also contained an unidentified amplicon and the sixth profile 
contained two Bif. adolescentis amplicons. One sample gave no PCR-DGGE product, two 



66 McCartney and Gibson 

of the remaining three samples contained Bif. longum, the final sample contained 
Bif. bifidum. This strategy was also employed to assess the fecal bifidobacterial diversity of 
10 babies. One sample gave no PCR product, 8/10 contained Bif. bifidum (one of which 
harbored a second unidentified amplicon) and the final sample comprised Bif. infantis, 
Bif. longum and an unidentified amplicon. 

Comparison of bifidobacterial enumerations obtained from plate counts and 
bifidobacterial-specific real-time PCR with either transaldolase gene primers or 16S 
rRNA primers has been performed (69). Good correlation was seen between all 
three enumeration methods when healthy adult samples were used (n = l). 
Correlation of bifidobacterial levels in infant samples (ft =10) was better between 
cultivation work and 16S rRNA gene real-time PCR than between cultivation work 
and transaldolase gene real-time PCR. Under-representation of the Bif. bifidum 
component of samples during transaldolase gene real-time PCR was largely 
responsible for this discrepancy. 

Probing Strategies 

As well as affording design of PCR primers for specific bacterial populations, the 
improved 16S rRNA gene sequence information has greatly enhanced the development of 
probing strategies for gut micro-organisms. Two probing strategies have generally been 
employed, namely, dot-blot hybridization and fluorescent in situ hybridization (FISH). 
The nature of the 16S rRNA gene of bacteria also enables development of oligonucleotide 
probes targeting different taxonomic levels, i.e., domain level (Bact 338), group level (e.g., 
Chis 150), genus level (e.g., Bif 164) or species level (e.g., Bdis 656) (57,70). The last 
5 years have seen enormous development and application of these strategies in gut 
microbiology (71-76). 

A longitudinal study was performed with nine healthy human volunteers (five males, 
four females) monitoring the fecal microbiota using FISH (72). The results demonstrated 
that 90-100% of 4', 6-diamidino-2-phenylindol dihydrochloride (DAPI)-stained cells were 
hybridized by the bacterial probe (Bact 338), and that the Clos. coccoideslEub. rectale 
group (Erect 482) and Bacteroides group (Bfra 602 and Bdis 656) represented almost 50% 
of the total bacteria of healthy humans. In addition, the Low G + C #2 group (Lowgc2P) 
comprised 12% of the total bacteria, and Bifidobacterium (Bif 164) 3%. Initial data 
indicated that the Clostridium lituseburense group (Clit 135), the Clostridium histolyticum 
group (Chis 150), and the StreptococcuslLactococcus group (Strc 493) all formed less than 
1% of the total bacteria and so were not included in the longitudinal study. In general, the 
fecal microbiota of individuals was shown to fluctuate during the 8-month study. 
Interestingly, the greatest variation was seen in the bifidobacterial component of the 
microbiota. A more recent study from the same laboratory group employed a set of 15 
probes to investigate the microbial composition of 1 1 healthy volunteers (73). Again, the 
Bacteroides group (27.7%) and the Clos. coccoideslEub. rectale group (22.7%) were seen 
to be the numerically predominant bacterial components. In addition, three other 
predominant groups were identified: Atopobium group (11.9%), Eubacterium low G + C 
#2/Fuso. prausnitzii group (10.8%), and Ruminococcus and relatives (10.3%). 
Bifidobacterium (4.8%), Eub. hallii and relatives (3.8%), Lachnospira and relatives 
(3.6%), and Eubacterium cylindroides and relatives (1.8%) were also dominant members 
of the microbiota. However, Enterobacteriaceae, the Lactobacillus! Enterococcus 
group, Phascolarctobacterium and relatives, and Veillonella were all subdominant (each 
forming 1%). Taken together, this afforded 90.5% coverage of the total bacteria hybridized 
with the Bact 338 probe. (N.B.: Eub. hallii and relatives, and Lachnospira and relatives are 



The Normal Microbiota of the Human Gastrointestinal Tract 67 

subsets of the Clos. coccoideslEub. rectale group, so were not included in summation). 
However, a large proportion of the DAPI-stained cells (~40%) were not accounted for by 
the Bact 338 probe. The question arises as to whether these cells are non- viable or 
metabolically inactive (low rRNA), impermeable, or represent novel bacterial groups 
whose 16S rRNA differs within the "conserved" region the Bact 338 probe targets or in the 
secondary structure surrounding it. 

Other research groups have developed and validated additional oligonucleotide 
probes suitable for FISH, for potentially important members of the human GI tract 
microbiota (75,76). Ruminococcus obeum-like bacteria have been frequently identified in 
ribosomal clonal libraries of human fecal samples and the development of probing 
strategies was thus considered pertinent (76). Following validation, the Urobe 63 probe 
was used to examine the Rum. obeum group in three healthy Dutch males (three samples 
were collected from each subject over one month). FISH enumeration was performed both 
by epifluorescent microscopy and by flow cytometry (which require different handling and 
thus different protocols). The two methods gave comparable results, demonstrating that 
Rum. obeum-like bacteria comprise ~2.5% of the total bacterial count (Bact 338). A 
further six individuals (two males, four females) provided stool samples and the results 
were consistent in all subjects. In addition, counts of the Clos. coccoideslEub. rectale 
group were made which indicated that the Rum. obeum group accounted for ~ 16% of 
this group (76). Similarly, the Fuso. prausnitzii cluster has been shown in numerous 
molecular analyses to be part of the dominant microbiota of healthy humans. As such, 
Suau, and coworkers (75) developed an oligonucleotide probe for this cluster which was 
applicable both for FISH and dot-blot hybridizations. Overall, 16.5% (range 5-28%) of the 
DAPI-stained cells hybridized with the Fprau 645 probe (n= 10 healthy adults). Samples 
from a further 10 healthy individuals were used for dot-blot analysis with the same probe 
and showed the Fuso. prausnitzii cluster accounted for 5.3% of the total bacterial 16S 
rRNA (range 1.5-9.5%). Unfortunately, these data are not comparable as different samples 
were used for each assay. In addition, the two assays provide distinctive enumeration: 
FISH provides counts of the number of cells in the sample (which can be represented as a 
percentage of total bacterial (Bact 338) cells or total cells (DAPI), whereas dot-blot 
provides an index of the percentage of total 16S rRNA the specific population forms. 
The index obtained by dot-blot is further complicated as it is not only proportional to the 
number of cells in the sample, but also the number of copies of the rRNA gene in each cell 
and the activity of the cells. 

Dot-blot analyses of the healthy human fecal microbiota using an array of probes 
have, once again, highlighted the inter-individual variation (71,74). Both studies 
employed six oligonucleotide probes to monitor the predominant bacterial groups. The 
work by Sghir and colleagues (74) (n = 21 healthy adults; 13 males, 14 females) was 
consistent with earlier work which showed that the Bacteroides group (including 
Bacteroides, Prevotella and Porphyromonas; 37%), the Clos. leptum subgroup (16%) 
and the Clos. coccoideslEub. rectale group (14%) were predominant, accounting for 67% 
of the total rRNA. Bifidobacterium and the enteric group each made up less than 1 % of 
the total rRNA, whilst the low-G + C Gram-positive group (including Lactobacillus, 
Streptococcus and Enterococcus) represented 1% (74). Marteau and coworkers (71) 
similarly demonstrated the predominant fecal rRNA (n = S healthy adults; four males, 
four females) corresponded to the Clos. coccoideslEub. rectale group (23%), the Clos. 
leptum subgroup (13%) and the Bacteroides group (8%) using the same probes as Sghir 
and colleagues (74). Although, using different probes, this later study indicated higher 
bifidobacterial and Lactobacillus! Enterococcus rRNA indices, 3% and 7%, respectively. 
Interestingly, Marteau and coworkers (71) compared the fecal rRNA indices of these 



68 McCartney and Gibson 

bacterial groups and E. coli species with cecal rRNA indices. Overall, the indices for the 
Bacteroides group and the Clos. leptum subgroup were significantly higher in fecal 
samples than cecal samples, and the Lactobacillus! Enterococcus fecal rRNA index was 
significantly lower than that of the cecum. The Clos. coccoides/Eub. rectale rRNA index 
was higher in fecal samples than cecal samples, but the inter-individual variation meant 
that this was not statistically significant. Concurrent cultivation analysis monitoring total 
anaerobes, facultative anaerobes, bifidobacteria and Bacteroides demonstrated signifi- 
cantly higher levels of total anaerobes, bifidobacteria, and bacteroides populations in 
fecal samples compared to cecal samples (71). 

Most recent developments in probing strategies include membrane- array and/or 
microarray methodologies (77,78). The results of these assays were in agreement with 
previous studies, demonstrating inter-individual variation in the fecal microbiota of 
different healthy human subjects. The predominant microbiota of healthy humans 
determined by the membrane-array technique (employing 60 oligonucleotide probes 
targeting 20 bacterial species) included Bacteroides species, Clos. clostridioforme, 
Clos. leptum, Fuso. prausnitzii, Pep. productus, Ruminococcus species, Bifidobacterium 
species and E. coli (78). In addition, analysis of the fecal microbiota of an individual 
suffering long-term diarrhea demonstrated a loss of a number of bacterial species common 
to the normal microbiota of healthy subjects. These results were replicated in a microarray 
study using the same probe array (77), where the probes were printed on aldehyde slides 
rather than applied to membranes. 

Overall, probing and PCR-based strategies have been shown to afford good coverage 
of the predominant microbiota of the GI tract. This situation is likely to improve with 
continued development of specific primer sets and/or oligonucleotide probes, particularly 
in the light of increased diversity as elucidated by community analysis work. Indeed, such 
community profiling studies provide excellent direction for the development of novel 
probes and primers. 



INVESTIGATIONS AT THE SUBSPECIES LEVEL 

One aspect of gut microbiology that is not amenable to current probing or PCR-based 
methodologies is subspecies differentiation (i.e., investigations of the microbial comple- 
xity and dynamics below the phylogenetic level of species). A number of studies have, 
however, demonstrated the importance of such research (48,51,52). One study monitored 
the composition of the bifidobacterial and lactobacilli populations of two healthy humans 
over a 12-month period (52). Overall, the bifidobacterial levels of both individuals were 
relatively stable throughout the study period [~ 10 10 /g of feces (wet weight)]. Lactobacilli 
levels were relatively constant in subject one (~10 9 /g) but fluctuated considerably in 
samples from subject two (10 6 -10 9 per gram). Bacteroides and enterobacterial levels were 
also examined during the study; the former remained stable for both individuals and the 
latter displayed marked variability (especially in subject two, for whom numbers were 
below the detection limit in weeks 17 and 20). Genetic fingerprinting techniques were used 
to differentiate the predominant bacterial isolates of the bifidobacterial and lactobacillus 
populations. Interestingly, two distinct bifidobacterial profiles were seen, with one 
individual harboring a simple, stable bifidobacterial population (five distinct strains of 
bifidobacteria were detected during 12 months, one of which was numerically 
predominant throughout the study). In contrast, the second subject harbored a more 
complex and dynamic bifidobacterial population (36 distinct strains were seen over the 



The Normal Microbiota of the Human Gastrointestinal Tract 69 

12 months, with between four and nine strains at any given time). The Lactobacillus 
populations of both subjects were simple and stable. None of the lactic acid bacterial 
strains isolated during the study were common to both individuals, further accentuating the 
inter-individual variations of these two microbial ecosystems (52). Subsequent work from 
the same laboratory extended these investigations to a further eight healthy humans (four 
males, four females) (51). Two separate samples were collected and processed for each 
individual. The results of this secondary study confirmed the findings of the former, with 
bifidobacterial levels remaining relatively stable and Lactobacillus numbers varying 
greatly. Again, each individual harbored unique bifidobacterial and Lactobacillus strains 
(at least in regard to the numerically predominant microbiota). Intriguingly, half of the 
subjects were shown to harbor a complex bifidobacterial microbiota (five or more 
predominant strains). 

McBurney and colleagues (48) examined the perturbation of the enterobacterial 
populations of the initial long-term study by McCartney and colleauges (52). Similar to the 
bifidobacterial populations of these two individuals, the enterobacterial population of 
subject one was relatively simple and stable (predominated by a single strain) whilst 
subject two harbored a diverse and dynamic enterobacterial biota (27 distinct strains were 
identified over 12 months). As mentioned previously, enterobacterial levels were below 
detection for subject two during weeks 17 and 20. This individual undertook a 7-day 
course of amoxicillin for a respiratory infection during weeks 21 and 22, after which time 
the enterobacterial population re-emerged. Most interesting, though, was the antibiotic- 
resistance profiles of this bacterial group before and after treatment. Strains isolated prior 
to antibiotic administration were susceptible to a wide range of antibiotics tested, whereas 
strains isolated following treatment were resistant to a number of antibiotics. Thirteen 
weeks after amoxicillin administration, multiple drug-resistant enterobacterial strains 
were still present. In the following 2 months, strains resistant to ampicillin were still 
harbored, and only after 25 weeks post-treatment did the predominant enterobacterial 
microbiota return to a simple, stable, susceptible composition. 

Taken together, the above-mentioned work clearly demonstrates the value of 
investigations at the subspecies level, as such studies afford more detailed analysis of the 
diversity and dynamics of the gut microbiota. Furthermore, such strategies allow the 
detection of microbial perturbations which are often not evident at the bacterial group, 
genus or species levels (79). 



CONCLUSION 

The normal microbiota of the human GI tract is a complex microbial community whose 
composition is defined by a number of factors (including host genomics, diet, age, 
bacterial succession, immune function and health status). In general, the predominant 
bacterial groups are relatively stable in healthy human adults. However, inter-individual 
variations are evident, reflecting the unique equilibrium of each person's GI ecosystem. In 
addition, examination of the microbial populations in more detail (i.e., investigations at the 
subspecies level) further demonstrates the complexity and dynamics of this bacterial 
community, and most probably reflects its adaptive nature. Interactions between the host 
and the gut microbiota have led some researchers to acknowledge that the human intestine 
is, indeed, "intelligent" — based on Alfred Binet's definition of intelligence: "intelligence 
is the range of processes involved in adapting to the environment" (13). 



70 McCartney and Gibson 

REFERENCES 

1. Simon GL, Gorbach SL. Intestinal flora in health and disease. Gastroenterology 1984; 
86:174-193. 

2. Macfarlane GT, Cummings JH. The colonic flora, fermentation, and large bowel digestive 
function. In: Phillips SF, Pemberton JH, Shorter RG, eds. The Large Intestine: Physiology, 
Pathophysiology, and Disease. New York: Raven Press Ltd, 1991:51-92. 

3. Tannock GW. An Introduction to Microbes Inhabiting the Human Body. London: Chapman & 
Hall, 1995. 

4. Mitsuoka T. Intestinal flora and human health. Asia Pacific J Clin Nutr 1996; 5:2-9. 

5. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Annu Rev Nutr 2002; 22:283-307. 

6. Holzapfel WH, Haberer P, Snel J, Schillinger U, Huis in't Veld JHJ. Overview of gut flora and 
probiotics. Int J Food Microbiol 1998; 41:85-101. 

7. Rabiu BA, Gibson GR. Carbohydrates: a limit on bacterial diversity within the colon. Biol Rev 
2002; 77:443-453. 

8. Guarner F, Malagelada J-R. Gut flora in health and disease. Lancet 2003; 361:512-519. 

9. Blaser MJ, Kirschner D. Dynamics of Helicobacter pylori colonization in relation to the host 
response. Proc Natl Acad Sci USA 1999; 96:8359-8364. 

10. Coconnier M-H, Lievin V, Hemery E, Servin AL. Antagonistic activity against Helicobacter 
infection in vitro and in vivo by the human Lactobacillus acidophilus strain LB. Appl Environ 
Microbiol 1998; 64:4573-4580. 

1 1 . Gibson GR, McCartney AL. Modification of the gut flora by dietary means. Biochem Soc Trans 
1998; 26:222-228. 

12. Marshall BJ. Helicobacter pylori. Am J Gastroenterol 1994; 89:S116-S128. 

13. Bourlioux P, Koletzko B, Guarner F, Braesco V. The intestine and its microflora are partners 
for the protection of the host: report on the Danone Symposium "The Intelligent Intestine," held 
in Paris, June 14, 2002. Am J Clin Nutr 2003; 78:675-683. 

14. Macfarlane GT, Macfarlane S, Gibson GR. Validation of three-stage continuous culture system 
for investigating the effect of retention time on the ecology and metabolism of bacteria in the 
human colon. Microb Ecol 1998; 35:180-187. 

15. Suau A, Bonnet R, Sutren M, et al. Direct analysis of genes encoding 16S rRNA from complex 
communities reveals many novel molecular species within the human gut. Appl Environ 
Microbiol 1999; 65:4799-4807. 

16. Moore WEC, Holdeman LV. Discussion of current bacteriological investigations of the 
relationships between intestinal flora, diet, and colon cancer. Cancer Res 1975; 
35:3418-3420. 

17. Salminen S, Bouley C, Boutron-Ruault M-C, et al. Functional food science and gastrointestinal 
physiology and function. Br J Nutr 1998; 80:S147-S171. 

18. Metges CC. Contribution of microbial amino acids to amino acid homeostasis of the host. 
J Nutr 2000; 130:1857S-1864S. 

19. Gordon JI, Hooper LV, McNevin MS, Wong M, Bry L. Epithelial cell growth and 
differentiation. III. Promoting diversity in the intestine: conversations between the microflora, 
epithelium, and diffuse GALT. Am J Physiol 1997; 273:G565-G570. 

20. Borruel N, Casellas F, Antolin M, et al. Effects of nonpathogenic bacteria on cytokine secretion 
by human intestinal mucosa. Am J Gastroenterol 2003; 98:865-870. 

21. Perdigon G, Maldonado Galdeano C, Valdez JC, Medici M. Interaction of lactic acid bacteria 
with the gut immune system. Eur J Clin Nutr 2002; 56:S21-S26. 

22. Tannock GW. The acquisition of the normal microflora of the gastrointestinal tract. In: 
Gibson SAW, ed. Human Health: The Contribution of Microorganisms. London: Springer- 
Verlag, 1994:1-16. 

23. Bezirtzoglou E. The intestinal microflora during the first weeks of life. Anaerobe 1997; 
3:173-177. 



The Normal Microbiota of the Human Gastrointestinal Tract 71 

24. Mountzouris KC, McCartney AL, Gibson GR. Intestinal microflora of human infants and 
current trends for its nutritional modulation. Br J Nutr 2002; 87:405-420. 

25. Salminen S, Gibson GR, McCartney AL, Isolauri E. Influence of mode of delivery on gut 
microbiota composition in seven year old children. Gut 2004; 53:1388-1389. 

26. Biavati B, Sgorbati B, Scardovi V. The Genus Bifidobacterium. In: Balows A, Triiper HG, 
Dworkin M, Harder W, Schleifer KH, eds. The Prokaryotes. A Handbook on the Biology of 
Bacteria: Ecophysiology, Isolation, Identification, Applications. 2nd ed., Vol. 1. New York: 
Springer- Verlag, 1992:816-833. 

27. Scardovi V. Genus Bifidobacterium Orla-Jensen 1924, 472 AL . In: Sneath PHA, Mair NS, 
Sharpe ME, Holt JG, eds. In: Bergey's Manual of Systematic Bacteriology, Vol. 2. Baltimore: 
Williams and Wilkins, 1986:1418-1434. 

28. Edwards CA, Parrett AM. Intestinal flora during the first months of life: new perspectives. Br 
J Nutr 2002; 88:S11-S18. 

29. Salminen S, Bouley C, Boutron Ruault M-C, et al. Functional food science and gastrointestinal 
physiology and function. Br J Nutr 1998; 80:S147-S171. 

30. Kleessen B, Bunke H, Tovar K, Noack J, Sawatzki G. Influence of two infant formulas and 
human milk on the development of the faecal flora in newborn infants. Acta Paediatrica 1995; 
84:1347-1356. 

31. Mitsuoka T, Kaneuchi C. Ecology of the bifidobacteria. Am J Clin Nutr 1977; 30:1799-1810. 

32. Stark PL, Lee A. The microbial ecology of the large bowel of breast-fed and formula-fed 
infants during the first year of life. J Med Microbiol 1982; 15:189-203. 

33. Yoshioka H, Fujita K, Sakata H, Murono K, Iseki K. Development of the normal intestinal flora 
and its clinical significance in infants and children. Bifidobacteria Microflora 1991; 2:33-39. 

34. Boehm G, Lidestri M, Casetta P, et al. Supplementation of a bovine milk formula with an 
oligosaccharide mixture increases counts of faecal bifidobacteria in preterm infants. Arch Dis 
Child 2002; 86:178-181. 

35. Harmsen HJM, Wildeboer-Veloc ACM, Raangs GC, et al. Analysis of intestinal flora 
development in breast-fed and formula-fed infants by using molecular identification and 
detection methods. J Pediatr Gastroenterol Nutr 2000; 30:61-67. 

36. Drasar BS, Jenkins DJA. Bacteria, diet, and large bowel cancer. Am J Clin Nutr 1976; 
29:1410-1416. 

37. Finegold SM, Sutter VL. Fecal flora in different populations, with special reference to diet. Am 
J Clin Nutr 1978; 3LS116-S122. 

38. Hill MJ, Drasar BS, Aries V, Crowther JS, Hawksworth G, Williams REO. Bacteria and 
aetiology of cancer of large bowel. Lancet 1971; 1:95-100. 

39. Aries V, Crowther JS, Drasar BS, Hill MJ, Williams REO. Bacteria and the aetiology of cancer 
of the large bowel. Gut 1969; 10:334-335. 

40. Moore WEC, Cato EP, Holdeman LV. Some current concepts in intestinal bacteriology. Am 
J Clin Nutr 1978; 3LS33-S42. 

41. Finegold SM, Attebery HR, Sutter VL. Effect of diet on human fecal flora: comparison of 
Japanese and American diets. Am J Clin Nutr 1974; 27:1456-1469. 

42. Holdeman LV, Good IJ, Moore WEC. Human fecal flora: variation in bacterial composition 
within individuals and a possible effect of emotional stress. Appl Environ Microbiol 1976; 
31:359-375. 

43. Hentges DJ, Maier BR, Burton GC, Flynn MA, Tsutakawa RK. Effect of a high-beef diet on the 
fecal flora of humans. Cancer Res 1977; 37:568-571. 

44. Cummings JH. Dietary fibre and the intestinal microflora. In: Hallgren B, ed. Nutrition and the 
Intestinal Flora. Stockholm: Almqvist and Wiksell International, 1983:77-86. 

45. Bornside GH. Stability of human fecal flora. Am J Clin Nutr 1978; 3LS141-S144. 

46. Hartley CL, Clements HM, Linton KB. Escherichia coli in the faecal flora of man. J Appl 
Bacteriol 1977; 43:261-269. 

47. Mason TG, Richardson G. A review. Escherichia coli and the human gut: some ecological 
considerations. J Appl Bacteriol 1981; 51:1-16. 



72 McCartney and Gibson 

48. McBurney WT, McCartney AL, Apun K, McConnell MA, Tannock GW. Perturbation of the 
enterobacterial microflora detected by molecular analysis. Microb Ecol Health Dis 1999; 
11:175-179. 

49. Shooter RA, Bettelheim KA, Lennox-King SMJ, O'Farrell S. Escherichia coli serotypes in the 
faeces of healthy adults over a period of several months. J Hyg 1977; 78:95-98. 

50. Wallick H, Stuart CA. Antigenic relationships of Escherichia coli isolated from one individual. 
J Bacteriol 1943; 45:121-126. 

51. Kimura K, McCartney AL, McConnell MA, Tannock GW. Analysis of fecal populations of 
bifidobacteria and lactobacilli and investigation of the immunological responses of their human 
hosts to the predominant strains. Appl Environ Microbiol 1997; 63:3394-3398. 

52. McCartney AL, Wenzhi W, Tannock GW. Molecular analysis of the composition of the 
bifidobacteria! and lactobacillus microflora of humans. Appl Environ Microbiol 1996; 
62:4608-4613. 

53. Matsuki T, Watanabe K, Fujimoto J, et al. Development of 16S rRNA-gene-targeted group- 
specific primers for the detection and identification of predominant bacteria in human feces. 
Appl Environ Microbiol 2002; 68:5445-5451. 

54. Tannock GW. Analysis of the intestinal microflora using molecular methods. Eur J Clin Nutr 
2002; 56:S44-S49. 

55. Vaughan EE, Schut F, Heilig HGHJ, Zoetendal EG, de Vos WM, Akkermans ADL. A 
molecular view of the intestinal microflora. Curr Issues Intest Microbiol 2000; 1:1-12. 

56. Zoetendal EG, Akkermans ADL, de Vos WM. Temperature gradient gel electrophoresis 
analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities 
of active bacteria. Appl Environ Microbiol 1998; 64:3854-3859. 

57. Blaut M, Collins MD, Welling GW, Dore J, van Loo J, de Vos W. Molecular biological 
methods for studying the gut microflora: the EU human gut flora project. Br J Nutr 2002; 
87:S203-S211. 

58. Wilson KH, Blitchington RB. Human colonic flora studied by ribosomal DNA sequence 
analysis. Appl Environ Microbiol 1996; 62:2273-2278. 

59. Konstantinov SR, Fitzsimons N, Vaughan EE, Akkermans ADL. From composition to 
functionality of the intestinal microbial communities. In: Tannock GW, ed. Probiotics and 
Prebiotics. Where are we Going?. Norfolk, U.K.: Caister Academic Press, 2002:59-84. 

60. Satokari RM, Vaughan EE, Akkermans ADL, Saarela M, de Vos WM. Bifidobacterial diversity 
in human feces detected by genus-specific PCR and denaturing gradient gel electrophoresis. 
Appl Environ Microbiol 2001; 67:504-513. 

61 . Walter J, Hertel C, Tannock GW, Lis CM, Munro K, Hammes WP. Detection of Lactobacillus, 
Pediococcus, Leuconostoc, and Weissella species in human feces by using group-specific PCR 
primers and denaturing gradient gel electrophoresis. Appl Environ Microbiol 2001; 
67:2578-2585. 

62. Zoetendal EG, Akkermans ADL, Akkermans, van Vliet WM, de Visser JAGM, de Vos WM. 
The host genotype affects the bacterial community in the human gastrointestinal tract. Microb 
Ecol Health Dis 2001; 13:129-134. 

63. Favier CF, Vaughan EE, de Vos WM, Akkermans ADL. Molecular monitoring of succession of 
bacterial communities in human neonates. Appl Environ Microbiol 2002; 68:219-226. 

64. Heilig HGHJ, Zoetendal EG, Vaughan EE, Marteau P, Akkermans ADL, de Vos WM. 
Molecular diversity of Lactobacillus spp. and other lactic acid bacteria in the human intestine 
as determined by specific amplification of 16S ribosomal DNA. Appl Environ Microbiol 2002; 
68:114-123. 

65. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben- Amor K, Akkermans ADL, de 
Vos WM. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly 
distributed along the colon and differ from the community recovered from faeces. Appl 
Environ Microbiol 2002; 68:3401-3407. 

66. Wang R-F, Cao W-W, Cerniglia CE. PCR detection and quantitation of predominant anaerobic 
bacteria in human and animal fecal samples. Appl Environ Microbiol 1996; 62:1242-1247. 



The Normal Microbiota of the Human Gastrointestinal Tract 73 

67. Germond J-E, Mamin O, Mollet B. Species specific identification of nine human 
Bifidobacterium spp. in feces. Syst Appl Microbiol 2002; 25:536-543. 

68. Matsuki T, Watanabe K, Tanaka R, Fukuda M, Oyaizu H. Distribution of bifidobacterial 
species in human intestinal microflora examined 16S rRNA-gene-targeted species-specific 
primers. Appl Environ Microbiol 1999; 65:4506-4512. 

69. Requena T, Burton J, Matsuki T, et al. Identification, detection, and enumeration of human 
Bifidobacterium species by PCR targeting the transaldolase gene. Appl Environ Microbiol 
2002; 68:2420-2427. 

70. Harmsen HJM, Welling GW. In: Tannock GW, ed. Probiotics and Prebiotics. Where are we 
going?. Norfolk, U.K.: Caister Academic Press, 2002:41-58. 

71. Marteau P, Pochart P, Dore J, Bera-Maillet C, Bernalier A, Corthier G. Comparative study of 
bacterial groups within the human cecal and fecal microflora. Appl Environ Microbiol 2001; 
67:4939-4942. 

72. Franks AH, Harmsen HJM, Raangs GC, Jansen GJ, Schut F, Welling GW. Variations of 
bacterial populations in human feces measured by fluorescence in situ hybridization with 
group-specific 16S rRNA-targeted oligonucleotide probes. Appl Environ Microbiol 1998; 
64:3336-3345. 

73. Harmsen HJM, Raangs GC, He T, Degener JE, Welling GW. Extensive set of 16S rRNA-based 
probes for detection of bacteria in human feces. Appl Environ Microbiol 2002; 68:2982-2990. 

74. Sghir A, Gramet G, Suau A, Rochet V, Pochart P, Dore J. Quantification of bacterial groups 
within human fecal flora by oligonucleotide probe hybridization. Appl Environ Microbiol 
2000; 66:2263-2266. 

75. Suau A, Rochet V, Sghir A, et al. Fusobacterium prausnitzii and related species represent a 
dominant group within the human fecal flora. Syst Appl Microbiol 2001; 24:139-145. 

76. Zoetendal EG, Ben-Amor K, Harmsen HJM, Schut F, Akkermans ADL, de Vos WM. 
Quantification of uncultured Ruminococcus obeum-\ike bacteria in human fecal samples by 
fluorescent in situ hybridization and flow cytometry using 16S rRNA-targeted probes. Appl 
Environ Microbiol 2002; 68:4225-4232. 

77. Wang R-F, Beggs ML, Robertson LH, Cerniglia CE. Design and evaluation of oligonucleotide- 
microarray method for the detection of human intestinal bacteria in fecal samples. FEMS 
Microbiol Lett 2002; 213:175-182. 

78. Wang R-F, Kim S-J, Robertson LH, Cerniglia CE. Development of a membrane-assay method 
for the detection of intestinal bacteria in fecal samples. Mol Cell Probes 2002; 16:341-350. 

79. Tannock GW. Molecular assessment of intestinal microflora. Am J Clin Nutr 2001; 
73:410S-414S. 



4 

The Intestinal Microbiota of the Elderly 



Fang He 

Technical Research Laboratory, Takanashi Milk Products Co., Ltd, Yokohama, 
Kanagawa, Japan 



INTRODUCTION 

With the significant progress in medical science and health care, the average life expectancy 
has increased by nearly three decades over the last century (1). The old (>65 years) and 
the "oldest" ( > 85 years) age groups are the fastest growing subpopulation in the world, 
especially in industrialized societies referred to as "aged societies." World Health 
Organization (WHO) figures indicate there are currently about 580 million people in the 
world aged 60 or older, and this figure is expected to rise to over a billion within the next 
20 years (2). 

It has been well known that many physiological functions, such as immunity and gut 
function, in humans usually decline progressively with age after approximately the 30th 
birthday (1). The elderly are an increased-risk population with high rates of morbidity 
and mortality due to their susceptibility to degenerative and infectious diseases. A major 
consequence of people living longer is an increased incidence in health problems. In fact, 
industrial societies are now suffering from a sharp increase in medical costs to the age- 
related infectious and autoimmune diseases, malignancies, allergies, and digestive problems. 
Therefore, effective measures to redress the age-related decline (or imbalance) in 
physiological function should be much sought. 

The intestinal microbiota mediates many crucial events towards the protection or 
alteration of health. This chapter summarizes the current knowledge and findings about the 
intestinal microbiota in the elderly, although a limited but growing body of literature on 
this subject is available. 



COLONIZATION AND SUCCESSION OF HUMAN INTESTINAL 
MICROBIOTA WITH AGE 

The gastrointestinal tract (GI tract) serves as one of the biggest interfaces between the 
body and the external environment (3). This GI tract is a highly specialized organ system 
that allows us to consume food in discrete meals as well as a very diverse array of 
foodstuffs to meet our nutrient needs. The organs of the GI tract include the oral cavity, 

75 



76 He 

esophagus, stomach, small and large intestine; in addition, the pancreas and liver secrete 
into the small intestine. The system is connected to the vascular, lymphatic, and nervous 
systems to facilitate regulation of the digestive response, delivery of absorbed compounds 
to organs of the body, and regulation of the food intake. 

One of the characteristic aspects of the GI tract is the presence of numerous 
endogenous microbes colonizing the surface of the GI tract throughout the life of the host. 
It consists of a complex community inside the host, known as the intestinal microbiota. 
In healthy adults, the intestinal microbial cells have been estimated to outnumber the 
host's somatic and germ cells by a ratio of 10:1 (4). The development of this microbiota is 
initiated during the birth process. The fetus exists in a sterile environment until birth. After 
being born, the infant is progressively colonized by bacteria from the mother's vagina and 
feces and from the environment. As long as nutrients and space are not limited, the 
commensals with high division rates predominate, e.g., enterobacteria (Escherichia coli) 
and Enterococcus appear. The succession of microbes in an infant' s intestinal tract also 
depends on the feeding mode. The fecal microbiota of breast-fed babies has been found to 
be relatively simple, usually exclusively dominated by Bifidobacterium (5). However, 
recent comparative studies showed that bifidobacteria were the predominate fecal bacteria 
in both group of infants (6,7). In bottle-fed infants, the count and frequencies of occurrence 
of Bacteroides, Enterobacteriaceae and streptococci were significantly higher than those 
in the breast-fed infants (6,7). After weaning, when solid food is consumed, the stools of 
infants begin to shift to an adult-like microbiota: bifidobacteria decrease remarkably and 
constitute only 5% to 15% of total microbes. The number of Bacteroidacecea, eubacteria, 
Peptococcaceae and usually Clostridia outnumber bifidobacteria, while aerobic bacteria 
such as E. coli and streptococci, which have been regarded as the predominant species are 
always detected, but account for less than 1% of the total bacterial count. Lactobacilli, 
Megasphaerae and Veillonellae are often found in adult feces, but the counts are usually 
less than 10 7 per gram of feces. By the end of the secondary year of life, the microbiota 
becomes more stable and resembles that of an adult (see also the chapter by McCartney 
and Gibson). As the microbial population increases nutrients become scarce and the 
intestinal niches become occupied with more specialized species with an advanced 
symbiotic relationship between the host and microbiota. Once the climax microbiota has 
become established, the major bacterial groups in the intestine of an adult usually remain 
relatively constant over time. 

The habitats of the intestinal microbiota vary in different parts of the human GI tract (8). 
In healthy persons, acid stomach contents usually contain few microbes. Immediately after 
a meal, counts of around 10 5 bacteria per milliliter of gastric juice can be recorded: 
bacteria including streptococci, enterobacteriaceae, Bacteroides and bifidobacteria 
derived from the oral cavity and the meal. The microbiota of the small intestine is 
relatively simple and no large numbers of organisms are found. Total counts are generally 
10 4 or less per milliliter, except for the distal ileum, where the total counts are usually 
about 10 6 /ml. In the duodenum and jejunum, streptococci, lactobacilli and Veillonellae are 
mainly found. Towards the ileum, E. coli and anaerobic bacteria increase in number. In 
the caecum, the composition suddenly changes and is similar to that found in feces, and the 
concentration may reach 10 11 per gram of content. 

As more than 400 species have been estimated to reside in the colon of healthy 

1 ^) 

adults, which may attain population levels nearly as high as 10 /g in the colon and may 
make up almost half the content by weight (8,9). This bacterial community is dominated 
by strict anaerobes, and contains less facultative anaerobes with a rate of anaerobes and 
aerobes as 1000:1. In accordance with the metabolic activity, the major bacteria present in 
the intestinal microbiota of the healthy adult can be divided roughly into three groups (10). 



The Intestinal Microbiota of the Elderly 77 

Group one is lactic acid-producing bacteria including Bifidobacterium, Lactobacillus and 
Streptococcus (including Enter ococcus), which may possess a symbiotic relationship with 
the host. Group two includes putrefactive bacteria such as Clostridium prefringens, 
Clostridium spp. Bacteroides, Peptococcaceae, Veillonella, E. coli, Staphylococcus and 
Pseudomonas aeruginosa. Others are like Eubacterium, Ruminococcus, Megasphaera, 
Mitsuokello, C butyricum and Candida, group three. Normally, near-stability exists in 
these habitats and each person has an individually fixed microbiota as far as qualitative 
composition is concerned. 

The intestinal microbiota play an important role in normal bowel function and 
maintenance of host health, through the formation of short chain fatty acids, modulation of 
immune responses, and development of colonization resistance (8,10). These functions of 
the intestinal microbiota are the consequence of the activities of the numerous intestinal 
bacteria as a whole community with a well-organized structure built on the balance among 
the various bacterial members. Therefore, the functions of the intestinal microbiota are 
very sensitive to factors that can alter the structure of the intestinal microbiota 
qualitatively and quantitatively such as aging, physiological state, disease, medication, 
diet, and stresses. 



Age-Altered Aspects of the Intestinal Microbiota 

Normal aging is associated with significant changes in the function of most organs and 
tissues, such as decreased taste thresholds, hypochlorhydria due to atrophic gastritis, and 
decreased liver blood flow and size (11). The GI tract is no exception, and there is 
increased evidence of impaired gastrointestinal function with aging (3,11-13). In the 
GI tract of the elderly, the age-related changes include decreased acid secretion by the 
gastric mucosa, and greater permeability of mucosal membranes which have been linked 
to increase in circulating antibodies to components of the intestinal microbiota in elderly 
subjects. Therefore, certain microbes which can take advantage of new ecological niches 
are assumed to become predominant inhabitants, leading to a dramatic shift in the 
composition of the gut microbiota upon age. 

Although the knowledge about the age-related alteration of the human intestinal 
microbiota is still limited, the structure of the intestinal microbiota in the healthy elderly 
has been suggested to be different from that of the healthy adults. This phenomenon is 
considered to be a result of aging, but it may also accelerate senescence. 

As early as in the 1960s, the scientific attention has been focused to characterize the 
intestinal microbiota of the elderly. In several works conduced in the different geographic 
regions, reduced presence of bifidobacteria was often observed in the fecal microbiota of 
the elderly compared to that of the healthy adults, as well as more putrefactive bacteria 
Enter obacteriaceae, Streptococcus, Staphylococcus, Proteus and C. perfringens (14-17). 

Mitsuoka and his colleagues (18,19) analyzed the composition of the intestinal 
microbiota in the various stages of life and observed an age-dependent change in the 
composition of the fecal microbiota. Bifidobacteria were less present in the fecal 
microbiota of senile (65- to 85-year-old) persons than in those of younger adults, while 
more lactobacilli and Clostridia were found in the fecal samples of the elderly. 

Mitsuoka and coworkers (20) compared the fecal flora of the elderly (61-95 years 
old) with healthy adults (31.8 + 6.6 years old) using optimized culture procedures for 
members of the anaerobic microbiota. Total bacterial count, Bidifobacterium Veillonella, 
Eubacterium were decreased significantly, whereas C perfringens and Lactobacillus were 
increased significantly. Furthermore, the frequencies of occurrence in Bifidobacterium, 



78 He 

Micrococaceae was decreased, while those of C perfringens, other Clostridium sp., and 
yeasts were increased significantly in the elderly compared with that of the healthy adults. 

Using the same method, Benno and Mitsuoka (21) did not find significant differences 
in Bacteroidaceae, Eubacterium, Peptostreptococcus and Megasphaera between the 
healthy elderly and healthy adults. However, Bifidobacterium (Bif. adolescentis and 
Bif. longum), Enterococcus were less in the elderly compared with the healthy adults, while 
lecithinase-negative Clostridium and C paraputrificum were increased in the elderly. 

Recently, non-culture-dependent molecular methods have been used to investigate 
the intestinal microbiota (22). The advent of these molecular methods, which do not rely 
on our ability to culture bacteria prior to quantification, allow additional information to be 
gained on the gut microbiota as a whole. Another method that allows ecological analysis 
without the need to culture the organism is that of community cellular fatty acid (CFA) 
analysis. Numerous environmental factors affect bacterial fatty acid synthesis, but certain 
signature fatty acids have been used to indicate the presence of specific groups of 
organisms in soil and marine environments, and have also been used to study community 
structure in human fecal samples. 

Direct polymerase chain reaction analysis was performed on elderly persons' fecal 
samples (22). Over 280 clones were generated and characterized by sequence analyses, 
providing a molecular taxonomic inventory. Phylogenetic analysis showed that the 
microbiota of the elderly was more diversified than that of younger adults. The proportion 
of unknown molecular species was very high among the clones derived from fecal samples 
of elderly persons. It is evident from this study that the fecal microbiota of the elderly 
person is very complex. The microbial diversity of the intestinal microbiota appears to be 
increased with age. This is in contrast to the microbial diversity of babies, which was 
found to be extremely low: only nine species were detected within each clone's library. 

Hopkins et al. (23) studied fresh fecal samples obtained from seven adults, five 
elderly individuals, and four geriatric patients diagnosed with C difficile-associated 
diarrhea. Selected fecal bacteria were investigated using viable counting procedures, 16S 
rRNA abundance measurements and community CFA profile. The principal micro- 
biological differences between adults and the elderly were the occurrence of higher 
numbers of enterobacteria and a lower number of anaerobe populations in the elderly 
group. Another important finding of this study was the lower number of bifidobacteria 
observed in the group of elderly patients. 

Hopkins and Macfarlane (24) isolated bacteria from fecal samples of healthy young 
adults, elderly subjects, and elderly patients with Clostridium difficile -associated diarrhea 
(CD AD). The isolated bacteria were identified to species level on the basis of their CFA 
profiles with Microbial Identification System (MIDI, Inc., Newark, DE) (MIDI). While 
Bacteroides species diversity increased in the feces of the elderly individuals, 
bifidobacteria diversity dramatically decreased with age. 

These observations indicate that aging may diminish bifidobacteria, and 
significantly increase Clostridia, including C. perfringens, and allow a slight increase of 
lactobacilli, streptococci, and enterobacteriacea. The total bacterial counts and 
anaerobes/aerobes in the intestinal microbiota of the elderly are relatively lower than 
those of the healthy infants and young adults. 

Historically, bifidobacteria have been considered to be the most important 
organisms for infants while lactobacilli, especially L. acidophilus, were considered the 
predominant beneficial bacterium for adults (5). However, bifidobacteria have recently 
been suggested to be more important throughout life as beneficial intestinal bacteria than 
lactobacilli (25,26). These ecological studies on the intestinal microbiota of the elderly 
indicate that bifidobacteria rather than lactobacilli are often decreased upon age. Although 



The Intestinal Microbiota of the Elderly 79 

some changes also happen upon age in other groups of bacteria, they are non-specific, not 
constant, and very individual. Furthermore, a decrease in bifidobacteria has often been 
observed in the intestinal microbiota of various young patients (23,27,28). Therefore, a 
decrease in bifidobacteria in the intestinal microbiota could be considered as an important 
hallmark for aging and disease of the human GI tract. 



BIFIDOBACTERIA IN THE ELDERLY 

Taxonomic Species Placement of Bifidobacteria! Microbiota with Age 

Bifidobacteria have been known since Tissier (5) first described a species from the feces of 
breast-fed infants, which was later named as Lactobacillus bifidus by Orla- Jensen (29). 
Since that time numerous studies have been published concerning the ecology and 
importance of bifidobacteria in the intestine of humans, especially in infants. 

A new taxonomic system was established by creating the genus Bifidobacterium 
with the description of several new species besides B. bifidum, which was the only existing 
species at that time (30,31). This was followed by an increasing number of new species 
isolated from humans and animals (32-35). The new concept of the genus Bifidobacterium 
taxonomy including 24 taxonomic species was summarized in special chapters of 
Bergey's Manual (1986). Currently, a total of 26 well-established taxonomic species have 
been described, among which are nine species which have been found to be exclusive 
residents of the human intestine. These are B. bifidum, B. longum, B. infantis, B. breve, 
B. adolescentis, B. angulatum, B. catenulateum, B. pseudocatenulatum, and B. dentium. 
Bifidobacteria appear between the 2nd and 5th day of life and continue to be one of the 
most numerous bacteria, amounting to about 10 10 /g of wet feces. Many studies indicated 
that each healthy adult has and maintains its own specific composition of Bifidobacterium 
microbiota during his/her life (33). 

Interestingly, the bifidobacterial composition of a human can progressively vary 
with aging, both qualitatively as well as quantitatively. The predominant species of 
bifidobacteria in the human GI tract can be differentiated to indicate various stages of life. 
However, lactobacilli, another of the important genus of endogenous bacteria considered 
to contribute to host health and well being, are changing only quantitatively, and do not 
express an apparent species diversity upon aging of the host. 

Bifidobacterium was found as one of the predominate bacteria in the intestinal 
microbiota of both infants and adults (19,32). However, the species and biotype 
composition of the fecal bifidobacteria progressively varied with increasing age. Species 
typical for infants were B. bifidium, B. infants, B. breve, and B. parvulorum. Typical for 
adults were four different biovars of B. adolescentis. B. bifidum and B. longum could often 
be found in both age groups, but in lower numbers. B. adolescentis biovar b was the 
most common Bifidobacterium in the microbiota of the elderly. The frequency of the 
occurrence of B. longum was 71% for infants, 62% and 33% for adults and the elderly, 
respectively. B. adolescents occurred 100%, 91%, and 79% in the elderly, adults, and 
infants, respectively. These results have been supported by studies conducted by other 
research groups (36,37). It was found that B. adolescentis and B. longum dominated the 
bifidobacteria of healthy adults, which is different from the bifidobacteria composition 
of infants. 

Mitsuoka (20) consistently observed that B. adolescentis biovar. b was significantly 
higher in the elderly, even when Bifidobacterium counts were similar among children, 
adults, and the elderly. The number of B. adolescentis and B. longum in healthy adults was 
significantly higher than those in aged persons. From 1829 fecal bacterial isolates from 15 



80 



He 



healthy adults, B. adolescentis, B. longum and B. bifidum were found to be the 
predominant species of bifidobacteria of these healthy adults (21). 

He and coworkers (38) isolated 51 Bifidobacterium strains from the feces of healthy 
adults (30-40 years old) and seniors (older than 70 years of age). The isolates were 
identified to species level based on the phenotypic characteristics. The isolates from the 
adults belonged to B. adolescentis, B. longum, B. infantis, B. breve, while those from the 
elderly were B. adolescentis and B. longum. 

Studies with molecular methods indicate a similar distribution of bifidobacteria 
species in the various stages of life. In a study using a non-culture-based method using 
PCR and denaturing gradient gel electrophoresis, B. adolescentis was found to be the most 
common species in feces of adult subjects as earlier indicated in the studies with traditional 
culture-based methods (39). 

Fecal bacteria from healthy young adults, elderly subjects, and elderly patients 
with CD AD were identified to species level on the basis of their CFA profiles with 
MIDI. Species diversity was found to decrease with age. B. angulatum was the most common 
bifidobacterial isolate in the healthy young adults. B. bifidum, B. catenulateum, 
B. pseudocatenulatum and B. infantis were not detected in the feces of the elderly 
subjects (24). 

Human Bifidobacterium species were identificated by Mullie and coworkers (40) 
with three multiplex PCRs. B. bifidum, B. longum and B. breve species were commonly 
recovered in infants, while B. adolescentis B. catenulateum/B. pseudocatenulatum and 
B. longum were predominant in adults. 

Matsuki and coworkers (41) applied species- and group-specific PCR directly to 
fecal samples and found B. catenulatum (B. catenulatum and B. pseudocatenulatum) 
in 92% of adult fecal samples and B. longum, B. adolescentis and B. bifidum in 65, 60, 
and 38% of the samples from adults, respectively. Comparison of species-species PCR 
method with the classical culture method revealed that some species, most frequently 
B. adolescentis, were detected by the direct PCR method but not by culturing followed 
by species-specific PCR of the isolates. 

The bifidobacteria in the intestinal microbiota of the healthy elderly is characterized 
by a reduced species diversity as well as quantitative decrease. The bifidobacteria in the 
elderly are characterized by B. adolescentis as the predominant species as well as a 
quantitative decrease within the whole intestinal microbiota. 



Mucus Adhesion of Bifidobacteria 

The reason for the age-related decrease in bifidobacteria numbers is still not well under- 
stood. Adhesion to the intestinal mucosa is regarded as a prerequisite for colonization by 
microbes and induction of the healthy promotion by them. It has therefore been proposed 
as one of the selection criteria for probiotic strains (42-45). 

Ouwehand and coworkers (46) tested four Bifidobacterium strains for adhesion to 
mucus isolated from subjects of different age groups including healthy newborns, 2- and 
6-month-old infants, adults (25-52 years) and elderly (74-93 years). The tested 
bifidobacteria adhered less to the mucus isolated from the elderly subjects compared to 
those from healthy infants and adults. The results suggest that the physiological condition 
of the mucus could be altered by aging, which can reduce the affinity spectrum of 
the mucus to bifidobacteria from various origins. This may be a factor involved in the 
decreased colonization of the elderly subjects by bifidobacteria and fewer species of 
Bifidobacterium present. 



The Intestinal Microbiota of the Elderly 81 

Twenty-four Bifidobacterium strains were examined for their ability to bind to 
immobilized human and bovine intestinal mucus glycoproteins (47). Each of the tested 
bacteria exhibited its characteristic adhesion to human and bovine fecal mucus. No 
significant differences were found among the taxonomic species. Among the tested 
bacteria, B. adolescentis, B. angulatum, B. bifidum, B. breve, B. catenulatum, B. infantis, 
B. longum and B. pseudocatenulatum adhered to human fecal mucus better than bovine 
fecal mucus, while the binding of B. animalis and B. lactis was not preferential. These 
results suggest that the mucosal adhesive properties of bifidobacteria may be a strain 
dependent feature, and the mucosal binding of the human bifidobacteria may be more host 
specific. 

Fifty-one Bifidobacterium strains were isolated from the feces of healthy adults 
(30-40 years old) and seniors (older than 70 years of age) and were tested for their ability 
to adhere to the mucus isolated from the healthy adults (30-40 years of age) (38). The 
strains isolated from healthy adults, and especially B. adolescentis, bound better to 
intestinal mucus than those isolated from seniors. These results indicate that the 
bifidobacteria isolated from the healthy elderly may pose a reduced affinity to the intestinal 
mucus from healthy adults. These results suggest that the poor colonization of 
bifidobacteria in the intestinal microbiota of the elderly may also be related to the 
development of a less adherent Bifidobacterium population as well as the reduced ability 
of mucus from this age group to facilitate Bifidobacterium adhesion. 

Laine and coworkers (48) investigated 30 Bifidobacterium strains isolated from the 
feces of the healthy elderly (> 80 years of age) Japanese and Finnish subjects. These 
strains were tested for their ability to adhere to the mucus only isolated from their own 
feces. The better mucus adhesion was observed in the combination of Bifidobacterium 
from the elderly and their fecal mucus rather than that of probiotic bifidobacteria from 
adults and the mucus from the elderly. 

The enhanced adhesion of B. adolecentis from the elderly to their mucus may, at the 
least, partly explain that B. adolescentis is a predominant species in the fecal 
Bifidobacterium microbiota. Therefore, there may be an advanced symbiotic relationship 
between B. adolescentis and the elderly. The replacement of the predominant species of 
bifidobacteria upon aging of the host may be one of the important events by which the 
intestinal microbes affect the homeostasis of physiological functions on the basis of 
the important contribution of bifidobacteria to human health and well being. 

Influence of Age-Related Decline in Immune Function and Influence 
on Intestinal Bifidobacteria! Microbiota 

Immunosenescence is defined as the state of deregulated immune function that contributes 
to the increased susceptibility of the elderly to infection and, possibly, to autoimmune 
diseases and cancer (49,50). When immunosenescence appears, the functional capacity of 
the immune system of the host gradually declines with age. The most dramatic changes in 
immune function with age occur within the T cells compartment, the arm of the immune 
system that protects against pathogens and tumors (51-54). The fact that T lymphocytes 
are more severely affected than B cells or antigen-presenting cells is mainly due to the 
involution of the thymus, which is almost complete at the age of 60. The host is then 
dependent on the T cells of various specificities, which eventually leads to changes in the 
T cell repertoire. CD45RA + "native" cells are replaced by CD45RA — "memory" cells, and 
a T cell receptor oligoclonality develops. At the same time, T cells with signal transduction 
defects accumulate. Age-related T cell alterations lead to a decreased clonal expansion 
and a reduced efficiency of T cell effectors functions, such as cytotoxicity or B cell 



82 



He 



functionality. Decreased antibody production and a shortened immunological memory are 
the consequence. Efficient protection of elderly individuals by suitable vaccination 
strategies is therefore a matter of great importance (51,55). Perhaps of greater 
consequence to interpretation of immunosenescence in the elderly is the decline in cell- 
mediated immunity (CMI). This is particularly important with respect to combating 
infectious disease, but also to tumor surveillance, since anti-tumor effects of the immune 
system are almost exclusively governed by the cell-mediated component. 

Interleukin (IL)-12 is a cytokine produced by mononuclear phagocytes and dendritic 
cells that serves as a mediator of the innate immune response to intracellular microbes and 
is a key inducer of cell-mediated immune responses towards microbes (56). IL-12 
activates natural killer (NK) cells, promotes interferon (IFN)-y production by NK cells 
and T cells, enhances cytotoxic activity of NK cells and cytolytic T lymphocytes, and 
promotes the development of TH1 cells. IL-10 and IL-12 are two cytokines secreted by 
monocytes/macrophages in response to bacterial products which have largely opposite 
effects on the immune system. IL-12 activates cytotoxicity and IFN-y secretion by T cells 
and NK cells, whereas IL-10 inhibits these functions. 

Many studies indicate that Gram-positive bacteria and their cell wall components 
are potent inducers of IL-12 for human monocytes, while the Gram-negative bacteria can 
promote more IL-10 (57-60). Karlsson and coworkers (61) also reported that Gram- 
positive bacteria B. adolescentis, Enterococcus fecalis, Lactobacillus plantarum, 
Streptococcus mitis can induce more IL-12 production by mononuclear cells from cord 
and adult blood compared to the gram-negative bacteria, Bacteroides vulgatus, 
Escherichia coli, Pseudomonas aeruginosa, Veillonella parvula and Nerisseria sicca. In 
contrast, more IL-10 was secreted by the stimulation of mononuclear from cord and adults 
with Gram-negative bacteria instead of gram-positive bacteria. 

Furthermore, He and coworkers (62,63) characterized the ability of bifidobacteria to 
affect the production of macrophage-like derived cytokines with a murine macrophage- 
like cell line, J774.1 (Fig. 1 and Table 1). B. adolescentis and B. longum, known as 
adult-type bifidobacteria, induced significantly more pro-inflammatory cytokine secretion, 
IL-12, and TNF-a by the macrophage-like cells than did infant-type bifidobacteria, 



c 



50.00 r 



40.00 - 



30.00 - 



20.00 - 



10.00- 



0.00 



□ Adult type bifidobacteria 
■ Infant type bifidobacteria 




p<0.01 



TNF-a 



IL-12 



Tested cytokine 



Figure 1 Cytokine production by a murine macrophage cell line J774.1 after exposure to adult- 
type bifidobacteria (B. adolescentis and B. longum) and infant-type bifidobacteria (B. bifidum and 
B. breve, B. infantis). Abbreviations: IL, interleukin; TNF, tumor necrosis factor. 



The Intestinal Microbiota of the Elderly 



83 





£2* 




1 




"oft 




c_ 




(N 




^H 




1 




l-H 




^ 




| 




W) 




Oh 




O 




^H 




1 




l-H 




^-^ 




£ 




W> 




3 




VO 




i 




l-H 




^^ 




| 




"oft 




sS? 




CQ. 




T-H 




d 






C 




•l-H 


/ y— v 


03 


1— * 


•fa 


1 


on 


W> 


g 


_C 


s 


« 


*»»«ii 


| 


k 


Ph 


vnj 


z 




H 


"Q 




o 




~v3 








CQ 




O 




■4— > 




<D 




5-H 

3 


g 


(Z3 


W> 


O 




Oh 

X 


o 


PJ 




?H 




<D 




<4-^ 




«c 




< 




<Z) 




i « 




^■^ 




<D 




u 




'-< 


d 


^t 


c. 


r-» 


a 


r-- 


•^ 


•— s 


H 


>^ 


■4— > 

OO 


X) 




C 




o 








•4-i 




<& 




u 




o 




D 




00 




C/3 


<d 


0) 


i 


• 1-H 


03 


M 


C 


o 


M 


-4—" 


<d 


>^ 


• ^H 


u 


o 

CD 




D- 




00 







(0 


d 


1- 


Z 



hw^tnoo)fn^H^ H H\qooH\q^f f iNt s ;oo(nn\o^;Ni s ; 
cocnrHcJriHNHrirHincnoo^o^^cndcoNdodcii-rHrHd 

+1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 

CN CN CN CN CN CN ^h^h^h^h ,— i CN cn CN 



cn rf o 



^ 



o i> o — cn m on 

^h f-- CN (N CN i— i i— i 

+1 +1 +1 +1 +1 +1 +1 

in oo in vo oo r- oo 



O <N (N 



. in 

VO VO VO On 
<N -h 



oc 



00 



r^ininr^cncnr^in 
■* ^ h \d n vi in <n 

+1 +1 +1 +1 +1 +1 +1 +1 
in cn vo in cn 



CN 



rf r- 



cn 



in 



voincnincN^t-r-vo 

h h ^ -t cn cn cn 



CN On cn vO On 00 CN 

^ i ^c^^o\a;in^qoofnrHio^fnQ'H'H| r jvqt^^a;f f |oo r j cs j 
^ododH^in'in'incoin'^oc'iHHciHr^tsHoit^Hoo^ONHH 

+1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 
ONCNcNO\cNONvor^^o\cncNcocNt^cnooovooNO^l-'xi-r---oocNvci 
voo^ooocnincNOoovooNcn»nocNcNcnvocn^cncno^vot^r^ 

md^^oidmONhain'oddhNWin^^hri^mooind 
r~cNr--'^--^-incncninin'^--HOcN'— ioONcnooooooor~- i or--oo'0 



•n ^ o in o 

H H rH H (S 

+1 +1 +1 +1 +1 

»n in r-; cn r-; 

CN 00 t-^ 00 CN 



00 


r- 


*-H OO 


m 


On 


On 


o 


— 


in cn 


00 


cn 


— H 


+1 


+1 1 


1 +1 +1 


+1 


+1 


+ 1 


00 


T— 1 


'sj- cn 


in 


T—t 


rh 


VO 


o 


in cn 


00 


T ~ ' 


On 



CN 

NO ^ t-^ 00 On 

in ^ cn cn cn 

+1 +1 +1 +1 +1 

cn cn cn oq r~; 

cn 'vj- -^j- cn — < 



^ 


CN 


00 


O- 


CN 


cn 


i— i 


CN 


+1 


+1 


+1 1 




f- 


^H 


ON 


^" 


m 


VO 


cn 


i 


' 


— -i 


i— i 


00 

a. 



Tt oo 



OO On 



^HOOONcncnoooocnooNNOin^i-ON'sfNO^© 



vo^HT^-cNCNcNcncNcNOcNin-^-'vt-^tTfcncncn'^i-'sf^tOcNCN^Hcn 

+1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 +1 



VO 



— -cncnvocNONcnvooocNONr--invo^i-in— ^ooNcncnvovo 
r-vocnoooo\OOooooN'^-r^cn — ooncnonvooo-^--— • 



— < 00 On 

in ^ oo 



cnONONOOONOoooo-^-O'^i- 
i—i cn cn cn ^ cn cn ^ — < 



cN-HOcnoocNr~-oooN— ■ 

NO)M(Shhhhh(n| 



vo in r- Ov o 
i-H i— i m cn , ^- 



3'3<££££<£££<£ £ £ £ £ £ £ 3 3 «S « «S 

C3 C3 "H -i-l --H .i-l •— --H .iH -iH O O •— •-* '-H •— O O O •— "H O W C5 •— •" "-I 

>^ >. .a .a .a .a .a .a .a .a «S ^2 >,>,>,>. ^2 <£ & >> >» «s >^ ^ .a .a .a 

•S -S bJ)0D0Dti00X)0i)ti00X)-^^-£'S-S-£ -^ ^ -^ -S « ->. -S -S OOODOO 

1313 <u <u i) i> d a> <u o .a ,a T3 ^ i 13 .a .a .a s s .a « s i> d <u 
ffiK<<<<<<<<QQffiKffiffiQQQKKQKffi<<< 



^t-inooo— -cnvor~oooN-^^^ooinvor~r~t^oovooor~-oooN^t-in'^i- 
oo^cNCNcNcncncncnooo-H^H^-.o-H^-ooooo-H^cN 
i^r--r-t^t^r^i^(^ONt--^H^H^--H^H^.cNCNCNONONVovovovovoin 
cNcNcNcNcNcNcNcNcNCNincncncncncncncncncNcNcNcNcNcNcNcn 

OUUUOUUOUOUUUUUOUOUUUUvJUUvJO 



6a 


6a 


6a 


6a 


>I 


6a 


6a 


6a 


6a 


6a 


*~- 


** 


v* 


+■* 


■*-* 


** 


*~- 


** 


•>-- 


*-i 


K 


e 


K 


s 


e 


C 


K 


C 


C 


K 


^j 


^ 


'i. 


^3 


^ 


^3 


^j 


*j 


^3 


^j 


VJ 


VJ 


VJ 


VJ 


VJ 


o 


VJ 


: ^ 


VJ 


VJ 


6a 


6a 


6a 


ua 


6a 


6a 


6a 


6a 


>i 


ua 


^ 


*) 


^j 


*J 


'^j 


^ 


^ 


'^ 


<*} 


<W 



A J5 Jvj ^ _o o 



"« "Q ~« "^3 "Q >• > 



Q "3 Q v^ "3 Q v3 Q Q Q Q-C5-Cj-Ci-C5-Ci-C5-C)-C>-5s 






&a 



6a 

:^2 






a s s s s 

60 50 OO Oo OO 

K s c s e 

VJ VJ VJ VJ VJ 



CQI^CQCQI^I^CQI^CQCQI^CQI^I^I^I^I^I^CQI^I^CQCQ 



2 *, 

& 2 

. o 

CQ U 



(Ncn^j-invor-oooNO 



cNcn^i-invor~-oooNO'-HCNcn'^-invor- 

hhhhhhhhMMOCJMMNM 



a- 

(N 



7D 



CN 



60 



r^ 



a. 
cn 
V 

a 

i 

HH 



7D 
C 



vo 

col 

i 

HH 

I— I 

•— 

a 



— - 
O 

— ' 

Q 



c3 
• — 



c 

— < 

o 

t__ 

CQ 

C 
'— 

O 
CD 

c 



C 

s 

CD -^ 

*-» 3 

^ •« ^ 

fa ° 

CD 5 M 



09 

C3 



•— 

W9 



13 
o 



cd L. -jo 

oc E; ^ 



6a 
K 

*3 



r^ 



84 



He 



m 



| 

o 

CO 

QJ 

• i-H 

o 

qj 

Oh 

o 

• i-H 

§ 

> 



QJ 

=1 

o 

Oh 
X 

W 

< 

CO 

U 



f- 

X) 

c 
o 

• i-H 

QJ 

o 

QJ 
C/3 

or 
QJ 
C 

• i-H 

o 



CM 

0) 
.Q 



(N 

i-H 



o 

i-H 






00 

w 
CO. 



J 



61) 
8 

Ph 

z 



c 

• i-H 

■<— ' 

CO 



QJ 

• i-H 

O 

QJ 
Oh 
CO 





V) 




Tt" 




• 

I-H 




On 




co 


I-H 


I-H 




+1 




+1 


+ 1 


+ 


on 


On 


^t 


,— i 


o 


CO 


CN 


^H 


VO 


^H 


On 


CN 


r^ 


^O 


(N 


NO 


r-» 


o 


i-H 




1-H 






CN 



On 



d °^ m' 



1 




+1 
CN 


i-H 


m 


i-H 








+ 1 


+1 -II 






m 


0> 

in 


NO 

in 


no 

CN 


m 

i-H 






^d 


CO 


i-H 
i-H 


On 


no 

CN 


| 


























(N 

i-H 














c3 


oq 

CO 

in 




oo 

CO 


On 
CO 


OO 

d 


m 


| 


+1 




+1 


+1 


+1 +1 




(N 

ON 

od 
o 


CO 

On 

On 


CN 

On 


CN 


no 
oo 


no 

• 

CN 


i-H 


i-H 










r "~ l 


fib 

a- 















b0 














D- 














f- 








Tt 






_^" 


o 




CO 


d 


1 — 1 


'-J 


1 


^ 




^r' 


i-H 


r^ 


r- 


W) 


+1 


| 


+1 h-I 


+1 -II 


Ph 


in 




^f 


VO 


m 


(N 


CO 


^o 




VO 


>n 


CN 


06 


V 


^ 




in 


i> 


On 




8 
1 

z 

H 

a 

(N 

i-H 


CN 










O 


O 


^T* 




NO 

• 


00 


m 

• 


CN 


l-H 


i-H 




^t 


CN 


O 


i-H 


NO 


+1 




+1 -II || || 


cd 


^O 


CN 


^ 


>n 


I-H 


O 


1 


CN 


O 


On 


O 


CO 


in 


J 


00 


2 


On 


\6 


On 


00 


l-H 


CN 






i-H 




CN 


-: Detectable limit foi 
r necrosis factor. 


o 


i-H 


m 


CO 


(N 


NO 

















an (SD); - 
TNF, turn 


























0) . . 














S .9 














*> -3 














* i 














T3 1h 














QJ S 














1/3 _- 














K 














a) .a 














*H 
















£ 












QJ >3 


Su 


03 










S K 




• »■« 


3 




i 


GO 


ts we 
iatio 


| 






£ 


K 

►s 


~£ ^ 


Q 


Q 


*0. 


"O. 


C/3 ~ 

0) "Q 


CQ 


CQ 


CQ 


CQ 


CQ 


CQ 





C^l 



The Intestinal Microbiota of the Elderly 



85 





OX) 




Oh 


a 


a 


•c 


o 


+-> 
O 
03 


— ' 

O 
D 


CO 


C/5 



3 

< 

4— » 

o 

c3 



•— 
3 
cc 


o- 

X 

PJ 

s- 
<L> 

<£ 
03 

CO 

a) 
U 

CN 

i 
C 

o 

u 

x> 

a 
_o 

<D 
'— 
O 

a) 

oo 

I 

>— < 

73 
3 

c3 
i 

73 

a 

c3 

OS 

U 

(N 
i 

C 

o 

03 

u 



"C 

4-J 
O 

c3 

CQ 
73 

O 

< 

4— ' 

o 

c3 



3 

_o 

"co 

<D 

73 

< 



CO 


.Q 

CO 



00 

I 



OD 

3 

o 

•<— I 

aj 

•— 
o 

i 



a) 

o 

73 

4—1 

o3 
_> 

■4— ' 

o 
cd 

c 



o 

1 

> 



co 

a) 

o 

CD 

4— l 

03 
_> 

'■4—' 

o 

o3 

3 



D 

O 

> 



>> 



■8 

> 

<D 
-C 
73 

< 



g 
'5b 
'G 
O 



E 

03 
d 

cD 

Oh 
C/3 



c 
o 



03 

•— 

*— i 

C/3 



o 


73 


in 


3 


co 




^^^ 


+■*> 


+1 




VO 


O 



CO 



CO — - 

vd in 
co r- 

+1+1 

CN O 

CO "~ 



+1 

CO 



o 

+1 

© 



r-- 

© 



+1 +1 



+1 + 

+ 



+1 +1 



+1 + 
+ 
+ 



+1 +1 +1 +1 



+1 



+1 + +1 
+ 



73 

C 

e 

_S-C 

'o3 
Q 



73 

C 

'o3 
Q 



73 

C 

_S-H 

'o3 

Q 



73 T3 

O O 

J-H '— 

c3 03 

Q Q 



73 
O 

,o 

S- 

03 

Q 



-a 

o 

,o 

<4-H 

S- 

03 

Q 



a) <u 

3 a 

4—1 ' 

C/5 <*) 

CD fli 

4— < 4— < 

c c 

O c 

03 03 

s s 



o 

C 

• pH 

4—1 



| 

3 

X 



d 

4—1 

o 



c 

03 
I 

X 



6<a 

I 






CO 
CO 

o 



65 

s 

I 



6? 

5j 



65 



65 
S 

65 






65 
65 



65 

s 

65 

S 

a 






s 



•s 



6rj 



6«] 



65 



c 

■ -H 



3 
73 

< 



65 



7§ 

55* 



CD 

d 

■ -H 
4—1 



3 
73 

< 



<D 







3 

•—> 




3 

• ^~ 
4—1 


C/3 


73 

O 


4—1 


3 


,0 


3 


•^H 


4h 


""" 


4—* 


>^ 


4—1 

r 


3 


S-H 


03 


73 


03 


«H-H 


< 


Q 


I— I 



*-- 
K 








^ 


^ 


65 




CJ 





"*^ 


65 


65 


65 


a 




■s 


■S 






Q 


a 


Q 



^J 



in 

co 

o 






•~J 



CN 

o 
o 
U 



Kj hj 



On CO 

O O 

Tt in 

o o 

U U 



«>J 



in 

o 



-J 



in 

o 



Kj Kj 



CO 

o 

CO 

in 



o 
o 



-J 



CN 

o 

o 



^ 



CO 

o 
o 



CQ CQ 



CQ 






Bo 






o 

in 

—1 

U 
U 

H 
< 



o o 

CN CN 

U U 

% % 



CQ CQ 



1 vO 

o o 

^h on 

in <N 

U U 



86 



He 



7? 

^< 

*-. 

s 

3 

•c 

j— - 

CJ 

PQ 

73 
O 

< 
O 

4-J 

o 

c3 



O 

CO 

O 

X 

W 



•— 


00 


s 


Oh 


r±i 


s *^ / 




d 




o 


CO 


•«-H 




■4— 1 


•— -^ 


<D 


<1) 


Vh 


u 


o 




CO 


CN 


VO 


O 

o 


1 


rt 


HH 


U 





C 

o 

4-J 

u 

00 

00 
i 

»— < 

73 
C 

c3 

N£> 

i 

H-l 

I— ( 

73 
C 

c3 

CO 

13 
U 

(N 

6 
o 

a 

U 



c3 

•c 

4—1 
O 

73 

o 

< 

CJ 

• y—t 

4— > 
CJ 



c 
o 

CO 
O 

< 



CO 
0) 

.Q 

re 



60 

O- 

d 
o 

•f-H 

4— < 

o 

■— 

o 

o 
;o 

00 
I 

-J 



CO 

13 

o 

-a 

CD 

4— < 

> 

4—1 

CJ 

d 



co 

13 

CJ 

CD 

03 

> 



CO 

13 

CJ 

73 

CJ 

4— I 

> 

•»-H 

4— < 

CJ 

C3 

d 



CO 

13 

CJ 

CD 

03 
> 



>. 



•S 

CJ 

> 

CO 

CJ 

-C 

•a 
< 



a 

•i-H 

W) 
s— 

o 



CJ 

E 

c3 

a 

CO 

CJ 

CD 

CJ 
Oh 

CO 



co 
en 

+1 



CN 

m 



in 

+1 

oo 

in 



no 
o 

+1 

in 

ON 



CO 

en 

+1 

ON 
ON 



o 


OO 






+1 


+ 


oo 


CN 


CN 


CN 

nC 



o 


o 


(N 




+1 


+1 


in 


r-- 


On 


oo 

<N 



+1 +1 



+1 + + 
+ + 



+1 + 
+ 



+1 +1 +1 +1 +1 +1 



CJ 

d 

■ -H 
4—1 

CO 

CJ 



c 

.03 



73 

O 

,o 

5^ 

!— 

03 

Q 



a) 

d 
• — 

4—1 

CO 
CJ 



3 
-a 

< 



(1) 




(D 


d 

4— < 

CO 
CJ 

■J * 


73 
O 


d 
• — 

co 

CJ 

4—1 


r 


O 


d 


. — 


<-f— i 


•■■" 


-4—* 


>> 


4—- 

r- 


d 




03 


73 


03 


'■4— 


< 


Q 


>— < 



0) CD 

d d 

4— I — ' 

CO CO 

CD CD 

4— ' 4— > 

d d 



d d 

<4— <4-< 

d d 



CD 

d 

■ -H 
4—1 

CO 



d 
.3 



CJ 

d 

■ -H 
4—1 

CJ 



d 
.03 



73 

O 

u 
03 

Q 



73 

O 

=42 

■— 
03 

G 



73 
O 

b 

03 

G 



CD 

4—1 

CD 



d 

d 



&5 



oq 



B 


g 


~ 


g 


g 


g 


g 


g 


a 


s 


s 


a 


S 


s 


s 


s 


c^ 


zc 


CC 73 


73 


'^3 


73 


'73 


--- 


a 
o 

">~1 










-C3 


-C) 


-Ci 


-Ci 


-Ci 


-G 


cq 


CQ 


CQ 


CQ 


CQ 


CQ 


CQ 



w 


^ 


^ 


^ 


'- 


■r- 


■r- 


^ 


^ 


^ 


^ 


^ 


^. 


k, 


k, 


k, 


-Ci 


-C} 


-Ci 


-C} 



s 

■S 

s 

ft, 



CQ CQ CQ CQ 



i 


OO 


r- 


oo 


On 


—- < 


oo 


in 


NO 


o 


6 
d 


o 


o 


o 


O 


O 


o 


— 


— — 


— — 


ON 


vo 


V£> 


N£> 


— H 


— 


— 


^h 


— — 


(N 


(N 


(N 


(N 


en 


en 


en 


en 


en 


d 


U 


U 


u 


U 


U 


U 


U 


U 


U 


C3 


§ 


S 


S 


^ 


s s 


s s 


S 


/) 


H 


H 


H 


h- 


H 


H 


H 


H 


H 



o 

en 
U 



H 



CN 

en 



oo 
en 



U U 



H 



H 



On 
CN 

en 

U 



CQ 



H 

o 

o 

CN 



U 



CO 

s 








C^ 








•^H 








g 








b0 








« 








c 








c 








s- 


# 






CJ 


o 








o 

CM 






'-4-H 

c 


d. 






o 






C 


— 






c 








• ^H 








4— 1 


^" "N 






CJ 

_CJ 


+ 






O 


+ 






u 


+ 






CJ 


N ^ 






CO 


. r 






CJ 


O 






d 


in 






od 


l 






Dh 


i 






03 


CN 






S 


+ 






u 


+ 






d 


N ' 






c 


O 






4-i 


CM 






u 


I 






CJ 
















^■^ 








O 


X~s 






u 


+1 






CJ 


^s 






I- 




# 




s 


O 




w 


3 

u 


^ 


E 


. — 


CJ 

o- 


CO 


d 
o 


d 


H 


73 

13 

C4d 


4—> 

CJ 
CJ 

4— ' 


o 

4— i 

CJ 
CJ 


c3 

CJ 

• ^H 


CJ 

"B. 


CJ 

3 


4— • 

CJ 

3 


CJ 

E 


c 

CJ 
c* 


| 


| 


< 


c 

Uh 


«ao 

D- 


"oo 


>. 


CJ 


Q- 


U 
U 


1 


c5 


O 




d 

CJ 


V 


V 



6 

u 



CO 

c 
-d 

CJ 

E 
o 

73 
co d 

73 O 
O <N 

Ph .S 

^3 CD 
^5 cj 



CN 
i 

C 
CJ 

c3 



.d 
co 

c3 

d r ^ 

^ 2 

t" 1 -a 

*8 § 

CD -d 

CD CD 

~d O 

3 -a 

MM 



Q Q 

c S 

e3 03 

CD CD 

s s 

co co 

o3 03 

73 TD 

CD CJ 

co co 

co co 

CD CJ 

9- & 

X X 

CD CD 

co co 

03 03 

CO CO 

13 13 

CD CJ 

CN CN 



d 
d 

CJ 



O O 
CJ o 

03 03 CD 

u u -s 

^ X) j 

d | -' 

o •• 



d 
O 



CD CD 
CD CD 



cd cj -a 



co co 
\C OO 






The Intestinal Microbiota of the Elderly 87 

B. bifidum, B. breve, and B. infantis. In contrast, B. adolescentis did not stimulate the 
production of anti-inflammatory IL-10 as the other tested bacteria did. At the same time, 
neither the adult-type nor the infant-type bifidobacteria were found to be likely to 
trigger inflammatory responses in human enterocytes (Table 2) (64). The results suggest 
that the adult-type bifidobacteria, especially, B. adolescentis, may be more potent to 
amplify but less able to down-regulate the inflammatory responses (Table 3). 

The intestinal microbiota of elderly people is in general different from those in 
infants; the former is more diverse and stable (8). One of the distinct age-related events 
in intestinal microbiology is the increase in numbers of facultative anaerobic Gram- 
negative bacteria (8); these bacteria may be opportunistic infective agents. They have been 
found to be the triggers of anti-inflammatory cytokine-production by macrophages and 
monocytes (60,61). The anti-inflammatory effects of these bacteria are believed to be one 
of the strategies required for their successful colonization of the host's intestine, 
overcoming the natural defense barrier, including inflammation. Therefore, an increase in 
bacteria, including bifidobacteria, which can enhance the intestinal inflammatory response 
in aged people, can be considered beneficial to counterbalance the age-related changes in 
their intestinal microecology. This may contribute to the homeostasis of the local 
immunity by preventing local inflammation from being oversuppressed. These results 
suggest that the dominance of the intestinal bifidobacteria by B. adolescentis may be one 
of the events in the intestinal environment in response to the aging of the host. 

These results can lead to a hypothesis that the age-related changes of the 
predominant species of bifidobacteria in the human intestine is a kind of well-acquired 
adaptation of the host to the changes in the intestinal microbiota, localizing the beneficial 
microbes such as B. adolescentis to enhance the colonization resistance against the 
exogenous infectious agents. For more information on the influence of the normal 
intestinal microbiota on the immune system, see the chapter by Moreau. 

Effects of Bifidobacteria! Probiotics on Immunosenescence 

Probiotics have been defined as a live microbial food ingredient that are beneficial to the 
health of the host (65,66). Most current probiotics are lactic acid bacteria, especially 
Lactobacillus and Bifidobacterium species (66). Among the proposed health-promoting 
effects of the probiotic strains are the enhancement of cell-mediated immune responses of 
the host by stimulating the pro-inflammatory cytokine, particularly IL-12 (67,68). The 
cell-mediated immune response, enhanced by the pro-inflammatory cytokine IL-12, has 
been considered as one of the most important underlying mechanisms contributing to the 
self-defense of the host a against tumors and allergy (56). Therefore, probiotics strains 
with the ability to stimulate IL-12 secretion can exhibit apparent anti-tumor and anti- 
allergic effects (69-71). Considering the fact that the reduced cell-mediated immune 
response is the main component of immunosensence of the elderly, such probiotics can 
be expected to benefit the elderly. After consumption of a probiotic B. lactis, increase in 
the proportion of the total CD + 4 and CD25 T lymphocytes and NK cells in the blood 
were observed (72,73). The ex vivo phagocytic and tumoricidal activity capacity of 
polymorphonuclear and mononuclear cells were increased by an average of 101 and 62%, 
respectively. These increases were significantly correlated with age, with volunteers 
older than 70 years experiencing significantly greater improvement than those younger 
than 70. In sight of the fact that the intestinal bifidobacteria are usually dominated by 
B. adolescentis with an advanced affinity specific to the mucus from the elderly and the 
ability to promote IL-12 production, B. adolescentis from the intestine of the healthy 
elderly may be a more reasonable candidate for use as probiotics to help the seniors to 



88 



He 



combat immunosenescence. Compared to other predominant species of bifidobacteria in 
infants and young adults, B. adolescentis is usually less quantitatively. Therefore, a 
strategy to increase senior-specific bifidobacteria, including B. adolescentis in the elderly 
could be a more practical way to improve the immunomodulatory effect of the intestinal 
flora. For more information on probiotics, see the chapter by Ouwehand and Khedkar. 



CONCLUSION 

With the progress in nutrition and medicine, the life-expectancy of people has increased. 
In industrialized societies this has led to increasing costs and spending for health care and 
medical treatment of their senior citizens. Growing scientific evidence suggests that aging 
alters the intestinal microbiota qualitatively and quantitatively, generating a different 
microbial community with an aberrant structure. The intestinal microbiota in the elderly is 
colonized by fewer bifidobacteria, and more potentially infectious microbes compared to 
infants and young adults. Furthermore, there is a decrease in the species diversity in 
bifidobacterial population of the elderly which is dominated by Bifidobacterium 
adolescentis and B. longum. The advanced affinity of B. adolescentis to mucus both 
isolated from the elderly suggests a deep symbiotic relationship between this microbe and 
host. The elevated ability of B. adolescentis to enhance the production of pro- 
inflammatory cytokine, particularly IL-12, by macrophages and monocytes suggests 
that this endogenous bacterium may play an important role in the maintenance of the CMI 
which can be impaired by age-related immunosenescence. This evidence can be used as 
the basis to consider B. adolescentis from the healthy elderly as a reasonable probiotic 
candidate for targeting the elderly, a growing subpopulation more prone to infection and 
autoimmune disease. 



REFERENCES 

1. Saunier K, Dore J. Gastrointestinal tract and the elderly: functional foods, gut microflora and 
healthy ageing. Dig Liver Dis 2002; 34:19-24. 

2. Kalache A. Active ageing makes the difference. Bull. WHO 1999; 77:299. 

3. Schneeman BO. Gastrointestinal physiology and function. Br J Nutr 2002; 88: 159—163. 

4. Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 1997; 
31:107-133. 

5. Tissier H. Recherches sur la flora intestinal des nourrisson. Theses Paris 1900; 1-253. 

6. Benno Y, Sawada K, Mitsuoka T. The intestinal microflora of infants: composition of fecal 
flora in breast-fed and bottle-fed infants. Microbiol Immunol 1984; 28:975-986. 

7. Yuhara T, Isojima S, Tsuchiya F, Mitsuoka T. On the intestinal flora of bottle-fed infant. 
Bifidobact Microflora 1983; 2:33-39. 

8. Mitsuoka T. A Color Atlas of Anaerobic Bacteria. Tokyo: Sobunsya, 1980. 

9. Mitsuoka T. Intestinal flora and aging. Nutr Rev 1992; 50:438-446. 

10. Mitsuoka T. Significance of dietary modulation of intestinal flora and intestinal environment. 
Biosci Microflora 2000; 19:15-25. 

11. Russell RM. Changes in gastrointestinal function attributed to aging. Am J Clin Nutr 1992; 
55:1203S-1207S. 

12. Gill HS, Darragh AJ, Cross ML. Optimizing immunity and gut function in the healthy. J Nutr 
Health Aging 2001; 5:80-91. 

13. Holt PR. Gastrointestinal disease in the elderly. Curr Opin Clin Nutr Metab Care 2003; 
6:41-48. 



The Intestinal Microbiota of the Elderly 89 

14. Haenel H. Uber die Mikrookologie alter Menschen. Zentralbl Bakteriol Hyg I Orgi 1963; 
188:219-230. 

15. Haenel H. Gesetzma(3igkeiten in der Zusammensetzung der fakalen Mikroflora. Eubiose und 
Dysbiose der menschlichen Darmbesiedlung. Ernahrungsforschung 1965; 10:289-301. 

16. Jantea F, Nicolae D, Bad-Oprisescu D, Voina P. Beitrag zur Darmmikroflora bei Menschen im 
Alter von 45-100 Jahren. Ernahrungsforschung 1965; 10:352-362. 

17. Gorbach SL, Nahas L, Lerner P, Weinstein L. Studies of intestinal microflora 1. Effects of diet, 
age, and periodic sampling on number of fecal microrganisms in man. Gastroenterology 1967; 
53:845-855. 

18. Mitsuoka T, Hayagawa K. Die Faecalflora bei Menschen I: Mitteilung: Die Zusanmmenset- 
zung der Faecalflora der verschiedenen Altersgruppen. Zbl Bakt Hyg I Abt Org 1972; 
223:333-342. 

19. Mitsuoka T, Hayagawa K, Kimura N. Die Faekalflora bei Menschen II: Mitteilung: Die 
Zusammensetzung der Bifidobakterienflora der verschiedenen Altersgruppen. Zbl Bakt Hyg I 
Abt Org 1973; 226:469-478. 

20. Mitsuoka T, Ohno K, Benno Y, Suzuki K, Namba K. Die Faekalflora bei Menschen IV. 
Mitteilung: Vergleich des neu entwickelten Verfahrens mit dem bisherigen ublichen Verfahren 
zur Darmfloraanalyse. Zbl Bakt Hyg I Abt Org 1976; 224:219-233. 

21. Benno Y. Mitsuoka Development of intestinal microflora in human and animal. Bifdobact 
Microflora 1986;5:13-25. 

22. Blaut M, Collins MD, Welling GW, Dore J, van Loo J, de Vos W. Molecular biological 
methods for studying that gut microbiota: the EU human gut flora project. Br J Nutr 2002; 
87:S203-S211. 

23. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related changes in intestinal bacterial 
population assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid 
profiles. Gut 2001; 48:198-205. 

24. Hopkins MJ, Macfarlane GT. Changes in predominant bacterial population in human faces 
with age and with Clostridium difficile infection. J Med Microbiol 2002;448-454. 

25. Hopkins MJ, Sharp R, Macfarlane GT. Variation in human intestinal microbiota with age. Dig 
Liver Dis 2002; 34:512-518. 

26. Mitsuoka T. Intestinal Bacteria and Health. Japan: Harcourt Brace Jovanovich, 1978. 

27. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic Estonian and 
Swedish 2-year-old children. Clin Exp Allergy 1999; 29:342-346. 

28. Kuvaeva IB, Orlova NG, Veselova OL, Kuznezova GG, Borovik TE. Microecology of the 
gastrointestinal tract and the immuneological status under food allergy. Nahrung 1984; 
28:689-693. 

29. Orla-Jensen S. La classification des bacteria lactiques. Lait 1924; 4:468-474. 

30. Reuter G. Vergleichende Untersuchungen uber die Bifidus-Flora im Sauglings- und 
Erwachsenenstuhl. Zbl Bakt I Orig 1963; 191:486-507. 

31. Reuter G. Designation of type strains for Bifidobacterium-species. Int J Syst Bact 1971; 
21:273-275. 

32. Mitsuoka T. Vergleichende Untersuchungen iiber die Bifidobakterien aus dem Verdauung- 
strakt von Menschen und Tieren. Zbl Bakt I Orig 1969; 210:52-64. 

33. Mitsuoka T. Taxonomy and ecology of bifidobacteria. Bifidobacteria Microflora 1984; 
3:11-28. 

34. Lauer E, Kandler O. DNA-DNA homology, murein type and enzyme patterns in the type 
strains of the genus Bifidobacterium. Syst Appl Microbiol 1983; 4:42-62. 

35. Scardovi V. Genus Bifidobacterium. Orla-Jensen 1924. In: Sneath et al., eds. Bergey's Manual 
of Systematic Bacteriology: Vol. 2 Section 15, Philadephia: The Williams & Wilkins 
Company, 1986; 1418-1434. 

36. Moore WE, Holdeman LV. Human fecal flora: the normal flora of 20 Japanese-Hawaiians. 
Appl Microbiol 1974; 27:961-979. 

37. Finegold SM. Normal human intestinal flora. Ann 1st Super Sanita 1986;73 1-737. 



90 



He 



38. He F, Ouwehand AC, Isolauri E, Hosoda M, Benno Y, Salminen S. Differences in composition 
and mucosal adhesion of bifidobacteria isolated from healthy adults and healthy seniors. Curr 
Microbiol 2001; 43:351-354. 

39. Satokari RM, Vaughan EE, Akkermans ADL, Saarela M, de Vos WM. Bifidobacterial diversity 
in human feces detected by genus-speciic PCR and denaturing gradient gel electrophoresis. 
Appl Environ Microbiol 2001; 67:504-513. 

40. Mullie C, Odou MF, Singer E, Romond MB, Izard D. Multiplex PCR using 16 S rRNA gene- 
targeted primers for the identifyication of bifidobacteria from human origin. FEMS Microbiol 
Lett 2003; 222:129-136. 

41. Matsuki T, Watanabe K, Fujimoto J, et al. Development of 16 S rRNA-gene-targeted group- 
species primers fro the detection and identification of predominant bacteria in human feces. 
Appl Environ Microbiol 2002; 68:5445-5451. 

42. Isolauri E, Stitas Y, Kankaanpaa P, Arvilommi H, Salminen S. Probiotics: effects on immunity. 
Am J Clin Nutr 2001; 73:444S-450S. 

43. Ouwehand AC, Tuomola EM, Lee YK, Salminen S. Microbial interactions to intestinal 
mucosal models. Methods Enzymol 2001; 337:200-212. 

44. Kohler H, McCormick BA, Walker WA. Bacterial-enterocyte crosstalk: cellular mechanisms 
in health and disease. J Pediatr Gastroenterol Nutr 2003 Feb; 36:175-185. 

45. Salminen S, Laine M, von Wright A, Vuopio-Varkila J, Korhonen T, Mattila-Sandholm T. 
Development of selection criteria for probiotic strains to acess their potential I n functional 
food. A Nordic and European approach. Biosci Microflora 1996; 2:23-28. 

46. Ouwehand AC, Isolauri E, Kirjavanen PV, Salminen SJ. Adhesion of four Bifidobacterium 
strains to human intestinal mucus from subjects in different age groups. FEMS Microbiol Lett 
1999; 172:61-64. 

47. He F, Ouwehand AC, Hashimoto H, Isolauri E, Benno Y, Salminen S. Adhesion of 
Bifidobacterium spp. to human intestinal mucus. Microbiol Immunol 2001; 45:259-262. 

48. Laine R, Salminen S, Benno Y, Ouwehand AC. Performance of bifidobacteria in oat-based 
media. Int J Food Microbiol 2003; 83:105-109. 

49. Pawelec G. Immunosenescence: impact in the young as well as the old? Mech Ageing Dev 
1999; 108:1-7. 

50. Mishto M, Santoro A, Bellavista E, Bonafe M, Monti D, Franceschi C. Immunoproteasomes 
and immunosenescence. Ageing Res Rev 2003; 2:419-423. 

51. Ginaldi L, Martinis MD, D' Ostilio A, Marini L, Loreto MF, Quaglino D. Immunological 
changes in the elderly. Ageing Clin Exp Res 1999; 11:281-286. 

52. Lesourd B, Mazari L. Nutrition and immunity in the elderly. Proc Nutr Soc 1999; 58:685-695. 

53. Castle SC. Clinical relevance of age-related immune dysfunction. Clin Infect Dis 2000; 
31:578-585. 

54. Effros RB. Ageing and immune system. Novartis Found Symp 2001; 235:130-145. 

55. Grubeck-Loebenstein B. Changes in the ageing immune system. Biologicals 1997; 
25:205-208. 

56. Peakman M, Vergani D. Basic and Clinical Immunology. Hong Kong: Churchill Livingstone, 
1997:1-388. 

57. Fujimoto T, Duda RB, Szilvasi A, Chen X, Mai M, O'Donnell MA. Streptococcal preparation 
OK-432 is a potent inducer of IL-12 and T helper cell 1 dominant state. J Immunol 1997; 
158:5619-5629. 

58. Haller D, Blum S, Bode C, Hammes WP, Schiffrin EJ. Activation of human peripheral blood 
mononuclear cells by nonpathogenic bacteria in vitro: evidence of NK cells as primary targets. 
Infect Immun 2000; 68:752-759. 

59. Hessle C, Honson LA, Wold AE. Lactobacilli from human gastrointestinal mucosa are strong 
stimulators of IL-12 production. Clin Exp Immunol 1999; 116:276-282. 

60. Hessle C, Andersson B, Wold AE. Gram-positive bacteria are potent inducers of monocytic 
interleukin-12 (IL-12) while gram- negative bacteria preferentially stimulate IL-10 production. 
Infect Immun 2000; 68:3581-3586. 



The Intestinal Microbiota of the Elderly 91 

61. Karlsson H, Hessle C, Rudin A. Innate immune responses of human neonatal cells to bacteria 
from normal gastrointestinal flora. Infect Immun 2002; 70:6688-6696. 

62. He F, Morita H, Ouwehand AC, et al. Stimulation of the secretion of pro-inflammatory 
cytokines by Bifidobacterium strains. Microbiol Immunol 2002; 46:781-785. 

63. He F, Isolauri E, Morita H, et al. Bifidobacteria isolated from allergic and healthy infants: 
differences in taxonomy, mucus adhesion and immune modulation effects. Microecol Ther 
2002;29:103-108. 

64. Morita H, He F, Fuse T, et al. Adhesion of lactic acid bacteria to Caco-2 cells and their effects 
on cytokine secretion. Microbiol Immunnol 2002; 46:293-297. 

65. Fuller R. Probiotics in man and animals. J Appl Bacteriol 1989; 66:365-378. 

66. Salminen S, Ouwehand A, Benno Y, Lee YK. Probiotics: how should they be defined? Trends 
Food Sci Technol 1999; 10:107-110. 

67. Gill HS. Stimulation of the immune system by lactic culture. Int Dairy J 1998; 8:535-544. 

68. Borchers AT, Keen CL, Gershwin ME. The influence of yogurt/ 'Lactobacillus on the innate and 
acquired immune response. Clin Rev Allergy Immunol 2002; 22:207-230. 

69. Matsuzaki T, Yamazaki R, Hashimoto S, Yokokura T. The effect of oral feeding of 
Lactobacillus casei strain Shirota on immunoglobulin E production in mice. J Dairy Sci 1998; 
81:48-53. 

70. Aso Y, Akaza H, Kotake T, Tsukamoto T, Imai K, Naito S. BLP Study Group. Preventive 
effects of a Lactobacillus casei preparation on the recurrence of superficial cancer in a double- 
blind trial. Eur Urol 1995; 27:104-109. 

71. Cross ML, Gill HS. Can Immunoregulatory lactic acid bacteria be used as dietary supplements 
to limit allergies? Int Arch Allergy Immunol 2001; 125:112-119. 

72. Gill HS, Rutherfurd KJ, Cross ML. Dietary probiotics supplementation enhances natural killer 
cell activity in the elderly: an investigation of age-related immunological changes. J Clin 
Immunol 2001; 21:264-271. 

73. Gill HS, Rutherfurd KJ, Cross ML, Gopal PK. Enhancement of immunity in the elderly 
supplementation with the probiotic Bifidobacterium lactis HN019. Am J Clin Nutr 2001; 
74:833-839. 



5 



Immune Modulation by the Intestinal 
Microbiota 



Marie-Christiane Moreau 

French Institute of Agronomical Research (INRA), Nancy, France 



INTRODUCTION 

Today, there is a growing interest in the intestinal microbiota and its relationship with the 
host's immunity. This is mainly due to two causes: first, the results obtained with 
probiotics, which have been defined as live micro-organisms that confer a health benefit on 
the host when consumed in adequate amounts (1), have shown interesting immunomo- 
dulatory properties in humans (1-3). Second, the studies by Dutchmann and coworkers 
(4) demonstrated for the first time, some years ago, that we are tolerant to our own 
digestive flora. A breakdown of this state leads to inflammatory bowel diseases (IBD). 
Consequently, the digestive flora can be considered as an organ belonging to the host's just 
as the spleen, heart, or brain. It plays an important role in the host's protection, especially 
by its actions on the immune system. 

The overall importance of the intestine, relating to health, is still not completely 
understood. It is an extremely complex organ, which has to assure the function of 
digestion of foods and absorption of nutrients. In addition to this, the intestine is the 
largest lymphoid organ in the body by virtue of lymphocyte number and quantity of 
immunoglobulin produced. It also harbors a huge reservoir of bacteria that colonize it 
very early after birth and which is called "the commensal or resident or autochthonous 
digestive microflora," and more recently the "intestinal microbiota." The relationships 
between the intestinal microbiota and intestinal immune system (IIS), described in some 
reviews (5-7) can be viewed in terms of "symbiosis" or "mutualism," which is the 
association of symbiosis and commensalism as explained by Hooper and Gordon (8). 
Indeed the IIS does not mount immune responses toward the intestinal microbiota that, in 
turn, exert many effects on the immune system. These effects can be characterized as 
activation, modulation, and regulation of immune responses and are effective at both 
intestinal and peripheral levels. 

In this chapter, effects of the intestinal microbiota on the host's immunity will be 
described, and in some cases effects of probiotic bacteria will also be discussed. 

93 



94 Moreau 

BRIEF REVIEW OF THE INTESTINAL MICROBIOTA 

From birth to death, the gut is colonized by a diverse, complex, and dynamic bacterial 
ecosystem that constitutes the intestinal microbiota. In newborns, it develops sequentially 
according to the maturation of intestinal mucosa and dietary diversification. In healthy 
conditions, the human baby's intestine is sterile at birth but, within 48 hours, 10 to 10 
bacteria can be found in 1 g of feces (9-11). The bacteria colonizing the baby's intestine 
come from the environment, where maternal vaginal and fecal microbes represent the 
most important source of bacterial contamination. However, the infant conducts an initial 
selection, since, out of all the bacteria present, only the facultative anaerobic bacteria such 
as Escherichia coli and Streptococcus will be able to colonize the intestinal tract, whatever 
the diet. Conditions under which this initial selection is operated have yet to be fully 
elucidated. They are related to endogenous factors, such as maturation of intestinal 
mucosa, mucus, growth promoters or inhibitors present in the meconium, or exogenous 
factors such as delivery conditions (natural childbirth, caesarean section), mother's status 
(antibiotic intake), and quality of the bacterial environment. Subsequently, obligate 
anaerobes such as Bacteroides, Clostridium, and Bifidobacterium colonize over the first 
week of life, following a second selection in which the diet factor plays a fundamental 
role. It has long been known that Bifidobacteria are predominant in exclusively breast-fed 
babies, while in bottle-fed babies it is not always present, or present at fluctuating levels 
and, in contrast to breast-fed babies, often associated with other anaerobic bacteria such as 
Bacteroides and Clostridia. Breast milk contains oligosaccharides enabling development 
of Bifidobacterium and may also function as receptor analogues of the mucus influencing 
the strains able to colonize the intestinal tract (12). A bacterial balance is obtained towards 
the end of the second week of life in which Bifidobacterium and E. coli predominate in 
exclusively breast-fed infants, while a more diverse microbiota, rich in E. coli, 
Bacteroides and, possibly Clostridium, Bifidobacterium, Staphylococcus, and other 
Enterobacteriaceae, is found in formula-fed infants. Thus the bacterial balance of the 
infantile microbiota mainly depends on two important factors: bacterial environment at 
birth and diet. During the last decade, some modifications of the microbiota balance in 
babies whatever the feeding have been observed, namely, dominance of Staphylococcus, 
low levels of E. coli, delayed colonization with anaerobic bacteria and absence or low 
levels of bifidobacteria (MJ Butel, personal communication). Excessive aseptic conditions 
present at birth, maternal antibiotic intake immediately before parturition or during 
childbirth could be, among other factors, responsible for such differences (13). Because of 
the fragility of the baby's digestive microbiota, which is poorly diversified, with about 10 
bacterial species of micro-organism versus over 400 in adults, the consequences of its 
modification have to be considered in terms of health. For example, recent studies suggest 
that some infancy pathologies, such as food allergy, could be due to the modifications of 
the intestinal microbiota of newborns. The latter will be discussed in chapter 10. 

Thereafter, according to dietary diversification, the digestive microbiota, enriched 
by the development of other strictly anaerobic bacteria, becomes more and more complex. 
It is considered to have assumed adult characteristics toward the age of 2 years (9-11). 

In adults, a complex and diverse digestive microbiota is present, mainly in the distal 
parts of the gut. In the duodenum, the number of bacteria is approximately 10 4 bacteria/g 

"7 R 

of intestinal content while in the ileum, the number reaches 10 -10 . The large intestine is 
the most densely colonized region (10-10 bacteria/g of content), essentially because of 
digestive stasis. Bacterial species established at levels over 10 -10 bacteria/g 
characterize the predominant microbiota, whereas those below such a threshold compose 
the subdominant microbiota. In fact, it is proposed that only predominant bacteria are able 



Immune Modulation by the Intestinal Microbiota 95 

to exert a measurable function. The dominant microbiota of human feces is mainly 
composed of strict anaerobic and extremely oxygen-sensitive bacteria. According to 
several authors, 30% to 70% of the microbiota is not identified because it is uncultivable 
with current techniques. The predominant species commonly isolated from the human 
feces belong to the genera Bacteroides, Eubacterium, Bifidobacterium, Ruminococcus, 
and Clostridium, and subdominant species include enterobacteria, particularly Escherichia 
coli and streptococci (14). Lactobacilli are frequently subdominant in humans or cannot 
even be detected. Some studies suggest that they may be abundant in the ileum. 

Very little data exists on the evolution of intestinal microbiota in the elderly. 
Nonetheless, bifidobacteria have been reported to decrease at old age, which may be 
related to a reduced adhesion to the intestinal mucus (15). 

Currently available molecular biology techniques should bring additional and 
complementary approaches to those offered by the usual culture techniques. Recent 
molecular methods have shown that every individual has his/her own gut microbial 
balance, which has been described to be stable (studies over a period of 6-9 months) (16). 

In conclusion, depending on the intestinal sites (duodenum, ileum, and colon) and 
the various periods of life, childhood, adulthood, aging, the human's intestinal microbiota 
also varies. This is discussed further in more detail in chapter 3 by McCartney and Gibson. 



BRIEF REVIEW OF IMMUNE RESPONSES 

Innate Immunity 

Cells responsible for the innate immunity provide the first line of host defense: monocytes/ 
macrophages, dendritic cells (DC), natural killer cells (NK), and neutrophils. These are the 
body's sentinels, able to detect danger and signal it to other cells, by the synthesis of 
molecules, such as NO (nitric oxide) which displays antibacterial activity, cytokines, and 
chemokines, which are small peptides acting by means of specific receptors expressed at 
the surface of targeted cells. Some of them have pro-inflammatory properties, and increase 
expression of surface markers on some cells allowing migration into neighboring 
lymphoid organs. DCs and macrophages are able to display a phagocytic activity, and by 
production of inflammatory chemokines and cytokines, to modulate other cells such as 
neutrophils, polymorphonuclear cells and eosinophils in the case of hypersensitivity, which 
increase the inflammatory action, and ultimately B and T cells, which will set up an 
"acquired" immune response (see below). NK cells contribute to antitumor activity. 

Innate immunity is fast, non-specific, and not endowed with memory. It also plays an 
important role in acquired immunity by the process of antigen (Ag) presentation to T cells 
and through the synthesis of some cytokines, which play an important role in the 
orientation of the specific immune responses. Thus, innate immunity is the first to 
intervene following exposure to an Ag. It also plays a fundamental role in acquired 
immunity, as described below. 

Macrophages and DCs are able to recognize "danger" by way of receptors called 
Toll-like receptors (TLRs), which respond to several bacterial components (17,18). To date, 
at least 10 TLRs have been found. TLR2 and TLR4 recognize cell wall structures: 
peptidoglycan of Gram-positive bacteria and lipopolysaccharides (LPS) of Gram- 
negative bacteria, respectively. TLR3 is found specifically in DCs, TLR5 is reported to 
recognize bacterial flagella and TLR9 recognizes pro-inflammatory CpG dinucleotide 
(cytosine phosphoryl guanine non-methylated) only found in the bacterial genome (19). 
Another surface receptor that binds LPS, CD 14, is expressed on the surface of monocytes 
and macrophages. In addition to macrophages and DCs, mucosal epithelial cells also 



96 Moreau 

express TLR2 and TLR4 (20). TLRs play an important role in the initiation of innate 
responses and hence in acquired immunity. The binding of bacterial molecules such as LPS, 
peptidoglycan and CpG motifs to TLRs results in the activation of the nuclear factor kP (NF- 
kP) pathway. NF-kP is a transcriptional factor that intervenes in the synthesis of the pro- 
inflammatory cytokines, TNF-a, IL-1 and IL-6, by cells of the innate immune system and 
intestinal enterocytes; it further stimulates phagocytosis and adhesion molecule expression, 
NO production and synthesis of IL-1 2 (21). In addition, NF-kP activation has an important 
role in regulating the expression of anti-apoptotic proteins and affecting the susceptibility of 
cells to apoptosis (22). Because of the importance of inhibiting the NF-kP pathway in certain 
circumstances, such as those found in the gut, this pathway is regulated by several processes 
as described elsewhere (21-25). 



Acquired Immunity 

Acquired responses consist of the Ag-specific humoral and cell-mediated immune 
responses. They express by synthesis of antibodies (Abs) and cellular responses, 
respectively. They involve three kinds of cells: Ag-presenting cells (APC) (mainly 
macrophages and DCs), T cells, and B cells. For cellular responses only APC and mainly 
CD8 + T cells are involved, while another population of T cells, CD4 + T cells, and B cells 
are needed for Ab synthesis. Antibodies can belong to several kinds of immunoglobulin 
isotypes: IgD, IgM, IgG, IgE, and IgA, and different subclasses including IgGl, IgG2, 
IgG3, and IgG4 or IgAl, and IgA2 in humans. After an initial contact with the Ag, 
acquired responses are slowly established (7-10 days) but are endowed with memory 
enabling a very rapid response after a further contact with the same Ag (within one day). 
The first step in the induction of the immune response is the presentation by APC, and 
recognition of the epitope (small part of an Ag), associated with major histocompatibility 
complex (MHC) molecules of class I or II, to the epitope-specific T cell receptor. In 
addition, the binding of co-stimulation molecules and equivalent receptors expressed by 
both APCs and T cells (CD40 and CD40 ligand, B7.2 and CD28, etc.), leads to the full 
activation and proliferation of naive T cells. Subsequently, a large proportion of those 
activated cells will die of apoptosis, the others surviving in the form of memory T cells. 
All those "lock-and-key" mechanisms are important and greatly contribute to 
modulate the immune responses. It has been shown that DCs play a key role in the 
acquired immune responses. They exist in an immature form in tissues. The mature form 
is obtained following contact with an Ag and phagocytosis. Mature DCs are able 
to synthesize cytokines and migrate into the neighboring draining lymph nodes in order to 
supply Ag information to the T cells. DC populations are heterogeneous (26) and, as will 
be further described, subsets of intestinal DCs display specific properties in terms of Ag 
presentation and cytokine secretion. 



Th1/Th2 Balance 

Several years ago Mosmann and coworkers (27) described different subsets of CD4 + T 
cells that differ by the cytokine profiles produced after activation (Fig. 1). Three kinds 
of T cells are now described from progenitor type helper T cells (ThO). The type 1 
helper T cells (Thl) mainly secrete IFN-y, a pro-inflammatory cytokine, and IL-2. Thl 
induce a weak synthesis of Abs by B cells (subclass IgG2a) and are recruited more in the 
event of a cell-mediated response. In contrast, activation of type 2 helper T cells (Th2) 
induces synthesis of cytokines IL-4, IL-5, IL-10, and IL-13, which have anti- 



Immune Modulation by the Intestinal Microbiota 



97 



Macrophages, 

Dendritic cells /" X 
( NK ) 




Cytokines: IFN-yJL-2 



I 



Weak antibody 

Production 

lgG2a 



Cytokines: IL-4, IL-5, IL-10. 



1 



Important antibody 

Production 

lgG1, lgG2b, IgE, IgA 



Figure 1 Schematic representation of the Thl/Th2 balance. Abbreviations: IFN, interferon; Ig, 
immunoglobulin; IL, interleukin; NK, natural killer cells; Th, T-helper cell. 

inflammatory properties. They induce a large production of Abs by B cells belonging to 
the isotypes and subclasses of IgGl, IgG2b, IgA, and IgE, the latter being involved in 
allergy. Activation of one population inhibits that of another. One of the major 
determinants of the Thl/Th2 differentiation is the cytokine environment at initial 
sensitization. Indeed the transition from ThO to Thl or Th2 depends on environmental 
factors, among which the innate immune cells, macrophages, DCs, and NK cells, play a 
considerable role through synthesis of some cytokines, especially IL-12, and IFN-y, 
acting on the orientation toward a Thl profile (Fig. 1). 

Another subset of T helper cells has been described in mice, the Th3 cells. They 
could play an important role in tolerance by suppressing the immune response through 
production of transforming growth factor- P (TGF-P) after Ag-specific triggering (28). 

The Thl/Th2 balance is an example of the complexity of the host's immune system, 
which has to respond to various immune stimuli by an appropriate immune response. In 
fact, according to the situation, an inflammatory immune response involving Thl and/or 
CD8 + T cells will be activated in intracellular infections needing cell-mediated responses. 
In contrast, a Th2 response producing a low inflammatory response with marked synthesis 
of IgGl or IgA Abs, will be more activated in other situations. With regard to the IgE 
response (Th2 response), it must remain moderate in order to not give rise to adverse 
allergic reactions. A balance between 11-4 and IL-10 may intervene in that regulation, in 
which IL-10 is believed to play a very important anti-inflammatory role (29). 



THE INTESTINAL IMMUNE SYSTEM 



The IIS is a particular immune system anatomically and functionally distinct from that 
present at the peripheral level (30-32). It is in contact with an enormous quantity of Ags, 
food proteins and intestinal bacteria, and does not mount an inflammatory response 
against them. At the same time, it has to protect against enteric pathogens and toxins. 

The IIS is mainly located in the small intestine and colon with differences in the 
anatomical patterning and physiological functions. It is important to be aware of the 
compartmentalization of the intestine even if the IIS associated with the small intestine 



98 



Moreau 



has been subject to more studies and is the most widely described. According to the 
compartment, differences in immune regulations in response to local Ags can be easily 
understood: food Ags are more numerous in the small intestine while in the ileum and 
colon they have essentially been digested and absorbed. In contrast, commensal microbes 
are scarce in the duodenum but more numerous in the ileum and above all in the colon. 

Three lines of defense are present: (i) natural defenses: stomach acidity, bile salts, 
mucus, motility, permeability, (ii) innate immune responses: Ag capture, cytokine 
secretion, TLRs, and (iii) acquired immune responses namely oral tolerance (OT) and 
secretory IgA (slgA) response. All of them interact together. 

Many results presented in this review are derived from studies with mice. Note for 
some results, it is not certain whether they reflect what is happening in humans (33,34). 

Anatomy 

The immune system associated with the small intestine is currently described according to 
two compartments: (i) the inducing sites, named the gut associated lymphoid tissue 
(GALT), consisting of organized aggregated lymphoid tissue, scattered small nodules, 
Peyer's patches (PPs), and mesenteric lymph nodes (MLN); and (ii) the effector sites, i.e., 
the lamina propria tissue where numerous mature B and T small lymphocytes (60% 
CD4 + T cells), plasma cells of which about 90% synthesize IgA are present, and the 
epithelium richly endowed with intra-epithelial lymphocytes (IEL) (CD8 + T cells) 
present between the tight junctions of some enterocytes (Fig. 2) (30-32,35). 

PPs are the first important inductive sites. They are macroscopic lymphoid 
aggregates that are found in the submucosa along the length of the small intestine. 
They consist of large B-cell follicles and intervening T-cell areas which are separated from 
the single layer of intestinal epithelial cells, known as the follicle-associated epithelium 
(FAE), by the subepithelial dome region where APCs are numerous (31). An important 
feature of the FAE is the presence of microfold (M) cells, which, in contrast to enterocytes, 
lack the surface microvilli, the normal thick layer of mucus, and cellular lysosomes. 
Thus, M cells are distinctive epithelial cells that occur only in the FAE. It is believed that 
they play a central role in the initiation of mucosal immune responses by transporting Ags, 
and microorganisms, to the underlying organized lymphoid tissue within the mucosa. 



© Peyer's Patch 



Villus 



2) Lamina propria 




©Epithelium 



-®GALT= PP and MLN 

-©LP 

-(§) Epithelium 



o T and B lymphocytes 
# IgA Plasmocytes 
O CD4+Tcell 

c£iEL(CD8 + ) Mesenteric 

t-> Antigen-presentating cell , 

a Mastocytes Lymph Node 

Goblet cell 

Figure 2 Schematic representation of the intestinal immune system. Abbreviations: GALT, gut- 
associated lymphoid tissue; IEL, intraepithelial lymphocytes; LP, lamina propria; MLN, mesenteric 
lymph nodes; PP, Peyer's patches. 



Immune Modulation by the Intestinal Microbiota 99 

Most of the mature cells found in the effector sites, T cells, plasma cells, epithelium 
CD8 a-P thymus-dependent IEL, derive from PPs. After oral Ag stimulation, the Ag- 
activated immature T and B cells present in PPs leave the PP, and migrate into the 
systemic compartment via the MLN, and the lymph, then enter the bloodstream via the 
thoracic duct. Subsequently the expression of a4P7 integrin, expressed at the surface of 
cells, allows them to bind the gut-specific vascular addressin, MadC AM- 1 , which is 
expressed at high levels by the vasculature of mucosal surfaces, inducing the cells to 
migrate across the endothelium into the lamina propria. Within the intestinal lamina 
propria, B cells differentiate into IgA-secreting plasma cells with a half-life of about 4!/ 2 
days, and most of the T cells undergo apoptosis. This fact has been suggested to be 
important to maintain the gut homeostasis preventing immune responses to luminal Ags 
(36). This cellular traffic, between the PP and lamina propria, has been particularly 
described for IgA plasmocytes. After antigenic stimulation at the PP, B cells undergo Ig 
class switching from expression of IgM to IgA which is under the influence of several 
factors, including cytokines, TGF-P, IL-4, and IL-10, and cellular signals delivered by DC 
and T cells present in PPs. After returning to the lamina propria, IgA plasmocytes 
synthesize and assemble two IgA units and the J chain. Then, a polymeric-Ig receptor 
(plgR) expressed by enterocytes allows selective transcytosis through the epithelial cells, 
and dimeric IgA are excreted in the lumen associated with the secretory component, a 
protein derived from the plgR, which confers to slgA interesting properties such as 
resistance to proteolytic enzymes present in the intestinal lumen (37). 

The physiological significance of the entero-enteric cell circulation is important. 
The induction of an immune response at a PP level propagates distally relative to the 
induction site, not only throughout the intestine but also to other mucosa. It has been 
shown that T and B cells, which have been activated in the GALT, are able to reach 
other mucosal surfaces, which together compose the mucosa-associated lymphoid tissue 
(MALT; vagina, breast during pregnancy and lactation, respiratory tract) by the way of 
homing receptors. This is known as the "common immune system of the mucosa." The 
cycle also shows that there are relationships between the IIS and systemic compartment, 
even though they have, as yet, not been fully elucidated. 

The APCs play a crucial role in the initiation and regulation of the immune 
responses, and are present in all the parts of IIS. In PPs we found immature DCs located in 
close proximity to M cells, which have the capacity to migrate into the interfollicular areas 
of the PP (T areas) and also via the lymphatic to T-cell areas of MLN, thereby stimulating 
T cells in both locations. In all these locations where T cells are stimulated by a given gut 
Ag, the resulting blasts have the capacity to move via the lymph to the thoracic duct and 
the blood to finally home in the gut wall. Ag-specific effector and memory cells thereby 
become disseminated along the whole gut wall, in the lamina propria, and the epithelium. 

Several and unusual subsets of DCs have been described in the murine PP. They are 
located either in the subepithelial dome (CDllb + /CD8 — ), or in the intrafollicular 
regions (CDllb-/CD8 + ) or at both sites (CDllb-/CD8-) (38). It has been described 
in mice that CD1 lb + /CD8 — DC, present in the subepithelial dome of PP, have unusual 
functional characteristics and differ from their peripheral counterparts. Upon antigenic 
stimulation, they secrete IL-10 and induce naive T cells to differentiate into Th2 with IL-4 
and IL-10 production (38). In contrast, in the spleen, the same DC subset secretes IL-12 
after antigenic stimulation under the same experimental conditions, and consequently 
drives the immune response to a Thl orientation with production of IFN-y. However the 
authors showed that the double negative population CD1 lb — /CD8— of DCs, is capable 
of secreting 11-12 upon recognition of microbial stimuli. These functional differences in 
the different PP DC populations may come from the type of Ag stimulation. Indeed, T cells 



100 Moreau 

in PP of mice immunized orally with live Salmonella typhimurium secrete large quantities 
of IFN-y (39). In these studies, it also seems to be important to consider the intestinal site 
from which PPs originate. In fact recent studies have shown that the presence of intestinal 
bacteria in the ileum influence the cytokine profile secreted by DCs in PPs (40). 

DCs are also found in the intestinal villi at the subepithelial level in lamina propria. 
When activated, they can penetrate the epithelium, and send dendrites to the epithelium 
surface, thus being able to directly sample luminal Ags and to present them to IEL and 
lamina propria lymphocytes (31,35). Unusual subsets of DCs are also found, including 
some that are similar to the IL-10 inducing DCs that have been described in PPs. This 
characteristic constitutes a particularity of DCs present both in PPs, lamina propria, and 
epithelium, with functional consequences as presented below for the section on oral 
tolerance. 

Another characteristic of the IIS is the presence of large numbers of activated 
memory CD4+ and CD8 + T cells throughout the lamina propria, expressing the 
chemokine receptor CCR5, probably because of the continuous exposure to environmental 
antigens (41). By contrast, the majority of CD4 + T cells in the peripheral blood and nodes 
are naive T cells (lack of CCR5 expression). It has been reported that PP contains naive T 
cells, expressing chemokine receptor CXCR4, but also activated and memory T cells, a 
phenomenon which is not found in other inductive lymphoid tissue such as MLN or 
peripheral lymph nodes (42). The reasons for this are unknown. 



Physiology 

The IIS generates two important immune functions. First is a suppressive function, also 
termed oral tolerance (OT), characterized by regulatory mechanisms avoiding local and 
peripheral immune responses to harmless environmental Ags present in the intestine, such 
as dietary proteins and bacterial Ags of the intestinal microbiota. Second is the immune 
exclusion performed by slgA Abs to protect the mucosa against pathogen microorganisms 
but also against bacterial translocation of commensal bacteria. Now, it is still unclear 
whether OT induction is accompanied by local slgA production or not. The knowledge of 
regulatory mechanisms that govern the IIS functions are important to understand. When 
the IIS is not functioning well, diseases can develop: enteric and/or systemic infections, 
hypersensitivities to dietary proteins, and IBD. 

Tolerance to Soluble Proteins: Oral Tolerance 

In healthy conditions, the IIS does not mount immune responses to food proteins and 
commensal bacterial Ags. Because two kinds of studies have been reported in the literature 
dealing with the mechanisms involved in OT either to food proteins or to intestinal 
microbiota, we distinguish the mechanisms described, and postulate that they may be 
different according to soluble proteins, such as food proteins, or to bacterial component 
Ags present in the intestinal microbiota. 

Oral tolerance is defined by the state of both systemic and mucosal immune 
unresponsiveness induced after soluble protein feeding. It is a long-lasting phenomenon, 
which affects suppression of both cellular and humoral Ag-specific immune responses. 
Despite the absence of direct evidence in infants, it is believed that OT, which has been 
shown to exist in adult humans (43), certainly plays an important role in the protection 
against hypersensitivity reactions to food proteins [hypersensitivities type I and IV, either 
IgE Abs (allergy) and cellular immune responses, respectively]. Studies on mice have 
shown that suppression of cellular responses lasts up to 17 months after one feeding of 



Immune Modulation by the Intestinal Microbiota 101 

20-mg ovalbumin (OVA) and the suppression of the IgG antibody response lasts more than 
3-6 months (44). 

A number of factors affect OT induction or its persistence (7,31,45). Briefly, they are 
linked to the Ag (nature, doses), the host (genetic, age, inflammatory diseases which affect 
the permeability of intestinal mucosa), intestinal microbiota (described below), and 
bacterial toxins (7). 

The sites where OT is generated, the different mechanisms, and the conditions in 
which they are operating are still a matter of debate (31). Discussion persists as to where 
primary immune responses are initiated: the PP, lamina propria or MLN. It has been 
assumed for many years that PPs are the principal site in which T cells encounter Ags 
derived from food and presented by several distinct potential APCs: macrophages, B 
lymphocytes and DCs. However, some studies suggest that M cells and PPs might not be 
necessary for the uptake and processing of Ags in the induction of OT. For instance, study 
in deficient uMT mice which do not possess B cells, M cells, and PPs because of the lack 
of B cells, showed that these mice are nevertheless able to induce a normal suppressive T 
cell response to oral Ag at the systemic level (46). They concluded that systemic T cell 
responses to orally administered soluble Ags requires neither the specialized Ag 
presentation properties by B cells, nor the microenvironment provided by M cells nor PPs, 
but most likely, are due to characteristics of professional APCs, especially DCs. In recent 
years, some in vitro studies on intestinal epithelial cell-lines have shown that Ags may be 
incorporated into MHC class-II positive exosomes derived from enterocytes (31). These 
vesicles, also called "tolerosomes," can be found in the bloodstream after Ag feeding, and 
are able to induce systemic tolerance when transferred into naive recipients. The 
mechanisms by which exosomes are able to tolerize T cells are under investigation. It has 
been postulated that exosomes can transmit MHC class II/peptide complexes to APCs such 
as DCs. Indeed, incubation of free exosomes bearing MHC class II complexes with DCs 
resulted in a highly efficient stimulation of specific T cells (47). 

Mechanisms implicated in OT are not completely elucidated (7,30,32,45). Studies in 
mice have supported the important roles of intestinal regulatory T cells (reg T cells) and 
DCs in the OT process. The key role of Ag presentation by DCs was provided by 
Viney and coworkers (48). In the study, they showed, in vivo, that administration of 
a hemopoietic growth factor, Flt3 ligand, to mice dramatically expands the number 
of functionally mature DCs in intestine and other lymphoid organs, and increases the 
susceptibility to induction of tolerance by feeding OVA. DCs recruited by Flt3L express 
only low levels of co- stimulatory molecules, supporting the view that intestinal DCs may 
normally be in a resting state without the ability to prime T cells. This mechanism has been 
called "anergy" and it was postulated that only high doses of Ag given to normal mice 
induce this mechanism. 

In addition to the OT, other active suppressor mechanisms, globally named Ag- 
driven suppression, or bystander suppression, have been described. They involve several 
subsets of reg T cells. Indeed, repeated oral administration of low-dose Ag leads to the 
development of Th2 CD4 + T cells secreting IL-4 and IL-10 and Th3 CD4 + T cells 
secreting TGF-(3 cytokines, with anti-inflammatory and suppressive properties. In 
addition, two other reg T cell subsets have recently been described: CD4 + CD25 + reg 
T cells, which could have an important role to prevent intestinal inflammation diseases and 
another reg T cell subset, named Trl, which has been demonstrated to suppress Ag- 
specific immune responses and actively down-regulate a pathological immune response in 
vivo, through production of 11-10 (49). This last finding suggests that Ag-specific Trl are 
capable of producing suppressor cytokines which exert an effect through a local bystander 
suppression. It has been shown that Trl reg T cells can be generated from repetitive 



102 Moreau 

stimulation of CD4 + T cells in the presence of IL-10 (49). Intestinal unusual subsets of 
DC-secreting-IL-10 present in both PP and lamina propria could be implicated in the 
genesis of some of these reg T cells. Indeed, they could drive the T cells towards 
suppressive Trl and reg Trl cells in the intestine and may be crucial for the induction 
of OT. 

Tolerance to the Intestinal Microbiota 

Tolerance to our intestinal microbiota is important to prevent IBD. Some of the OT 
mechanisms may play a role in the tolerant state, but evidence is scarce. It has been 
described that intestinal CD4 + T cells normally recognize the local commensal bacteria, 
but that their responses are inhibited by local reg T cells in an IL-10 and/or TGF-(3- 
mediated manner (50). CD4 + CD25 + reg T cells also play an important role to suppress 
immune responses to bacterial Ags. However, other regulatory mechanisms, involving the 
regulation of immune responses specifically directed towards bacterial components, are 
now suggested. They mainly concern the regulation of the NF-kB pathway, as described 
previously, and where several inhibitory molecular mechanisms intervene (21-25). 

Recently, it has been shown that the functionality of intestinal macrophages and DCs 
is different from that of the peripheral compartment. In humans, and under physiological 
conditions, neither macrophages nor enterocytes express CD 14, a surface receptor 
involved in the response to bacterial LPS, and CD89, the receptor for IgA (51). 
Consequently, they do not respond to LPS by inflammatory cytokine production. The 
absence of CD89 on lamina propria macrophage down-regulates IgA-mediated 
phagocytosis, an activity that normally induces the release of pro-inflammatory mediators 
including reactive oxygen intermediates, leukotrienes, and prostaglandins. This fact 
contributes to maintain the low inflammatory level in normal human intestinal mucosa. 

Modifications of the intestinal homeostasis may modify the inhibitory factors of the 
NF-kB pathway leading to secretion of pro-inflammatory cytokines (20), and/or up- 
regulated CD 14 expression. During the inflammatory process in the intestinal mucosa, 
CD 14 + blood monocytes are probably recruited to the mucosal increasing inflammatory 
reactions. This is the situation prevailing in IBD, in which intestinal tolerance of its 
microbiota has been shown to be deficient (4). 

Antibody slgA Responses 

Another important function elicited by the IIS is the secretion of slgA Abs, which 
represent the most prominent Ab class at the mucosal surface. Secretory IgM Abs can also 
contribute to surface protection in the case of selective IgA deficiency. Secretory IgA 
perform "immune exclusion," which is a non-inflammatory immune response playing an 
important protective role against enteropathogenic opportunistic microorganisms 
(rotavirus, Salmonella, Shigella, Toxoplasma, etc.) for which the intestine constitutes an 
important portal of entry. Thus, they prevent microbial adhesion, especially in the 
duodenum where some pathogenic bacteria such as enterogenic E. coli can adhere. 
Furthermore, they prevent viral multiplication in enterocytes and perform neutralization of 
toxins. They also prevent the translocation of pathogenic and non-pathogenic bacteria 
towards the systemic compartment and concomitantly prevent any damage to the 
epithelium (52). Recently, it has been shown in mice, that dimeric IgA, when bound to the 
secretory component (SC), are more efficient in protection against bacterial respiratory 
infection (53). This effect is due to an appropriate tissue localization of slgA to mucus, 
conferred by carbohydrate residues present in SC. This feature results in an optimal 
protective effect of slgA at mucosal surface by immune exclusion. 



Immune Modulation by the Intestinal Microbiota 103 

In mice, a dual origin for IgA plasma cells in the small intestine has been shown. IgA 
plasma cells originate from two lineages of B cells designated B-l and B-2, which differ 
according to their origins, anatomical distribution, cell surface markers, Ab repertoire and 
self-replenishing potential. B-l cells are maintained by self-renewal of cells resident in the 
peritoneal cavity, and they utilize a limited repertoire that is mostly directed against 
ubiquitous bacterial Ags. B-2 cells, originated from bone-marrow precursors, are present 
in organized follicular lymphoid tissues, within PP, as precursors of plasma cells, and use a 
large repertoire of Abs. Thus the slgA response to specific proteins Ags requires a classical 
costimulation by Ag-specific T cells, an entero-enteric cycle as described previously, and 
are secreted by IgA plasma cells derived from B2 lineage precursors in the PP. By contrast, 
slgA Abs against Ags from commensal bacteria are T cell independent, polyspecific, and 
are secreted by IgA plasma cells derived from the peritoneal cavity B-l cells (54). They 
protect the host from the penetration of commensal bacteria. In mice, B-l lineage could 
represent 40% of total IgA plasma cells. The contribution of peritoneal B cells to the 
intestinal lamina propria plasma cell population in humans is still a matter of debate (33). 

In conclusion, IIS have some phenotypic and functional characteristics, which 
profoundly differ from those found in the peripheral immune system. An important 
finding, which has emerged from recent studies, is the importance of the MLN in the 
induction of both OT and active immunity (slgA secretion), where trafficking of DCs from 
PP and lamina propria, after being loaded with Ag, could prime naive T cells. Indeed, total 
and specific IgA-Ag responses, as well as OT induction are absent in mice that lack MLNs 
(31). Many studies are, however, needed to get a better understanding of the mechanisms 
involved in intestinal immune responses, and the conditions in which they are elicited. 
They are important for the maintenance of the intestinal homeostasis, and are based on 
a continual cross talk between all the immune cells of both IIS (including enterocytes) and 
peripheral immune system and external events in which the digestive microbiota plays an 
important role. 



RELATIONSHIPS BETWEEN THE INTESTINAL IMMUNE SYSTEM 
AND INTESTINAL MICROBIOTA 

The intestinal microbiota has marked influences on the intestinal and peripheral host's 
immunity. In some cases, the effects are produced by the whole intestinal microbiota, 
whereas in other cases only one predominant bacterium is capable of producing a certain 
immunostimulatory effect that is as effective as that of the whole microbiota. Moreover, 
the post-natal period seems to play a crucial role in the cross talk between the intestinal 
microbiota and the development of some important immunoregulatory processes, 
especially those involved in the suppressive responses. 

Most of the data come from original experimental animal models of germ-free (GF) 
mice and gnotobiotic mice, i.e., GF mice colonized with known bacteria. The role of 
intestinal microbiota in humans has largely been extrapolated from studies conducted on 
probiotic bacteria, mainly Bifidobacterium and Lactobacillus strains, and from 
epidemiological studies. 

The intestinal microbiota acts on the three lines of defense of IIS. Recently, 
very interesting papers have been published on the role of intestinal bacteria on natural 
defenses, which are more or less related to innate defenses, especially on epithelium, which 
belong to the IIS. Thus intestinal microbiota should act on: intestinal permeability (55), 
production of fucosylated glycoconjugates (56), glycosylation of the intestinal cell layer 
which is involved in resistance or susceptibility to intestinal infections by the presence or 



104 Moreau 

absence of appropriately glycosylated receptors (57) and, expression of angiogenins, 
especially angiogenin 4 which may have microbiocidal properties (58). These results and 
others, showing that the intestinal microbiota influence the gene expression in epithelial 
cells (59), give new insights in the wonderful cross talk existing between bacteria 
and epithelium. 

The intestinal microbiota also interacts with the other lines of defense, innate and 
acquired immunities. These effects can be of particular importance during the early 
postnatal life that is a period of high risk for intestinal disorders due to enteric pathogens 
and/or food hypersensitivities. During the neonatal period, mammalian species exhibit 
some degree of reduced immunocompetence that could be attributed to a functional 
immaturity in cells involved in immune intestinal responses. It could be also attributed to 
the lack of bacterial stimulation given by the intestinal microbiota which is absent during 
the fetal life. After birth, a well-balanced bacterial colonization will "educate" the IIS in 
a good manner allowing immunoregulatory mechanisms governing IIS functions to 
operate rapidly. 

As already mentioned in the introduction, the activation, modulation and regulation 
of the IIS are the main effects exerted by the intestinal microbiota. Gnotobiotic animal 
models are useful in analyzing such effects of intestinal microbiota on IIS activities. 

Experimental Animal Models: Gnotobiotic Mice 

In experimental studies, the role of the digestive microbiota is determined by comparison 
between GF and conventional (CV) animals, or GF mice colonized with a human fecal 
microbiota, the humanized-mice. Several results show that the human fecal microbiota 
reproduces the same immunostimulatory effects as those produced by the mouse intestinal 
microbiota (60,61), and consequently, this mouse model is a very interesting tool for human 
studies. The first step is the demonstration of the effect of the entire intestinal microbiota on 
a specific immune response by comparison between GF and CV or humanized-mice. The 
second step is to determine the bacteria that are responsible for the immunomodulatory 
effect observed. For this purpose, GF mice are colonized with only one or several known 
bacteria originated from mice or human microbiota. These "gnotobiotic mice," such as GF 
mice, are reared in isolators under microbial controlled conditions. After oral colonization, 
the bacteria expand rapidly to colonize the intestine to a very high level within one day. 
A period of 3 weeks is estimated to be the time required for an optimal stimulatory effect of 
the intestinal microbiota. Thus gnotobiotic models allow in vivo analysis of the specific 
role played by the various bacteria composing the intestinal microbiota with respect to 
immune responses. This has enabled demonstration that the bacterial immunomodulatory 
effect is sometimes "strain-dependent." A more detailed discussion on the use of GF in the 
study of the intestinal microbiota is described in chapter 15 by Norin and Midtvedt. 

Activation of the Intestinal Immune System 

It has been shown that the presence of intestinal microbiota plays an important role in the 
development and activation of IIS even if many effects are still ignored. Its role may be of 
particular importance in the neonatal period and could determine many of the outcomes 
in later life. 

As newborns, GF animals exhibit an underdeveloped IIS, which can be normalized by 
bacterial colonization of the intestine with the fecal microbiota from a CV animal or human, 
within 3 weeks. In GF mice, PPs are poorly developed, and germinal centers are absent. The 
absence of digestive microbiota only affects some subsets of thymus-dependent IEL, the 



Immune Modulation by the Intestinal Microbiota 



105 



single positive thymo-dependent CD4 + or CD8 + af3 IEL, the other thymo-independent 
homodimeric aa CD8 + subpopulations of IEL (all the y5-IEL and part of the aP IEL) 
being always present in GF mice (35). Cellularity of the LP is greatly reduced in GF mice 
and it has been demonstrated that the intestinal microbiota is the major target of the IgA 
plasmocyte development. 



IgA-Secreting Cells 

As in the neonate, the intestinal IgA-secreting cell (IgA-SC) number is much reduced in 
adult GF mice. Three weeks after bacterial colonization of the intestine, GF mice have 
an IgA-SC number equivalent to that found in CV mice. In the young, the adult number of 
IgA-SC is reached at the age of 6 weeks in mice and between 1 and 2 years in babies (7). 
This important delay might be attributed to the immaturity of the IIS of the newborn and/or 
the suppressive effect of Abs present in the mother's milk. However, it might also be 
due to the stimulatory effect of the intestinal microbiota that has been established 
according to a sequential manner from birth to after weaning as described previously. To 
test the later hypothesis, several models of adult gnotobiotic mice were colonized by the 
entire digestive microbiota obtained from growing CV mice from one day after birth to 
25 days of age (i.e., 6 days after weaning; 62). In these experimental adult models, the 
effect of maternal milk, and the possible immaturity of the neonate were excluded, and 
only the stimulatory effect of the digestive microbiota was tested. After 4 weeks, adult 
recipients were sacrificed, and the immunostimulatory effect of the digestive microbiota 
evaluated by the IgA-SC numbers present in intestinal villi by immunohistochemical 
observations. Digestive microbiota of mice 3 to 21 days old exerted only a partial 
stimulatory effect on the intestinal IgA-SC number in gnotobiotic recipients (Table 1). 
However, gnotobiotic recipients colonized with the digestive microbiota of 25-day-old 
mice had a similar IgA-SC number to that found in adult CV mice. 

These results obviously show the important role played by the sequential 
establishment of the digestive microbiota in full development of the intestinal IgA-SC 
number and the pivotal role played by the bacterial diversification present after weaning in 
this process. Results have been confirmed by other studies (7). Moreover, taking into 
account the 3 -week delay between the bacterial stimulus and the intestinal IgA-SC 
response, these results showed that the neonate is capable of developing a slgA response at 
birth, the intensity of which depends on the stimulatory capacity of the intestinal bacteria 
present in the intestine. It is tempting to project such results onto infants where the full 
development of the intestinal IgA-SC number observed at 2 years of age is correlated to 
the stabilization of the intestinal microbiota. 



Table 1 Effect of the Sequential Establishment of Intestinal Microbiota of Growing CV Mice on 
the Maturation of Intestinal IgA Plasma Cells Measured in Gnotobiotic Mice 



Gnotobiotic mice harboring the digestive flora of: 



IgA plasma cell number/villus 



Adult conventional mice 

Adult germ-free mice 

Growing conventional mice 1-4 days old 

Growing conventional mice 7-23 days old 

Growing conventional mice 25 days old 



41 + 1 
4 + 0.5 

15 + 2 
23 + 1 
43 + 1 



Source: From Refs. 62, 63. 



106 Moreau 

Attempts have been made to elucidate the role played by individual bacterial strains 
present in the digestive microbiota of CV growing mice (63). Results showed that some 
Gram-negative bacteria such as E. coli or Bacteroides play an important adjuvant role on 
this immunological non-specific effect, probably due to the LPS present in the cell wall of 
these bacteria (7). These studies have shown the importance of the intestinal microbiota 
diversification on the complete development of IIS in young. They promote insight into 
the close correlation between dietary modification and intestinal microbiota diversification 
and consequently its effect on the infantile IIS. Excessively early or late dietary 
modification may have consequences on quality of the intestinal microbiota equilibrium 
and, consequently, may affect the development of the IIS. 

Dendritic Cells 

As described above, the intestine is populated by some characteristic subsets of DCs, 
which are believed to play a pivotal role in the orientation of the acquired immune 
responses towards tolerance. Is the intestinal microbiota the main factor that determines 
such characteristics? Currently, only few studies exist in this field. 

From some studies, it appears that inflammatory stimuli are very important for 
maturation of DCs in GF mice as well as in neonates, and the intestinal microbiota could 
afford such stimuli. It has been demonstrated that the rapid and constitutive trafficking of 
DCs from the IIS to the MLNs can be increased by the presence of inflammatory stimuli, 
such as LPS (64). Other studies have shown that it is possible to increase the rate of 
postnatal development of the intestinal DC population in rats by intra-peritoneally 
administration of IFN-y (65). We can conclude that these inflammatory factors are 
physiologically important to maintain activation of DCs and the intestinal microbiota may 
have an important part in this process. 

Another question concerns the specific functions of intestinal DCs. Are there 
specific distinct lineages of DCs attracted into the intestinal mucosa under the control of 
specific chemokines or adhesion molecules, or are precursor DCs modified after their 
arrival in the tissue? In his interesting review (31), Mowat explains that, given the 
plasticity of DCs in other tissues, it is reasonable to believe the latter hypothesis, and 
mucosal DCs are the cells that integrate the genetic and environmental factors to shape 
T-cell responses to local Ags in ways such that homeostasis is maintained. Intestinal 
epithelial cells, by the ability to constitutively produce TGF-(3 and by the regulatory 
factors controlling inflammatory cytokine secretion, could be the first level of regula- 
tory control. Moreover, recent studies have shown that lamina propria stromal cells 
constitutively produce cyclo-oxygenase 2 (COX2)-dependent protaglandin E2 (PGE2) 
under the influence of the physiological levels of LPS that are absorbed from intestinal 
microbiota. These metabolites act as down-regulators of the immune response to dietary 
Ags (66). Moreover, DCs themselves might also express COX2 and produce PGE2 in 
response to LPS. As PGE2 is known to polarize DC differentiation towards an IL-10- 
producing inhibitory phenotype, this would explain the prevalence of such DCs in the 
normal gut (67). 

The subunit p40 is present in IL-12 and IL-23, which are both Thl -inducing 
cytokines. In a elegant study, Becker and coworkers (40) using transgenic mice expressing 
a reporter under the control of the IL-12p40 subunit promoter, showed that some subsets of 
lamina propria DCs, present in the small intestine but not in the colon, constitutively 
exhibited transgene expression. This expression was restricted to the ileum, associated 
with the intracellular nondegraded bacteria as revealed by fluorescent in situ hybridization 
(FISH), and was not found in the ileum of GF mice. In addition to supporting literature 



Immune Modulation by the Intestinal Microbiota 107 

elsewhere (68), these results obviously show how the presence of the intestinal microbiota, 
which become more abundant in the ileum, can influence the immune responses elicited 
at this specific area of the intestine. They afford new data on the compartmentalization of 
the IIS, which have to be considered carefully to avoid erroneous conclusions. 

In conclusion, GF, and gnotobiotic animal models are very useful tools to gain new 
insight into the fundamental role played by the intestinal microbiota on the complete 
activation of the IIS, with functional consequences. In certain aspects, adult GF mice, in 
which the IIS is poorly developed, may be considered as similar to that of the neonate and 
immunological immaturity of neonates can be questioned. 

Modulation of Specific Immune Responses: 
The IgA Anti-Rotavirus Response 

Little information is available regarding the role of intestinal microbiota composition on 
the modulation of the specific slgA Ab response against enteropathogens. Indeed, it can be 
assumed that, according to the composition of the digestive microbiota and the presence, 
or not, of some bacteria in the dominant microbiota, the specific immune responses might 
be different. 

This fact is of particular importance in babies where the poorly diversified intestinal 
microbiota is strongly influenced by the type of milk. Indeed, it is well known that breast- 
fed babies are more resistant to enteric infections than formula-fed babies (69,70). Human 
breast milk contains abundant bioactive components that may provide direct protective 
effects to infants against enteric pathogens (71), but breast-feeding also influences the 
intestinal microbiota composition enhancing Bifidobacterium development. To test the 
influence of the intestinal microbiota on the modulation of a specific intestinal slgA-Ab 
response, a slgA anti-rotavirus response was established in a mouse model. This involved 
an original model of adult gnotobiotic mice colonized with the fecal microbiota of a 
breast- or a bottle-fed infant and then orally inoculated with a heterologous simian 
rotavirus strain S A- 1 1 . As previously described, the adult mouse model described here 
excluded breast milk effects and the possible immaturity of the neonate immune system 
[(72) and manuscript in preparation]. 

Bacterial strains found in the dominant fecal microbiota of a breast- or formula-fed 
baby were isolated and inoculated in the digestive tract of the gnotobiotic mice. They 
established in a similar manner as in babies. "Breast-fed mice" were colonized with 
Bifidobacterium, Escherichia coli and Streptococcus, while only two Gram-negative 
bacteria, E. coli and Bacteroides, colonized the digestive tract of "formula-fed" mice. The 
two groups of gnotobiotic mice were similar in all respects except for the intestinal 
microbiota and especially by the presence or absence of Bifidobacterium. They were orally 
inoculated with rotavirus 3 weeks after bacterial colonization to allow the bacteria time to 
affect the immune system of the host. The kinetics of slgA anti-rotavirus Ab responses 
were measured in feces by enzyme linked immunosorbent assay (ELISA) over a one 
month period of time and at sacrifice, numbers of slgA-anti-rotavirus secreting cells were 
evaluated in the small intestine by solid phase enzyme-linked immunospot (ELISPOT) 
assay. 

Kinetics of the slgA anti-rotavirus response were similar in the two groups of 
gnotobiotic mice, but the maximal level, that was reached 20 days after viral inoculation, 
was approximately 4-fold higher in "breast-fed" than in "formula-fed" gnotobiotic 
mice (Fig. 3). The same difference was measured for the slgA-anti-rota virus secreting cell 
numbers. To assess the respective immunomodulatory role of two bacteria present 
in the baby's intestine, Bifidobacterium bifidum (Gram-positive bacteria) and E. coli 



108 



Moreau 





Gnotobiotic mice : « Breast-fed baby » 

(Bifidobacterium, Escherichia coii, 
Streptococcus) 



Gnotobiotic mice « Formula-fed baby » 
(Escherichia coii, Bacteroides) 



Oral Inoculation with rotavirus 



Three weeks later; level of anti-rotavirus slgA antibodies in feces 
18 AU 5AU 

Abbreviation: AU, arbitrary units 

Figure 3 Adjuvant effect of the fecal microbiota of breast-fed babies on the intestinal anti- 
rotavirus antibody response measured in gnotobiotic mice. Source: From Ref. 72. 



(Gram-negative bacteria), two other groups of gnotobiotic mice were created. Results 
presented in Table 2 obviously show the adjuvant capacity of the strain of Bifidobacterium 
bifidum on the intestinal slgA anti-rotavirus response while, in contrast, E. coii exerted a 
suppressive effect, as compared with GF mouse response. These results show how the 
presence of Bifidobacterium bifidum in the fecal microbiota of babies modulates the 
suppressive effect exerted by the presence of E. coii. Given the importance of rotavirus 
infections as a cause of infantile diarrhea worldwide, the presence of Bifidobacterium in 
the intestinal microbiota of babies is of great interest to stimulate this protective Ab slgA 
response. These results can be compared to those found previously, which showed that a 
strain of Lactobacillus rhamnosus GG, used as a probiotic, and given to babies suffering 
from rotavirus diarrhea, shortened the diarrhea duration, and stimulated the specific IgA 
anti-rotavirus response (73,74). Other studies have shown an enhancement of serum or 
intestinal Ab response to orally administered Ags by Gram-positive bacteria (75), 
especially lactic acid producing bacteria used as probiotics (76). 

These results also showed that GF mice are able to mount a slgA anti-rotavirus 
response while its IIS is poorly developed suggesting a lack of correlation between the 
non-specific IgA response induced after bacterial colonization and the specific anti- 
rotavirus Ab response. The latter findings confirm previous results from Cebra and 
coworkers (77). Such data have also been described in humans where one- week-old 
babies are capable of developing protective immunity following oral vaccination with 
poliovirus or hepatitis B virus while the complete development of natural slgA is only 
achieved several months later (78). Consequently, the ability to give a highly specific 
slgA anti-rotavirus Ab response could be correlated with the modulatory effect of 
intestinal bacteria rather than with the development of IIS. Mechanistic studies are 
required to clarify the molecular basis upon which some digestive bacteria modulate the 
slgA Ab response to enteric pathogens. 

The adjuvant effect of Bifidobacterium sp. may be strain-dependent. In a recent study 
we have shown that four different species of Bifidobacterium isolated from the fecal 



Immune Modulation by the Intestinal Microbiota 109 

Table 2 The Gut Colonization of Different Bacterial Strains Modulates the Intestinal 
Anti-rotavirus IgA Antibody Response Measured in Gnotobiotic Mice 

Anti-rotavirus slgA antibody 
Intestinal microflora of gnotobiotic mice level (AU/g of feces) 

Bifidobacterium bifidum (from baby) 3 1 + 7 a f 

Bifidobacterium DN 173 010 (a commercial strain) 21 ±3 a t 

Germ-free (control) 11+2 

Bifidobacterium infantis + B. pseudocatenulatum + 4 ± 1 a | 

B. angulatum + B. sp (from human adult) 

E. coli (from infants) or Bacteroides vulgatus (from 4 + l a J, 

human adult) 

a Significant difference with germ-free mice (/?<0.01). 
Abbreviation: AU, arbitrary units. 
Source: From Refs. 72, 79. 



microbiota of an adult human lacked the adjuvant ability to stimulate the slgA anti- 
rotavirus response in gnotobiotic mice but, on the contrary, exerted a suppressive effect as 
do E. coli (Table 2) (79). Thus, the modulating effect of Bifidobacterium is strain- 
dependent, as it has also been described for different Lactobacillus strains used as 
probiotics in other mice studies (80). Taken together, these data suggest that it is important 
to define the modulatory effect of the strains of bifidobacteria either normally colonizing 
the digestive tract of babies after birth or given as probiotics, to modulate in a good 
protective way a specific intestinal immune response. 

In conclusion, and on the basis of the experimental and clinical data, we may 
consider that the presence of certain bacterial strains in the infantile intestinal microbiota, 
namely some strains of Bifidobacterium, or some transiting strains of probiotics, enable 
activation of the mechanisms that result in optimization of the anti-rotavirus protective 
IgA Ab response. Elucidation of the immunomodulatory mechanisms must now 
be pursued. 



Regulation of the Immune Responses 

Tolerance to Soluble Proteins: Oral Tolerance 

The role of the intestinal microbiota on the OT process has been demonstrated by various 
experimental studies using GF mice. Results depend on the immune response considered, 
oral Ag, and experimental schedule used. In these experiments, immune responses to 
a specific Ag are compared in two groups of mice: the tolerant group where mice are fed 
with an Ag prior to the peripheral immunization with the same Ag, and the control group 
fed with only the buffer before the same peripheral immunization. Specific immune 
responses to the Ag used are then evaluated (Ab responses in serum or cellular response 
by delay ed-type hypersensitivity) in both groups. The tolerant state is present when 
peripheral immune responses to the Ag are abolished or significantly decreased in the 
group Ag-fed as compared with the control group. 

In an initial study, Wannemuehler and coworkers (81) showed that, in contrast to 
what is observed with the CV mice, gavage of GF mice with a particular antigen, sheep red 
blood cells (SRBC), does not enable suppression of immune responses to SRBC in serum. 
However, the OT process was re-established when LPS was administered orally prior 
to gavage. The authors concluded that Gram-negative bacteria play a fundamental role in 



110 Moreau 

the mechanisms responsible for OT. Subsequently, other experiments using adult GF mice 
fed with a soluble protein, OVA, in order to study the immune suppression of anti-OVA 
serum IgG response, demonstrated that it was possible to induce OT in GF mice. However, 
in contrast to what is observed with CV mice, the suppression was of very short duration, 
about 10-15 days, versus more than 5 months in CV mice (82). Similar results were 
obtained in human-microbiota-associated gnotobiotic mice (60). Colonization of the 
intestinal tract with E. coli alone prior to gavage was sufficient to restore lasting 
suppression (83), and the same results were obtained with another Gram-negative bacteria, 
Bacteroides (unpublished personal data), while in our experimental conditions, adult GF 
colonized with the strain of Bifidobacterium bifidum isolated from a baby's feces, had no 
effect on the serum IgG anti-OVA suppression (83). 

Recently, in their experimental conditions, Sudo and coworkers (84) showed that in 
OVA-fed mice, the GF state does not allow suppression of the systemic anti-OVA IgE 
response in serum in contrast to what is observed with CV mice. Colonization of the 
intestinal tract by a strain of Bifidobacterium infantis restored the suppression but only 
when the strain colonized the intestinal tract of the mouse from birth. The importance of 
the presence of intestinal bacteria from birth in the optimization of the immune processes 
has also been suggested in a more recent study (60). 

It is interesting to compare these experimental results to those described in human 
neonates by Lodinova-Zadnikova and coworkers (85). In their study, they colonized 
the digestive tract of babies just after birth with a given strain of E. coli. In these conditions 
E. coli is able to establish durably in the digestive tract of newborns as described 
previously (86). After 10 years (preterm infants) and 20 years (full-term infants), 
differences in occurrence of food allergies between colonized and control subjects were 
statistically significant; 21% versus 53%, and 36% versus 51% respectively. Furthermore, 
recent clinical trials using ingestion of a strain of probiotic, Lactobacillus rhamnosus GG, 
during the last month of pregnancy to women and after birth to babies during 6 months, 
reduced the incidence of atopic eczema in at-risk children during the first 4 years of 
life (87). However, in this case, IgE levels were not decreased in the treated group as 
compared with the placebo group. The protective mechanisms of these interventions are 
not elucidated. 

All these experimental data show the importance that a single bacterial strain present 
in the intestinal digestive microbiota of infants may have with respect to the establishment 
of tolerance mechanisms. Are there E. coli, Bacteroides or some strains of Bifidobacterium 
which play this important role? First, as suggested by previous studies, it is not sure 
whether the mechanisms are the same for suppression of the various isotypes IgG and IgE 
(45,88), and consequently that the same bacteria are operating on them. Secondly, as 
described previously, all the strains belonging to the same bacterial genus have not the 
same immunoregulatory properties and it is conceivable that some Bifidobacterium strains 
may have regulatory properties on suppressive immune processes. 

The cellular ways by which the bacteria are acting, and the exact bacterial 
components involved are not known. However, from an ecological point of view, it is 
important to note that some experimental data point out the importance of the neonatal 
period with respect to the ability to recognize bacterial messages. 



Tolerance to the Intestinal Microbiota 

An important question is why the intestinal microbiota does not mount an inflammatory 
response in the gut while this state is broken in pathologic conditions such as IBD? 



Immune Modulation by the Intestinal Microbiota 111 

The mechanisms by which commensal and non-pathogenic bacteria are tolerated 
by the IIS is beginning to be understood and may result from a cross-talk between 
bacteria, epithelium, and immune cells. In an interesting experimental study, Neish 
and co-workers (89) demonstrated, using an in vitro model of cultured human intestinal 
epithelial cells, that a non-pathogenic strain of Salmonella directly influenced the 
intestinal epithelium to limit inflammatory cytokine production. They showed that the 
immunosuppressive effect was due to the inhibition of the NFk-B activation pathway by 
blockage of IkB-oc degradation. Another interesting conclusion from this study was that 
non-pathogenic bacteria, which do not belong to the commensal intestinal microbiota, 
are unable to induce inflammatory responses. Another study converges to an opposite 
conclusion (90). In several intestinal epithelial cell lines, the authors demonstrated that 
a commensal bacterial strain, Bacteroides vulgatus, was able to activate the NF-kB 
signaling pathway through IkB-gc degradation and RelA phosphorylation. However, the 
presence of TGF-P1 cytokine inhibits B. vw/ga to-mediated NF-kB transcriptional 
activity showing that the responsiveness of intestinal epithelial cells to luminal enteric 
bacteria depends on a network of communication between immune and epithelial cells 
and their secreted mediators. 

Recently, it was shown in vivo in mice, that the intestinal microbiota itself plays 
a regulatory role with respect to inhibition of the NFk-B activation pathway, by the way of 
another inhibitory factor, the peroxisome proliferator-activated receptor (PPARy) (61). 
The latter is highly expressed in the colon and its activation has anti-inflammatory effects, 
with protection against colitis. PPARy activators are able to limit inflammatory cytokine 
production through the inhibition of the NF-kB pathway. It has been suggested that PPARy 
could play an important role in homeostasis of the gut, especially in the colon. In patients 
with IBD, impaired expression of PPARy in colon epithelial cells was observed (61). 
In the same work, in vivo observations showed that the intestinal microbiota and TLR-4 
regulates PPARy expression by epithelial cells of the colon. Indeed, it is highly expressed 
in CV mice while it is barely detectable in GF mice. When TLR-4 transfected CaCo-2 cells 
were incubated with LPS, an increase of PPARy expression was observed showing 
the involvement of TLR-4 in this process and suggesting that PPARy may be a regu- 
latory factor able to shut down the TLR-4 signaling given by bacterial LPS abundant in 
the colon (61). 

Taken together, these data provide evidence that the cross-talk existing between the 
IIS and intestinal microbiota pass through regulatory processes preventing inflammatory 
responses induced by activation of some nuclear factors, such as NF-kB, which could be 
different, or predominant, according to the intestinal site. They are mediated through the 
actions of commensal bacteria, but also through exogenous non-pathogenic bacteria action 
and this data is of importance in terms of nutrition. Indeed, we can ingest billions of 
exogenous bacteria in some foods such as fermented milks and some cheeses, without 
detrimental consequences. In terms of pathology, a lot of other questions concerning the 
mechanisms and origin of IBD have yet to be answered. Why is an activation of the NF-kB 
pathway observed in IBD? Is it due to some subsets of the intestinal microbiota, which are 
suddenly dominant in an unbalanced microbiota? Is it due to enteropathogens which can 
interact with the NF-kB pathway during infection? Or, is it due to a decrease and 
modification of mucus secretion allowing excessive adhesion of commensal bacteria? 
All these factors, and others, may be responsible. 

It is interesting to give recent clinical results concerning oral administration of 
probiotics on the maintenance of the remission phase in IBD, either the use of a mixture 
of 8 strains of lactic-acid bacteria used as probiotics (VSL#3) in chronic pouchitis (91), or 
a yeast strain, Saccharomyces boulardii (92) or the E. coli Nissle 1917 (93) in ulcerative 



112 Moreau 

colitis. The mechanisms underlying such beneficial effects are still not known and they are 
multifactorial. From experimental data it has been suggested that a stimulation of the non- 
inflammatory IL-10 cytokine production by ingestion of probiotics may be involved 
in such protective effect (94). Further experimental and clinical studies need to be 
conducted to further elucidate the mechanisms involved in the epithelium-bacterial 
cross talk. 



RELATIONSHIPS BETWEEN THE PERIPHERAL IMMUNE SYSTEM 
AND INTESTINAL MICROBIOTA 

Activation of the Immune System 

Innate immunity plays a very important role in the activation of the immune system and 
the ability to develop specific acquired immune responses. Through their Ag-presenting 
activity and the synthesis of numerous pro-inflammatory chemokines and cytokines (IL-8, 
IL-1, IL-6, TNF-a, and IL-12), macrophages, and DCs play a key role in the regulation 
of immune responses. They are the gatekeepers of the host, generating innate resistance to 
pathogens, and specific immune responses by the stimulation of T-cell-acquired immunity 
and regulation of the TH1/Th2 balance. 

It has been postulated that the immune defects in neonates may result from 
a developmental immaturity of APC functions (78), and bacterial components resulting 
from intestinal colonization could be an important factor for maturation of APCs (95). 
Recently, Sun and coworkers (96) investigated the ontogeny of peripheral DCs and their 
capacity to provide innate responses to microbial stimuli in early life. They show that 
neonatal murine spleen DCs have intrinsic capacity to produce bioactive IL-12. Moreover, 
after microbial stimulation given in vitro by LPS, they are able to up-regulate MHC and 
costimulatory molecule expression required for productive interaction with naive T cells. 
Thus, neonatal DCs could be fully competent in their innate functions but they need to be 
activated, through TLR recognition as described previously, by bacterial stimuli afforded 
by the intestinal microbiota. Another interesting study supports this hypothesis. Nicaise 
and coworkers (97) demonstrated that the presence of the intestinal microbiota underlies 
IL-12 synthesis by macrophages derived from splenic precursors. 

On the basis of those experimental data, one can wonder whether the first bacteria 
colonizing the intestinal tract, E. coli, rich in LPS, and subsequently bifidobacteria rich 
in peptidoglycan and CpG dinucleotides, do not play such crucial activating roles? It is 
conceivable that in newborns, the abrupt colonization of the intestinal tract by the 
microbiota may induce a physiological inflammatory reaction with, as a consequence, an 
increase in intestinal permeability, bacterial translocation and systemic activation of 
immune cells, especially APCs. Experimental evidence supports that hypothesis. Studies 
in mice have shown that the presence of the intestinal microbiota induces the synthesis of 
pro-inflammatory cytokines IL-1, IL-6, and TNF-a by peritoneal macrophages. Such 
effects can be reproduced in gnotobiotic mice colonized with E. coli alone while 
a Bifidobacterium bifidum strain isolated from baby's feces had no effect (Table 3) (98). 

Other non-specific resistance factors play an important role in host defense 
mechanisms to infection. GF and gnotobiotic animal models have showed that some 
functional parameters involved in innate immunity, phagocytosis, complement system, 
and opsonins, are expressed to a lesser extent than in CV animals (99). 



IL-6 


TNF-a 


6,33 


72 


2,62 a 


<50 a 


2,46 a 


<50 a 


7,24 b 


108 b 



Immune Modulation by the Intestinal Microbiota 113 

Table 3 Influence of Intestinal Bacteria on the Inflammatory Cytokine Production by Peritoneal 
Macrophages 

Gnotobiotic mice Cytokines (units/ml) 

IL-1 
Conventional 18200 

Germ-free 8300 a 

Bifidobacterium bifidum 8000 a 

Escherichia coli 15350 

a Significant difference with conventional mice (/?<0.01). 

Not significant. 
Abbreviations: IL, interleukin; TNF, tumor necrosis factor. 
Source: From Ref. 98. 



Modulation and Regulation of Immune Responses 

Balance Thl/TKl 

Experimental results, epidemiological studies and clinical trials strongly argue for the fact 
that bacterial environment plays a crucial role in the Thl/Th2 balance via different 
mechanisms of which cytokine synthesis by innate immune cells, especially IL-1 2, and 
IFN-y, could play a decisive role. 

The prenatal period and early childhood are considered to be critical for the 
establishment and maintenance of a normal Thl/Th2 balance. It has been described that 
the immune context at birth is mainly Th2, while Thl responses are partially suppressed, 
enabling non-rejection of the fetus during gestation. After birth, neonates must rapidly 
restore the balance by developing the potential to induce Thl -type responses (100). 
Various studies have shown that, in atopic infants, the switch does not occur, and the infant 
is in a context of an imbalance toward Th2 with a predisposition to development of IgE 
responses (101,102). The neonatal period is thus considered to be extremely important in 
enabling regulation of the Thl/Th2 balance to become operative, and the switch could 
occur during the first 5 years of life especially during the first year of life (103). 

The Th2— >Thl switch is dependent on multiple factors whose relative importance 
has yet to be elucidated. Bacterial stimuli are considered to play a considerable role, and 
some years ago it had been claimed that infections might prevent the development of atopic 
diseases. This is referred to as the "hygiene hypothesis" (13), but it is now a matter of 
debate. From a recent study (104), authors did not find any evidence that exposure 
to infections in infants reduces the incidence of allergic disease, but, in contrast, exposure to 
antibiotics may be associated with an increased risk of developing allergic disease. Today, 
accumulating evidence suggests that rather than infections, alteration of the composition of 
the intestinal microbiota early in life may be an important determinant of atopic status 
(13,105). Experimental studies have supported this hypothesis. Thus, in one-week-old rats, 
peripheral immunization leads to a Th2-biased memory response. However, when the rats 
are concomitantly administered a bacterial extract by the oral route with immunization, the 
memory response switches to both Thl and Th2 (106). Another study showed how, in three- 
week-old mice, the disturbance in intestinal bacterial equilibrium following ingestion of an 
antibiotic, kanamycin, promoted a shift in the Thl/Th2 balance toward a Th2-dominant 
immunity, while it became Thl and Th2 in non-treated growing CV mice (107). Ingestion 
of intestinal bacteria such as Enterococcus faecalis five days after antibiotic treatment 
again permitted the shift back towards the Thl/Th2 balance (108). 



114 Moreau 

From an epidemiological point of view, very interesting studies argue in favor of 
the important role of the bacterial environment in the first year of life in order to ensure the 
good orientation of immune responses preventing the short- and long-term development of 
atopic diseases (13,101,103,109-111). Recent comparative studies have been conducted 
in children living in the same allergenic environment but under different life- style 
conditions, urban and farming environments. Results showed that substantial protection 
against development of asthma, hay fever, and allergic sensitization was seen only in 
children exposed to stables, farm raw milk, or both in their first year of life (103). Authors 
also found that prenatal exposure of women had a substantial protective effect. 

Bacteria that are responsible for such effects are not known. Gram-negative bacteria 
rich in LPS have been suggested to be important in that phenomenon (85,109,1 12) but it is 
also possible that Gram-positive bacteria, such as bifidobacteria and Lactobacillus, are 
involved. The comparative study between Swedish and Estonian children (105) has 
suggested a specific role of the intestinal microbiota, regarding its nature, diversity and 
changes with time. Besides genetic factors, which are known to play an important role in 
the development of allergic diseases, all these data suggest that the infant intestinal 
microbiota normally rich in Gram-negative (LPS-producing) and Gram-positive bacteria 
may not be well-balanced in atopic children. Depending on the microbial environment 
associated with the life-style, especially during the first year of life, a restoration of the 
normal balance could be achieved. 

Clinical trials using probiotics to treat or prevent atopic eczema in infants have also 
generated arguments suggesting that the infantile intestinal microbiota balance plays an 
important role in the good orientation of immune responses. In a recent double-blind trial, 
Kalliomaki and coworkers (87) have shown that the supplementation of pregnant women 
one month before delivery followed by 6 months post-parturition (mother or baby) with 
a probiotic strain, Lactobacillus rhamnosus GG, lead to a significant decrease in the 
incidence of atopic eczema in babies with a family history of atopic disease. At two years 
of age, atopic eczema was diagnosed in 23% of treated babies versus 46% in the placebo 
group. The preventive effect of L. rhamnosus GG extends to the age of 4 years follow-up 
treatment (87). The mechanisms involved in such a protection are unknown. Indeed, the 
frequencies of positive skin-prick test reactivity (measuring the specific IgE levels) were 
comparable between treated and placebo groups. Further studies are necessary to elucidate 
the mechanisms responsible for these interesting protective effects. 

On the basis of all the above data, questions arise with respect to delivery conditions, 
infant feeding, and antibiotic treatments to be administered during infancy in order to 
enable and optimally establish and maintain integrity of the intestinal microbiota. 
Probiotics may also be considered as good palliative agents with respect to impaired 
equilibrium of the intestinal microbiota. Knowledge of the immunoregulatory 
mechanisms driven by the intestinal microbiota of infants, as well as the bacterial 
components which are involved, are crucial to prevent some pathologies which are 
dramatically increasing today. 

Natural IgG 

In the absence of immunization, there is a natural level of immunoglobulins (Ig) in serum 
named "natural Ig" or "natural Abs." The roles of those Abs in the immune responses have 
yet to be completely elucidated but it is known that they play important regulatory roles in 
humoral immune responses, especially in immune responses to self-Ag (113). It has also 
been demonstrated in mice that they intervene with the development of the B 
repertoire at peripheral level (spleen), enabling expansion of the Ab response towards 



Immune Modulation by the Intestinal Microbiota 115 

thymo-dependant Ags (114,115). In man, the role of these natural Abs is under 
investigation in the context of research on certain autoimmune disease (116). 

Intrinsic and extrinsic factors, especially the intestinal microbiota, act on the natural 
Ig levels, depending on isotypes and sub-classes. Thus, GF mice had normal serum IgM 
levels, but IgG, and IgA levels are approximately 5% of conventionally reared littermates 
(1 14). It has been established in mice that one of the roles of the natural IgG is to expand 
B cell repertoire. The latter can be evaluated through the expression of some genes coding 
for the variable part of the heavy chain of Ig (VH gene) using probes. Analysis of a VH 
gene expression has provided a quantitative tool for the global assessment of Ab 
repertoire, and a preferential use of the gene means that the repertoire is poorly diversified. 

Early in ontogeny, a high frequency of B cells could bind to multiple Ags, among 
which auto-Ags are found, in neonatal CV mice. This fact has been correlated with 
preferential use of VH gene family, namely VH7183. In CV adult mice these multi- 
reactive B cells are much less frequent coinciding with a random usage of VH genes, as 
seen by the decreased utilization of VH7173 gene family, showing a diversified repertoire. 
Thus, there is a maturation of the immune system of adult CV mice. This fact is not present 
in adult GF mice where a high percentage of B cells expressing VH 7138 genes is found as 
in neonatal CV mice (115). The injection of purified natural IgG Ig from serum adult CV 
mice into GF mice reduced the use of the VH7183 gene family in the peripheral B-cells, as 
in CV mice (115). From these data authors concluded that if a genetic program leading to 
non-random position-dependent preference of rearrangement and expression initially 
controls the establishment of the VH repertoire, a broader utilization of the B-cell 
repertoire is thereafter stimulated by environmental Ags and Igs. The finding that GF mice 
maintain a "fetal-like" VH repertoire that can be modified by the administration of pooled 
Igs from normal unimmunized CV mice establishes the crucial role of the intestinal 
microbiota in this function. 

This data may have clinical relevance. Many reports have described the beneficial 
results of intravenous injection of normal human IgG in treatment of autoimmune 
disease (116). 

The mechanism by which exogenous antigenic stimulation can influence the 
expression of VH gene remains unclear. Exogenous Ags may play an important role in 
the final modulation of the expressed repertoires either by direct stimulation of Ag-specific 
clones or indirectly by idiotype interactions mediated by the Abs produced in those 
responses (113-115). 



Autoimmune Diseases 

One example of the regulatory effect exerted by intestinal microbiota on an autoimmune 
disease has been reported by Van der Broek and co-workers (117). Streptococcal cell wall 
(SCW)-induced arthritis is a chronic erosive polyarthritis, which can be induced in 
susceptible rats by a single intra-peritonal injection of a sterile aqueous suspension of 
SCW. The acute phase of the disease develops within a few days, the second, chronic 
phase, which mainly involves peripheral joint inflammation, develops from 10 days after. 
The second phase is dependent on functional T lymphocytes. F344 rats are genetically 
described as resistant to the second chronic phase, while in contrast another strain of rats, 
Lewis rats, are described as susceptible. These data suggest that a T-cell unresponsiveness 
due to immune tolerance to SCW may be the mechanism underlying resistance to SCW- 
induced arthritis of F344 rats, while Lewis rats are defective in their tolerance. When 
F344 rats are reared in GF conditions, they become susceptible to SCW-induced arthritis 
as are Lewis rats. There was a correlation between the susceptibility of the disease and the 



116 Moreau 

T cell proliferation response to SCW measured in vitro. In CV Lewis and GF-F344 rats, 
a proliferation was measured while it was not present in CV F-344 rats. This concept that 
disease might result from a similarity between naturally occurring cell surface Ags of the 
host and those expressed on some commensal or pathogenic micro-organisms have been 
referred to as the "molecular mimicry hypothesis." Mono-association of GF F344 rats 
with E. coli resulted in resistance, which equaled that in CV F344 rats whereas 
mono-association with a Lactobacillus strain did not really affect susceptibility. Thus, 
in CV F-344 rats, a state of tolerance to arthritogenic epitopes is induced during the 
neonatal period of life and maintained through life by the bacterial microbiota, resulting 
in resistance to SCW-induced arthritis. In Lewis rats, this tolerant state is deficient and/or 
easily broken. 

Bacterial effects have been suggested in other autoimmune diseases. Thus, oral 
antibiotic treatment after adjuvant-induced arthritis (AIA) induction in rats significantly 
decreased clinical symptoms of AIA while, concomitantly, E. coli levels increased in the 
distal ileum of antibiotic-treated rats (118). In addition, it has been described that 
Mycobacterial infections profoundly inhibit the development of diabetes in non-obese 
diabetic (NOD) mice (119). 



CONCLUSION 

From all the experimental epidemiological and clinical results presented here, the 
digestive microbiota can be considered as an organ: it is specifically tolerated by 
the host and in turn, it exerts many continuous regulatory effects on intestinal and 
peripheral host's immune responses. Consequently, it plays fundamental roles in health. 
It is very important to develop knowledge about its composition, the bacterial components 
and metabolites that participate to such immunoregulatory effects, and the exact 
mechanisms involved. 

Studies from GF animals have demonstrated the importance of the digestive 
microbiota on intestinal and peripheral immune systems. In some cases, the entire 
digestive microbiota is needed to obtain the complete effect while other immunoregulatory 
effects can be reproduced with only one bacterium and sometimes with only specific 
strains. Because the intestinal microbiota is a dynamic community which modifies from 
birth to old age in predominant bacteria composition, specific targeted interests have to be 
defined for the study of relationships between the intestinal microbiota and the host, 
according to age. Indeed, bacterial species found in the predominant microbiota are not 
constantly the same throughout life and several studies have demonstrated the strain- 
dependant immunomodulatory effect of bacteria. For instance, some strains of 
bifidobacteria, such as B. breve, are more commonly found in infants but less in adults 
(120). Other studies from adult GF animals have demonstrated that some bacterial effects 
are only obtained when the bacteria colonized the intestinal tract from birth indicating that 
the bacterial effects need some characteristics of the neonate immune system. A number of 
indirect findings converge toward the idea that the neonatal period is crucial for the infant 
with respect to setting up the regulatory mechanisms which will play an important role in 
the good orientation of immune responses throughout life. Because of the long-term 
consequence of the establishment of appropriate immunoregulatory networks, it is very 
important to develop knowledge on the cross-talk between the intestinal microbiota and 
immune system early in life. In this context, recent studies of the innate responses to 
bacterial constituents should generate decisive information in support of the role of the 
intestinal microbiota. 



Immune Modulation by the Intestinal Microbiota 117 

In adults, regulation of immune responses seems to be constantly reshaped by 
persistent interactions between the host and its digestive microbiota. 

Today, an increasing challenge for researchers studying immunity (IIS as well as 
oral or peripheral immune responses after Ag vaccination, pro-, and prebiotic effects) is 
that the intestinal microbiota of experimental rodents used is not defined and can differ 
between breeders because of the great variety in housing conditions. Since the 
development of knock-out mice, which are very sensitive to infections, the microbial 
status required by experimenters has led to the production of highly clean animals which 
carry a commensal microbiota with reduced diversity. This fact has probably a significant 
impact on the development of the immune responses. Thus, because results could not 
reflect the exact conditions of microbial stimulation, the interpretation of experiments 
may be completely different according to different laboratories. Some controversial 
results obtained in mice and humans might also be explained by such paucity of mouse 
microbiota existing in pathogen-free mouse breeding-care units. Now, it is crucial to 
develop animal models in which the commensal microbiota will be better defined and 
designed to allow the maintenance of biological features relevant in the field of 
immunological investigations. 

A more comprehensive understanding of the relationships between the intestinal 
microbiota and innate and acquired immune systems should offer new approaches for the 
therapy of some diseases such as allergies and IBD and for the design of oral vaccinations, 
and the maintenance of health. Beneficial micro-organisms such as probiotics, and dietary 
ingredients such as prebiotics, that act on the digestive microbiota, show promise for 
treatment in these immune-related intestinal disorders. Researchers addressing those 
subjects have to consider the digestive microbiota in their investigations. 

All of the studies presented here clearly indicate the close relationship between the 
prokaryotic and eucaryotic worlds, and the intricacy and complexity of the relationships. 
Much work remains to be done and much is left to discover about our intestinal microbiota 
and immunity. It is to be hoped that the current enthusiasm with respect to the interest in 
the action of intestinal microbiota on immunity will continue to increase. The practical 
applications that can emerge in terms of human health can be highly significant. 



REFERENCES 

1. Sanders ME. Probiotic bacteria: implications for human health. J Nutr 2000; 130:384S-390S. 

2. Isolauri E, Sutas Y, Kankaanpaa P, Arvilommi H, Salminen S. Probiotics: effects on 
immunity. Am J Clin Nutr 2001; 73:444S-450S. 

3. Erickson KL, Hubbard NE. Probiotic immunomodulation in health and disease. J Nutr 2000; 
130:403S-409S. 

4. Dutchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer zum Buschenfelde KH. 
Tolerance exists towards commensal intestinal flora but is broken in active inflammatory 
bowel disease. Clin Exp Immunol 1995; 102:448-455. 

5. Guaner F, Malagelada JR. Gut flora in health and disease. The Lancet 2003; 360:512-519. 

6. Umesaki Y, Setoyama H. Structure of the intestinal flora responsible for development of the 
gut immune system in a rodent model. Microbes Infect 2000; 2:1343-1351. 

7. Moreau MC, Gaboriau-Routhiau V. Influence of commensal intestinal microflora on the 
development and functions of the gut-associated lymphoid tissue. Microb Ecol Health Dis 
2001; 13:65-86. 

8. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science 2001; 
292:1115-1118. 

9. Rubaltelli FE, Biadaioli R, Pecile P, Nicoletti P. Intestinal flora in breast-fed and bottle-fed 
infants. J Perinat Med 1998; 26:186-191. 



118 Moreau 

10. Raibaud P. Factors controlling the bacterial colonization of the neonatal intestine. In: 
Hanson LA, ed. Biology of human milk. New- York Nestec Ltd.: Vevey /Raven Press, 
1988:205-219. 

1 1 . Favier CF, Vaughan EE, DeVos WM. Akkermans ADL. Molecular monitoring of succession 
of bacterial communities in human neonates. Appl Environ Microbiol 2002; 68:219-226. 

12. Kelly D, Coutts AGP. Early nutrition and the development of immune function in the neonate. 
Proc Nutr Soc 2000; 59:177-185. 

13. Wold AE. The hygiene hypothesis revised: is the rising frequency of allergy due to changes 
in the intestinal flora ? Allergy 1998; 46:20-25. 

14. Salminen S, Bouley C, Boutron-Ruault MC, et al. Functional food science and gastrointestinal 
physiology and function. Br J Nutr 1998; 80:S147-S171. 

15. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related changes in intestinal bacterial 
populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid 
profiles. Gut 2001; 48:198-205. 

16. Tannock GW. Molecular assessment of intestinal microflora. Am J Clin Nutr 2001; 
73:410S-414S. 

17. Vasselon T, Detmers PA. Toll receptors: a central element in innate immune responses. 
Inf Immun 2002; 70:1033-1041. 

18. Uderhill DM, Ozinsky A. Toll-like receptors: key mediators of microbe detection. Curr Opin 
Immunol 2002; 14:103-110. 

19. Hemmi H, Takeuchi O, Kawai T, et al. A toll-like receptor recognizes bacterial DNA. Nature 
2000; 408:740-745. 

20. Cario E, Rosenberg IM, Brandwein SL, Beck PL, Reinecker HC, Podolsky DK. 
Lipolopysaccharide activate distinct signaling pathways in intestinal epithelial cell lines 
expressing toll-like receptors. J Immunol 2000; 164:966-972. 

21. Neish AS. The gut microflora and epithelial cells: a continuing dialogue. Microbes Infec 
2002; 4:309-317. 

22. Wu MX, Ao Z, Prasad KV, Wu R, Schlossman SF. IEX- 1L, an apoptosis inhibitor involved in 
NF-KB-mediated cell survival. Science 1998; 281:998-1001. 

23. Xavier RJ, Podolsky DK. How to get along-friendly microbes in a hostile world. Science 
2000;289:1483-1484. 

24. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator- 
activated receptor gamma is a negative regulator of macrophage activation. Nature 1998; 
391:79-82. 

25. Tato CM, Hunter CA. Host-pathogen interactions: subversion and utilization of the NF-kappa 
B pathway during infection. Inf Immun 2002; 70:331 1—3317. 

26. Rescigno M, Granucci F, Cittero S, Foti M, Ricciardi-Castagnoli P. Coordinated events during 
bacteria-induced DC maturation. Immunol Today 1999; 20:200-204. 

27. Mosmann TR, Cherwinski H, Bond W, Giedlin A, Coffman RL. Two types of murine helper T 
cell clone. Definition according to profiles of lymphokine activities and secreted proteins. 
J Immunol 1986; 136:2348-2357. 

28. Miller A, Lider O, Roberts AB, Spom MB, Weiner HL. Suppressor T cells generated by oral 
tolerization to myelin basic protein suppress both in vitro and in vivo immune reponses by the 
release of transforming growth factor (3 after antigen-specific triggering. PNAS USA 1992; 
89:421-425. 

29. Pretolani M, Goldman M. IL-10: a potential therapy for allergic inflammation? Immunol 
Today 1997; 6:277-280. 

30. Mowat AMc, Viney JL. The anatomical basis of intestinal immunity. Immunol Rev 1997; 
156:145-166. 

31. Mowatt AMc. Anatomical basis of tolerance and immunity to intestinal antigens. Nat Rev 
Immunol 2003; 3:331-341. 

32. Brandtzaeg P. Development and basic mechanisms of human gut immunity. Nutr Rev 1998; 
56:S5-S18. 

33. Boursier L, Farstad IN, Mellembakken JR, Brandtzaeg P, Spencer J. IgVH gene analysis 
suggests that peritoneal B cells do not contribute to the gut immune system in man. 
Eur J Immunol 2002; 32:2427-2436. 



Immune Modulation by the Intestinal Microbiota 119 

34. MacDonald TT, Monteleone G. IL-12 and Thl immune responses in human Peyer's patches. 
Trends Immunol 2001; 22:244-247. 

35. Guy-Grand D, Vassali P. Gut intraepithelial lymphocyte development. Curr Opin Immunol 
2002; 14:255-259. 

36. Bu P, Keshavarzian A, Stone DD, et al. Apotosis: one of the possible mechanisms that 
maintains unresponsiveness of the intestinal mucosal immune system. J Immunol 2001; 
166:6399-6403. 

37. Johansen FE, Braathen R, Brandtzaeg P. The J chain is essential for polymeric Ig recepetor- 
mediated epitheliall transport of IgA. J Immunol 2001; 167:5185-5192. 

38. Iwasaki A, Kelsall BL. Unique functions of CDllb + CD8 + , and double negative Peyer's 
patch dendritic cells. J Immunol 2001; 166:4884-4890. 

39. George A. Generation of gamma interferon responses in murine Peyer's patches following 
oral immunisation. Infect Immun 1996; 64:4606-4611. 

40. Becker C, Wirtz S, Blessing M, et al. M.F. Constitutive p40 promoter activation and IL-23 
production in the terminal ileum mediated by dendritic cells. J Clin Invest 2003; 1 12:693-706. 

41. Veazey RS, Marx PA, Lacker AA. The mucosal immune system primary target for HIV 
infection and AIDS. Trends Immunol 2001; 22:626-633. 

42. Jump RL, Levine AD. Murine Peyer's patches favor development of an IL-10 secreting, 
regulatory T cell population. J Immunol 2002; 168:6113-6119. 

43. Husby S, Mestecky J, Moldoveanu Z, Holland S, Elson CO. Oral tolerance in humans — T cell 
but not B cell tolerance after antigen feeding. J Immunol 1994; 152:4663-4670. 

44. Strobel S, Ferguson A. Persistence of oral tolerance in mice fed ovalbumin is different for 
humoral and cell-mediated immune responses. Immunology 1987; 60:317-318. 

45. Strobel S, Mowat AMc. Immune responses to dietary antigens: oral tolerance. Immunol 
Today 1998; 19:173-181. 

46. Alpan O, Rudomen G, Matzinger P. The role of dendritic cells, B cells, and M cells in gut- 
oriented immune responses. J Immunol 2001; 166:4843-4852. 

47. Vincent-Schneider H, Stumptner-Cuvelette P, Lankar D. Exosomes bearing HLA-DR1 
molecules need dendritic cells to efficiently stimulate specific T cells. Int Immunol 2002; 
14:713-722. 

48. Viney JL, Mowat AM, O'Malley JM, Williamson E, Fanger NA. Expanding dendritic cells 
in vivo enhances the induction of oral tolerance. J Immunol 1998; 160:5815-5825. 

49. Groux H, O'Garra A, Bigler M, et al. A CD4 + T-cell subset inhibits antigen-specific T-cell 
responses and prevent colitis. Nature 1997; 389:737-742. 

50. Khoo UY, Proctor IE, Macpherson AJ. CD4- Tcells down-regulation in human intestinal 
mucosa: evidence for intestinal tolerance to luminal bacterial antigens. J Immunol 1997; 
158:3626-3634. 

51. Smith PD, Smythies LE, Mosteller-Barnum M, et al. Intestinal macrophages lack CD 14 and 
CD89 and consequently are down-regulated for LPS- and IgA-mediated activities. J Immunol 
2001; 167:2651-2656. 

52. Macpherson AJ, Hunziker L, McCoy K, Lamarre A. IgA responses in the intestinal 
mucosa against pathogenic and non-pathogenic microorganisms. Microbes Infect 2001; 
3:1021-1035. 

53. Phalipon A, Cardona A, Kraehenbuhl JP, Edelman L, Sansonetti PJ, Corthesy B. Secretory 
component: a new role in secretory IgA-mediated immune exclusion in vivo. Immunity 2002; 
17:107-115. 

54. Macpherson AJ, Gatto D, Sainsbury E, Harriman GR, Hengartner H, Zinkernagel RM. 
A primitive T cell-independant mechanism of intestinal mucosal IgA response to commensal 
bacteria. Science 2000; 288:2222-2226. 

55. Madsen K, Cornish A, Soper P, et al. Probiotic bacteria enhance murine and human intestinal 
epithelial barrier function. Gastroenterology 2001; 121:580-591. 

56. Bry L, Falk PG, Midtvedt T, Gordon JL A model of host-microbial interactions in an open 
mammalian ecosystem. Science 1996; 273:1380-1383. 

57. Freitas M, Axelsson LG, Cayuela C, Midtvedt T, Trugnan G. Microbial-host interactions 
specifically control the glycosylation pattern in intestinal mouse mucosa. Histochem Cell Biol 
2002; 118:149-161. 



120 Moreau 

58. Hooper LV, Stappenbeck TS, Hong CV, Gordon JI. Angiogenins: a new class of microbicidal 
proteins involved in innate immunity. Nature Immunol 2003; 4:269-273. 

59. Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular 
analysis of commensal host-microbial relationships in the intestine. Science 2001; 
291:881-884. 

60. Gaboriau-Routhiau V, Raibaud P, Dubuquoy C, Moreau MC. Colonization of gnotobiotic 
mice with human microflora at birth protects against Escherichia coli heat-labile enterotoxin- 
mediated abrogation of oral tolerance. Pediatr Res 2003; 54:739-746. 

61. Dubuquoy L, Jansson EA, Deeb S, et al. Impaired expression of peroxisome proliferator- 
activated recepetor y in ulcerative colitis. Gastroenterology 2003; 124:1265-1276. 

62. Moreau MC, Raibaud P, Muller MC. Relation entre le developpement du systeme 
immunitaire intestinal a IgA et l'etablissement de la flore microbienne dans le tube digestif 
du souriceau holoxenique. Ann Immunol (Inst Pasteur) 1982; 133D:29-39. 

63. Moreau MC, Ducluzeau R, Guy-Grand D, Muller MC. Increase in the population of duodenal 
IgA plasmocytes in axenic mice monoassociated with different living or dead bacterial strains 
of intestinal origin. Infect Immun 1978; 21:532-539. 

64. Macpherson GG, Liu MM. Dendritic cells and Langherans cells in the uptake of mucosal 
antigens. Curr Top Microbiol Immunol 1999; 236:33-53. 

65. Mac William AS, Holt PG. Mucosal dendritic cells in the respiratory tract. Mucosal Immunol 
Update 1997; 5:21-25. 

66. Newberry RD, Stenson WF, Lorenz RG. Cyclooxygenase-2-dependent prostaglandin E2 
production by stromal cells in the murine small intestine lamina propria: directing the tone of 
the intestinal immune response. J Immunol 2001; 166:4465-4472. 

67. Harizi H, Jusan M, Pitard V, Moreau JF, Gualde N. Cyclooxygenase-2- issued prostaglandine 
E2 enhances the production of endogenous IL-10, which down-regulates dendritic cell 
functions. J Immunol 2002; 168:2255-2263. 

68. Uhlig HH, Powrie F. Dendritic cells and the intestinal bacterial flora: a role for localized 
mucosal immune responses. J Clin Invest 2003; 112:648-651. 

69. Wold AE, Hanson LA. Defense factors in human milk. Curr Opin Gastroenterol 1994; 
10:652-658. 

70. Mastretta E, Longo P, Laccisaglia A, et al. Effect of Lactobacillus GG and breast-feeding in 
the prevention of rotavirus nosocomial infection. J Pediatr Gastroenterol Nutr 2002; 
35:527-531. 

7 1 . Goldman AS. Modulation of the gastrointestinal tract of infants by human milk. Interfaces and 
interactions. An evolutionary perspective. J Nutr 2000; 130:426S-431S. 

72. Moreau MC. Effet immunomodulateur des bacteries intestinales: role des bifidobacteries. 
J Pediatr Puericulture 2001; 14:135-139. 

73. Isolauri E, Juntenen M, Rautanen T, Sillanaukee P, Koivula T. A human Lactobacillus strain 
{Lactobacillus casei sp strain GG) promotes recovery from acute diarrhea in children. 
Pediatrics 1991; 88:90-97. 

74. Kaila M, Isolauri E, Soppi E, Virtanen V, Laine S, Arvilommi H. Enhancement of the 
circulating antibody secreting cell response in human diarrhea by a human Lactobacillus 
strain. Pediatr Res 1992; 32:141-144. 

75. Herias MV, Midtvedt T, Hanson LA, Wold AE. Increased antibody production against gut- 
colonizing. E. coli in the presence of the anaerobic bacterium Peptostreptococcus. Scand 
J Immunol 1998; 48:277-282. 

76. Flo J, Goldman H, Roux ME, Massoud E. Oral administration of a bacterial immunomodulator 
enhances the immune response to cholera toxin. Vaccine 1996; 14: 1 167—1 173. 

77. Cebra JJ, Bos NA, Cebra ER, Kramer DR, Kroese FGM, Schrader CE. Cellular and molecular 
biologic approaches for analyzing the in vivo development and maintenance of gut mucosal 
IgA responses. In: Mestecky et al. eds. Advances in Mucosal Immunology. New- York: 
Plenum press, 1995:429-434. 

78. Bona C, Bot A. Neonatal immunoresponsiveness. Immunologist 1997; 5:5-9. 

79. Moreau MC, Gaboriau-Routhiau V, Guiard G, Bouley. Strain-dependent immunomodulatory 
properties of lactic acid bacteria: experimental data from Bifidobacterium strains and 
Lactobacillus strains. 17th International Congress of Nutrition, Modern aspects of nutrition, 
Vienna, 27-31 August 2001. 



Immune Modulation by the Intestinal Microbiota 121 

80. Maassen CBM, Van Holten-Neelen C, Balk F, et al. Strain-dependent induction of cytokine 
profiles in the gut by orally administered Lactobacillus strains. Vaccine 2000; 18:2613-2623. 

81. Wannemuehler MJ, Kiyono H, Babb JL, Michalek SM, McGhee JR. Lipopolysaccharide 
(LPS) regulation of the immune response: LPS converts germfree mice to sensitivity to oral 
tolerance induction. J Immunol 1982; 129:959-965. 

82. Moreau MC, Gaboriau-Routhiau V. The absence of gut flora, the doses of antigen ingested, 
and aging affect the long-term peripheral tolerance induced by ovalbumin feeding in mice. 
Immunol Res 1996; 147:49-59. 

83. Moreau MC, Gaboriau-Routhiau V, Dubuquoy C, Bisetti N, Bouley C, Prevoteau H. 
Modulating properties of intestinal bacterial strains, Escherichia coli, and Bifidobacterium, on 
two specific immune responses generated by the gut, i.e., oral tolerance to ovalbumin, and 
intestinal IgA anti-rotavirus response, in gnotobiotic mice. The 10th International Congress of 
Immunology, New-Dehli. In: Talwar GP, Nath I, Ganguly NK, Rao KVS, eds. Bologna: 
Monduzzi Editore, 1998:407-411. 

84. Sudo N, Sawamura SA, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal 
bacterial flora for the development of an IgE production system fully susceptible to oral 
tolerance induction. J Immunol 1997; 159:1739-1745. 

85. Lodinova-Zadnikova R, Cukrowska B, Tlaskalova-Hogenova H. Oral administration of 
probiotic Escherichia coli after birth reduces frequency of allergies and repeated infections 
later in life (after 10 and 20 years). Int Arch Allergy Immunol 2003; 131:209-211. 

86. Duval-Iflah Y, Ouriet MF, Moreau MC, Daniel N, Gabilan JC, Raibaud P. Implantation 
precoce d'une souche de Escherichia coli dans l'intestin de nouveau-nes humains: effet de 
barriere vis-a-vis de souches de E. coli antibioresistantes. Ann Microbiol (Inst Pasteur), 133 A 
1982; 133A:393-408. 

87. Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of 
atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 2003; 
361:1869-1871. 

88. McMenamin C, McKersey M, Kuhnlein P, Hunig T, Holt PG. Gamma-delta T cells down- 
regulate primary IgE responses in rats to inhaled soluble protein antigens. J Immunol 1995; 
154:4390-4394. 

89. Neish AS, Gewirtz A, Zeng H, et al. Procaryotic regulation of epithelial responses by 
inhibition of iKB-aubiquition. Science 2000; 289:1560-1563. 

90. Haller D, Russo MP, Sartor RB, Jobin C. IKK beta and phosphatidylinositol 3-kinase/Akt in non- 
pathogenic Gram negative enteric bacteria-induced RelA phosphorylation and NF-kB activation 
in both primary and intestinal epithelial cell lines. J Biol Chem 2002; 277:38168-38178. 

91. Gionchetti P, Amadini C, Rizello F, Venturi A, Poggioli G, Campieri M. Probiotic for the 
treatment of postoperative complications following intestinal surgery. Best Pract Res Clin 
Gastroenterol 2003; 17:821-831. 

92. Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative 
colitis. Eur J Gastroenterol Hepatol 2003; 15:697-698. 

93. Rembacken BJ, Snelling AM, Hawkey P, Chalmers DM, Axon TR. Non pathogenic 
Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. 
Lancet 1999; 354:635-639. 

94. Madsen K, Doyle JS, Jewell LD, Tavernini M, Fedorak RN. Lactobacillus species prevents 
colitis in interleukin-10 gene-deficient mice. Gastroenterology 1999; 116:1107-1114. 

95. Ridge JP, Fuchs EJ, Matzinger P. Neonatal tolerance revisited: turning on newborn T cells 
with dendritic cells. Science 1996; 271:1723-1726. 

96. Sun CM, Fiette L, Tanguy M, Leclerc C, Lo-Man R. Ontogeny and innate properties of 
neonatal dendritic cells. Blood 2003; 102:585-591. 

97. Nicaise P, Gleizes A, Sandre C, et al. The intestinal microflora regulates cytokine production 
positively in spleen-derived macrophages but negatively in bone marrow-derived 
macrophages. Eur Cytokine Net 1999; 10:365-372. 

98. Nicaise P, Gleizes A, Forestier F, Sandre C, Quero AM, Labarre C. The influence of E. coli 
implantation in axenic mice on cytokine production by peritoneal and bone marrow-derived 
macrophages. Cytokine 1995; 7:713-719. 

99. Podoprigora G. The role of microbial factors in non-specific resistance of the host to infection. 
Microecol Ther 1996; 24:207-217. 



122 Moreau 

100. Adkins B, Bu YR, Guevara P. Murine neonatal CD4 + lymph nodes are highly deficient in the 
development of antigen-specific Thl function in adoptive adult hosts. J Immunol 2002; 
169:4998-5004. 

101. Renz H, vonMutius E, Mi S, Wolkers F, Hirsh T, Wieland SK. T(H)1/T(H)2 immune 
responses profiles differ between atopic children in eastern and western Germany. J Allergy 
Clin Immunol 2002; 109:338-342. 

102. Marodi L. Down-regulation of Thl responses in human neonates. Clin Exp Immunol 2002; 
128:1-2. 

103. Riedler J, Braun-Fahrlander C, Eder W, et al. The ALEX study team. Exposure to farming in 
early life and development of asthma and allergy: a cross-sectional survey. Lancet 2001; 
358:1129-1133. 

104. McKeever TM, Lewis SA, Smith C, et al. Early exposure to infections and antibiotics and the 
incidence of allergic disease: a birth cohort study with the West Midlands general practice 
research database. J Allergy Clin Immunol 2002; 109:43-50. 

105. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic Estonian 
and Swedish 2-year-old children. Clin Exp Allergy 1999; 29:342-346. 

106. Bowman LM, Holt PG. Selective enhancement of systemic Thl immunity in immunologically 
immature rats with an orally administered bacterial extract. Infect Immun 200 1; 69:371 9-3727 . 

107. Oyama N, Sudo N, Sogawa H, Kubo C. Antibiotic use during infancy promotes a shift in the 
Thl/Th2 balance towards Th2-dominant immunity in mice. J Allergy Clin Immunol 2001; 
107:153-159. 

108. Sudo N, Yu XN, Aiba Y, et al. An oral introduction of intestinal bacteria prevents the 
development of a long-term Th2-skewed immunological memory induced by neonatal 
antibiotic treatment in mice. Clin Exp Allergy 2002; 32:1112-1116. 

109. Braun-Fahrlander C, Riedler J, Herz U, et al. Environmental exposure to endotoxin and its 
relation to asthma in school-age children. N Engl J Med 2002; 347:869-877. 

110. Strannegard O, Strannegard IL. The causes of the increasing prevalence of allergy: is atopy 
a microbial privation? Allergy 2001; 56:91-102. 

111. Von Mutius E. Environmental factors influencing the development and progression of 
pediatric asthma. J Allergy Clin Immunol 2002; 109:525S-532S. 

112. Cukrowska B, Lodinova-Zadnikova R, Enders C, Sonnenborn U, Schulze J, Tlaskalova- 
Hogenova H. Specific proliferative and antibody responses of premature infants to intestinal 
colonization with non-pathogenic E. coli strain Nissle 1917. Scand J Immunol 2002; 
55:204-209. 

113. Avrameas S. Natural antibodies: from "horror autotoxicus" to "gnothi seauton". Immunol 
Today 1991; 123:154-159. 

1 14. Bos NA, Meeuswen CG, Wostmann BS, Pleasants JR, Benner R. The influence of exogenous 
stimulation on the specificity repertoire of background immunoglobulin-secreting cells of 
different isotypes. Cell Immunol 1988; 112:371-380. 

115. Freitas AA, Viale AC, Sunblad A, Heusser C, Coutinho A. Normal serum immunoglobulins 
participate in the selection of peripheral B-cell repertoires. PNAS 1991; 88:5640-5644. 

116. Kaveri SV, Lacroix-Desmazes S, Mouthon L, Kazatchine MD. Human natural antibodies: 
lessons from physiology and prospects for therapy. Immunologist 1998; 6:227-233. 

117. Van der Broek MF, Van Bruggen MCJ, Koopman JP, Hazenberg MP, Van den Berg WB. Gut 
flora induces and maintains resistance against streptococcal cell wall-induced arthritis in F344 
rats. Clin Exp Immunol 1992; 88:313-317. 

118. Nieuwenhuis EES, Visser MR, Kavelaars A, et al. Oral antibiotics as novel therapy for arthritis. 
Evidence for a beneficial effect of intestinal E. coli. Arthritis Rheum 2000; 43:2583-2589. 

119. Martins TC, Aguas A. Mechanisms of Mycobacterium aWwra-induced resistance against 
insulin-dependant diabetes melitus (DDM) in non-obese diabetic (NOD) mice: role of Fas and 
Thl cells. Clin Exp Immunol 1999; 115:248-254. 

120. Gavini F, Cayuela C, Antoine JM, et al. Differences on the distribution of bifidobacterial and 
enterobacterial species in human faecal microflora of three different (children, adults, elderly) 
age groups. Microb Ecol Health Dis 2001; 13:40-45. 



6 



Mucosal Interactions and Gastrointestinal 
Microbiota 



Wai Ling Chow and Yuan-Kun Lee 

National University of Singapore, Department of Microbiology, 
Faculty of Medicine, Singapore 



INTRODUCTION 

The human gut harbors a complex and diverse microbiota. The numbers of microorganisms in 
the upper gastrointestinal (GI) tract are kept low by the actions of gastric acid, pancreatic 
enzymes, bile, and a propulsive motor pattern. The colonic population of microbes is 
estimated to be 10 organisms/gram with at least 400 possible species. The above figure 
was obtained by traditional culture-based methods. Modern molecular methods such as 16S 
ribosomal RNA clone libraries that are discussed in Chapter 1 indicate that the number of 
species will be even higher. The composition of the intestinal microbiota varies from human 
to human. These differences in the composition of the microbiota are affected by 
physiological, chemical, and environmental factors. The common intestinal microbiota in 
humans includes predominantly members of genera Clostridium, Eubacterium, Bacter- 
oides, Atopobium and Bifidobacterium spp. and many others to a lesser extent. There is an 
approximation that almost 90% of the cells in our body are microbial, whereas only 10% 
are human. 

The bacteria that colonize the gut must be able to proliferate at a rate that resists 
washout. Adherence to the intestinal mucosal surface is an important factor in intestinal 
bacterial colonization. In healthy individuals, a layer of mucus is found to line the gut. It is 
composed mostly of glycoproteins and serves as a lubricant and a protective lining over 
the mucosa. Microbiota degradation of the mucin polymeric glycoprotein results in the 
release of monosaccharides such as N-acetylglucosamine and fucose amongst others, 
which the microbiota use to support their growth (2). Furthermore, under the mucus the 
surfaces of intestinal epithelial cells are covered with an abundance of terminally 
fucosylated glycoproteins and glycolipids which are induced by members of the intestinal 
microbiota (3). In particular, it was demonstrated that Bacteroides thetaiotaomicron 
cleaves L-fucose moieties from the host's surface and internalizes them for use as an 
energy source. This commensal microbe modulates the production of the fucose by the 
host with its requirement needs, which gives it a competitive colonization advantage 

123 



124 



Chow and Lee 



within the intestinal niche (68). Thus, the interaction of microorganisms with the mucosa 
is a complex one, which involves cross-talk between the microbes, and between the 
microbes and the host. 

In this chapter, we provide some insights about the development and regulation of 
the gastrointestinal microbiota as well as the interaction of the microbes with the intestinal 
mucosal layer. The majority of research on the molecular interactions between microbes 
and the mucosa relate to pathogen-enterocyte interaction, and consequently, this field is 
also occasionally referred to. 



FEATURES OF THE GASTROINTESTINAL TRACT 

Structure and Function of the Small Intestine 

The small intestine is the principal site of food digestion, nutrient absorption as well as 
endocrine secretion. It is the longest component of the alimentary tract, measuring over 
6 meters, and is divided into three anatomic regions: duodenum, jejunum and ileum. The 
duodenum begins at the pylorus of the stomach and is the proximal 20-25 cm of the small 
intestine. The jejunum spans about 2.5 meters in length. The ileum is approximately 
3.5 meters long and an extension of the jejunum. 

The absorptive surface area of the small intestine is greatly increased by tissue and 
cell specializations such as plicae circulares, villi and microvilli (Fig. 1). Plicae circulares 
are permanent transverse folds of the mucosa, forming semicircular or spiral elevations. 
They are abundant in the distal duodenum and beginning of the jejunum. Intestinal villi are 
finger-like outgrowths of mucosa protruding into the lumen of the small intestine. 
Microvilli are protrusions of the apical plasmalemma of the epithelial cells covering the 
intestinal villi, increasing the surface area of the small intestine 20 times. Therefore, these 
modifications immensely amplify the absorptive and interactive (with intestinal content, 
including the microbiota) surface area of the small intestine. 

The mucosa comprises the lining epithelium, a lamina propria that houses glands and 
muscularis mucosa. There are at least 5 types of cells found in the intestinal mucosal 



r 



Villi 



Lymphoid 

cell 



Paneth 
cell 



Mucous 
epithelium of 

pylorus 



Goblet cell 




Lamina 

propria 



Muscularis - 
mucosae 



Figure 1 Schematic diagram of the mucosa, villi, and component cells of the small intestine. 



Mucosal Interactions and Gastrointestinal Microbiota 125 

epithelium. They include enterocytes, goblet cells, Paneth cells, enteroendocrine cells and 
M cells (microfold cells). Both the enterocytes and the goblet cells line the villus and are the 
major cell types in the epithelium. The enterocytes are columnar in shape and have brush 
borders composed of microvilli which help to enhance the water ions and nutrient absorbing 
surface area. Goblet cells are unicellular mucin-secreting glands which produce mucinogen 
and mucin, a component of mucus. The number of goblet cells increases progressively down 
the gastrointestinal tract from the duodenum, to jejunum, ileum and colon, where they are 
most abundant. The Paneth cells' role is to maintain the innate immunity by secreting 
antimicrobial substances such as a-defensins (4,69). Enteroendocrine cells are present only in 
small numbers ( ~ 1 %) and their functions include the production of panacrine and endocrine 
hormones (5). M cells are modified enterocytes overlying the enlarged lymphatic nodules in 
the lamina propria. Their function is to phagocytose and transport antigens present in the 
intestinal lumen to the underlying macrophages and lymphoid cells, which then migrate to 
other compartments of the lymphoid nodes, where immune responses to foreign antigens are 
initiated (5). 

The lamina propria is rich in lymphoid cells, which will protect the intestinal lining 
from bacterial invasion. The loose connective tissue of lamina propria forms the main part 
of the villi, extending down to the muscularis mucosa. The epithelium may invaginate into 
the lamina propria between the villi to form glands, termed the crypts of Lieberkuhn. 
These tubular glands consist of enterocytes, goblet cells, regenerative cells, enteroendo- 
crine cells and Paneth cells. The rate at which the regenerative cells proliferate is high and 
they are capable of replacing other cell types in the intestinal epithelium. As mentioned 
above, the pyramidal-shaped Paneth cells secrete antibacterial agents, such as lysozyme 
and a-defensins or cryptdins, and internalized extracellular matter such as bacteria and 
immunoglobulin. Therefore, it is postulated that these cells help in regulation of the 
bacterial microenvironment in the small intestine. 



Structure and Function of the Large Intestine 

The large intestine is a continuation of the ileum and is usually divided into three 
regions: the colon, rectum and anal canal. The colon accounts for nearly the full length 
of the large intestine. The colon absorbs water and electrolytes (approximately 1400 ml 
per day). It also compacts and eliminates feces (about 100 ml per day). Feces are 
composed of water (75%), dead bacteria (7%), roughage (5%), inorganic substances 
(5%), and undigested protein, dead cells and bile pigment (1%). Bacterial products, 
including the vitamins riboflavin, thiamin, vitamin B12 and vitamin K, are also 
excreted in the feces (5). 

The colonic mucosal membrane does not have any folds due to an absence of villi 
(Fig. 2). The intestinal glands are long and characterized by a great abundance of goblet 
and absorptive cells, and a small number of enteroendocrine cells. The large intestinal 
epithelium is specialized for mucos secretion, salt and water absorption. 

The histology of the rectum is identical to that of the colon except that the crypts of 
Lieberkuhn are deeper and fewer in number. The rectum is about 12-18 cm in length and 
is continuous with the anal canal, which spans about 3 to 4 cm. The mucosa of the anal 
region displays a series of longitudinal folds, the rectal columns of Morgagni. These rectal 
columns meet one another to form pouch-like outpocketings, the anal valves with 
intervening anal sinuses. The anal valves assist in supporting the column of feces (5). The 
epithelial cells of the entire gastrointestinal tract are constantly shed. They are replaced 
with stem cells that have undergone mitosis. The high turnover rate of the epithelial cells 
may explain why the small intestine is affected rapidly by the administration of 



126 



Chow and Lee 








°0* O N * 






Colonic 
epithelium 



Lamina 
propria 




^ Muscularis 

mucosae 



•4| — Submucosa 




uscularis 
externa 



Figure 2 Schematic diagram of the colonic epithelium and associated cells. 



anti-mitotic drugs, as in cancer chemotherapy. The epithelial cells continue to be lost at the 
tip of the villi, but drugs inhibit cell proliferation (6). 



Mucus 

The gastrointestinal tract contains tremendous numbers of microorganisms and some of 
these microorganisms are pathogenic in nature under certain conditions. Therefore a 
function of the mucus is to protect the underlying epithelial cells by keeping the microbes 
and toxins at bay, on the outer mucosal surfaces. The mucus layer is comprised of various 
mucosal secretions including mucins, trefoil peptides, and surfactant phospholipids. 

Mucus occurs in two distinct physical forms: (1) a thin layer of stable, water 
insoluble mucus gel firmly adhering to the gastroduodenal mucosal surface, (2) and as 
soluble mucus which is quite viscous but mixes with the luminal juice (7). 

The layer of mucus that is bound to the surface of the gastrointestinal tract is 
resistant to its removal from the mucosa. It is approximately 50-450 Jim thick in humans 
and about twofold less in rats. This adherent mucus functions to support and define 
the mucosal ecosystem since it is the outermost sensory "organ" of the mucosal immune 
system. The mucus gel plays a role in providing surface neutralization by having the 
HCO^~ barrier to the gastric acid. The surfactant lipids maintain surface hydrophobicity on 
the mucus. The adherent mucus also serves as a stable protective barrier that prevents the 
entry of luminal pepsin to the underlying epithelial cells. 

The soluble mucus plays a role in maintaining the protective barrier because it is not 
physically attached to the mucosa and can be removed from the mucosa by gentle washing. 
Due to the viscous nature of the soluble mucus, the soluble mucus makes an excellent 
lubricant which allows easy movement of solid material in the lumen. This helps to 
prevent the damage to the underlying epithelial cells as well as minimize the tearing of the 
adherent layer of mucus from the mucosal surface (7). 



Mucosal Interactions and Gastrointestinal Microbiota 127 

The main structural component of the mucus layer are the mucins or glycoproteins 
of molecular weight ranging from one to several million daltons. When concentrated, 
these glycoprotein macromolecules (M r >2X10 6 ) polymerise to form gels. Mucin 
molecules consist of carbohydrate side chains (70-80%) bound to a protein skeleton. The 
O-linked oligosaccharide chains contain a restricted number of monosaccharides, 
including galactose, fucose, N-acetylgalactosamine, N-acetylglucosamine and often 
terminated with sialic acids or sulfate groups, which account for the poly anionic nature of 
mucins at a neutral pH (7,8). Oligosaccharides chains are successively added on to mucins 
specifically by membrane bound gly cosy ltransf erases. The biochemistry of the intestinal 
mucins confers their protective nature: the protein backbone has a high O-linked 
oligosaccharide content ( > 80% carbohydrate by mass) that provides lectin-binding 
capacity, whereas the ability of the protein core to form multimers (through disulphide 
bonds) causes polymerization into gels and bestows viscoelasticity and lubrication (9). 
The trefoil peptides also facilitate the mucins to confer visoelasticity on the mucus (10). 

The composition of the mucus is constantly regulated by the varying secretion rates 
of the mucin types, ions, lipids, proteins and water. The variation in the composition of the 
mucus is also dependent on the development stage of the host as well as the host's diet and 
the interaction of the commensals and pathogens (10). Commensals rapidly colonize the 
individual soon after birth and some play a role in inhibiting the growth of pathogenic 
bacteria. However, many commensals are capable of becoming opportunistic pathogens 
by overgrowing when the stable gastrointestinal ecosystem is disturbed. Thus, the mucus 
has to be continuously secreted and then shed, discarded, digested or recycled. This form 
of protective mechanism keeps the numbers of both pathogens and commensals in check 
by blocking the bacterial adherence to the epithelial cells. 



MICROBIOTA AND GASTROINTESTINAL SYSTEM 

Distribution of Microbiota 

The mucosal surface of the human body, including the gastrointestinal tract, the 
respiratory tract and the urogenital tract, has a total surface area of more than 400 m (11). 
The gastrointestinal tract's surface area is about 200-300 m 2 and is colonized by 10 13-14 
bacteria with hundreds of bacterial species and subspecies. 

The normal microbiota of the gastrointestinal tract has been grouped and defined into 
two categories, the autochthonous (indigenous) and the allochthonous (nonindigenous) 
species (12). The autochthonous microbes (1) are always present in the normal adult's 
gastrointestinal tract, (2) play a role in maintaining the stable bacterial populations in 
the gastrointestinal tract, (3) colonize particular parts of the tract, (4) can grow 
anaerobically, (5) colonize their habitats in succession in infants, and (6) often associate 
intimately with the gastrointestinal mucosal epithelium. 

On the other hand, allochthonous species are not characteristic of the normal habitat. 
Allochthonous microbiota is defined as transient microbes which will not be established 
but would just be passing through, having arrived in the habitat in food, in water, from 
another habitat in the gastrointestinal tract, or from elsewhere in the body. These microbes 
either cannot or find it very challenging to establish themselves since they cannot compete 
in the various niches or may be killed by host or bacterial factors. 

However, the allochthonous microorganisms might colonize the habitats vacated by 
the autochthonous microbes in the disturbed gastrointestinal system (13). This was 
evidently seen in the administration of antibiotics which caused severe disturbance in the 
gastrointestinal microbiota leading to undesirable effects, such as the overgrowth and 



128 Chow and Lee 

superinfection with allochthonous microorganisms like yeast (14,15); see also chapter 18 
by Sullivan and Nord in this book. 

Thus, the main difference between autochthonous and allochthonous species is that 
an autochthonous microbe naturally colonizes the habitat, whereas an allochthonous one 
cannot colonize it except under abnormal or atypical situations (13). 

In a steady gastrointestinal ecosystem, all the niches are probably occupied by 
indigenous microbes. The number of microorganisms in the stomach and the upper 
two-thirds of the small intestine is very scarce: a maximum of 10 4 per milliliter of intestinal 
contents. The relatively low number of microbes is due to the low pH (approximately pH 2) 
of the intestinal contents resulting from gastric acid production and the relatively swift flow 
(transit time of 4-6 hours) of digesta through the stomach and small intestine. Culturing 
studies indicate that lactobacilli and streptococci are commonly found microbes in the small 
intestine (16). Unlike the bulk of the microbes within the gastrointestinal tract, both the 
lactobacilli and streptococci are acid-tolerant bacteria, and are capable of surviving the 
passage through the stomach. 

The ileum contains larger numbers of microbes (10 - 1 bacteria per ml of intestinal 
contents) in comparison to the upper regions of the gastrointestinal tract. The higher 
bacterial numbers in the ileum are the result of a lower peristalsis and low 
oxidation-reduction potential. Therefore, lactobacilli, streptococci, enterobacteriacae 
and anaerobic bacteria are able to establish themselves in the distal region of the small 
intestine. The main site of microbial colonization in the gastrointestinal tract is the colon. 
The slow intestinal motility in the colon with a transit time of up to 60 hours and low 
oxidation-reduction potential are responsible for the large numbers of bacteria present. 
The colon contains 10 -10 bacteria per gram of intestinal contents. More than 99% of 
the colonic microbiota are obligate anaerobes such as Bacteroides spp., Eubacterium, 
Bifidobacterium and Clostridium spp. (17). 



Enteric Pathogens 

Most intestinal bacterial infections are caused by enteric pathogens. The clinical 
symptoms usually associated with the intestinal infections include fever, abdominal pain 
and diarrhea. Enteric bacteria are capable of evading host defense factors such as gastric 
acidity, intestinal motility, the normal indigenous microbiota, mucus secretion, and 
specific mucosal and systemic immune mechanisms. 

In order for ingested pathogenic bacteria to infect the colon, they produce virulence 
factors. Enteric bacteria can be divided into four main categories based on the virulence 
factors that enable them to overcome the host defense. The first group of bacterial 
pathogens consists of Campylobacter jejuni, Yersinia enterocolitica, Shigella and 
Salmonella species. Their mechanism of virulence involves the mucosal invasion with 
intraepithelial cell multiplication resulting in cell death. The second group comprises 
enteric pathogens that produce cy to toxins which will in turn cause cell injury and 
inflammation. Microorganisms that produce cytotoxins include Clostridium difficile, 
enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC). The 
third class of pathogens secretes enterotoxins which will alter intestinal salt and water 
balance without affecting mucosal morphology. Vibrio cholerae, Shigella and 
enterotoxigenic E. coli produce such enterotoxins. The last category of enteric pathogens 
can only cause disease when they tightly adhere to the intestinal surface. The classic 
enteropathogenic E. coli as well as the enteroadherent E. coli is typical of this group. Both 
the small intestine and colon are primary sites for enteroadhesion (18). 



Mucosal Interactions and Gastrointestinal Microbiota 129 

DEVELOPMENT OF Gl TRACT NORMAL MICROBIOTA IN HUMANS 

The fetus in utero is sterile until birth. Colonization of the human body with a heterogenous 
collection of microorganisms from the birth canal begins at delivery. The Lactobacillus 
species constitute the major population of the vaginal microbiota and thus provide the initial 
inoculum to the infant during birth. In the case of caesarean section or premature infants, 
most microbes that are transferred to the newborn can be traced from the environment, i.e., 
from other infants via the air, equipment and nursing staff (19). Therefore, the type of 
delivery (passage through the birth canal versus caesarean section) as well as the type of diet 
(breast versus formula feeding) might affect the pattern of microbial colonization. 

The general pattern observed was that the facultative microorganisms appeared first 
and were subsequently followed by a limited number of anaerobes during the first two 
weeks (20). The types of bacterial strains that are capable of populating the GI tract are 
regulated through the limitation of the intestinal milieu, which changes with the successive 
establishment of the different bacteria. Hence, bacteria that are capable of oxidative 
metabolism, such as enterobacteria, streptococci and staphylococci, are among the first to 
proliferate in the gut. As the numbers of the facultative bacteria increase, they consume 
oxygen and lower the redox potential to negative values. These conditions are favorable 
for the anaerobic bacteria to multiply and reach much higher levels than that of the first 
week. Populations of bifidobacteria, Bacteroides and Clostridia, the commonly found 
anaerobes, increase with subsequent change of conditions in the GI tract. By the fourth 
week, the fecal microbiota of the breast-fed infants consists mainly of bifidobacteria and 
other groups to a lesser extent including enterobacteria, Clostridia, and Bacteroides. 
However, in formula-fed infants, bifidobacteria do not beome so dominant and a more 
complex microbiota develops. The differences between the breast-fed and formula-fed 
infants gradually disappear with the intake of solid food. By the twelfth month, the number 
of facultative anaerobes declines as the anaerobes begin to increase and form a stable 
population, resembling that of adults in numbers and in composition. By the age of two, 
the profile resembles that of an adult (19). In adults, the ratio of anaerobic to aerobic 
bacteria is 1000:1 (21). 

Adhesion of Bacteria 

The colonization of microorganisms in various niches is dependent on their ability to 
adhere to surfaces and substratum. Adhesion or adherence is defined as the measurable 
union between a bacterium and substratum. A bacterium is considered to have adhered to a 
substratum when energy is required to separate the bacterium from the substratum (22). 
Adhesion of a bacterium to a substratum, its colonization and finally possible invasion 
of the tissue is a multi-step process. It usually involves two or more kinetic steps. Firstly, the 
bacterium approaches the substratum via long distance interactions, such as van der Waals 
forces and electrostatic forces and becomes loosely attached (22). Complementary 
adhesion-receptor interaction leads to the formation of a bacterium-cell complex: 

Bacteria + Intestinal cell <=^ Bacterium — Intestinal cell complex (1) 

where k t and k_! are dissociation constants for the above reaction. At equilibrium, the 
concentration of the adhered bacteria (e x ) can be expressed as: 

e x = e m • x/(k x + x) (2) 

where e m is the maximum value of e x at saturated bacterial concentration (23). The value 



130 Chow and Lee 

of e m is equivalent to the concentration of adhesion sites on the mucosal surface and x is the 
concentration of bacterial cells present around the adhesion site. The dissociation constant, 
k x determines the affinity the bacterial cells have for the adhesion sites on the mucosal 
surfaces. Thus, the adhesion of a bacterium to the substratum is determined by two major 
properties: the concentration of the bacterium in the vicinity of the cell receptor (x in the 
above equation) and the affinity of the bacterium for the receptor (k x in the equation). 

Bacterial adhesion is crucial for invasive pathogenic microbes and may be important 
for certain commensals, prior to colonization of the intestinal mucosa. The receptors for 
bacterial adhesins are found in three groups of membrane consitituents: integral, peripheral 
and cell surface coat components. These receptors are chemically proteins, glycoproteins or 
glycolipids. They fulfill the criteria of a biological receptor because they exhibit specific 
binding followed by physiologically relevant responses. An example would be membrane- 
associated fibronectin acting as a receptor molecule for streptococci (22). 

Bacterial adhesion to substrata receptors could involve the specific adhesin-receptor 
interaction and non-specific interactions. The specific adhesion is defined as the association 
between the bacteria and substratum that requires rigid stereochemical constraints (22). 
Many bacteria have the ability to produce lectins (24), carbohydrate-specific proteins, 
which are usually expressed on the bacterial surfaces. Lectins are a subset of adhesins that 
recognize and bind to a defined carbohydrate sequence present on host glycoproteins. 
Previous studies reported that there were three main types of adhesin-receptor interactions. 
The first type was based on the carbohydrate-lectin recognition, the second kind involved 
protein-protein interaction and the third class, which is the least characterized, involved the 
binding interactions between hydrophobic moieties of proteins and lipids (25). 
A well-established example is the type 1 fimbriae (carrying adhesins) of E. coli which 
recognize D-mannose as the receptor site on the host mucosal surface (26). Binding 
of some Lactobacillus to human colonic cells is a mannose-specific adherence mechanism 
(27,28). Their similarity in binding specificity may contribute to competitive exclusion of 
enteropathogens by some strains of probiotic lactic acid bacteria. Lactic acid bacteria 
have been shown to exclude enteropathogens from the mucosal surface in in vitro 
studies (29-32). 

On the other hand, the non-specific adhesion is also an association between a 
bacterium and substratum that may involve the same forces involved in the specific 
adhesion. However, in non-specific adhesion, a precise stereochemical fit is not necessary. 
Non-specific interaction comprises the physiochemical forces such as van der Waals, 
electrostatic forces (33), hydrogen bonding (34), and hydrophobic interactions (35). 

The synthesis of adhesins can be switched on and off by the bacteria, depending on 
the environmental conditions, a process called phase variation (36). Phase variation has 
been demonstrated in Gram-negative bacteria. However, the environmental regulation of 
adhesin expression is likely to be present in some commensal and lactic acid bacteria also, 
since bacteria that are unable to regulate their adhesin expression are often inefficient 
colonizers (37,38). It has been suggested that the mucosal adhesive properties of the 
lactic acid bacteria is strain and host dependent, and the mucosal binding of human lactic 
acid bacteria are strain- and host specific (39,40). The adhesion and colonization of 
bifidobacteria have been suggested to be disease (allergy, cancer) dependent (41,42). The 
adhesion to the intestinal mucus of the fecal bifidobacteria from healthy infants was 
significantly higher than for allergic infants, suggesting a correlation between allergic 
disease and the composition of the bifidobacteria (41). Surprisingly, bifidobacteria, 
amongst other bacteria, were generally positively associated with increased risk of colon 
cancer in a study involving native Japanese and African patients (42). The ability of 
intestinal bacteria to persist on the intestinal mucosal surface may ultimately be determined 



Mucosal Interactions and Gastrointestinal Microbiota 131 

by their doubling time in the intestine to maintain a high local concentration. Slowly- 
dividing bacteria would be expected to be out-competed or washed-out with the intestinal 
contents (43). 



CROSS-TALK BETWEEN BACTERIA AND INTESTINAL 
EPITHELIAL CELLS 

As discussed in chapter 5, some ingested probiotic bacteria have shown immunomo- 
dulatory properties (44-46). Both commensal and pathogenic bacteria possess recognized 
structures named pathogen-associated molecular patterns (PAMPS). These recognized 
structures are essential for the microbe, mostly constitutively expressed and shared by the 
same group of microorganisms. PAMPS that are characterized to date include 
N-formylated peptide (47), lipopolysaccharides (LPS) (48), and lipopeptides (49), more 
recently described PAMPS are flagellin (50) and unmethylated segments of CpG DNA 
(51). Even though unmethylated segments of CpG DNA are not a cell surface structure, it 
serves to differentiate the microorganism from the host. Therefore, they epitomize the 
ideal targets for the innate immune system to identify the presence of infectious agents 
with a limited numbers of receptors. 

The best studied of the PAMPS is the glycolipid LPS, an important component of the 
outer membrane of Gram-negative bacteria. LPS is recognized by Toll-like receptor 
(TLR) 4, the first described member of the family of transmembrane TLR molecules that 
play a central role in the transcription activation of host defense mechanisms, such as 
chemokine and cytokine secretion, and the expression of costimulatory molecules (52). 
TLRs are transmembrane receptors defined by the presence of leucine-rich repeats in the 
extracellular portion of the molecule and a Toll/IL-IR/resistance (TIR) cytoplasmic 
domain. The extracellular leucine-rich repeats are thought to function in ligand recognition, 
whereas the TIR domain works in signaling. Leucine-rich repeat domains are common to 
proteins that are involved in the recognition of foreign proteins. There are currently 10 
identified members of the mammalian TLR family (52). From recent publications (53), it 
has been shown that some types of intestinal epithelial cells express TLR 4. 

Upon activation of TLR 4 by LPS, a series of events lead to the activation of 
ubiquitin ligase TRAF6 by a unique self-polyubiquitination reaction. TRAF6 then 
activates the TAK1 complex (54). This step leads to the phosphorylation and activation of 
mitogen-activated protein kinase and the inhibitor kB kinase (IKK) complex (54,55). The 
IKK complex comprises two kinases, IKKa and IKKP, and one protein, NEMO. When 
activated, IKKP phosphorylates IkBoc, triggering its polyubiquitination and degradation 
(56,57). In the unstimulated state, the IkBoc interacts and traps NFkP in the cytosol. 
Degradation of IkBoc releases the NFkP to translocate into the nucleus and to activate 
proinflammatory and prosurvival gene expression. Therefore, TLR 4 activates multiple 
signaling pathways which will eventually lead to the production of cytokines and other 
factors to protect the host against infection (58). The expression level of TLR 4 in the 
intestine of patients with inflammatory bowel disease was found to be strongly 
up-regulated compared to the TLR 4 expression in healthy individuals. 

As for the other PAMPS such as N-formylated peptides, the cell surface receptors 
that recognized them are the heterotrimeric G-protein coupled receptors (59). 
N-formylated peptides play an important role in recruiting and activating inflammatory 
cells (60). They will eventually activate the NFkP pathway the same way as the TLR. 

On the other hand, enteric pathogens have also evolved mechanisms to evade the 
immune recognition and defense. Helicobacter pylori, the etiological agent of gastritis and 



132 Chow and Lee 

stomach cancer, expresses hypoacylated LPS to avoid recognition by the human 
TLR4/MD2 module (61). Other pathogens like Yersinia pseudotuberculosis have 
developed ways to down-regulate TLR 4 signaling by injecting proteins to abolish the 
signaling leading to NFk(3 activation (52). 

At the beginning of the chapter, we mentioned that the gastrointestinal tract is 
colonized by huge, complex and dynamic populations of microorganisms. Hence, the 
molecular pattern recognition of the epithelial cells of the gastrointestinal mucosa needs to 
be tightly regulated so as to avoid an extreme immune response and uncontrolled 
inflammatory reaction. The exact mechanism by which they do this still remains to be 
elucidated. However, recent studies have shed light into this area of interest. The 
mechanism by which one TLR, TLR 5, achieved this feat is due to the fact that gut 
epithelial cells express TLR 5 only on their basolateral surfaces. Therefore only those 
bacteria that breached the epithelial cells or have translocated flagellin across the epithelia 
will activate the receptor (62). 

Using a gnotobiotic mouse model it was shown that Bacteroides thetaiotaomicron is 
able to induce the production of oc-L fucose on intestinal epithelial cells via a regulator, 
FucR, as a molecular sensor of L-fucose availability (3,68). FucR coordinates expression 
of an operon encoding enzymes in the L-fucose metabolic pathway in the bacteria with 
expression of another locus that regulates production of fucosylated glycans in the 
intestinal enterocytes. By tightly coordinating presentation of host-derived fucose with 
the rate of fucose utilization, an excess of epithelial fucose is avoided. This may minimize 
the risk of encroachment by pathogens that use fucosylated glycans as receptors for their 
adhesins (69). 

Certain pathogenic bacteria require intimate contact with the host to cause disease. 
E. coli (EPEC) is one such pathogen which requires intimate attachment to the host cells for 
maximum virulence to occur. There are a few factors which facilitate the cross-talk between 
the microorganism and the host epithelial cells and this involves the EPEC-secreted 
proteins, the type-three secretion system and the expression of outer membrane protein, 
intimin (64,65). The release of extracellular protein via the type-three secretion system is 
necessary for the formation of attaching lesions by EPEC. The attachment of bacteria is by 
means of intimin binding to a 90 kDa tyrosine phoshorylated protein in the host membrane. 
This receptor is known as translocated intimin receptor (Tir) and is of bacterial origin; it is 
translocated on to the host membrane where its tyrosine residues become phosphorylated 
and binds to intimin. Subsequent signal transduction events that occur within the host cells 
are the activation of protein kinase C, inositol triphosphate and calcium release. This leads 
to the formation of an actin-rich pedestal that forms a dome-like anchoring site for the 
bacteria which is an essential feature of EPEC pathogenesis (63). 

There is evidence to suggest that in some strains of Lactobacillus reuteri, mucus- 
binding adhesion could be induced by the presence of mucin glycoproteins and solid 
substratum (66). 



CONCLUSION 

The gastrointestinal tract is a highly dynamic ecosystem where interaction of the 
microbiota with the host mucosa plays an important role. Thus, it not only functions to 
digest food and absorb nutrients; it is also the major site where communication between 
microbes, and also between microbiota and their host takes place. 

Probiotics and prebiotics offer dietary means to support the balance of intestinal 
microbiota. They may be used to counteract local immunological dysfunctions, to stabilize 



Mucosal Interactions and Gastrointestinal Microbiota 133 

the gut mucosal barrier function, to prevent infectious succession of pathogenic 
microorganisms or to influence intestinal metabolism. However, many of the proposed 
mechanisms still need to be validated in human clinical trials (67). Future research on 
commensal microbiota interactions with mucosal surfaces of the host should focus on the 
cross-talk and determining the signaling mechanisms involved. 



REFERENCES 

1 . Mitsuoka T. The human gastroinstestinal tract. In: Brian JB Wood, ed. In: Lactic Acid Bacteria, 
Vol. 1. London and New York: Wood Publisher Elseivier Applied Science, 1992:69—1 14. 

2. Hoskins LC. Mucin degradation in the human gastrointestinal tract and its significance to 
enteric microbial ecology. Eur J Gastroenterol Hepatol 1993; 5:205. 

3. Bry L, Falk PG, Midtvedt T, Gordon JI. A model of host- microbial interactions in an open 
mammalian ecosystem. Science 1996; 273:1380-1383. 

4. Ross MH, Kaye GI, Pawlina W. Digestive system III: esophagus and gastrointestinal tract. 
Histology: A Text and Atlas. Philadelphia: Lippincott Williams & Wilkins, 2003 pp. 474-531. 

5. Gartner LP, Hiatt JL. Digestive system III — alimentary canal. Color Textbook of Histology. 
Philadelphia: W.B. Saunders Company, 1997 pp. 325-335. 

6. Junqueria LC, Carneiro J, Contopoulos AN. Digestive tract. In: Junqueria LC, Carneiro J, 
Contopoulos AN, eds. Basic Histology: Text and Atlas. Los Altos. CA: Lange Medical 
Publications, 1998 pp. 272-303. 

7. Allen A, Carroll NJ. Adherent and soluble mucus in the stomach and duodenum. Dig Dis Sci 
1985; 30:55S-62S. 

8. Forstner JF, Oliver MG, Sylvester FA. Production, structure, and biologic relevance of 
gastrointestinal mucins. In: Blaser MJ, Smith PD, Ravdin JI, Greenberg HB, Guerrant RL, eds. 
Infections of the Gastrointestinal Tract. New York: Raven Press, 1995:71-88. 

9. Belley A, Keller K, Gottke M, Chadee K, Gottke M. Intestinal mucins in colonization and host 
defense against pathogens. Am J Trop Med Hyg 1999; 60:10-15. 

10. Cone RA. Mucus. In: Orga PL, Mestecky J, Lamm ME, Strober W, Bienenstock J, McGhee JR, 
eds. Mucosal Immunology. New York: Academic Press, 1999:43-60. 

1 1 . Kraehenbuhl JP, Neutra MR. Molecular and cellular basis of immune protection of mucosal 
surfaces. Physiol Rev 1992; 72:853-879. 

12. Savage DC. Microbial ecology of the gastrointestinal tract. Ann Rev Microbiol 1977; 
6:155-178. 

13. Alexander M. Microbial Ecology. New York: Wiley, 1971 pp. 3-21. 

14. Nord CE, Hermdah I A, Kager L. Antimicrobial induced alteration of the human oropharyngeal 
and intestinal microflora. Scand J Infect Dis Suppl 1986; 49:64-72. 

15. Trenschel R, Peceny R, Runde V, et al. Fungal colonization and invasive fungal infections 
following allogeneic BMT using metronidazole, ciprofloxacin and fluconazole or ciprofloxacin 
and fluconazole as intestinal decontamination. Bone Marrow Transplant 2000; 26:993-997. 

16. Tannock GW. Normal Microflora: An Introduction to Microbes Inhabiting the Human Body. 
U.K.: Chapman and Hall, 1995. 

17. Herbert MK, Holzer P, Roewer N. Problems of the Gastrointestinal Tract in Anesthesia, the 
Perioperative Period, and Intensive Care. New York; Berlin: Springer, 1999. 

18. Cohen MB, Giannella RA. Bacterial infections: pathophysiology, clinical features and 
treatment. In: Philips SF, Permberton JH, Shorter RG, eds. The Large Intestine: Physiology, 
Pathophysiology and Disease. New York: Raven Press, 1991:395-428. 

1 9. Conway PL. Microbial ecology of the human large intestine. In: Gibson GR, Macfarlane GT, eds. 
Human Colonic Bacteria: Nutrition, Physiology, and Pathology. Boca Raton: CRC, 1995:1-24. 

20. Stark PL, Lee A. The microbial ecology of the large bowel of the breast-fed and formula-fed 
infants during the first year of life. J Med Microbiol 1982; 15:189-203. 

21. Berg RD. The indigenous gastrointestinal microflora. Trends Microbiol 1996; 4:430-435. 



134 Chow and Lee 

22. Ofek I, Doyle R. Recent developments in bacterial adhesion to animal cells. In: Ofek I, 
Doyle R, eds. Bacterial Adhesion to Cells and Tissues. London: Chapman and Hall, 
1994:321-512. 

23. Lee YK, Lim CY, Teng WL, Ouwehand AC, Tuomola EM, Salminen S. Quantitative approach 
in the study of adhesion of lactic acid bacteria to intestinal cells and their competition with 
Enterobacteria. Appl Environ Microbiol 2000; 66:3692-3697. 

24. Mirelman D, Ofek I. Microbial lectins and agglutinins. In: Mirelman D, ed. Microbial Lectins 
and Agglutinins. New York: J. Wiley and Sons, 1986:1-19. 

25. Abraham SM, Sharon N, Ofek I. Adhesion of bacteria to mucosal surfaces. In: Orga PL, 
Mestecky J, Lamm ME, Strober W, Bienenstock J, McGhee JR, eds. Mucosal Immunology. 
New York: Academic Press, 1999:31-42. 

26. Schembri MA, Kjaergaard K, Sokurenko EV, Klemm P. Molecular charcterisation of the 
Escherichia coli FimH adhesion. J Infect Dis 2001; 183:S28-S31. 

o 

27. Adlerberth I, Ahrne S, Johansson ML, Molin G, Hanson LA, Wold AE. A mannose-specific 
adherence mechanism in Lactobacillus plantarum conferring binding to the human colonic cell 
line HT-29. Appl Environ Microbiol 1996; 62:2244-2251. 

28. Neesen J-R, Granato D, Rouvet M, Servin A, Teneberg S, Karlsson K-A. Lactobacillus 
johnsonii Lai shares carbohydrates-binding specificities with several enteropathogenic 
bacteria. Glycobiology 2000; 10:1193-1199. 

29. Bernet MF, Brassart D, Neeser JR, Servin AL. Lactobacillus acidophilus LAI binds to cultured 
human intestinal cell lines and inhibits cell attachment and cell invasion by enterovirulent 
bacteria. Gut 1994; 35:483-489. 

30. Chauviere G, Coconnier MH, Kerneis S, Darfeuille-Michaud A, Joly B, Servin AL. 
Competitive exclusion of diarrheagenic Escherichia coli (ETEC) from human enterocyte- 
like Caco-2 cells by heat-killed Lactobacillus. FEMS Microbiol Lett 1992; 70:213-217. 

3 1 . Gopal PK, Prasad J, Smart J, Gill HS. In vitro adherence properties of Lactobacillus rhamnosus 
DR20 and Bifidobacterium lactis DR10 strains and their antagonistic activity against an 
enterotoxigenic Escherichia coli. Int J Food Microbiol 2001; 67:207-216. 

32. Tuomola EM, Ouwehand AC, Salminen SJ. The effect of probiotic bacteria on the 
adhesion of pathogens to human intestinal mucus. FEMS Immunol Med Microbiol 1999; 
26:137-142. 

33. Busscher HJ, Weerkamp AH. Specific and non-specific interactions in bacterial adhesion to 
solid substrata. FEMS Microbiol Rev 1987; 46:165-173. 

34. Pimentel GC, McClellan AL. In: Freeman WH, ed. The Hydrogen Bond. San Francisco; New 
York: Reinhold Pub. Corp, 1960. 

35. Reid G, Servin AL, Bruce AW, Busscher HJ. Adhesion of three lactobacillus strains to human 
urinary and intestinal epithelial cells. Microbios 1993; 75:57-65. 

36. Klemm P. Fimbrial adhesions of Escherichia coli. Rev Infect Dis 1985; 7:321-340. 

37. Adlerberth I. Establishment of a normal intestinal microflora in the newborn infant. In: 
Hanson L, Yolken RH, eds. Probiotics, Other Nutritional Factors, and Intestinal Microflora. 
Philadelphia: Vevey/Lippincott-Raven Publishers, 1999:63-78. 

38. Duguid JP, Old DC. Adhesive properties of enterobacteriaceace. In: Beachey EC, ed. In: 
Bacterial Adherence, Receptors and Recognition, Vol. 6. London: Chapman and Hall, 
1980:185-216. 

39. He F, Ouwehan AC, Hashimoto H, Isolauri E, Benno Y, Salminen S. Adhesion of 
Bifidobacterium spp. to human intestinal mucus. Microbiol Immunol 2001; 45:259-262. 

40. Kirjavainen PV, Ouwehand AC, Isolauri E, Salminen SJ. The ability of probiotic bacteria to 
bind to human intestinal mucus. FEMS Microbiol Lett 1998; 167:185-189. 

41. He F, Ouwehand AC, Isolauri E, Hashimoto H, Benno Y, Salminen S. Comparison of mucosal 
adhesion and species identification of bifidobacteria isolated from healthy and allergic infants. 
FEMS Immunol Med Microbiol 2001; 30:43-47. 

42. Moore WE, Moore LH. Intestinal floras of populations that have a high risk of colon cancer. 
Appl Environ Microbiol 1995; 61:3202-3207. 



Mucosal Interactions and Gastrointestinal Microbiota 135 

43. Lee YK, Ho PS, Low CS, Arvilommi H, Salminen S. Permanent colonization by Lactobacillus 
casei is hindered by low rate of cell division in mouse gut. Appl Environ Microbial 2004; 
70:670-674. 

44. Link-Amster H, Rochat F, Saudan KY, Mignot O, Aeschlimann JM. Modulation of a specific 
humoral immune response and changes in intestinal flora mediated through fermented milk 
intake. FEMS Immunol Med Microbiol 1994; 10:55-63. 

45. Schiffrin EJ, Rochat F, Link-Amster H, Aeschlimann JM, Donnet-Hughes A. Immunomodula- 
tion of human blood cells following the ingestion of lactic acid bacteria. J Dairy Sci 1995; 
78:491-497. 

46. Haller D, Blum S, Bode Ch, Hammes WP, Schiffrin EJ. Activation of human PBMC by non- 
pathogenic bacteria in vitro: evidence of NK cells as primary targets. Infect Immun 2000; 
68:752-759. 

47. Anton P, O' Conell J, O'Conell D, et al. Mucosal binding sites for the bacterial chemotactic 
peptide, formyl-methionyl-leucyl-phenylalanine (FMLP). Gut 1998; 42:374-379. 

48. Poltorak A, He X, Smirnova I, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr 
mice: mutations in Tlr4 gene. Science 1998; 282:2085-2088. 

49. Takeuchi O, Sato S, Horiuchi T, et al. Cutting edge: role of Toll-like receptor 1 in mediating 
immune response to microbial lipoproteins. J Immunol 2002; 169:10-14. 

50. Gerwitz AT, Simon PO, Jr., Schmitt CK, et al. Salmonella typhimurium translocates flagellin 
across intestinal epithelia inducing a proinflammatory response. J Clin Invest 2001; 
107:99-109. 

51. Di Martino P, Girardeau JP, Der Vartanian M, Joly B, Darfeuille-Michaud A. The central 
variable V2 region of the CS31A major subunit is involved in the receptor-binding domain. 
Infect Immun 1997; 65:609-616. 

52. Backhed F, Hornef M. Toll-like receptor 4-mediated signaling by epithelial cell surfaces: 
necessity or threat? Microbes Infect 2003; 5:951-959. 

53. Abreu MT, Arnold ET, Thomas LS, et al. TLR4 and MD-2 expression is regulated by 
immune-mediated signals in human intestinal epithelial cells. J Biol Chem 2002; 
277:20431-20437. 

54. Wang C, Deng L, Hong M, Akkaraju GR, Inoue J, Chen ZJ. TAK1 is a ubiquitin-dependent 
kinase of MKK and IKK. Nature 2001; 412:346-351. 

55. Ninomiya-Tsuji J, Kishimoto K, Hiyama A, Inoue J, Cao Z, Matsumoto K. The kinase TAK1 
can activate the NIK-I kappa B as well as MAP kinase cascade in the IL-1 signaling pathway. 
Nature 1999; 398:252-256. 

56. Delhase M, Hayawaka M, Chen Y, Karin M. Positive and negative regulation of IkappaB 
kinase activity through IKKbeta subunit phosphoryation. Science 1999; 284:309-313. 

57. Hu Y, Baud V, Delhase M, et al. Abnormal morphogenesis but intact IKK activation in mice 
lacking the IKKalpha subunit of IkappaB kinase. Science 1999; 284:316-320. 

58. Zhang Y, Bliska JB. Role of toll-like receptor signaling in the apoptotic response of 
macrophages to Yersinia infection. Infect Immun 2003; 71:15 13—1519. 

59. Radel SJ, Genco RJ, De Nardin E. Structural and functional characterization of the human 
formyl peptide receptor ligand-binding region. Infect Immun 1994; 62:1726-1732. 

60. Snyderman R, Fudman EJ. Demonstration of a chemotactic factor receptor on macrophages. 
J Immunol 1980; 124:2754-2757. 

61. Chaudhary PM, Ferguson C, Nguey V, et al. Cloning and characterization of two 
Toll/Interleukin- 1 receptor-like genes TIL3 and TIL4: evidence for multi-gene receptor 
family in human. Blood 1998; 91:4020-4027. 

62. Gewirtz AT, Liu Y, Sitareman SV. Madara. Intestinal epithelial pathobiology: past, present, 
future. Best Pract Res Clin Gastroenterol 2002; 16:851-867. 

63. Lu L, Walker WA. Pathologic and physiologic interactions of bacteria with the gastrointestinal 
epithelium. Am J Clin Nutr 2001; 73:1124S-1130S. 

64. Luo Y, Frey EA, Pfuetzner RA, et al. Crystal structure of enteropathogenic Escherichia coli 
intimin-receptor complex. Nature 2000; 405:1073-1077. 



136 Chow and Lee 

65. Sinclair J, O' Brien AA. Cell surface-localized nucleolin is a eukaryotic receptor for the 
adhesion intimin-y of enterohemorrhagic Escherichia coli 0157: H7. J Biol Chem 2002; 
277:2876-2885. 

66. Jonsson H, Strom E, Ross S. Addition of mucin to the growth medium triggers mucus-binding 
activity in different strains of Lactobacillus reuteri in vitro. FEMS Microbiol Lett 2001; 
204:19-22. 

67. Holzapfel WH, Haberer P, Snel J, Schillinger U, Huis in't Veld JHJ. Overview of gut flora and 
probiotics. Int J Food Microbiol 1998; 41:85-101. 

68. Hooper LV, Xu J, Falk PG, Midtvedt T, Gordon JI. A molecular sensor that allows a gut 
commensal to control its nutrient foundation in a competitive ecosystem. Proc Natl Acad Sci 
U.S.A. 1999; 96:9833-9838. 

69. Ayabe T, Satchell DP, Wilson CL, Parks WC, Selsted ME, Ouellette AJ. Secretion of 
microbicidal alpha-defensins by intestinal Paneth cells in response to bacteria. Nat Immunol 
2000; 1:113-118. 



7 

The Metabolism of Nutrients and Drugs 
by the Intestinal Microbiota 



Barry R. Goldin 

Department of Public Health and Family Medicine, Tufts University School of Medicine, 
Boston, Massachusetts, U.S.A. 



INTRODUCTION 

The intestinal microbiota of humans is comprised of a complex ecosystem of metabolically 
active microorganisms that reside close to the mucosal surface of the intestine. The bacteria 
of the intestine can interact with substrates introduced orally or compounds entering the 
intestinal lumen via the bile, mucosal secretions, or systemically from the circulatory 
system. This chapter will review the bacterial reactions performed on nutrients and drugs 
entering the intestine. The composition and distribution of the intestinal microbiota will not 
be discussed, and the readers are referred to other chapters in this book and review articles 
that address this topic (1-4). It is, however, important to note that the intestinal microbiota at 
any given time weighs approximately 1 10 to 200 grams and consists of at least 400 different 
species. The number of bacterial cells is approximately ten times greater than the total 
number of cells comprising the human body. Although the mass of the intestinal 
microbiome is equivalent to that of a single kidney, the number and diversity of species 
affords the microbiota a diverse metabolic role in the human body. This chapter will review 
some of these reactions and implications of these transformations to the host; however, no 
attempt will be made to exhaustively review all known reactions carried out by the 
microorganisms that inhabit the gastrointestinal tract of humans and animals. 



GENERAL METABOLISM AND FUNCTION OF THE MICROBIOTA 

The bacteria of the intestinal microbiota are predominantly anaerobic with a small 
percentage of facultative anaerobes. Therefore, intestinal bacteria do not use oxygen as a 
terminal election acceptor, and derive their energy from anaerobic respiration or substrate 
level phosphorylation. The magnitude of energy derived is the difference in redox 
potential between the substrate, and the products formed (5,6). The major overall balance 
of the intestinal microbiota derives from the ability to convert available substrates, 
principally originating from oral ingestion by the host of nutrients, fiber, and intestinal 

137 



138 



Goldin 



secretions or endogenously host synthesized compounds entering the intestine via the bile 
into the biomass that makes up the microorganisms in the intestine. The total biomass is 
principally controlled by space constraints, transit time of the digesta, and substrate 
availability. In general, approximately 50% of the fecal mass is composed of intestinal 
microorganisms. In addition to the utilization of substrates derived from the host, the 
intestinal microbiota can provide the host with energy mainly in the form of short chain 
fatty acids, and nutritive benefit by producing certain vitamins. 

Metabolic Reactions of the Intestinal Microbiota 

In Table 1 the major chemical reactions performed by the microbiota are listed. Most of 
the bacterial reactions can be classified as reductive, hydrolytic, or removal of functional 
groups such as dehydroxylation and decarboxylation. These reactions are often catalyzed 
by specific bacterial enzymes. 



NUTRIENTS AND DIETARY PLANT COMPOUNDS 

Fermentation of Carbohydrates 

Carbohydrate fermentation is a major source of energy for the intestinal microbiota. It has 
been estimated based on the biomass of the microbiota in the intestine that 20-70 grams of 
carbohydrate or equivalent substrates based on similar energy density would be required to 
be fermented to provide a biomass steady state (5-8). This calculation takes into account 



Table 1 Reactions Performed by the Intestinal Microflora 



Types of reaction 



Example of substrate 



Hydrolytic reactions 

Glucuronides 

Glycosides 

Amides 

Esters 

Sulfamates 

Nitrates 
Reductive reactions 

Nitrocompounds 

Azocompounds 

Double bonds 

Aldehydes 

N-oxides 
Nitrosation 

Amines 
Removal of functional group 

C-hydroxy 

N-hydroxy 

Carboxyl 

Methyl 

Amine 

Chlorine 



Phenolphthalein-glucuronide 

Cellobiose 

Methotrexate 

Acetyldigoxin 

Amygdalin 

Pentaerythritol trinitrate 

1-nitropyrene 
Direct red 2 

Polyunsaturated fatty acids 
Benzaldhydes 
4-Nitroquinoline- 1 -oxide 

Dimethylamine 

Bile acids 

N-hydroxyfluorenyl-acetamide 

Amino acids 

Biochanin A 

Amino acids 

DDT 



Abbreviation: DDT, dichloro-diphenyl-trichloroethane. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 139 

that bacteria are excreted daily, and that the normal intestinal transit time varies between 
48 and 72 hours. In "Western societies," such as the United Kingdom, the major intestinal 
bacterial carbohydrate substrates available are non-starch polysaccharide 12 grams, 
oligosaccharides 5 grams, simple sugars less than 5 grams, resistant starch 4 grams, and 
fermentable polysaccharides from intestinal mucus unknown (5,9). The amount of 
carbohydrate derived from colonic mucus available for bacterial fermentation is limited 
based on the fact that elemental diets support a very low bacterial biomass (10,1 1). All of 
these sources of carbohydrates are not readily digested and absorbed by humans, and thus 
arrive intact in the colon. 

Bacteroides, the most abundant bacterial genus in the ileum and colon can degrade, 
and ferment a number of different polysaccharides, including xylan, psyllium 
hydrocolloid, and numerous other plant polysaccharides (12,13). Bacteroides can also 
degrade host derived glycans such as chondroitin sulfate, mucin, heparin, hyaluronate, 
and glycosphingolipids. 

The fact that non-absorbable polysaccharides would not provide energy for the host 
adds a function to intestinal bacterial carbohydrate fermentation, namely salvaging energy. 
The major end products of bacterial fermentation in the intestine are the short chain fatty 
acids, acetate, propionate, and butyrate (14). For humans approximately 20-70 grams of 
carbohydrate would normally be fermented by intestinal flora per day. This would translate 
into 30-105 kcal per day or between 1.5 and 5% of typical human caloric intake. This 
percentage of caloric requirements varies greatly with the amount of fiber and other non- 
absorbable polysaccharides consumed per day. In developing countries, populations may 
derive larger benefits from bacterial metabolism in the intestine, as a result of greater 
consumption of plant fiber. 

Intestinal Bacterial Protein, Amino Acid, and Nitrogen Metabolism 

In monogastric animals there are several sources of nitrogen containing compounds that 
enter the large intestine, and thus are substrates for metabolic action by the microflora. The 
sources include incompletely digested dietary protein, protein from intestinal epithelial 
cells, and digestive secretions including digestive enzymes, glycoprotein mucins, free 
amino acids, and peptides including those derived from a bacterial origin. In addition, 
ammonia, urea, and nitrate are found in the ileal effluent. In terms of amounts and 
composition of nitrogen containing compounds entering the large intestine, it has been 
estimated in humans that 12-18 grams of protein enter the cecum from the ileum per day, 
and 2-3 grams per day of nitrogen (15). The approximate relative amount of nitrogen 
containing compounds in the large intestine is 48-51%, 34-42% peptides, and 10-15% 
urea/ammonia/nitrate, and free amino acids (15). The nitrogen sources in ileal effluent are 
primarily pancreatic enzyme protein and dietary protein residue. In contrast, in the feces the 
nitrogen compounds are more than 50% of bacterial origin (16). Therefore, although the 
balance of nitrogen is relatively well maintained between the amount entering and leaving 
the colon, the bacteria change the nature of the nitrogen containing compounds by utilizing 
these nitrogen compounds, and to large extent converting them into bacterial protein, which 
is found in the feces as intact bacteria, and as products of the lysed microorganisms. 

There are five major bacterial pathways for deaminating amino acids; four are 
designated as direct pathways, and one is considered an indirect pathway. The direct 
pathways are: reduction resulting in saturated fatty acid production; oxidation resulting in 
the formation of keto acids; hydrolysis causing the formation of an alpha-hydroxy fatty acid; 
and removal of the elements of ammonia, producing an unsaturated fatty acid (17). A fifth 
deamination pathway is known as the Strickland reaction, and is carried out by Clostridia 



140 Goldin 

that have little or no capacity to degrade single amino acids. As consequence the Clostridia 
degrade amino acids in pairs by a coupled oxidation-reduction reaction forming a keto acid 
and a saturated fatty acid. Reduction reactions are the major pathway for the degradation of 
amino acids in the intestine. The reduction products of the action of intestinal anaerobic 
organisms include: acetic, propionic, butyric, and isovaleric, isobutyric, and 2-methylbu- 
tyric acids (18). Other reductive products are ammonia, amines, carbon dioxide, and 
hydrogen (19). Some of the products that result from reductive degradation of aromatic 
amino acids include phenol, p-cresol, phenylactic acid, phenylpropionic acid, indole, 
indoleacetic acid, and indolepropionine acid. 

Decarboxylation is a second class of reactions that the intestinal microbiota perform 
in the course of the intestinal amino acid degradation (20). Bacterial decarboxylases act on 
amino acids to form amines and carbon dioxide. Many of these decarboxylases are 
specific, acting only on a single amino acid. There are a number of different genera of 
intestinal bacteria that have decarboxylase activity including: enterobacteria, enterococci, 
lactobacilli, Clostridia, Bacteroides, and bifidobacteria (19). Some of the specific products 
formed from bacterial decarboxylation are the formation of cadavarine from lysine, 
putrescine from ornithine, histamine from histidine, and tyramine from tyrosine. 

Intestinal bacteria can assimilate ammonia from the surrounding environment (20), 
and incorporate it into cell structures. Bacteria are also capable of ammonia production 
from peptides and amino acids (21). 



Bacterial Intestinal Lipid Metabolism 

In healthy humans, the vast majority of free fatty acid formed from dietary lipids is 
absorbed in the small intestine. The anaerobic bacterial microflora have the capability to 
hydrate, and hydrogenate double bonds found in unsaturated fatty acids (22,23). This is 
evidenced by the presence of 10-hydroxy stearic acid in human feces. The limited amounts 
of fatty acids that are transported to the lower intestinal tract relegate intestinal bacterial 
metabolism to minor significance in humans. 



Short Chain Fatty Acids 

Short chain fatty acids are not an important dietary nutrient, however, they are being 
discussed at this point because they are a significant end product of carbohydrate and 
amino acid bacterial metabolism. Short chain fatty acids are readily absorbed from the 
human colon, and facilitate the absorption of salt and water by the colon. Colonic 
epithelium derives 60-70% of its energy from short chain fatty acids with butyrate being 
the most important in this regard (24). Short chain fatty acids also stimulate mucosal 
growth in the colon. As stated previously, the major short chain fatty acids produced by 
intestinal bacterial fermentation are acetate, butyrate, and propionate. Additional end acid 
products include: lactate, succinate, and formate (25). The fate of these bacterially 
produced acid end products has been studied to varying extents. In humans, acetate is 
always found at a concentration of 50 micromolar in fasting venous blood. After a 
carbohydrate rich meal, these blood levels rise to 100 to 300 micromolar (5). The half-life 
of acetate in the blood is only a few minutes, and is taken up and metabolized in skeletal 
and cardiac muscle, brain, and adipocytes for lipogenesis (5). Acetate spares fatty acid 
oxidation but has only a small influence on glucose metabolism, and has no effect on 
insulin release in humans. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 141 

Intestinal Bacterial Synthesis and Metabolism of Vitamins 

The human intestinal bacteria can synthesize vitamin K, a member of the naphtoquinone 
family. The liver cannot synthesize the prothrombin complex, a blood-clotting factor, 
unless menaquinone, a substituted naphthoquinone, is present. The peptides that become 
the glycopeptides of the prothrombin complex require menaquinone for synthesis from the 
appropriate RNA codon. 

Bacteria found in the intestine can also synthesize homologues of menaquinone-7 
(vitamin K 2 ). The synthesized homologues range from the 6-isoprene unit side chain 
containing menaquinone-6 to menaquinone- 1 3 (26,27). The vitamin K bacterial reactions 
occur, in part, in the ileum, where the menaquinone is absorbed. The importance of bacterial 
synthesis of vitamin K has been demonstrated in human studies (28). Adult subjects 
maintained on a low vitamin K diet for several weeks did not develop a deficiency. When 
these subjects were treated with antibiotics such as neomycin that reduce the bacterial 
population of the intestine, a significant decrease in plasma prothrombin levels was noted 
(28,29). 

Most of the vitamin B 12 (cyanocobalamin) required by humans comes indirectly 
from the meat and milk of ruminants. The synthesis of B 12 in ruminants is exclusively of 
bacterial origin. The human intestinal microflora also synthesize vitamin B 12 as evidenced 
by the fecal secretion of approximately 5 micrograms per day. However, it appears most of 
the bacterially formed B i2 in humans occurs in the large bowel where absorption most 
likely does not occur due to lack of B 12 mucosal receptors. However, there is a study of 
healthy subjects from Southern India that reported the synthesis of vitamin B 12 in the 
jejunum and ileum, an area where absorption of the vitamin can occur (30). It was 
demonstrated that pseudomonas and klebsiella were two of the bacteria that synthesized 
B 12 in the small intestine. 

Biotin is synthesized by the human intestinal microflora. The administration of 
antibiotics can lower human urinary biotin levels. The importance of bacterial 
involvement in biotin synthesis has been demonstrated in germfree rats. The germfree 
animals require biotin in their diet; in contrast conventional rats can thrive without dietary 
biotin (22). 

Folic acid and thiamine B complex vitamins are also synthesized by bacteria in the 
intestinal tract. This synthesis does not solely provide for human requirements, and dietary 
sources of these vitamins are required to prevent deficiencies (31). 



Intestinal Bacterial Metabolism of Isoflavones and Lignans 

Dietary plant sources, such as vegetables, fruit, and cereals contain in addition to nutrients 
a large number of physiological active compounds. Many of these orally consumed 
compounds are transformed by the intestinal bacteria, which can result in either biological 
activation or deactivation of these substances. There are many plant-derived substances. In 
this section, two of these compounds of recent interest, the bacterial metabolism of the 
phytoestrogen compounds isoflavones and lignans are discussed. 

Isoflavones have weak estrogenic and antiestrogenic activities. Soybeans contain the 
highest levels of isoflavones in the human food chain. Other plant foods that contain 
isoflavones are pinto beans, navy beans, and chick peas, which have approximately two 
orders of magnitude lower levels. For populations consuming soy-based foods, the amount 
of isoflavones eaten daily is between 30 and 150 mgs. For daidzein, one of three major soy 
isoflavones, the intestinal bacteria can convert the parent compound into several end 
products. Among the end products are o-desmethylangolensin, equol, cis-4-equol, and 



142 Goldin 

dihydrodaidzein (32). There is a large individual variation in the ability of intestinal 
bacteria to metabolize daidzein. Studies have shown that production of equol does not 
occur in 30-40% of people fed soy isoflavones, and the remainder are active equol 
producers (33). The conversion of daidzein to equol can be of physiological importance 
since equol is a more potent estrogenic substance. The factors that control the extent of 
bacterial conversion of isoflavones in the intestine are unknown. 

Genistein, the isoflavone with the highest concentration in soy, is converted by 
intestinal bacteria to dihydrogenistein, and p-ethyl phenol. These reactions most likely 
lower or destroy the estrogenic activity of genistein. Glycetein, the third most prevalent 
isoflavone contained in the soybean, is bacterially converted to 5-hydroxy-, and 
5-methoxy-o-desmethylangolensin. There are other bacterial metabolites of isoflavones, 
and new end products are still being isolated. 

Lignans are found in relatively high concentrations in flaxseed, whole-grain 
products, vegetables, and sesame seeds (32). Lignans also exhibit weak estrogen and anti- 
estrogen activity, although these activities are lower than those found in isoflavones (32). 

The plant lignan precursors secoisolariciresinol and matainresinol are converted by the 
intestinal microflora to enterodiol and enterolactone, respectively (32). The physiological 
importance of these bacterial conversions are not clear. 



INTESTINAL BACTERIAL METABOLISM OF HOST ENDOGENOUSLY 
SYNTHESIZED COMPOUNDS 

Bacterial Cholesterol Metabolism 

The intestinal tract has a major impact on cholesterol metabolism (34-36). A major source of 
intestinal cholesterol comes from the de novo synthesis of the sterol compound. Cholesterol 
also can enter the intestine from dietary sources. It has been estimated that 34-57% of dietary 
cholesterol is absorbed from the intestine (37). In humans cholesterol synthesized by the 
intestinal cells is introduced into the lumen by exfoliation of these cells. An additional source 
of intestinal cholesterol is via biliary excretion. 

The fecal excretion of total neutral sterols in humans ranges from 350-900 mg/day, 
with a mean of 700 mg/day (38). Cholesterol accounts for about 20% of the total neutral 
sterols excreted in the feces, or about 150 mg/day. The normal range of cholesterol 
excreted by humans in the feces is between 75-200 mg/day. As discussed above, there are 
three sources of intestinal and fecal cholesterol: unabsorbed cholesterol from the diet 
which contributes 20%, bile which contributes 67%, and sloughed intestinal epithelial 
cells which contribute 13% of the total fecal cholesterol (39). 

The cholesterol that enters the intestine can be metabolized by bacterial microflora. 
Cholesterol is converted to 4-cholesten-3 one which is an intermediate formed by the 
oxidation of the 3 beta-hydroxyl group to a ketone, and isomerization of the 5-6 double 
bond to the 4-5 position. Coprostonone is formed by the reduction of the 4-5 double bond. 
The final reaction is the formation of coprostonal by reduction of the 3-beta to a hydroxyl 
group (34). 

The amounts of cholesterol and its metabolites found in feces are approximately 
20% cholesterol, 65% coprostonal, and 10% coprostanone (40). An additional 5% of fecal 
neutral sterols are made up of cholesterol, cholestanone, and epicoprostanol (40). 

Studies in Americans have shown that the majority of this population metabolizes 
cholesterol in the intestine (41). The distribution of intestinal bacterial conversion was 
bimodal. The majority of subjects converted 70-99% of cholesterol in their feces to 
metabolites, and a smaller group of individuals converted 0-19% of cholesterol (42). 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 143 

Intestinal Bacterial Metabolism of Bile Acids and Bile Pigments 

Cholesterol is a precursor of bile acids, and both are synthesized in the liver from two carbon 
units. Bile acids synthesized in the liver are conjugated through an amide bond to either 
glycine or taurine. The conjugated bile acids are deposited in the bile, and excreted into the 
upper small intestine. The bacterial conversion of bile acids primarily occurs in the distal 
ileum and colon. The bacterial reactions on bile acids include: the hydrolysis of the amide 
bond to release free bile acids from their corresponding glycine and taruine conjugates; an 
oxidoreduction of the hydroxyl groups at C3, C7, and C12 to form either oxo bile acids or 
alpha hydroxyl groups after the reduction of the beta groups (inversion products); and 
dehydroxylation at C7, and to a smaller extent at the C3 and C12 positions (43). 
The consequence of these reactions is the conversion of primary to secondary bile acids, and 
the re-absorption of free bile acids from the ileum, and to a lesser extent, from the colon. 
Only approximately 5% of bile acids are lost in the feces in each cycle as a result of bacterial 
deconjugation of bile acids (44). 



Bacterial Metabolism of Androgens and Estrogens 

Estrone, estradiol, and estriol are the three major estrogens that are excreted into the bile. 
These estrogens are conjugated to glucuronic acid and/or sulfate. Upon excretion of these 
conjugated estrogens from the bile into the small intestine the conjugates are available 
substrates for bacterial metabolism. The bacteria of the lower small intestine and colon can 
hydrolyze the estrogen conjugate releasing free estrogens (45). The nonconjugated 
estrogens are then subject to additional bacterial action. A major reaction involves 
oxidoreduction of the C17 position. Bacteria can convert estrone to estradiol, and the fecal 
flora can also convert 16 alpha hydroxy estrone to estriol (46). 

The intestinal bacteria can also modify androgens. The intestinal bacteria can 
reversibly oxidize and reduce the 3 -hydroxy group, and reduce steroid nuclear double bonds 
at the one and four positions. The latter reactions can result in several intercon versions of 
androgens (47). 

Other Steroid Hormone Bacterial Conversions 

Studies have shown that fecal organisms can modify corticosteroids. The corticosteroids 
undergo reduction in ring A, and undergo side-chain dehydroxylation separately or sequen- 
tially with the reduction (48). Cortisol is converted to 2 1-deoxycortisol, tetrahydrocortisol, and 
tetrahyro-21-deoxy Cortisol (48). Corticosterone is metabolized to tetrahydrocorticosterone, 
21 -deoxycorticosterone, and 3-alpha-hydroxy or 3 -beta-fry droxy epimers of tetrahydro-21- 
deoxycorticosterone (48). 

The intestinal bacteria can also transform progesterone similar to the reactions 
described above. Bacterial reduction of ring A can occur, as well as 16-alpha dehydroxylation, 
which can cause epimersation of the side chain (48). 



OTHER BACTERIAL REACTIONS 

Sulphate Metabolism 

The human colon contains Gram-negative anaerobes capable of reducing sulphates. The 
process is referred to as dissimilatory sulphate reduction, and results in the conversion of 
sulphates and sulphites to sulphides (49,50). The major bacterial genus that performs this 



144 Goldin 

reaction in the human colon is Desulfovibrio. Hydrogen gas in the colon is used as an 
electron donor in the formation of sulphides (50). The source of sulphates for bacterial 
reduction can come from food preservatives and drugs, and the levels of sulphides are 
highest in the sigmoid colon and rectum (51). Less than half of the human population 
appears to actively reduce sulphate in the large bowel (52). 

Aromatization 

Quinic acid is found in food products such as coffee, tea, fruits, and vegetables. Quinic 
acid has an aliphatic cyclic structure. Quinic acid is excreted in the urine as hippuric acid, 
an aromatic ring containing compound (53). Evidence that the intestinal bacteria are 
involved in the aromatization comes from the observation that hippuric acid is not formed 
when Quinic acid is given parenterally, and the formation of hippuric acid is inhibited 
when the antibiotic neomycin is given to humans (53). These findings strongly support the 
hypotheses that aromatization occurs as a result of intestinal bacterial action. 

Bacterial Carbon-Carbon Bond Cleavage 

The human intestinal flora has been shown capable of breaking the carbon bond between 
two of the rings of the product sennidin (54), which is found in senna and rhubarb. The 
product formed from this cleavage is rhein anthrone. The carbon-carbon cleavage is of 
physiological importance since this reaction is required for the observed laxative action of 
plant sennosides. 



BACTERIAL INTESTINAL FORMATION OF MUTAGENS 

In Table 2 are shown some of the mutagens formed as a result of intestinal bacterial 
reactions. The bacterial enzymes that catalyze the reactions that potentially can produce 
mutagens, carcinogens, and tumor promoters are also presented in Table 2. Some of the 
reactions discussed in this section also act on various drugs, and will again be discussed in 
the section on drug metabolism. 

Intestinal bacterial enzymes that have been implicated in the formation of 
carcinogens, mutagens, and tumor promoters include: beta-glucuronidase, beta-glucosidase, 
beta-galactosidase, nitroreductase, azoreductase, sulfatases, nitrosation, tryptophanase, 
1 -alpha-steroid dehydrogenase, and 7 -alpha-hydroxy steroid dehydroxylase (55). 

Table 2 Substrates Converted into Mutagens as a Result of Intestinal Bacterial Reactions 

Substrate Bacterial enzyme 

2-Nitrofluorene Nitroreductase 

Metronidazole Nitroreductase 

Trypan blue Azoreductase 

Ponceau 3R Azoreductase 

Cycasin Beta-glucosidase 

1 -Nitropyrene B eta-glucuronidase 

Cyclamate Sulfatase 

Dimethylamine Nitrosation 

Tryptophan Tryptophanase 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 145 

Glycosidase 

A classic example of the role of the intestinal flora in generating carcinogens is illustrated 
by the action of this bacterial enzyme on the plant derived compound cycasin (56). 
Cycasin is a naturally occurring beta-glucoside of methylazoxymethanol, extractable from 
the seeds and roots of cycad plants. It was observed that when Cycasin was fed to infant 
rats a number of different tumors developed. The Cycasin-induced tumors included 
hepatomas, renal sarcomas, squamous-cell carcinomas of the ear duct, and most frequently 
large bowel and duodenal adenocaricomas (56). The genetic strain of the rat did not appear 
to have a major influence on tumor development. It was, however, noted that the intestinal 
flora was required for tumorgenesis, since when Cycasin was given orally to germfree rats 
no tumors were observed (57). The discovery of the carcinogenicity of Cycasin led to 
experiments to test the precursor aglycones of Cycasin azoxymethane, azomethane, and 
dimethylhydrazine. These compounds were carcinogenic in conventional and germfree 
rats (58). The route of administration was not critical, and tumors developed after oral or 
subcutaneous administration (56). These results confirmed that the hydrolysis by the 
intestinal flora of the glycosidic bond was required for the activation of Cycasin. It was 
also observed that infant but not adult rats developed tumors when given Cycasin by 
intraperitoneal injection confirming the observation that tissue b-glucosidase disappeared 
in rats after 3 weeks of life (56). 

Many other plant natural products occur as glycosides. These glycosides do not 
demonstrate mutagenicity when tested in the Salmonella test; however, upon hydrolysis of 
the glucosidic linkages they become mutagenic. There have been several studies showing 
mixed fecal cultures or fecal isolates of Streptococcus faecium can convert non-mutagenic 
rutin (quercetin-3-D-beta-D-glucose-alpha-L-rhamnose) to quercetin (59). Quercetin has 
been shown to be mutagenic in the Ames salmonella assay. Red wine and tea contain 
glycosides of quercetin. 



Beta-Glucuronidase 

The formation of glucuronides in the liver is an important mechanism for detoxifying and 
enhancing excretion of a large number of orally ingested nutrients and their end products, 
other dietary compounds, and drugs, as well as endogenously synthesized compounds, 
such as estrogens. In humans many of these glucuronides depending on the structure of the 
aglycone, are excreted in the bile, and subsequently enter the duodenum. The glucuronides 
are then subject to bacterial deconjugation primarily in the ileum and colon. As a 
consequence of this bacterial deconjugation physiologically active, toxic, and 
carcinogenic compounds are regenerated. In addition to their formation in the intestine 
these compounds can be reabsorbed into the portal blood system. This results in recycling 
of these hydrolyzed glucuronides, and this process is referred to as the enterohepatic 
circulation. 

Several studies have shown that intestinal beta-glucuronidase can alter or amplify 
the biological activity of exogenous and endogenous compounds. 

The metabolism of the carcinogen N-hydroxyflourenylacetamide administered 
parenterally to conventional and germfree rats was studied by Weisburger et al. (60). 
Germfree rats excreted larger amounts of the glucuronides of N-hydroxyflourenylace- 
tamide in their feces compared to conventional animals. The cecal and fecal contents of 
conventional rats contained mostly unconjugated N- hydroxy flourenylacetamide, and its 
metabolites; in contrast most of these metabolites were glucuronide or sulfate conjugates 
in germfree animals. 



146 Goldin 

It has been shown that cell-free extracts derived from a number of different bacteria 
residing in the intestinal tract, including Bacteroides fragilis, Bacteroides vulgatus, 
Bacteroids thetaiotamicron, Eubacterium eligens, Peptostreptoccus, and Escherichia coli, 
were capable of increasing the mutagenic activity of bile from rats fed 1-nitropyrene via 
stomach tube. These extracts had beta-glucuronidose activity. Cell-free extracts of 
bacteria that were not able to enhance the mutangenicity of the bile did not possess beta- 
glucuronidose activity (61). These data support the hypothesis that glucuronides of 
1-nitropyrene metabolites entering the bile can be hydrolyzed by intestinal bacterial beta- 
glucuronidase to produce active deconjugated mutagenic products. 

Bacterial Azoreductase 

Azoreductase activity, which is of exclusively bacterial origin in the lumen of the 
intestine, catalyzes the reduction of the azo bond to cause the formation of aromatic 
amines. The highly reactive intermediates and end products have been shown to be 
mutagenic and carcinogenic. Azo dyes are used for coloring in the food industry, and as 
dyes and stains in textiles and other products. Water-soluble azo dyes are degraded by the 
intestinal microflora in the gastrointestinal tract (62). There is a 90% correlation between 
carcinogenicity and mutagenicity for aromatic amines and azo dyes tested by the Ames 
Salmonella test (63). The need for bacterial azoreductase and nitroreductase to activate 
mutagens, such as azocompounds in combination with intestinal mucosal microsomal 
enzymes has been demonstrated (64,65). 

The reduction of azocompounds by azoreductase is mediated through a free radical 
mechanism that produces intermediates that react with nucleic acids and proteins. The 
action of azoreductase on food dyes results in the release of phenyl-, and naphthyl- 
substituted amines. The amines generated in the lower intestine by bacterial action are 
probably oxidized by microsomal enzymes in the intestinal mucosa to carcinogens. 

Bacterial generation of mutagens from a number of azodyes has been demonstrated. 
Trypan blue, a widely used biologic stain, is converted to the mutagen O-toluidine by cell 
free extracts of Fusobacterium, an anaerobic organism found in the large intestine (66). 
Ponceau 3R, another biologic stain, is reduced Fusobacterium to 2, 4, 5-trimethylaniline 
which is mutagenic (67). Other azo dyes that have been shown to be transformed by 
bacterial reduction to mutagenic or carcinogenic products are direct black 38, direct red 2, 
and direct blue 15. Congo red lacks mutagenic activity, however, preincubation of dye 
with cecal bacteria generates mutagen-positive products (68). 

Bacterial Nitroreductase 

Nitroreductase similar to azoreductase is exclusively of bacterial origin in the lumen of the 
intestine. The enzyme is required for the mutagenic activity of nitrocompounds (64). 
Nitroreductase generates reactive nitroso and N-hydroxyintermediates in the course of 
converting aromatic amines. 1-nitropyrene is formed by the reaction of nitrogen oxides 
with the combustion product pyrene. The presence of 1-nitropyrene in diesel exhaust 
makes exposure to this compound a real risk. 1-nitropyrene is mutagenic in bacterial test 
systems, and carcinogenic when administered to the rat. When 1-nitropyrene was fed to 
conventional rats 5% to 6% of the dose was detected in the feces as 1-aminopyrene (69). 
When the same feeding experiment was performed with germfree rats no 1-aminopyrene 
was detected in the feces. The reduction of 1-nitropyrene to 1-aminopyrene is a carcinogen 
activation process, and the results cited above indicate that the intestinal microflora are 
important in the activation of 1-nitropyrene. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 147 

Mixed bacterial fecal specimens obtained from humans have been shown to reduce 
6-nitrochrysene to 6-aminochrysene, a compound that causes cancer in mice (70). The 
intermediate nitrosopolychic aromatic hydrocarbons generated in the conversion of the 
nitro groups to an amine are highly reactive compounds that can alter DNA. 

Bacterial Nitrosation 

Since the first report of the induction of liver cancer in rats fed dimethylnitrosamine (71), 
more than 80 different nitroso compounds have been identified as cancer-causing agents. 
The formation of nitrosamines results from the reaction of secondary amines with nitrite at 
acid pH. Nitrite is commonly added to cured meat and fish, and nitroso compounds have 
been measured in these foods (72). 

Bacteria have been implicated in the formation of N-nitroso compounds. Nitrite can 
be produced by the bacterial reduction of nitrate. High levels of nitrate are often present in 
leafy vegetables. The oral microbial flora of humans can reduce nitrate with the formation 
of nitrite. This reaction can raise nitrite levels in saliva to 6-10 ppm (73). 

It has been shown that when dimethylamine and sodium nitrite are incubated at pH 
7.0 under anaerobic conditions with rat intestinal microflora, the formation of 
dimethylnitrosamine was detected (74). These findings indicate that nitrosamines could 
be generated in the intestine, where the pH is nearly neutral, and the reaction would occur 
extremely slowly without bacterial enzyme catalysis. 

Bacterial Metabolism of Tyrosine and Tryptophan 

Tyrosine and tryptophan are amino acids that can be converted by bacterial reactions in 
toxins and carcinogens. The tryptophanase containing Bacteroides thetaiotamicron, an 
organism found in the intestine, can convert tryptophan to indole a compound with 
carcinogenic activity (75). 

Tyrosine is converted to phenol by aerobic intestinal bacteria, and to p-cresol by 
intestinal anaerobic bacteria. These metabolites of tyrosine are not found in the urine of 
germfree mice. Phenol and cresol have been shown to be tumor promoters in mice. 



BACTERIAL INTESTINAL DRUG METABOLISM 

In Table 3 are shown some representative natural and synthetic compounds that are or 
have been used as drugs that have been shown to be metabolized by the intestinal 
microflora. A description of the bacterial reactions involved for some of these drugs is 
given below. 

DOPA 

DOPA (3, 4-Dihydroxyphenylalanine) is used for the treatment of Parkinson's disease. 
DOPA replaces dopamine lost to Parkinson's disease because dopamine itself cannot cross 
the blood-brain barrier. Intestinal microbial metabolism of DOPA influences the dose 
required for the pharmacological action of this drug. The bacterial modification involves a 
dehydroxylation resulting in the removal of the hydroxyl group at the para position of the 
aromatic ring of phenylalanine (76). The product of this reaction, meta-hydroxylphenyl- 
acetic acid, is not active in the treatment of Parkinson's disease. In addition DOPA can be 
decarboxylated by intestinal bacteria forming inactive amines which can be detected in 



148 Goldin 

Table 3 Drugs, Supplements, and Additives Metabolized by the Intestinal Microflora 

Digoxin 

Diethylstilbesterol 

Estrogens 

Cyclamate 

Azulfidine 

3, 4-Dihydroxyphelalanine 

Amygdalin 

Metronidazole 

Caffeine 

Propachlor 

Morphine 

Buprenophine 

Oxazepum 

Phenolphthalein 

Warfarin 



urine. As a consequence of these bacterial reactions the dose of DOPA required to 
influence the symptoms associated with Parkinson's disease is greatly elevated. 



Salicylazosulfapyridine (Azulfidine) 

Azulfidine has been shown to be beneficial for the treatment and prevention of recurrence 
of ulcerative colitis. The drug structurally has sulfapyridine and aminosalicylate moieties 
attatched via an azo bond. The drug was originally designed to deliver the anti- 
inflammatory action of aminosalicylate, and the antimicrobial activity of sulfapyridine. 
The introduction of the azo bond linkage produced an unsymmetrical molecule that was 
non-absorbable in the upper intestine. 

It has been demonstrated that the azo bond of azulfidine is reductively cleaved by 
fecal bacterial cultures and that conventional but not germfree animals can also perform 
this cleavage reaction (77). The resultant products of the bacterial cleavage have been 
shown to have a different distribution (78). 5-aminosalicylate, because of its dual positive 
and negative charge is not absorbed from the colon, and is found almost exclusively in the 
feces. Sulfapyridine is readily absorbed from the intestine, and is excreted in the urine. 
This observation has been noted in humans and in rats (78). The evidence suggests that 
aminosalicylate is the active component for treating ulcerative colitis, and that the azo 
bond linkage affords an effective delivery system to the large intestine by being non- 
absorbable in the upper gastrointestinal tract, and then being slowly released by the action 
of bacteria in the lower ileum and large intestine. 



Metronidazole 

Metronidazole is an antibiotic which has a specificity against pathogenic anaerobes (79). 
Metronidazole structurally is a 5-nitroimidazole. The compound has been shown to be 
mutagenic in the Ames assay. This activity is lost when tester strains deficient in 
nitroreductase are used in the assay. The nitro group is reduced to amine group prior to ring 
cleavage which yields acetamide and N-(2-hydroxyethyl) oxamic acid. Therefore the 
amine intermediate generated by bacterial action is not stable, and breaks down to 
simpler metabolites. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 149 

Cyclamate 

Cyclamate (cyclohexylamine- N- sulfonate) was used as an artificial sweetening agent 
until it was banned. It had been reported that the intestinal flora can hydrolyze 
c-sulfonates, o-sulfonates, and N-sulfonates (54). Initially it was reported that Cyclamate 
could not be metabolized in the body. It was however, shown that Cyclamate could be 
converted to the bladder carcinogen cyclohexylamine as a result of the action of intestinal 
bacterial catalyzed N-sulfate ester hydrolysis (80). Cyclohexylamine was absorbed from 
the intestine, and excreted in the urine. Prolonged feeding of Cyclamate to rats increased 
the hydrolysis to the amine, and withholding cyclamate from the diet caused a decline in 
hydrolytic activity within 5 days (81). 



Digoxin 

The role of intestinal bacterial metabolism is important in the action of the cardiac 
glycoside drug digoxin (82). In order to form a pharmacologically active drug, the 
bacterial flora has to remove a trisacchride from the parent compound, releasing 
digoxigenin. The bacterial intestinal flora can further reduce the double bond in the lactone 
ring to form dihydrodigoxigenin (82). This compound is pharmacologically inactive. It 
was found that 36% of Americans in New York city given digoxin had the capability to 
reduce the double bond forming the inactive metabolite of digoxin (83). A total of 14% of 
New Yorkers excreted large amounts of metabolites of digoxin. These findings indicate at 
least 14%, and possibly a greater percentage of the population receiving digoxin will not 
achieve predicted serum levels resulting from the action of the intestinal microflora. 
Studies on a population residing in southern India indicated only 13.7% of those tested 
could reduce digoxin, and only 1% excreted large amounts of metabolites (83). These 
studies indicate that there are interethnic variations in the metabolic capacity of the 
intestinal microflora to reduce the double bond in the lactone ring of digoxin. This finding 
is not surprising based on the observation that Eubacterium lentum is exclusively 
responsible for the reductive reaction (82). 



Diethylstilbesterol 

Diethylstilbesterol is a highly active synthetic estrogen. This compound had been used 
prior to its being banned as a drug to prevent spontaneous abortions during pregnancy. It 
was subsequently discovered that this compound had serious side-effects, including 
reproductive problems, and vaginal cancer in the daughters of mothers given 
diethylstilbesterol during pregnancy. The metabolic fate of this compound has been 
studied (84). When diethylstilbesterol glucuronide was given orally to germfree rats, the 
compound was rapidly recovered in the feces. This results from poor absorption of the 
glucuronide from the intestine. In conventional rats the fecal recovery of diethylstilbes- 
terol is significantly reduced. The explanation for this finding is based on the ability of the 
beta-glucuronidase produced by intestinal microflora to generate the free compound from 
its glucuronide. Free diethylstilbesterol is more readily absorbed from the intestine. In 
conventional animals diethylstilbesterol makes approximately 1.5 passes through the 
enterohepatic circulation. The increased exposure resulting form the enterohepatic 
circulation can enhance the pharmacologic action, as well as the side-effects 
of diethylstilbesterol. 



150 Goldin 

Estrogens: Hormone Replacement Therapy and Birth Control 

Estrogens are used as a drug in a number of different human conditions. The most common 
are in hormone replacement therapy for treating menopausal symptoms and other 
consequences of aging in postmenopausal women, and for preventing conception in 
premenopausal women. 

The metabolism of estrogens involves an enterohepatic circulation that is dependent 
on intestinal bacterial deconjugation, and intestinal re-absorption similar to those of bile 
acids. Approximately 60% of circulating estrogens are conjugated in the form of 
glucuronides or sulfates, and are excreted in the bile (85-87). Deconjugation, a required 
step to cause intestinal mucosal cell re-absorption, is catalyzed by bacterial beta- 
glucuronidase and sulfatase. Approximately 97% of the estrogens excreted in the feces are 
in the deconjugated form, although virtually all of the estrogens in bile are conjugated. 

Another indicator of the involvement of the intestinal microflora in estrogen 
metabolism and pharmacokinetics is the observation that oral antibiotics exert an effect on 
the enterohepatic circulation of estrogens. It has been observed that urinary estriol 
concentration is decreased following oral administration of penicillin, ampicillin or 
neomycin (88). When antibiotics were given, fecal excretion of estrogens increased 
60-fold, and unconjugated estrogens increased 3-fold. 

These findings have a clinical significance. Failures of oral contraception pills have 
been associated with the use of oral antibiotics. Five pregnancies were reported among 88 
women receiving rifampin at the same time they were on oral contraceptive pills (89). 
Other antibiotics associated with birth-control failures are ampicillin, chloramphenicol, 
and sulfamethoxy-pyridazine (90). 



CONCLUSION 

This chapter has reviewed some of the important intestinal bacterial interactions with 
nutrients, endogenously synthesized hormones and other compounds, and orally ingested 
drugs. Since there is no available human germfree model to compare the magnitude of the 
importance of the intestinal flora in the various reactions cited in this chapter it is difficult 
to quantitatively evaluate. Based on animal models, the intestinal microflora are not an 
absolute requirement for survival, however, they do influence nutrient requirements, drug 
responses, and the effectiveness of various endogenously produced substances. Therefore, 
the metabolic potential of the intestinal microflora has to be considered in human 
biochemical and physiological activities and responses. 



REFERENCES 

1. Finegold SM, Atteberg HR, Sutter VL. Effect of diet on human fecal flora: comparison of 
Japanese and American diets. Am J Clin Nutr 1974; 27:1456-1469. 

2. Moore WEC, Holderman LV. Human fecal flora: the normal flora of 20 Japanese-Hawaiians. 
Appl Microbiol 1974; 27:961-979. 

3. Finegold SM, Sutter VL, Mathisen GE. In: Hentges DJ, ed. Human Intestinal Microflora in 
Health and Disease. New York: Academic Press, 1983:3-31. 

4. Goldin BR. Intestinal microflora: metabolism of drugs and carcinogens. Ann Med 1990; 
22:43-48. 

5. Cummings JT, Macfarlane GT. Role of intestinal bacteria in nutrient metabolism. J Parenter 
Enteral Nutr 1997; 21:357-365. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 151 

6. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Ann Rev Nutr 2002; 22:283-307. 

7. McNeil NI. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr 
1984; 39:338-342. 

8. Bergman EN. Energy contributions of volatile fatty acids from the gastrointestinal tract in 
various species. Physiol Rev 1990; 70:567-590. 

9. Cummings JH, Englyst HN. Fermentation in the large intestine and the available substrates. 
Am J Clin Nutr 1987; 45:1243-1255. 

10. McCamman S, Beyer PL, Rhodes JB. A comparison of three defined formula diets in normal 
volunteers. Am J Clin Nutr 1977; 30:1655-1660. 

1 1 . Winitz M, Seedman DA, Groff J. Studies in metabolic nutrition employing chemically defined 
diets. Am J Clin Nutr 1970; 23:525-545. 

12. Salyers AA, Gheruidini F, O'Brien M. Utilization of xylan by two species of human colonic 
bacteroides. Appl Environ Microbiol 1981; 41:1065-1068. 

13. Salyers AA, Harris CJ, Wilkins TD. Breakdown of psyllium hydrocolloid by strains of 
bacteroides ovatus from human intestinal tract. Can J Microbiol 1978; 24:336-338. 

14. Hoverstad T, Fausa O, Bjorneklatt A, Bohmer T. Short-chain fatty acids in the normal human 
feces. Scand J Gastroenterol 1984; 19:375-381. 

15. Chacko A, Cummings JH. Nitrogen losses from the human small bowel: obligatory losses and 
the effect of physical form of food. Gut 1988; 29:809-815. 

16. Stephen AM, Cumings JH. The microbial contribution to fecal mass. J Med Microbiol 1980; 
13:45-56. 

17. Barker HA. Amino acid degradation by anaerobic bacteria. Ann Rev Biochem 1981; 
50:23-40. 

18. Macfarlane GT, Allison C. Utilization of protein by human gut bacteria. FEMS Microbiol Ecol 
1986;38:19-24. 

19. Macfarlane S, Macfarlane GT. In: Gibson GR, Macfarlane GT, eds. Human Colonic Bacteria. 
Boca Raton: CRC Press, 1995:75-100. 

20. Metges CC, Petzke KJ, El-Khoury AE, Henneman L, Grant I. Incorporation of urea and 
ammonia nitrogen into ileal and fecal microbial proteins and plasma five amino acid in normal 
men and ileostomates. Am J Clin Nutr 1999; 70:1046-1058. 

21. Goldin BR, Lichtenstein AH, Gorbach SL. In: Shils ME, Olson JA, Moshe S, eds. Modern 
Nutrition in Health and Disease. 85th ed. Philadelphia: Lea and Febiger, 1994:569-582. 

22. Coates ME, Fuller R. In: Clarke RTJ, Bauchop T, eds. Microbial Ecology of the Gut Flora. New 
York: Academic Press, 1977:311-342. 

23. Cummings JH. Short chain fatty acids in the human colon. Gut 1981; 22:763-779. 

24. Cummings JH. In: Gibson GR, Macfarlane GT, eds. Human Colonic Bacteria. Boca Raton, FL: 
CRC Press, 1995:101-130. 

25. Hill MJ. In: Hill MR, ed. Microbial Metabolism in the Digestive Tract. Boca Raton, FL: CRC 
Press, 1986:31-41. 

26. Fernandez F, Collins MD. Vitamin K composition of anaerobic bacterial. FEMS Microbiol Lett 
1987;41:175-180. 

27. Ramotor K, Conly JM, Chubb H, Louie TJ. Production of menoquinones by intestinal 
anaerobes. J Infect Dis 1984; 150:213-218. 

28. Frick PG, Riedler G, Brogli H. Dose response and minimal daily requirement for vitamin K in 
man. J Appl Physiol 1967; 23:387-389. 

29. Udall JA. Human sources and absorption of Vitamin K in relation to anticoagulation stability. 
J Am Med Assoc 1965; 194:127-129. 

30. Albert MJ, Mathan VJ, Baber SJ. Vitamin B12 synthesis by human small intestinal bacteria. 
Nature 1980; 283:781-782. 

31. Wostmann BS. The germfree animal in nutritional studies. Ann Rev Nutr 1981; 1:257-279. 

32. Adlercreutz H, Mousovi Y, Loukovaara M, Hanaloimen E. In: Hochberg RB, Naftolin F, eds. 
The New Biology of Steroid Hormones, Vol. 72. New York: Raven Press, 1991:145-154. 



152 Goldin 

33. Rowland IR, Wiseman H, Saners TAB, Adlercreutz H, Bowery EA. Interindividual variation in 
metabolism of soy isoflavones and lignans: influence of habitual diet on equol production by 
the gut microflora. Nutr Cancer 2000; 36:27-32. 

34. Lichtenstein AH. Intestinal cholesterol metabolism. Ann Med 1990; 22:49-52. 

35. Eyssen HJ, Parmantier GG. Biotransformation of sterols and fatty acids by the intestinal 
microflora. Am J Clin Nutr 1974; 27:1329-1340. 

36. McNamara DJ, Prosa A, Miettinen TA. Thin layer and gas-liquid chromatographic 
identification of neutral steroids in human and rat feces. J Lipid Res 1981; 22:474-484. 

37. Grundy SM, Mok HYL. Determination of cholesterol absorption in man by intestinal perfusion. 
J Lipid Res 1977; 18:263-271. 

38. Grundy SM, Ahrens EH, Davignon J. The interaction of cholesterol absorption and synthesis in 
man. J Lipid Res 1969; 10:103-115. 

39. Ferezou J, Coste T, Chevalier F. Origins of neutral sterols in human feces studied by stable 
isotope labeling. Digestion 1981; 21:232-243. 

40. Reddy BS, Martin CW, Wynder EL. Fecal bile acids and cholesterol metabolites of patients 
with ulcerative colitis, a high risk group for development of colon cancer. Cancer Res 1977; 
37:1697-1701. 

41. Reddy BS, Wynder EL. Large-bowel carcinogenesis: fecal constituents of populations with 
diverse incidence rates of colon cancer. J Natl Cancer Inst 1973; 50:1437-1442. 

42. Wilkins TD, Hackman AS. Two patterns of neutral sterol conversion in the feces of normal 
North Americans. Cancer Res 1974; 34:2250-2254. 

43. Owen RW. In: Hill MR, ed. Microbial Metabolism in the Digestive Tract. CRC Press: Boca 
Raton, 1986:51-65. 

44. Plaa GL. In: Gillette JR, ed. Handbook of Experimental Pharmacology, Vol. 28. New York: 
Springer, 1975:139-140. 

45. Adlercreutz H, Martin F, Lehtinen T, Tilskanen MJ, Pulkkinen MO. Effect of ampicillin 
administration on plasma. Conjugated and unconjugated estrogen and progesterone levels in 
pregnancy. Am J Obstet Gynecol 1977; 128:266-271. 

46. Adlercreutz H, Martin F, Pulkknen M, et al. Intestinal metabolism of estrogens. J Clin 
Endocrinol Metab 1976; 43:497-505. 

47. Lombardi P, Goldin B, Boutin E, Gorbach SL. Metabolism of androgens and estrogens by 
human fecal microorganisms. J Steroid Biochem 1978; 9:795-801. 

48. Bolskenheuser VD, Winter J. In: Hentges DJ, ed. Human Intestinal Microflora in Health and 
Disease. New York: Academic Press, 1983:215-239. 

49. Gibson GR, Macfarlane S, Macfarlane GT. Metabolic interactions involving sulphate-reducing 
methanogenic bacteria in the human large intestine. FEMS Microb Ecol 1993; 12:117-125. 

50. Gibson GR, Cummings JH, Macfarlane GT. Use of a three-stage continuous culture system to 
study the effect of mucin on dissimilatory sulfate reduction and methanogenesis by mixed 
populations of human gut bacteria. Appl Environ Microbiol 1988; 54:2750-2755. 

51. McFarlane GT, Gibson GR, Cummings JH. Comparison of fermentation reductions in different 
regions of the human colon. J Appl Bacteriol 1992; 72:57-64. 

52. Pitcher MCL, Beatty ER, Gibson GR, Cummings JH. Incidence and activities of sulphate- 
inducing bacteria in patients with ulcerative colitis. Gut 1995; 36:A63. 

53. Asatoor MA. Aromatization of quinic acid and shikimic acid by bacteria and the production of 
urinary hippurate. Biochem Biophy Acta 1965; 100:200-202. 

54. Scheline RR. Metabolism of foreign compounds by gastrointestinal microorganisms. 
Pharmacol Rev 1973; 25:451-523. 

55. Goldin BR. In situ bacterial metabolism and colon mutagens. Ann Rev Microbiol 1986; 
40:367-393. 

56. Laqueur GL, Spatz M. Oncogenicity of cycasin and methylazoxy methanol. Gann Monograph 
Cancer Res 1975; 17:189-204. 

57. Laqueur GL, McDaniel EG, Matsumoto H. Tumor induction in germ free rats with 
methylazoxy methanol (MAM) and synthetic MAM acetate. J Natl Cancer Inst 1967; 
39:355-371. 



The Metabolism of Nutrients and Drugs by the Intestinal Microbiota 153 

58. Druckery H, Pressmann R, Matzkies F, Ivanovic S. Selective erzengung von darmkrebs bei 
ratten durch 1,2-dimethylhydrozin. Naturwissenschoften 1967; 54:285-286. 

59. Macdonald IA, Brussard RG, Hutchinson DM, Holdeman LV. Rutin induced beta-glucosidase 
activity in Streptococcus faecuim VGH-1 and streptococcus sp strain FRP-17 isolated from 
human feces: formation of the mutagen quercetin from rutin. Appl Environ Microbiol 1984; 
47:350-355. 

60. Weisburger JH, Grantham PM, Horton RE, Weisburger EK. Metabolism of the carcinogen 
N-hydroxy-N-2-fluorenylacetamide in germfree rats. Biochem Pharmacol 1970; 19:151-162. 

61. Morotomi M, Nanno M, Watanbe T, Sakurai T, Mutai M. Mutagenic activitaton of biliary 
metabolites of 1-nitropyrene by intestinal microflora. Mutat Res 1985; 149:171-178. 

62. Chung KT. The significance of azo-reduction in the mutagenesis and carcinogenesis of azo 
dyes. Mutat Res 1983; 114:269-281. 

63. Ames BN, McCann J, Yamoski E. Methods for detecting carcinogens and mutagens with 
salmonella/mammalian-microsome mutagenicity test. Mutat Res 1975; 31:347-364. 

64. McCoy EC, Rosenkranz HS, Mermelstein R. Evidence for the existence of a family of 
nitroreductase capable of activating nitrated polycyclics to mutagens. Environ Mutogen 1981; 
3:421-427. 

65. Karpinsky GE, Rosenkranz HS. The anaerobic-mediated mutagenicity of 2-nitrofluorene and 
2-aminofluorene for Salmonella typhimurium. Environ Mutogen 1980; 2:353-358. 

66. Hartman CP, Falk CE, Andrews AW. Azo reduction of trypan blue to a known carinogen by a 
cell-free extract of a human intestinal anoerobe. Mutat Res 1978; 58:125-132. 

67. Hartman CP, Andrews AW, Chung KT. Production of mutagen from ponceau 3R by a human 
intestinal anaerobe. Infect & Immunity 1979; 23:686-689. 

68. Reid TM, Morton KC, Wang CY, King CM. Conversion of congo red and 2-azoxyfluorence to 
mutagens following in vitro reduction by whole-all rat cecal bacteria. Mutat Res 1983; 
117:105-112. 

69. El-Bayoumy K, Fharma C, Louis Y, Reddy B, Hecht SS. The role of the intestinal microflora in 
the metabolic reduction of 1-nitropyrene to 1-aminopyrene in conventional and germfree rats 
and in humans. Cancer Lett 1983; 19:311-316. 

70. Manning BW, Campbell WL, Franklin W, Delclos KB, Cerniglia CE. Metabolism of 
6-nitrochyfesene by intestinal microflora. Appl Environ Microbiol 1988; 54:197-203. 

71. Magee PH, Barnes JM. The production of malignant hepatic tumors in the rat by feeding 
dimethylnitrosamine. Br J Cancer 1956; 10:1 14—122. 

72. Ender F, Ceh L. Occurrence of nitrosamines in foodstuffs for human and animal consumption. 
Food Cosmet Toxicol 1968; 6:569-571. 

73. Tannenbaum SR, Sinskey AJ, Weisman M, Bishop W. Nitrate in human saliva, its possible 
relationship to nitrosamine formation. J Natl Cancer Inst 1974; 53:79-84. 

74. Klubes P, Cerna I, Rabinowitz AD, Jondorf WR. Factors effecting dimethrylnitrosamine 
formation from simple precursors by rat intestinal bacteria. Food Cosmet Toxicol 1972; 
16:307-330. 

75. Chung KT, Fulk GE, Slein MW. Tryptophanase of fecal flora as a possible factor in the etiology 
of colon cancer. J Natl Cancer Inst 1975; 54:1073-1078. 

76. Goldin BR, Peppercorn MA, Goldman P. Contribution of host and intestinal microflora in the 
metabolism of L-DOPA by the rat. J Pharmacol Exp Ther 1973; 186:160-166. 

77. Peppercorn MA, Goldman P. The role of intestinal bacteria in the metabolism of 
salicylazosulfapyridine. J Pharmacol Exp Ther 1972; 181:151-162. 

78. Peppercorn MA, Goldman P. Distribution studies of salicylazosulfapyridine and its 
metabolities. Gastroenterology 1973; 64:240-245. 

79. Goldman P. Drug therapy: Metronidazole. N Engl J Med 1980; 303:1212-1218. 

80. Renwick AG, Williams RT. The fate of cyclamate in man and other species. Biochem J 1972; 
129:869-879. 

81. Wallace WC, Lethco ET, Brower EA. The metabolism of cyclamate in rats. J Pharmacol Exp 
Ther 1970; 175:325-330. 



154 Goldin 

82. Lindenbaum J, Rund DG, Butler VP, Tse-Eng D, Sala JR. Inactivation of digoxin by the gut 
flora: revised by antibiotic therapy. N Engl J Med 1981; 305:789-794. 

83. Muthan VI, Wiederman J, Dobkin JF, Lindenbaum J. Geographic differences in digoxin 
inactivation, a metabolic activity of the human anaerobic gut flora. Gut 1989; 30:971-977. 

84. Fisher LJ, Millburn P, Smith RL. The fate of 14 C stilleslerol in the rat. Biochem J 1966; 100:69. 

85. Eriksson H, Gustafsson JA. Excretion of steroid hormones in adults: steroids in feces from 
adults. Eur J Biochem 1971; 18:146-150. 

86. Sandberg AA, Slaunwhite WR, Jr. Studies on phenolic steroids in human subjects. II. The 
metabolic fate and hepato-biliary-enteric circulation of 14 C-estrone and 14 C-estradiol in 
women. J Clin Invest 1957; 36:1266-1278. 

87. Sandberg AA, Slaunwhite WR, Jr. Studies on phenobic steroids in human subjects. VII. 
Metabolic fate of estriol and its glucuronide. J Clin Invest 1965; 44:694-702. 

88. Adlercreutz H, Martin F, Tikkanen MJ, Pulkkinen MO. Effect of ampicillin administration on 
the excretion of twelve oestrogens in pregnancy urine. Acta Endocrinol 1975; 80:551-557. 

89. Reimer D. Rifampicin "pill" do not go well together. J Am Med Assoc 1974; 227:608. 

90. Dossetar J. Drug interaction with oral contraceptives. BMJ 1975; 4:467-468. 



8 

The Metabolism of Polyphenols 
by the Human Gut Microbiota 



Max Bingham 

Unilever Research and Development, Vlaardingen, The Netherlands 

SUMMARY 

Polyphenols are considered to be key active constituents of fruits and vegetables and 
responsible for many of the health protective effects of diets rich in these foods. While their 
structure varies considerably, following ingestion, most ( ~ 95%) persist to the colon where 
they encounter the human gut microbiota. Here they may undergo considerable structural 
alteration to compounds that may have enhanced biological properties or possibly degraded 
into inert metabolites and excreted. As such, the human gut microbiota may have a 
significant influence on the final outcomes of polyphenol ingestion. Moreover, inter- 
individual variation in the composition of the microbiota means that certain compounds are 
metabolized in different ways, and this is reflected in the considerable variability seen in 
excreted polyphenol metabolites. Consequently, polyphenols as active ingredients in 
functional foods may turn out to be beneficial for only a certain proportion of the 
population. Clearly, this may further have an impact on disease risk and health protection. 
This chapter considers the potential role of the human gut microbiota in polyphenol 
metabolism and highlights the level of current understanding of this process. 



INTRODUCTION 

Interest in the role of polyphenols in health has never been greater. Responsible for much 
of the flavor, texture, and appearance of fruits, vegetables, pulses, and grains, polyphenols 
are also considered to be largely responsible for many of the positive health effects of a 
diet rich in these particular food groups. In particular, epidemiologic studies suggest a 
protective effect of fruits and vegetables against cancer and coronary heart disease (1-3). 
In addition to antioxidant properties, polyphenols show a number of interesting activities 
in animal models and within in vitro systems. These effects include scavenging free 
radicals, nitric oxide regulation, apoptosis induction, inhibition of cell proliferation and 
angiogenesis and phytoestrogenic activity (4-7). As such these effects may contribute to 
their potentially protective role in cancer and coronary heart disease. And yet, the question 
remains of whether these types of studies are relevant in humans since substantial 
proportions of ingested polyphenols persist to the colon and may undergo extensive 
metabolism in the gut prior to absorption (8,9). This may further explain the failure of 

155 



156 Bingham 

many studies that sought to detect increases in antioxidant capacity in plasma following 
diets rich in polyphenols. 

Significantly, our understanding of these processes is limited but a considerable 
focus of attention on this issue has recently been established. This is following the 
realization that achievable concentrations of polyphenols in circulation may be 
significantly affected by the metabolic activities of the human gut microbiota (9,10). 
Overall, such processes may represent a significant factor in determining the final health 
outcomes of a diet rich in fruits, vegetables, pulses, and grains. We consider how the 
human gut microbiota can influence the bioavailability of polyphenols and establish the 
extent to which variations in microbiota composition between individuals may affect 
such processes. 



TYPES OF POLYPHENOLS AND MICROBIAL METABOLISM 

Polyphenol compounds are ubiquitous in the plant kingdom. They are secondary plant 
metabolites since they are not required in their primary metabolism. Rather, these 
compounds are essential for appearance, taste, stability, and often the protection of plant 
tissue. They have a wide variety of structures, chemical characteristics and to date several 
thousand compounds of this nature have been identified in higher plants (3). A more 
limited proportion of these compounds are present in edible food crops. The wide variety 
of compound characteristics means they are often separated into different classes 
according to their structural properties (Fig. 1). 

An important issue in the study of polyphenols in the diet is that the most commonly 
consumed ones are not necessarily the most active within the human body. There are several 
possible reasons for this: they may have low intrinsic activity, they may be poorly absorbed 
from the intestine, extensively metabolized in the intestine or rapidly eliminated. In 
addition, the metabolites circulating in blood or reaching target organs, and those that result 
from hepatic or digestive processes may differ from their original substances in biological 
activity. It is therefore crucial to have an extensive knowledge of the bioavailability of 
polyphenols if the true health effects of these compounds are to be understood. 

Even though polyphenols exhibit a large structural diversity, the metabolism of 
these compounds occurs via a common pathway (1 1). A limited proportion of polyphenols 
(mostly as aglycones) is absorbed intact in the small intestine. The balance, mostly present 
as glycosides, persists to the colon where they may undergo extensive metabolism and 
structural alteration by the colonic microbiota. A diverse range of smaller molecular 
weight compounds result and these can be detected in plasma, urine, and feces in various 
forms (10,12,13). A consistent observation in studies of polyphenol metabolism is that 
considerable inter-individual variation is seen in both the types and amounts of polyphenol 
metabolites that result from polyphenol ingestion (14-16). In many cases it is thought that 
compositional variations in the colonic microbiota are responsible for this. Many factors 
can influence the development and composition of the microbiota, including the overall 
diet, drugs, age, xenobiotics, and host factors such as gastric secretions and luminal pH 
(17-19). By proxy, the composition of the microbiota may have significant influence on 
the metabolism of polyphenols and thus the final health outcomes of diets rich in fruits, 
vegetables, cereals, and grains. In the light of this we consider the fate of a number of 
representative polyphenols classes (phenolic acids, flavonoids, anthocyanins, and 
proanthocyanidins) to illustrate an important aspect of the extent to which the colonic 
microbiota activity can impact on health. 



The Metabolism of Polyphenols by the Human Gut Microbiota 



157 



Hydroxycinnamic acids 



R, 



O 



OH 

R 1 = OH: Coumaric acid 
R 1 = R 2 = OH: Caffeic acid 
R 1 = OCH 3 , R 2 = OH: Ferulic acid 



Isoflavones 




R 1 = H: Daidzein 
R 1 = OH: Genistein 



Stilbenes 

HQ 



/ \ 



\ 



HO 



\\ / 



-OH 



Hydroxybenzoic acids 

\ 

o 



R, 



OH 



R, 



R 1= R 2 =OH, R 3 =H: Protocatechuic acid 
R 1= R 2 =R 3 =H: Gallic acid 



Flavonoids 




OH O 

e.g. Quercetin 



e.g. resveratrol 




e.g. secoisolariciresinol ""y" 'O 

OH CK 



Figure 1 Example structures of main polyphenol groups. 

PHENOLIC ACIDS— HYDROXYCINNAMATES 
AND HYDROXYBENZOATES 



The phenolic acids are generally the lowest molecular weight polyphenol compounds. 
Hydroxycinnamates (Fig. 2) are a core class of polyphenols and are central to the 
biosynthetic pathways of polyphenols. Caffeic and quinic acids combine to form 
chlorogenic acid, which is found in many types of fruit and in high concentrations in 
coffee (20). Ferulic acid is the most abundant phenolic acid found in grains and may 
constitute the main dietary source of this compound. Hydroxybenzoic acids (Fig. 2) are 
less abundant in plants but are often found in red fruits, black radishes and onions. Tea is 
an important source of gallic acid containing 4.5 g/kg fresh weight tea leaves (21), whilst 
ellagic acid is a major polyphenol in some berry fruits. Hydroxybenzoic acids are also 
important components of complex hydrolysable tannins such as gallotannins in mangoes 
and ellagitannins in red fruits, hazelnuts, walnuts, pomegranates, and oak aged wines 
(from the barrels) (22-25). 

Human bioavailability studies for hydroxycinnamates reveal that between 0.3 and 
25% of ingested dose is excreted in urine (12). Chlorogenic acid (ingested as coffee) has 
been detected at low concentrations in urine samples (26,27) along with a range of smaller 
molecular weight secondary metabolites including ferulic acid, isoferulic acid, 
dihydroferulic acid, vannilic acid, 3,4-dihydroxyphenylpropionic acid, 3 -hydroxy hippuric 
acid, and hippuric acid (27-29). One third of ingested chlorogenic acid is absorbed in the 



158 



Bingham 



OH 



.OH 



1 ,2-dihydroxybenzene 
catechol 




HO. 



HO 




HO 




O 
benzoic acid 



O 



2,5-dihydroxybenzene acid 



phenylacetic acid 




,OH 



O 



3-hydroxyphenylacetic acid 



4-hydroxyphenylacetic acid 



OH 



O 



.OH 



3,4-dihydroxyphenylacetic acid 





3,-phenylpropionic acid 
hydrocinnammic acid 



3-(4-hydroxyphenyl)-propionic acid 






3,4-dihydroxycinnamic acid 3,4-dihydroxyphenylpropionic acid 

caffeic acid 



4-hydroxy-3-methoxycinnamic 

acid 

ferulic acid 



Figure 2 Examples of phenolic acids and flavonoid metabolites (e.g., hydroxycinnamic acids and 
hydroxybenzoic acids). 



small intestine, leaving the balance to persist to the colon where it is exposed to the gut 
microbiota (26,30). Inter-individual variations in excretion profiles in these studies suggest 
that hydroxycinnamates are substantially metabolized by the colonic microbiota. In vitro 
studies have also revealed that chlorogenic acid is extensively metabolized by the colonic 
microbiota (31). Using inocula from different volunteers, it was clearly demonstrated that 
degradation rates of chlorogenic acid and the production of the metabolites 3,4- 
dihydroxyphenylpropionic acid and 3 -hydroxy phenylpropionic acid varied considerably 
between volunteers. Meanwhile in studies with healthy human volunteers, over 50% of the 
ingested dose was excreted as hippuric acid (a potential microbial metabolite of 
chlorogenic acid), whilst in the same study individuals without a colon excreted a much 
smaller amount of aromatic acids. The balance was not metabolized and excreted as 
chlorogenic acid in the latter volunteers. The absence of a colon and therefore a substantial 
colonic microbiota in the volunteers and the apparent excretion of intact chlorogenic acid 
effectively demonstrate the necessity for and the metabolic capacity of the colonic 
microbiota (27). 



The Metabolism of Polyphenols by the Human Gut Microbiota 159 

Successful attempts have been made to identify colonic microbiota species capable 
of metabolizing hydroxcinnamates. Inter-individual differences in excretion profiles of 
volunteers imply that the composition of the resident microbiota may be important in 
determining this final profile. Given that some of these metabolites are considered to be 
potentially protective of health, knowledge of the identity of species responsible for such 
metabolic activity is valuable. It has been demonstrated that at least three colonic 
microbiota species (Bifidobacterium lactis, Lactobacillus gasseri, and Escherichia coli) 
can release hydroxy cinnamates from chlorogenic acid in the gut (32) as well as diferulic 
acid being released in the colon as a result of metabolism by esterase activity of the colonic 
microbiota (33,34). Given that free hydroxycinnmates (including ferulic, caffeic, and 
p-coumaric acids) exhibit antioxidant and anticarcinogenic properties in vitro and in 
animal models, and that various microbial metabolites can be absorbed readily (35), this 
supports the notion that some beneficial effects of hydroxycinnamtes can be ascribed to the 
metabolic activities and products of the colonic microbiota. 

A more limited set of studies has been carried out for hydroxybenzoates. 
Ellagitannins are polyphenols made up of subunits of ellagic acid (a hydroxybenzoate) 
and are thought to possess chemopreventative properties that might contribute to health 
benefits in humans (36-38). Their fate has been studied in 40 volunteers consuming a 
variety of foodstuffs known to contain high levels of ellagitannins (39). In all cases the 
ellagitannin microbial metabolite urolithin B (which may be antiangiogenic) could be 
identified although inter-individual variation in excretion rates was large. Furthermore 
they were able to identify high and low excretors of this compound in much the same way 
that consumers of soya can be differentiated by their ability to excrete equol (40,41). Again 
this observation indicates that the gut microbiota is likely to be important in the 
bioavailability of these potentially health-promoting compounds and that variations in the 
composition of the microbiota may dictate the production of a potentially health-promoting 
metabolite. At present, we are unaware of any studies designed to identify components of 
the colonic microbiota that are potentially responsible for the metabolism of ellagitannins. 



FLAVONOIDS 

Flavonoids are the most important class of polyphenols in plants. Over 6000 flavonoids 
have been identified so far (3) and their structural variety is based on the flavan or 
2-phenyl-benzo-dihydropyrane skeleton. Flavonoids are further differentiated into 
subclasses (Fig. 3). The metabolism of two of these classes are discussed here — flavonols 
and flavan-3-ols. 

Flavonols 

Flavonols are the most ubiquitous flavonoids in plants, with the main representatives being 
quercetin, kaempferol, myricetin, and isohamnetin, which are predominantly present as 
glycosides bound to a variety of sugar moieties. The richest sources are onions, curly kale, 
leeks, broccoli, and blueberries and are present at levels of approximately 30 mg/kg fresh 
weight although in certain circumstances can reach in excess of 1.2 g/kg fresh weight. Red 
wine and tea are also rich sources. 

In contrast to other classes of polyphenols, flavonols such as quercetin and 
kaempferol have received a larger amount of attention in terms of bioavailability over the 
past few years. This is largely because of their ubiquitous nature in food crops but also 
because a great deal of their apparent in vitro effects on health parameters have failed to be 



160 



Bingham 



Anthocyanidins 



Flavanones 




R 1= R 2 =H: Perlargonidin 

R^OH, R 2 =H: Cyanidin 
R 1= R 2 =OH: Delphinidin 
R^OCHg, R 2 =OH: Petunidin 
R 1= R 2 =OCH 3 : Malvidin 




R 1= H: R 2 =OH: Narigenin 
R 1= R 2 =OH: Eridocytol 
R.,=OH: R 2 =OCH 3 : Hesperetin 



Flavanols 



Flavonols 




R 1= R 2 =OH: R 3 =H: Catechins 
R 1= R 2 =R 3 =OH: Gallocatechins 




OH O 



R 2 =OH: R 1 =R 3 =OH: Kaempferol 
R 1= R 2 =OH: R 3 =H: Quercetin 
R 1= R 2 =R 3 =OH: Myricetin 



Trimeric procyanidin 



Flavones 





R 1= H: R 2 =OH: Apigenin 
R 1= R 2 =OH: Luteolin 



Figure 3 Example structures of various flavonoid groups. 



repeated in vivo (13). The majority of these compounds exist as glycosides in their original 
food matrices and thus reach the colon intact following ingestion. Here they can serve as 
substrate for the microbiota. Associations between the urinary excretion of simple phenolics 
such as hydroxyhippuric acid, hydroxyphenylacetic acid and 3-(hydroxyphenyl)-propionic 
acid, and a high flavonoid intake have been observed in a number of human studies 
(28,29,40-48) indicating that a substantial proportion of polyphenols undergo metabolism 
in the gut. In addition, the microbiota has also been confirmed as the major site for the 
release of free flavonol agly cones from their conjugated forms following cleavage of ester or 



The Metabolism of Polyphenols by the Human Gut Microbiota 161 

glycosidic forms (49). Quercetin derivatives are deconjugated and converted to 
hydroxyphenylacetic acids by the colonic microbiota in vitro (50). Confirming these 
observations, recent in vitro studies revealed the production of 3 -hydroxyphenylacetic acid 
and 3-(hydroxyphenyl)-propionic acid from rutin (a representative glycoside of quercetin) 
in human gut microbiota fermentation studies (31). An important observation in these 
studies was that the pattern of degradation varied considerably between donor fecal 
microbiota samples and with concentration of the initial substrate. This is significant since 
many of the compounds produced in this degradative process may have enhanced biological 
properties. 3,4-dihydroxyphenylacetic acid and 4-hydroxyphenylacetic acid have more 
effective antiplatelet aggregation activity than their precursors rutin and quercetin (51). 

Compositional variations in the microbiota may have a significant impact on the 
final metabolic products of flavonol metabolism. Indeed reports of studies designed to 
confirm these observations are now appearing. Eubacterium ramulus is capable of 
metabolizing quercetin both in vitro and in rats associated with the organism (8). In both 
cases, the isolate was capable of releasing quercetin from its glycosidic form and was then 
able to cleave the ring system of quercetin and produce mainly 3,4-dihydroxyphenylacetic 
acid. Further studies in humans revealed that E. ramulus is a common member of the 
human gut microbiota (52); its resident population level is dependant on flavonoid intake 
and the production of secondary metabolites of flavonoids (such as 3,4-dihydroxyphe- 
nylacetic acid) was greatest when E. ramulus populations where increased (15). 
Meanwhile E. ramulus has also been tested for its abilities to degrade other structurally 
related flavonoids including other flavonols, flavones, flavan-3-ols, and flavonones, and in 
certain cases, significant metabolism can occur (53). Clostridium orbiscindens, which is an 
obligate anaerobe commonly found in the intestinal tract, is also capable of cleaving the 
C3-C4 bond of quercetin to give 3,4-dihydroxyphenylacetic acid (54). In recent studies, it 
was also shown to degrade a range of other flavonols and flavanones in vitro and that it was 
present in 8 of 10 volunteers at levels of 1.87 X 10 to 2.5 X 10 cells/ g (55). At present, these 
are the most extensively published reports on the influence of microbiota composition in 
polyphenol metabolism and set the benchmark for future studies in other polyphenol classes. 



Flavan-3-ols 

Flavan-3-ols are found in most plants and the stereo isomers ( + )-catechin and ( — )- 
epicatechin are the most common monomeric flavan-3-ols in fruits. ( + )-gallocatechin and 
( — )-epigallocatechin are their corresponding 0-3 gallates and are rarer but found in 
certain seeds of leguminous plants, in grapes and in tea. Catechins are found in many types 
of fruit and red wine but by far the most abundant sources are green tea and chocolate (56). 
The bioavailability of flavan-3-ols differs markedly among the different catechins 
and appears to be related substantially to structure and degree of galloylation (12). Again 
due to their structure, a substantial proportion of ingested flavan-3-ols may persist to the 
colon where they encounter the colonic microbiota. The colonic degradation of flavan- 
3-ols such as catechin, epicatechin, and epicatechin gallate have been investigated 
previously (14,44,57-59) revealing that in contrast to other similar structures the 
heterocyclic C-ring is not cleaved per se. The hydroxy lation pattern of flavan-3-ols 
(5,1,3,3' ,4'-) has instead been suggested to enhance the opening of the heterocyclic ring 
after hydrolysis (60,61) and this results in the production of a large number of metabolites 
from the colonic microbiota: 3,4-dihydroxyphenylacetic acid, 3 -hydroxyphenylacetic acid, 
homo vanillic acid and their conjugates are derived from the B-ring and phenolic acids from 



162 Bingham 

the C-ring (61). In animal studies, phenylvalerolactones, and phenylpropionic acids have 
also been identified as degradation products (61). 

Antibiotic treatment in rats significantly alters the metabolism of catechin and 
decreases the urinary elimination of many of the compounds of flavan-3-ol metabolism 
indicating that an intact microbiota is necessary for the production of many of these 
compounds (57). At present there are limited studies (described by 8; see Flavonols) that 
have investigated the specific species that may be responsible for the conversion of flavan- 
3-ols, and data is limited on any possible inter-individual variation in final metabolic 
profiles. However, green tea catechins have been shown to cause a shift in bacterial 
populations in humans (62), pigs (63) and chickens (64), and this may have relevance to 
the overall polyphenol metabolic capabilities of the resident microbiota. Structurally, 
these compounds may exhibit substantial anti-oxidant activities and thus the influence of 
the composition of the resident microbiota and associated metabolic variations could 
impact on the overall health impact of flavan-3-ol ingestion. 



ANTHOCYANIDINS 

The red to purple colored anthocyanidins are responsible for a good portion of color in 
fruits and flowers. They are only present as glycosides or anthocyanins and their color is 
pH dependent. In the human diet, anthocyanidins are present in red wine, certain varieties 
of cereals, certain leafy and root vegetables (e.g., aubergines, cabbage, beans, onions, and 
radishes) and most abundantly in fruit. The content is generally proportional to the color 
intensity and may reach values of 2-4 g/kg fresh weight in blackberries and black currants. 
They are found mainly in the skin, except where the flesh is also colored. 

Anthocyanidins and anthocyanins have been reported previously as having several 
positive effects on health (35,65-72). Much of this evidence has been derived in vitro and 
very little is known about their bioavailability in vivo. Previous human and rat studies have 
reported very low recoveries of intact anthocyanins in urine (73). Very little is known of 
the specific fate of the balance of these compounds. Given their structure, it is likely that 
they will undergo substantial metabolism by the human gut microbiota in much the same 
way as any other flavonoid structure. And yet, studies performed in the 1970s indicated 
that degradation of anthocyanins by the microbiota occurs to a much more limited extent 
than with other flavonoid structures (61). However recent studies investigated in vitro 
whether the anthocyanin glycosides, cyanidin-3-glucoside, and cyanidin-3-rutinoside 
were deglycosylated and whether the resulting aglycones were degraded further to smaller 
phenolic compounds by colonic bacteria (74). Cyanidin-3-glucoside and cyanidin 
aglycone were identified as intermediary metabolites of cyanidin-3-rutinoside. Proto- 
catechuic acid was identified as a major metabolite at early stages of the fermentations 
along with a variety of other low molecular weight metabolites suggesting that the 
anthocyanins were converted by the gut microbiota. However, protocatechuic acid was 
also formed in vitro with the simple incubation of cyanidin with rat plasma in the absence 
of colonic microbiota (75). These experiments, although far from conclusive indicate that 
bacterial metabolism of anthocyanins can occur and is likely to involve the cleavage of 
glycosidic links and the breakdown of the anthocyanidin heterocycle — thus having a 
potential impact on the bioavailability of these compounds in vivo. However, significantly 
more investigation is needed before the real extent of the involvement of the microbiota is 
uncovered in terms of metabolism and the bioavailability of anthocyanins. 



The Metabolism of Polyphenols by the Human Gut Microbiota 163 

PROANTHOCYANIDINS 

Proanthocyanidins are dimers, oligomers, and polymers of flavan-3-ols and are formed by 
enzymatic or chemical condensation. These so-called "condensed tannins" contribute to 
astringent tastes in fruits (e.g., grapes, peaches, apples, pears, berries etc.), beverages (e.g., 
wine, cider, tea, beer etc.) and chocolate. At a lower degree of polymerization they are 
colorless and bitter to taste, but with greater polymerization the taste becomes astringent 
and the color yellow to brown. Proanthocyanidins purely consisting of catechin and 
epicatechin monomers are called procyanidins, which are the most common type of 
proanthocyanidins. Less abundant are the prodelphinidins, which include both epicatechin 
and gallocatechin monomers. 

Previous studies in rats have indicated that the bioavailability of procyanidins is low 
and characterized by a very low urinary recovery (0.5% ingested dose) (76). Procyanidin 
consumption in rats and in humans is associated with the production of several aromatic 
compounds including derivatives of phenylpropionic, phenylacetic, and benzoic acids 
(77,78). More recent studies have also established that consumption of proanthocyanidins 
from grape seed extract can result in a consistent increase in urinary excretion of 
3 -hydroxy phenylpropionic acid and 4-O-methylgallic acid. Inter-individual variation in 
excretion of 3-hydroxyphenylproionic acid was significant (79). The microbial 
metabolism of proanthocyanidins has never been studied in humans but the microbial 
origin of these compounds was established in vitro following incubation of 
proanthocyanidins with rat cecal contents (80) and human fecal microbiota (81). These 
studies utilized 14 C labeled proanthocyanidin oligomers and led to the formation of 
m-hydroxyphenylpropionic acid, m-hydroxyphenylacetic acid and their /^-hydroxy 
isomers, m-hydroxyphenylvaleric acid, phenylpropionic acid, phenyl acetic acid and 
benzoic acid. Attempts have been made in the past to identify intestinal bacteria that can 
degrade proanthocyanidins (82,83) although these studies actually failed. The impact of 
proanthocyanidins on colonic microbiota populations has been investigated in rat studies 
and revealed that there was a shift in the predominant bacteria present towards Gram- 
negative Enterobacteriaceae and Bacteroides species (84). Furthermore, proanthocyanidin 
intestinal absorption and microbial metabolism of some of the above metabolites fell as 
the degree of polymerisation increased (77,81). Thus studies on antioxidant and biological 
effects of proanthocyanidins are only useful when targeted at compounds with a low 
degree of polymerization. Larger compounds do not appear to be able to reach systemic 
circulation or be available for microbial metabolism that would result in significant 
production of readily absorbable phenolic acid metabolites. However, this does highlight 
that at least some of the purported health effects of proanthocy anidin-rich diets may be due 
to secondary metabolites rather than the original ingested compounds. 



PERSPECTIVES 

The role of dietary polyphenols in health and disease continues to be the focus of much 
academic and commercial research. Consumption of diets rich in polyphenols is generally 
thought to be beneficial to health and this has led to great excitement over the potential of 
diets, supplements, and pro-drugs based on polyphenol compositions. As we have 
discussed, much of the latest research surrounds the question of bioavailability since they 
must reach target tissues in a form that is viable and can have the desired effects. A major 
obstacle for these compounds, though, is the microbial mass in the colon since in many 
cases they persist intact to the colon, and are structurally ideal for metabolism by the 



164 Bingham 

human gut microbiota. This review highlights the extent to which certain polyphenol 
classes undergo metabolism and structural alteration in the colon, and suggests that much 
of the prescribed in vivo health benefits of polyphenols may be due to secondary 
metabolites of polyphenols rather than the original compounds. 

Well-designed studies have evaluated the need for an intact microbiota in 
polyphenol metabolism, although this is not so for all classes. Perhaps one of the most 
consistent observations in human bioavailability studies of dietary polyphenol compounds 
is how striking the inter-individual variations are in the types and amounts of metabolic 
breakdown products seen following polyphenol ingestion. The reasons for this have at 
present not been rigorously investigated, but it seems very likely that variations in 
composition of the resident microbiota between individuals is key. Numerous factors can 
influence the composition of the microbiota (19) and this in turn may affect the overall 
metabolic capabilities of this system. 

Only a limited amount of research has been targeted at specifically trying to identify 
actual species of the human gut microbiota that are responsible or capable of metabolizing 
polyphenols. This is perhaps a reflection of the difficulties encountered in undertaking 
such an effort. Simply isolating single strains of bacteria anaerobically and carrying out 
suitable fermentation assays when presented with such a complex mixed culture of 
bacteria is an extreme challenge in terms of the laboratory time required. Furthermore 
much of the microbial mass in the colon remains to be described or cultured (85,86). In 
terms of the metabolic pathways that polyphenols follow during their degradation in the 
gut, they are fairly complex and multistaged, suggesting (although unconfirmed at this 
stage) that more than one species/strain may be required for the complete degradation of 
the original substrate. The application of culture independent molecular microbiology 
techniques (such as fluorescent in situ hybridization) (87) and modern analytical chemistry 
techniques (such as metabonomics in combination with pattern recognition techniques) 
means that an understanding can be gained of whether variable levels of target populations 
present in the gut are related to the production of specific metabolites. This may in turn 
have an impact on specific health outcomes (such as cardiovascular markers of health or 
the development of cancers). This is particularly so given that a number of the microbial 
metabolites are now thought to have specific activities related to health. 



REFERENCES 

1 . World Cancer Research Fund. Vegetables and fruit. In: Food, Nutrition and the Prevention of 
Cancer: A Global Perspective. Washington, D.C.; World Cancer Research Fund/ American 
Institute for Cancer Research 1997; 436-446. 

2. Law MR, Morris JK. By how much does fruit and vegetable consumption reduce the risk of 
ischemic heart disease? Eur J Clin Nutr 1998; 52:549-556. 

3. Arts IC, Hollman PC. Polyphenols and disease risk in epidemiologic studies. Am J Clin Nutr 
2005; 81:317-325. 

4. Higdon JV, Frei B. Tea catechins and polyphenols: health effects, metabolism and antioxidant 
functions. Crit Rev Food Sci Nutr 2003; 43:89-143. 

5. Yang CS, Landau JM, Huang MT, Newmark HL. Inhibition of carcinogenesis by dietary 
polyphenolic compounds. Annu Rev Nutr 2001; 21:381-406. 

6. Nijveldt RJ, van Nood E, van Hoorn DEC, Boelens PG, van Norren K, van Leeuwen PAM. 
Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin 
Nutr 2001; 74:418-425. 

7. Adlercreutz H, Mazur W. Phytoestrogens and western diseases. Ann Med 1997; 29:95-120. 



The Metabolism of Polyphenols by the Human Gut Microbiota 165 

8. Blaut M, Schoefer L, Braune A. Transformation of flavonoids by intestinal microorganisms. Int 
J Vitam Nutr Res 2003; 73:79-87. 

9. Clifford MN. Diet-derived phenols in plasma and tissues and their implications for health. Plant 
Med 2004; 70:1103-1114. 

10. Halliwell B, Rafter J, Jenner A. Health promotion by flavonoids, tocopherols, tocotrienols and 
other phenols: direct or indirect effects? antioxidant or not? Am J Clin Nutr 2005; 1:268-276. 

11. Spencer JP. Metabolism of tea flavonoids in the gastrointestinal tract. J Nutr 2003; 
1:3255-3261. 

12. Manach C, Williamson G, Morand C, Scalbert A, Remesy C. Bioavailability and bioefficacy of 
polyphenols in humans I. Review of 97 bioavailability studies. Am J Clin Nutr 2005; 
1:230-242. 

13. Williamson G, Manach C. Bioavailability and bioefficacy of polyphenols in humans II. Review 
of 93 intervention studies. Am J Clin Nutr 2005; 1:243-255. 

14. Mulder TP, Rietveld AG, van Amelsvoort JM. Consumption of both black tea and green tea 
results in an increase in the excretion of hippuric acid into urine. Am J Clin Nutr 2005; 
1:256-260. 

15. Simmering R, Pforte H, Jacobasch G, Blaut M. The growth of the flavonoids-degrading 
intestinal bacterium, Eubacterium ramulus, is stimulated by dietary flavonoids in vivo. FEMS 
Microb Ecol 2002; 40:243-248. 

16. Jenner AM, Rafter J, Halliwell B. Human fecal water content of phenolics: the extent of colonic 
exposure to aromatic compounds. Free Rad Biol Med 2005; 38:763-772. 

17. Rowland IR. Factors affecting metabolic activity of the intestinal microbiota. Drug Metab Rev 
1988; 19:243-261. 

18. Kirjavainen PV, Gibson GR. Healthy gut microbiota and allergy: factors influencing 
development of the microbiota. Ann Med 1999; 31:288-292. 

19. Mallett AK, Rowland IR. Factors affecting the gut microbiota. In: Rowland I, ed. Role of the 
Gut Flora in Toxicity and Cancer. London: Academic Press, 1998:347-382. 

20. Clifford MN. Chlorogenic acids and other cinnamates — nature, occurance and dietary burden. 
J Sci Food Agric 1999; 79:362-372. 

21. Tomas-Barberan FA, Clifford MN. Dietary hydroxybenzoic acid derivatives and their possible 
role in health protection. J Sci Food Agric 2000; 80:1024-1032. 

22. Clifford MN, Scalbert A. Elagitannins — nature, occurence and dietary burden. J Sci Food Agric 
2000;80:118-125. 

23. Laszlavik M, Gla L, Misik S, Erdei L. Phenolic compounds in two hungarian red wines 
matured in quercus robur and quercus petrea barrels: HPLC analysis and diode array detection. 
Am J Enol Vitic 1995; 46:67-74. 

24. Haslam E. Plant Polyphenols, Vegetable Tannins Revisitied. Cambridge, U.K.: Cambridge 
University Press, 1989. 

25. Gil MI, Tomas-Barberan FA, Hess-Pierce B, Holcroft DM, Kader A A. Antioxidant activity of 
pomegranate juice and its relationship with phenolic composition and processing. J Agric Food 
Chem 2000; 48:4581-4589. 

26. Olthof MR, Hollman PC, Katan MB. Chlorogenic acid and caffeic acid are absorbed in humans. 
J Nutr 2001; 131:66-71. 

27. Olthof MR, Hollman PC, Buijsman MN, van Amelsvoort JM, Katan MB. Chlorogenic acid, 
quercetin-3-rutinoside and black tea phenols are extensively metabolized in humans. J Nutr 
2003; 133:1806-1814. 

28. Rechner AR, Spencer JP, Kuhnle G, Hahu U, Rice-Evans CA. Novel biomarkers of the 
metabolism of caffeic acid derivatives in vivo. Free Rad Biol Med 2001; 30:1213-1222. 

29. Rechner AR, Pannala AS, Rice-Evans CA. Caffeic acid derivatives in artichoke extract are 
metabolized to phenolic acids in vivo. Free Rad Res 2001; 35:192-202. 

30. Gonthier MP, Verney MA, Besson C, Remesy C, Scalbert A. Chlorogenic acid bioavailability 
largely depends on the its metabolism by the gut microbiota in rats. J Nutr 2003; 
133:1853-1859. 



166 Bingham 

3 1 . Rechner AR, Smith MA, Kuhnle G, et al. Colonic metabolism of dietary polyphenols: influence 
of structure on microbial fermentation products. Free Rad Biol Med 2004; 36:212-225. 

32. Couteau D, McCartney AL, Gibson GR, Williamson G, Faulds CB. Isolation and 
characterisation of human colonic bacteria able to hydrolyse chlorogenic acid. J Appl 
Microbiol 2001; 90:873-881. 

33. Andreasen MF, Kroon PA, Williamson G, Garcia-Conesa MT. Intestinal release and uptake of 
phenolic antioxidant diferulic acids. Free Rad Biol Med 2001; 31:304-314. 

34. Konishi Y, Kobayashi S. Microbial metabolites of ingested caffeic acid are absorbed by 
monocarboxylic acid transporter (MCT) in intestinal Caco-2 cell monolayers. J Agric Food 
Chem 2004; 52:6418-6424. 

35. Tsuda T, Watanabe M, Ohshima K, et al. Antioxidative activity of the anthocyanin pigments 
cyanidin 3-O-p-D-glucoside and cyanidin. J Agric Food Chem 1994; 42:2407-2410. 

36. Gali HU, Perchellet EM, Klish DS, Johnson JM, Perchellet JP. Antitumour-promoting 
activities of hydrolyzable tannins in mouse skin. Carcinogenesis 1992; 13:715-718. 

37. Castonguay A, Gali HU, Perchellet EM, et al. Antitumourigenic and antipromoting activities of 
ellagic acid, ellagitannins and oligomeric anthocyanin and procyanidin. Int J Oncol 1997; 
10:367-373. 

38. Kresty LA, Morse MA, Morgan C, et al. Chemoprevention of esophageal tumourogenesis by 
dietary administration of lyophilised black rasperberries. Cancer Res 2001; 61:6112-6119. 

39. Cerda B, Tomas-Barberan FA, Espin JC. Metabolism of antioxidant and chemopreventative 
ellagitannins from strawberries, raspberries, walnuts and oak aged wine in humans: 
identification of biomarkers and individual variability. J Agric Food Chem 2005; 53:227-235. 

40. Rechner AR, Kuhnle G, Bremmer P, Hubbard GP, Moore KP, Rice-Evans CA. The metabolic 
fate of dietary polyphenols in humans. Free Rad Biol Med 2002; 33:220-235. 

41 . Setchell KDR, Brown NM, Lydeking-Olsen E. The clinical importance of the metabolite equol. 
A clue to the effectiveness of soy and its isoflavones. J Nutr 2002; 132:3577-3584. 

42. Booth AN, Emerson OH, Jones FT, DeEds F. Urinary metabolites of caffeic and chlorogenic 
acids. J Biol Chem 1957; 229:51-59. 

43. Booth AN, Murray CW, Jones FT, DeEds F. The metabolic fate of rutin and quercetin in the 
animal body. J Biol Chem 1956; 223:251-257. 

44. Bravo L, Abia R, Eastwood MA, Saura-Calixto F. Degradation of polyphenols (catechin and 
tannic acid) in the rat intestinal tract: effect on colonic fermentation and fecal output. Br J Nutr 
1994; 71:933-946. 

45. DuPont MS, Bennett RN, Mellon FA, Williamson G. Polyphenols from alcoholic apple cider 
are absorbed, metabolized and excreted by humans. J Nutr 2002; 132:172-175. 

46. Graefe EU, Veit M. Urinary metabolites of flavonoids and hydroxycinnamic acids in humans 
after application of a crude extract from Equisetum arvense. Phytomedicine 1999; 6:239-246. 

47. Gross M, Pfeiffer M, Martini M, Cambell D, Slavin J, Potter J. The quantitation of metabolites 
of quercetin flavonols in human urine. Cancer Epidemiol Biomarkers Prev 1996; 5:711-720. 

48. Pietta PG, Gardana C, Mauri PL. Identification of ginko biloba flavonol metabolites after oral 
administration to humans. J Chromatogr B 1997; 693:249-255. 

49. Plumb GW, Garcia-Conesa MT, Kroon PA, Rhodes M, Ridley S, Williamson G. Metabolism of 
chlorogenic acid by human plasma, liver, intestine and gut microbiota. J Sci Food Agric 1999; 
79:390-392. 

50. Aura AM, O'Leary KA, Williamson G, et al. Quercetin derivatives are deconjugated and 
converted to hydroxyphenylacetic acids but not methylated by human fecal flora in vitro. 
J Agric Food Chem 2002; 50:1725-1730. 

51. Kim DH, Jung EA, Sohng IS, Han JA, Kim TH, Han MJ. Intestinal bacterial metabolism of 
flavonoids and its relation to some biological activities. Arch Pharm Res 1998; 21:17-23. 

52. Simmering R, Kleessen B, Blaut M. Quantification of the flavonoid-degrading bacterium 
Eubacterium ramulus in human fecal samples with a species specific oligonucleotide 
hybridisation probe. Appl Environ Microbiol 1999; 65:3705-3709. 

53. Schneider H, Blaut M. Anaerobic degradation of flavonoids by Eubacterium ramulus. Arch 
Microbiol 2000; 173:71-75. 



The Metabolism of Polyphenols by the Human Gut Microbiota 167 

54. Winter J, Popoff MR, Grimont P, Bokkenheuser VD. Clostridium orbiscindens sp. Nov., a 
human intestinal bacterium capable of cleaving the flavonoid C-ring. Int J Syst Bacteriol 1991; 
41:355-357. 

55. Schoefer L, Mohan R, Scwiertz A, Braune A, Blaut M. Anaerobic degradation of flavonoids by 
Clostridium orbiscindens. Appl Environ Microbiol 2003; 69:5849-5854. 

56. Labenbrink C, Lapczynski S, Maiwald B, Engelhardt UH. Flavonoids and other polyphenols in 
consumer brews of tea and other caffeinated beverages. J Agric Food Chem 2000; 
48:1752-1757. 

57. Gott DM, Griffiths LA. Effects of antibiotic pretreatments on the metabolism and excretion of 
[U14C]( + )-catechin [U14C]( + )-cyanidanol-3 and its metabolite, 3 / -0-methyl-( + )-catechin. 
Xenobiotica 1987; 17:423-434. 

58. Meselhy MR, Nakamura N, Hattori M. Biotransformation of ( — )-epicatechin-3-0-gallate by 
human intestinal bacteria. Chem Pharm Bull 1997; 45:888-893. 

59. Wang LQ, Meselhy MR, Li Y, et al. The heterocyclic ring fission and dehydroxylation of 
catechin and related compounds by Eubacterium sp. Strain SDG-2, a human intestinal 
bacterium. Chem Pharm Bull 2001; 49:1640-1643. 

60. Spencer JP, Scroeter H, Rechner AR, Rice-Evans CA. Bioavailability of flavan-3-ols and 
procyanidins: gastrointestinal tract influences and their relevance to bioactive forms in vivo. 
Antioxid Redox Signal 2001; 3:1023-1039. 

61. Scheline RR. CRC Handbook of Mammalian Metabolism of Plant Compounds. Boca Raton, 
FL: CRC Press, 1991. 

62. Okubo T, Ishihara N, Oura A, et al. In vivo effects of tea polyphenol intake on human intestinal 
microbiota and metabolism. Biosci Biotechnol Biochem 1992; 56:588-591. 

63. Hara H, Orita N, Hatano S, et al. Effect of tea polyphenols on fecal flora and fecal metabolic 
products of pigs. J Vet Med Sci 1995; 57:45-49. 

64. Hara Y. Influence of tea catechins on the digestive tract. J Cell Biochem 1997; 27:52-58. 

65. Morazzoni P, Bombardelli E, Vaccinium myrtillus L. Fitoterapia 1996; 67:3-29. 

66. Renaud S, de Longeril M. Wine, alcohol, platelets, and the french paradox for coronary heart 
disease. Lancet 1992; 339:1523-1526. 

67. Havsteen B. Flavonoids, a class of natural products of high pharmacological potency. Biochem 
Pharmacol 1983; 32:1141-1148. 

68. Mian E, Curri SB, Lietti A, Bombardelli E. Anthocyanosides and the walls of micro vessels: 
further aspects of the mechanism of action of their protective effect in syndromes due to 
abnormal capillary fragility. Minerva Med 1977; 68:3565-3581. 

69. Kadar A, Robert L, Miskulin M, Tixier JM, Brechemier D, Robert AM. Influence of 
anthocyanoside treatment on the cholesterol-induced atherosclerosis in the rabbit. Paroi 
Arterielle 1979;5:187-205. 

70. Wang H, Nair MG, Strasburg GM, et al. Antioxidant and anti-inflammatory activities of 
anthocyanins and their aglycon, cyanidin, from tart cherries. J Nat Prod 1999; 62:294-296. 

71. Kamei H, Kojima T, Hasegawa M, et al. Suppression of tumor cell growth by anthocyanins 
in vitro. Cancer Invest 1995; 13:590-594. 

72. Wang H, Cao G, Prior RL. Oxygen radical absorbing capacity of anthocyanins. J Agric Food 
Chem 1997; 45:304-309. 

73. Felgines C, Texier O, Besson C, Fraisse D, Lamaison JL, Remesy C. Blackberry anthocyanins 
are slightly bioavailable in rats. J Nutr 2002; 132:1249-1253. 

74. Aura AM, Martin-Lopez P, O'Leary KA, et al. In vitro metabolism of anthocyanins by human 
gut microbiota. Eur J Nutr 2004; 44:133-142. 

75. Tsuda T, Horio F, Osawa T. Absorption and metabolism of cyanidin 3-0-(3-D-glucoside in rats. 
FEBS Lett 1999; 449:179-182. 

76. Baba S, Osakabe N, Natsume M, Terao J. Absorption and urinary excretion of procyanidin B 2 
[epicatechin-(43-8)-epicatechin] in rats. Free Rad Biol Med 2002; 33:142-148. 

77. Gonthier MO, Donovan JL, Texier O, Felgines C, Remesy C, Scalbert A. Metabolism of 
dietary procyanidins in rats. Free Rad Biol Med 2003; 35:837-844. 



168 Bingham 

78. Rios LY, Gonthier MP, Remesy C, et al. Chocolate intake increases urinary excretion of 
polyphenol-derived phenolic acids in healthy human subjects. Am J Clin Nutr 2003; 
77:912-918. 

79. Ward NC, Croft KD, Puddey IB, Hodgson JM. Supplementation with grape seed polyphenols 
results in increased urinary excretion of 3-hydroxyphenylpropionic acid, an important 
metabolite of proanthocyanidins in humans. J Agric Food Chem 2004; 52:5545-5549. 

80. Groenewoud G, Hundt HK. The microbial metabolism of condensed ( + )-catechins by rat - 
cecal microbiota. Xenobiotica 1986; 16:99-107. 

81. Deprez S, Brezillon C, Rabot S, et al. Polymeric proanthocyanidins are catabolised by human 
colonic microbiota into low molecular weight phenolic acids. J Nutr 2000; 130:2733-2738. 

82. Makkar HP, Becker K. Degradation of condensed tannins by rumen microbes exposed to 
quebrancho tannins (QT) in rumen simulation technique (RUSITEC) and effects of (QT) on 
fermentative processes in the RUSITEC. J Sci Food Agric 1995; 69:495-500. 

83. Makkar HP, Blummel M, Becker K. In vitro effects of and interactions between tannins and 
saponins and fate of tannins in the rumen. J Sci Food Agric 1995; 69:481-493. 

84. Smith AH, Mackie RI. Effect of condensed tannins on bacterial diversity and metabolic activity 
in the rat gastrointestinal tract. Appl Environ Microbiol 2004; 70: 1 104—1 115. 

85. McCartney AL. Application of molecular biological methods for studying probiotics and the 
gut flora. Br J Nutr 2002; 88:29-37. 

86. Manguin I, Bonnet R, Seksik P, et al. Molecular inventory of fecal microbiota in patients with 
Chrohn's disease. FEMS Microb Ecol 2004; 50:25-36. 

87. Namsolleck P, Thiel R, Lawson P, et al. Molecular methods for the analysis of gut microbiota. 
Microb Ecol Health Dis 2004; 16:71-85. 



9 



Molecular Analysis of Host-Microbe 
Interactions in the Gastrointestinal Tract 



Peter A. Bron 

Wageningen Centre for Food Sciences and NIZO Food Research, BA Fde, Wageningen, 
The Netherlands 

Willem M. de Vos 

Wageningen Centre for Food Sciences and Laboratory of Microbiology, Wageningen 
University Wageningen, The Netherlands 

Michiel Kleerebezem 

Wageningen Centre for Food Sciences and NIZO Food Research, BA Fde, Wageningen, 
The Netherlands 



INTRODUCTION 

From birth to death, the human gastrointestinal tract (GI tract) is colonized by a vast and 
complex consortium of mainly bacterial cells that outnumbers our somatic and germ cells 
(1). The microflora in this niche is estimated to be composed of at least 500 different 
species. However, this number is likely to represent a large underestimate, since it has 
been based on culturing studies that are known to be selective and notably underestimate 
the large number of Gram-positive intestinal bacteria. Molecular approaches, such as 
broad-range sequencing of 16S ribosomal RNA genes, have been used to monitor the 
composition of the dominant Gl-tract microbiota in different individuals at different points 
in their lives (see chapter 1). These approaches revealed a relatively stable composition in 
individual adults, but they appeared to be considerably variable when different individuals 
were compared (2,3). Moreover, host development (4,5), host genotype (6), and 
environmental factors (7) influence the composition of the microbiota, emphasizing 
how challenging it is to define and compare bacterial communities within and between 
specified intestinal niches of a given individual at a particular time point in his or her life. 
The fact that we have not yet been able to culture the majority of the members of this 
bacterial community further complicates studies on the activity of individual members of 
the Gl-tract consortium. An important development in this respect are the sophisticated 
enrichment strategies that have led to the isolation of new bacterial species from fecal 
samples [(8) and see chapter 1]. 

Several biological barriers are met by bacteria during residence in and travel through 
the different parts of the host's GI tract, such as the gastric acidity encountered in the 
stomach, the presence of bile salts in the duodenum and stress conditions associated with 

169 



170 Bronetal. 

oxygen gradients that are steep at the mucosal surface, while the colon lumen is virtually 
anoxic. Moreover, considerable bacterial competition is encountered throughout the 
intestinal tract and is most prominent in the colon where bacterial density is highest. There 
are many functions that can be ascribed to the bacterial Gl-tract communities, including 
the processing of undigested food, the stimulation of the host's immune system, and 
providing colonization resistance to pathogens (9). However, it seems that we are only 
beginning to understand the dimensions of these interactions. This is evident from the 
major impact that bacterial colonization seems to have on the host and the presently known 
response of intestinal bacteria that are reviewed below. 



BACTERIAL RESPONSES TO THE HOST 

In Vitro Approaches 

Due to the complex nature of host-specific and chemical stress conditions that are met by 
bacteria in the GI tract many studies describe the in vitro response of intestinal bacteria to a 
simplified model that mimics (a component of) the stress encountered in the host's GI tract. 

Historically, these studies have been performed in pathogens, including studies 
describing the response towards acid stress in enteropathogenic bacteria such as Salmonella 
and Escherichia coli, which revealed that RpoS, Fur, PhoP, and OmpR are important 
pH-response regulators (10). More recent studies describe food-grade bacteria and their 
tolerance to acid stress. These studies have focused mainly on physiological aspects such as 
determination of levels of acid-tolerance (11,12). Changes in protein synthesis during acid 
adaptation have been studied in Propionibacterium freudenreichii using 2D-gel 
electrophoresis, indicating an important role in the early acid tolerance response for 
a biotin carboxyl carrier protein and enzymes involved in DNA synthesis and repair, as well 
as a role in the late response for the universal chaperones GroEL and GroES (13). 

Several studies describe the defense mechanisms of Gram-negative enteric bacteria 
towards bile acids, which include the synthesis of porins, transport proteins, efflux pumps 
and lipopoly saccharides (14). In addition, a few genome- wide approaches aiming at the 
identification of proteins important for bile salt resistance in Gram-positive bacteria have 
been described. In Propionibacterium freudenreichii, Listeria monocytogenes and 
Enterococcus faecalis differential proteome analysis using 2D-gel electrophoresis led to 
the identification of several proteins that were expressed at a higher level in the presence of 
bile salts relative to control conditions lacking bile salts (15-17). In Propionibacterium 
freudenreichii these bile-induced proteins were further analyzed by N-terminal 
sequencing and peptide mass fingerprinting, leading to the identification of 1 1 proteins 
important in bile stress response. The induced proteins include general stress proteins such 
as ClpB and the chaperons DnaK and Hsp20 (16). Analogously, a subset of the proteins 
identified in E. faecalis appeared to be inducible by multiple sublethal stresses, including 
heat, ethanol, and alkaline pH (18). The fact that these general stress proteins are induced 
by bile is in agreement with the cross protection against bile after thermal or detergent 
pre-treatment that has been observed in several bacteria, including Enterococcus faecalis, 
Listeria monocytogenes and Bifidobacterium adolescentis (15,19,20). Moreover, in 
Escherichia coli an rpoS mutant failed to develop starvation-mediated cross protection 
after in vitro mimicking of osmotic, oxidative, and heat stresses (21). Two other bile- 
induced proteins in Propionibacterium freudenreichii are the superoxide dismutase and 
cysteine synthase, which could be involved in the protection against the oxidative stress 
imposed on Propionibacterium freudenreichii by bile. In addition, other studies describe 
the oxidative stress response of Gl-tract organisms, including Campylobacter coli, 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 



171 



Escherichia coli and several Shigella species (21-23). A deletion mutant in the gene 
encoding superoxide dismutase in Campylobacter coli displayed poor survival and 
colonization during infection of an animal model (23). Moreover, proteins involved in 
signal sensing and transduction, and an alternative sigma factor appeared to be bile- 
inducible (16). Next to these proteomic approaches, random gene disruption strategies 
have been applied to Listeria monocytogenes and Enterococcus faecalis, resulting in 
strains that are more susceptible to bile salts than the wild-type strains. Subsequent genetic 
analysis of the mutants revealed that the disrupted genes encode diverse functions, 
including an efflux pump homologue (19) and genes involved in oxidative stress response, 
and cell wall and fatty acid biosynthesis (24). In Lactobacillus plantarum a genetic screen 
resulted in the identification of 3 1 genes of which the expression appeared to be induced by 
bile. In analogy with the random gene disruption strategies applied in other species, this 
genetic screen in L. plantarum led to the observation that efflux pumps and changes in the 
architecture in the cell envelope are important for bile resistance of these bacteria (25). 
Moreover, these findings are in agreement with several physiological studies in Gl-tract 
bacteria such as L. plantarum, Propionibacterium freudenreichii and L. reuteri that 
demonstrated that bile salts induce severe changes in the morphology of the cell 
membrane and/or cell wall of these organisms (Fig. 1) (16,25,26). 

Overall, the aforementioned in vitro experiments have provided insight in the 
response of specific bacteria towards components of the complex mixture of stress 
conditions that is met by these bacteria during residence in or transit through the GI tract of 
their hosts. Although these approaches have helped to unravel the response of specific 
micro-organisms towards certain Gl-tract conditions, they will not suffice to describe their 
behavior in the GI tract. The full response repertoire will only be triggered in vivo, where all 
physicochemical conditions are combined with specific host-microbe and microbe-microbe 
interactions. Therefore, more sophisticated approaches have aimed at the development of 
tools that allow the in vivo identification of genes that are important in the GI tract. 

Overview of In Vivo Strategies 

Three main strategies have been developed for the identification of genes that are either 
highly expressed, differentially expressed or specifically required in vivo (Fig. 2). These 



(A) 



(B) 




Figure 1 Exemplary representation of the morphological changes induced by bile. L. plantarum 
cells were grown on laboratory media without (A) or with (B) 0. 1 % of porcine bile and the bacterial 
cells were investigated by scanning electron microscopy. Source: From Ref. 25. 



172 



Bron et al. 



(A) 



-/ 



unique tag sequence 



transposon 



Introduce transposon pool into bacteria 



T 



Organize 



mutants 



individual signature tagged mutants 



o 


♦ 


o 


o 


o 


o 


♦ 


o 


♦ 


♦ 


• 


o 


o 


o 


• 



Pool mutants 



Ia 



Administer to 
animal model 




input pool 



recovered pool 



i 



Hybridize to replicate filters 



H 



o 


♦ 


o 





o 


o 


• 


o 


• 


o 


♦ 


o 


o 


o 


♦ 



o 


/ 


o 


o 


o 


°/ 


k 


o 


• 


• 


i 


o 


o 


o^ 





Identify attenuated mutants 



(B) 



Administer to 
animal model 




Isolate in vivo and in vitro 

bacterial mRNA and 

use as template for 

cDNA synthesis 



Hybridize 
to xDNA 



i 



Identify mRNAs only 
present in vivo 



(C) 



antibiotic 
resistance 

gene 




in vitro 
reporter gen 
(lacZ) 



in vivo reporter gen 
(purA, cat, hly, tnpR) 



random xDNA fragments 



I 



Administer plasmid library to animal model 




Recover in vivo active sublibrary 



J 



♦ ♦ 



♦ 
♦ 



Identify in vitro 
-^-> inactive clones 



as in vivo induced 



Figure 2 Schematic representation of the basic principles of STM (A), SCOTS (B) and 
(R-)IVET (C). Abbreviations: IVET, in vivo expression technology; SCOTS, selective capture of 
transcribed sequences; STM, signature tagged mutagenesis. 

strategies have mainly been applied for the identification of genes from pathogens which 
are important during infection of their animal host. Signature tagged mutagenesis (STM) 
utilizes a negative selection strategy in which an animal host is infected with a pool of 
sequence-tagged insertion mutants. Mutated genes represented in the initial inoculum but 
not recovered from the host are essential for growth in the host (27,28). A major advantage 
of STM is that this type of screen provides direct proof for the importance of the mutated 
genes in the relevant niche. Unfortunately, only limited numbers of mutants can be 
screened per animal model. Therefore, large scale animal experiments are required for 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 173 

genome-wide mutant screens and for this reason STM screens are labor-intensive. In 
addition, mutants that are slow-growing, contain mutations in genes encoding redundant 
functions, or that can be complemented in a mixed population remain undetected or are at 
least underrepresented (29). Moreover, mutants for genes that are essential in the 
laboratory can never be obtained and, therefore, their importance for persistence in vivo 
cannot be investigated using this technique. Nevertheless, the STM strategy has been 
applied successfully to identify genes important in Gl-tract colonization by at least six 
enteric pathogens, including Klebsiella pneumoniae, Vibrio cholerae, and Escherichia 
coli (27). Lipopoly saccharides have been recognized as an important factor in Gl-tract 
persistence and colonization of several Gram-negative bacteria, as they have emerged as a 
common theme in the STM-based studies. In addition, the importance of the global 
regulator of anaerobic metabolism Fnr was highlighted by several STM screens, which is 
not surprising considering the low oxygen tension in the colon. Moreover, the alternate 
sigma factor RpoN was found in several of the STM screens and is likely to associate with 
RNA polymerase to promote the transcription of genes that are specifically required in the 
Gl-tract niche. Finally, STM studies revealed the importance of specific adhesins, 
including the type IV pili of Vibrio cholerae and Citrobacter rodentium (27). 

A second strategy that has been applied for the identification of in vivo transcribed 
genes is selective capture of transcribed sequences (SCOTS). cDNA is prepared from total 
RNA isolated from infected cells, or tissue samples. cDNA mixtures obtained are then 
enriched for sequences that are transcribed preferentially during growth in the host, using 
hybridizations to biotinylated bacterial genomic DNA in the presence of cDNA similarly 
prepared from bacteria grown in vitro. This strategy is very effective for the identification 
of highly abundant genes in situ which are also expressed to a lower level in the laboratory. 
In contrast to the STM strategy, genes that are essential in the laboratory can be 
investigated for their importance in Gl-tract colonization. Nevertheless, major 
disadvantages of SCOTS are the instability of bacterial mRNA for the construction of 
cDNA libraries, the low abundance of mRNA from transiently or lowly expressed genes, 
and the technical difficulty in isolation of sufficient high-quality mRNA from small 
populations of bacteria in vivo (29). SCOTS has only been applied in a limited number of 
studies and the majority of these screens was performed to identify bacterial genes 
expressed within macrophages (30-33). More recently, the first SCOTS strategy utilizing 
an animal model to identify genes important during infection was performed (34). This 
approach resulted in the identification of Escherichia coli genes of which the expression is 
either relatively abundant or induced in vivo. Similar to the STM approaches described 
above, this SCOTS approach revealed the induction of expression of genes involved in 
pilus formation and lipopolysaccharide (LPS) biosynthesis. Other genes identified 
included iron-responsive and plasmid- and phage-encoded genes (34). 

The third strategy that has been used to identify genes that are specifically induced or 
required during infection is in vivo expression technology (IVET). Similar to SCOTS, the 
IVET strategy is capable of identifying genes that are non-essential or redundant, while in 
an STM approach genes are only identified that are essential in vivo. An important 
difference between IVET and SCOTS lies in the fact that SCOTS is capable of identifying 
genes that are active in the laboratory, but, nevertheless, are induced in the host, while 
IVET only identifies in vivo induced genes that are very lowly or not expressed in the 
laboratory. The IVET approach relies on the generation of transcriptional fusions of 
genomic sequences to a reporter gene encoding an enzymatic activity. Nowadays, four 
variations of IVET utilizing different reporter genes have evolved as discussed in the 
section below. 



174 Bronetal. 

In Vivo Expression Technology Approaches 

The original IVET approach involves a tandem set of two promoterless reporter genes, 
namely pur A and lacZ, which were used to identify promoters that are specifically switched 
on in Salmonella typhimurium during infection (35). Purine auxotroph mutants (A/?wrA) of 
Salmonella typhimurium were only able to survive in a mouse model system when 
complemented in trans with a plasmid encoded pur A copy. The promoterless pur A gene 
was thereby utilized as a reporter for the identification of chromosomal fragments that are 
capable to complement the mutants, thereby strongly selecting for chromosomal fragments 
which harbor promoter elements that are active in the mouse model system. Subsequently, 
the in vivo active promoters are tested for the absence of promoter activity in vitro utilizing 
the second reporter gene (lacZ). The second variation of IVET is based on selection of an 
antibiotic resistance gene as selectable marker. One obvious disadvantage of this second 
variation of IVET is that the antibiotic must be administered to the host animal, which will 
certainly disturb the naturally occurring microflora in the GI tract. Therefore, the screening 
conditions assessed with this variant of IVET significantly differ from the native, in vivo 
situation. On the other hand, the addition of different levels of the selective antibiotic 
allows for selection of in vivo induced genes in a wider range of promoter activities. The 
third type of IVET selection uses a single gene as a dual reporter. The first example of such 
a dual reporter was hly, encoding the pore-forming haemolysin listeriolysin O (LLO) of 
Listeria monocytogenes (36). LLO mediates lysis of the phagosomal membrane in 
macrophages following infection. This reporter provides an in vivo selection for active 
fusions that allow for escape from the phagosomal compartment and subsequent 
multiplication. Moreover, a convenient screen on blood agar plates can be performed to 
identify inactive fusions in vitro, since clones harboring such fusions do not display 
haemolysis on these plates. The major drawback of the three aforementioned IVET 
variations is that the experimental set-up is designed in such a way that gene activity is 
required throughout the residence of the bacteria in the host. Hence, genes that are weakly 
expressed in the laboratory or transiently expressed only in a specific compartment of the 
host's GI tract slip through the selection procedure without being noticed. The fourth IVET 
variation circumvents this disadvantage by using the irreversible enzymatic activity of 
resolvases as reporter gene. Recombination-based IVET (R-IVET) is the only IVET 
approach that functions as a genetic screen. An antibiotic resistance marker flanked by two 
resolvase-recognition sites is integrated into the chromosome of the bacterium of interest. 
Subsequently, a promoterless copy of a resolvase-encoding gene, typically the tnpR gene 
from Tny5, is introduced on a plasmid and used to trap transcriptional activation by 
monitoring changes in the antibiotic resistance phenotype. Importantly, this approach does 
not rely on selective pressure during the animal experiments, as promoter activations are 
irreversibly trapped by the excision of the antibiotic resistance marker and can be identified 
after recovery of the bacterium under investigation from the host. 

In the first decade, (R-)IVET was extensively utilized for the identification of genes 
important during infection of at least 15 different pathogens, including Klebsiella 
pneumoniae, Salmonella enterica, and Listeria monocytogenes (29,37). Thereby, (R)-IVET 
is the most extensively applied screen for the identification of in vivo-induced genes 
during infection in animal models. The number of genes that are identified with an individual 
(R)-IVET screen varies strongly and ranges from 1 to approximately 100 genes (37). 

Several of these screens identified genes that were already known to be involved in 
virulence and this observation was considered an intrinsic validation of these (R-)IVET 
screens (29). An exemplary finding along these lines is the identification of agrA 
using R-IVET in Staphylococcus aureus (38). This gene encodes a quorum-sensing 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 175 

transcriptional activator and agrA mutants constructed in this organism prior to the 
R-IVET screen had already been shown to display a virulence defective phenotype (39). In 
general, regulators are one of the predominant classes of genes identified with (R-)IVET 
(29). Another frequently encountered class of in vivo induced genes in pathogenic bacteria 
are involved in the uptake of divalent cations, including many examples of Fe 
transporters (29). The harsh conditions these pathogens encounter when they transit from 
rich laboratory media to the host's GI tract apparently results in the induction of this group 
of genes. This suggestion is further supported by the observation that several in vivo 
induced genes were demonstrated to be similarly regulated under low Fe concentrations 
in vitro (40-42). Other genes that frequently arise from (R-)IVET screens have functions 
in a variety of generally recognized functional categories, including cell metabolism, 
DNA repair and general stress response. 

Recently, the first two reports appeared that describe the utilization of (R-)IVET 
strategies in food-grade or commensal micro-organisms in order to determine the specific 
induction of gene expression in these bacteria after introduction in the GI tract of animal 
models. In L. reuteri an IVET strategy based on in vivo selection of an antibiotic resistant 
phenotype (the aforementioned second variation of IVET) led to the identification of three 
genes important for this organism during colonization of the GI tract of Lactobacillus-free 
mice (43). One of these genes encodes a peptide methionine sulfoxide reductase (msrB) 
which has previously been identified using IVET in the non-food-associated Streptococcus 
gordonii during endocarditis (44). Although not noticed by the authors at that time, this 
was an important clue suggesting an overlap in the genetic response triggered in the 
pathogenic and non-pathogenic world following contact with the host. The second report 
dealing with in vivo induction of genes in food-associated microbes describes a R-IVET 
approach in L. plantarum (45). Previously, the resolvase-encoding tnpR-res system (46) 
has been applied to trap promoter activities in R-IVET experiments in several pathogenic 
bacteria. Therefore, initial attempts aimed at implementation of this system in 
L. plantarum. A res-ery-res cassette was successfully integrated into the chromosome 
of this bacterium and a promoterless copy of the tnpR gene was cloned on a low-copy 
plasmid. Despite the successful cloning of the endogenous, highly active IdhLl promoter 
upstream of tnpR, excision of the ery gene from the chromosome of L. plantarum was 
never observed (Bron et al. unpublished data). These experiments strongly suggest that the 
tnpR resolvase is not functional in L. plantarum under the conditions applied during 
the experiments. Therefore, an alternative strategy was chosen to implement R-IVET in 
L. plantarum, which involved the cre-loxP system (47). This system was previously 
demonstrated to be functional in another lactic acid bacterium (LAB), Lactococcus lactis 
(48). Hence, a loxP-ery-loxP cassette was integrated into the chromosome of L. plantarum 
and a promoterless copy of ere was cloned on a low-copy vector. This system appeared 
to be functional in L. plantarum, as IdhLl -promoter driven expression of the ere gene 
led to the irreversible excision of the loxP-ery-loxP cassette from the chromosome. 
Subsequently, a library containing L. plantarum chromosomal fragments upstream of ere 
was constructed and administered to mice. The library was recovered from fecal samples 
and analyzed for L. plantarum colonies that had lost their erythromycin resistant 
phenotype during passage through the animal model. These erythromycin sensitive 
colonies potentially harbor chromosomal fragments of which the expression was in 
vivo induced. Using this strategy, 72 L. plantarum genes were identified as being in vivo 
induced (ivi genes) during host Gl-tract transit (45). The distribution over the generally 
recognized classes of main biological functions appeared to be random. A slight 
overrepresentation of R-IVET genes is observed around the origin of replication as 
compared to the rest of the genome (Fig. 3). However, the significance of the latter 



176 Bronetal. 




Figure 3 Using R-IVET 72 L. plantarum genes could be identified as in vivo induced (ivi) during 
passage of the mouse GI tract. The chromosomal localization of these ivi genes is represented in the 
inner circle, while the outer two circles represent the ORFs on the positive (outer circle) and negative 
(middle circle) chromosomal DNA strand. 



observation is unclear. Nine of the 72 ivi genes appeared to encode sugar-related functions, 
including genes involved in ribose, cellobiose, sucrose, and sorbitol transport. Another 
nine genes encode functions involved in acquisition and synthesis of amino acids, 
nucleotides, cofactors, and vitamins, indicating their limited availability in the GI tract. 
Four genes involved in stress-related functions were identified, reflecting the harsh 
conditions that L. plantarum encounters in the GI tract. Another four genes encoding 
extracellular proteins were identified that could mediate interactions with host Gl-tract 
epithelial cells. Remarkably, the protein encoded by one of the hypothetical proteins 
identified in this study in L. plantarum is a homologue (32% identity) of the only 
conserved hypothetical protein that was identified with I VET in L. reuteri (43). Moreover, 
a large number of the functions and pathways identified in L. plantarum have previously 
been identified in pathogens as being important in vivo during infection (45). This striking 
amount of parallels between the pathogenic and non-pathogenic in vivo response suggests 
that survival rather than virulence is the explanation for the importance of these genes 
during host residence. Recently, nine of the L. plantarum ivi genes were selected, mainly 
focusing on genes that encode proteins with a predicted role in cell envelope functionality, 
stress response and regulation, for the construction of isogenic gene replacement mutants. 
Quantitative polymerase chain reaction (PCR) experiments were performed to monitor the 
relative population abundance of the group of L. plantarum replacement mutants in fecal 
samples after competitive passage through the GI tract of mice. These experiments 
revealed that after Gl-tract passage the relative abundance of three of the ivi gene 
mutants was 100- to 1000-fold reduced as compared to other mutant strains, suggesting 
an important role for these three ivi genes, encoding the IIC transport component 
of a cellobiose phosphotransferase system (PTS), an extracellular protein that contains an 
LPQTNE motif, and a copper transporting ATPase, in the functionality of L. plantarum 
during passage of the GI tract (49). 



INSIGHTS FROM GENOMICS 

Nowadays more and more bacteria are undergoing genome sequencing and as a result over 
130 completed bacterial genomes have become available in the public domain. Following 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 177 

the first example of Haemophilus influenzae in 1995 (50) the major focus of these efforts 
has initially been on pathogenic bacteria and includes the completion of several genome 
sequences of food-borne pathogens, including Bacillus cereus (51), Salmonella 
typhimurium (52), and Listeria monocytogenes (53). Over the last years sequencing of 
the genomes of food-associated, non-pathogenic bacteria has received considerable 
attention, including the elucidation of the complete genome sequence of Bacillus subtilis 
in 1997 (54). Moreover, the first complete LAB genome sequence published was that of 
Lactococcus lactis subspecies lactis strain IL1403 (55). To date, only two other high- 
fidelity genome sequences of LAB, L. plantarum strain WCFS1 (56) and L. johnsonii 
strain NCC533 (57), have been published. An additional number of LAB genomes is 
nearing completion and draft genome information has become available in the public 
domain in 2002 with the publication and appearance of genome sequences for LAB 
provided by the Joint Genome Institute (http://genome.jgi-psf.org/microbial/) in 
collaboration with the lactic acid bacteria genomics consortium (58,59). Next to this 
large amount of sequence data from food-associated LAB, successful efforts have been put 
in determination of the (complete) genome sequences of members of our normal colonic 
microbiota, in particular Bacteroides thetaiotaomicron (60) and Bifidobacterium longum 
(Fig. 4) (61). 

L. plantarum is a versatile and flexible organism that is able to grow on a wide variety 
of sugar sources. This phenotypic trait is reflected in the genome sequence of L. plantarum, 
which harbours a remarkably high number of 25 complete PTS enzyme II complexes as well 
as several incomplete complexes. This high number of PTS systems is far more than that 
found in other complete bacterial genomes, and similar only to Listeria monocytogenes (53) 
and Enterococcus faecalis (62). In addition to these PTS systems, the L. plantarum genome 
encodes 30 transporters that are predicted to be involved in the transport of carbon sources. 
This high sugar uptake flexibility has also been observed in the genomes of other LAB, such 
as L. johnsonii (57) and L. acidophilus (http://www.calpoly.edu/~rcano/Lacto_genome. 
html). Moreover, a remarkably high percentage of regulatory genes (8.5%) appeared to be 



'Escherichia coli (4.6) 

Bifidobacterium breve (2.4) 

'Bifidobacterium longum (2.3) 



■ Pediococcus pentosaceus (2.0) 
' Lactobacillus rhamnosus (2.4) 



Propionibac terium freudenreichii (2.6) 
Brevibacterium linens (3.0) 

Bacteroides thetaiotaomicron (6.3) 



j — Lactobacillus casei (2.6) 

Lactobacillus sakei (1 .9) 



-£ 



-Lactobacillus plantarum (3.3) 

— Lactobacillus brevis (2.0) 
Lactobacillus alimentarius 



rLactobacillus johnsonii (2.0) 

Lactobacillus gasseri (1.8) 

Lactobacillus delbrueckii (2.3) 

| Lactobacillus acidophilus (2.0) 

Lactobacillus helveticus (2.4) 



-Enterococcus faecalis (3.2) 

-Bacillus subtilis (4.2) 



J Bacillus cereus (4.2) 

Bacillus anthracis (5.2) 

Listeria monocytogenes (2.9) 

-Lactococcus lactis (2.3) 

-Streptococcus thermophilus (1.8) 



-Leuconostoc mesenteroi des (2.0) 



Oenococcus oeni (1.8) 



-Clostridium perfringens (3.0) 



0.10 



Figure 4 Phylogenetic relationship based upon the neighbor-joining method of partial 16S rDNA 
sequences (Escherichia coli positions 107 to 1434). It should be noted that for some species the 
genome sequence has (partially) been determined for multiple strains. LAB genomes are 
underscored, and published, complete genomes are shown in bold. The estimated genome sizes 
are indicated between brackets. 



178 



Bron et al. 



encoded in the L. plantarum genome. Similar percentages were found in Listeria 
monocytogenes, in which 7.3% of all the encoded genes were predicted to be involved in 
regulatory functions. This could be a reflection of the many different environmental 
conditions that these bacteria face. Moreover, these sophisticated regulatory systems enable 
these organisms to adapt quickly to changes in the sugar composition of the host's diet 
during residence in the proximal parts of the GI tract (Fig. 5). 



o 



o 



o 




Ingestion of carbohydrates by the host; 
high diet-dependent variation 





f" 



Uptake of simple carbohydrates 
by bacteria and the host 



Small intestine 



<^> 



Bacteria mediated 
production and liberation 
of host-specific glycans 




Bacteria breakdown of 
complex polysaccharides 



<~D> 




OO 
O " 




Uptake of simple carbohydrates 
by bacteria and the host 



Large intestine 



Figure 5 Molecular model of bacterial sugar utilization in the GI tract. In the small intestine 
mono- and disaccharides are rapidly consumed by the host. Typically, bacteria that live in this niche 
display highly flexible sugar utilization capacities, allowing them to quickly adapt to changes in the 
carbon source availability that is determined by the host's diet. This high sugar flexibility is required 
to compete with the host for carbon acquisition. In the large intestine more complex oligo- and 
polysaccharides are the only available C-source. Therefore, bacteria in this niche are usually able to 
hydrolyse complex dietary polysaccharides and host-derived glycoproteins and glycoconjugates. 
Subsequently, the released, simpler sugars are utilized as C-source by the host and the bacteria 
residing in the colon. Source: From Ref. 63. 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 179 

The genomes of B. thetaiotaomicron and Bifidobacterium longum encode an 
elaborate apparatus for acquiring and hydrolysing otherwise indigestible dietary 
polysaccharides (60,61). In B. thetaiotaomicron this "colonic substrate dependence" is 
associated with an environment-sensing system consisting of a large repertoire of 
extracytoplasmic function sigma factors and one- and two-component signal transduction 
systems (60). In contrast, genes involved in sugar transport and hydrolysis in 
Bifidobacterium longum are organized in operons which are predominantly regulated by 
Lacl-type, sugar responsive repressors (61). The tight regulation of sugar utilization 
observed in these bacteria allows a stringent response to environmental changes and is in 
accordance with the fact that Bifidobacterium longum and B. thetaiotaomicron need to 
adapt to wide fluctuations in substrate availability in the colon (60,61). It is speculated that 
the mode of regulation via repression of genes could allow a quicker response in 
Bifidobacterium longum (61). Similarly, an operon in L. acidophilus involved in utilization 
of the prebiotic compound fructooligosaccharide contains a Lad type repressor. 
Moreover, the expression of this operon is subject to global catabolite repression in the 
presence of readily fermentable sugars (64). Another interesting finding in the genome of 
B. thetaiotaomicron is that it appears to encode the capacity to use a variety of host- 
derived glycoproteins and glycoconjugates. Sixty-one percent of its glycosylhydrolases 
are predicted to be located in the periplasm, outer membrane, or extracellularly. This 
suggests that these enzymes are not only important for fulfilling the needs of 
B. thetaiotaomicron but may also help shape the metabolic milieu of the intestinal 
ecosystem in ways conducive to maintaining a microbiota that supplies the host with 10 to 
15% of our daily calories as fermentation products of dietary polysaccharides (Fig. 5) (60). 
Similarly, the genome sequence of Bifidobacterium longum revealed insights into the 
interaction of bifidobacteria with their host, as genes encoding polypeptides with 
homology to glycoprotein-binding fimbriae are present in the genome. Moreover, a 
eukaryotic-type serine protease inhibitor is encoded in the genome and could be involved 
in the reported immunomodulatory activity of bifidobacteria (61). 

Recently, the complete genomes of L. plantarum (3.3 Mbp) and L. johnsonii 
(2.0 Mbp) were compared, revealing that these genomes have only 28 regions with 
conservation of gene order, encompassing approximately 0.75 Mbp (65). Notably, these 
regions are not co-linear, indicating major chromosomal rearrangements. Moreover, 
metabolic reconstruction indicated many differences between these two lactobacilli, as 
numerous enzymes involved in sugar metabolism and the biosynthesis of amino acids, 
nucleotides, fatty acids and cofactors are lacking in L. johnsonii. Interestingly, major 
differences were also seen in the number and types of putative extracellular proteins, 
which could play a role in host-microbe interactions in the GI tract. The differences 
between L. plantarum and L. johnsonii, both in genome organization and gene content, are 
exceptionally large for two bacteria of the same genus, emphasizing the complexity and 
diversity of the Lactobacillus genus (65). 

Overall, the availability and comparison of bacterial genome sequences and their 
annotated functions provides valuable clues towards the survival strategy of these bacteria 
during their residence in the human GI tract. Additionally, these complete genome 
sequences are powerful tools for the convenient and effective interpretation of the data 
generated by the in vitro and in vivo screening procedures described above. Moreover, 
comparative genomics can provide important insight in diversity, evolutionary 
relationship and functional variation between bacteria, which might eventually generate 
a comprehensive view of the behavior of microbes during residence in the human 
Gl-tract. 



180 Bronetal. 

IN SITU PROFILING OF TRANSCRIPTION IN THE Gl TRACT 

As soon as sequence data is available for a few genes in a bacterium of interest, one could think 
of several sophisticated tools that allow investigation of the in situ expression levels of specific 
genes. One example of such an approach is the implementation of quantitative reverse 
transcriptase polymerase chain reaction (qRT-PCR) in the gram-negative bacterium 
Helicobacter pylori (66). This study describes the assessment of gene expression in this 
pathogen within the mouse and human gastric mucosae. Three genes, encoding urease, 
catalase, and a putative adhesin specific for adherence to human gastric mucosa, were selected 
for analysis, as their role during host residence was already established. Using minute 
quantities of mRNA isolated from human and mouse infected mucosae, the in situ expression 
of these three genes could be established. Moreover, the results of this study indicate that the 
relative abundance of transcripts was the same in the human and mouse model system. Hence, 
this study demonstrates that qRT-PCR is a powerful tool for the detection and quantification 
of bacterial gene expression in the GI tract (66). Similar experiments were performed in 
L. plantarum. An in vitro screen and a R-IVET screen were already performed in this LAB to 
identify genes of which the expression is induced in vitro by bile or in situ in the GI tract of 
a mouse model system, respectively (25,45). Matching of the results obtained in these two 
screening procedures revealed two genes, encoding an integral membrane protein and an 
argininosuccinate synthase that appeared to be induced by bile in vitro as well as in vivo in the 
GI tract of a mouse model system. Therefore, the expression of these two genes was assessed 
using qRT-PCR followed by S YBR green fluorescence detection. As the duodenum is the site 
of bile release, expression in this specific region of the host' s GI tract was investigated. 
The results confirmed that the expression levels of these two genes were significantly higher in 
L. plantarum cells isolated from the mouse duodenum relative to cells grown in standard 
laboratory media (25). Current studies aim at the confirmation of gene-induction of several 
other L. plantarum genes initially identified with the R-IVET screen (Marco et al. unpublished 
data). Moreover, transcription profiling under a variety of in vitro conditions could identify 
more matches with the R-IVET screen and these genes could subsequently be analyzed with 
qRT-PCR. These experiments might reveal the specific environmental cue involved in in situ 
induction in the GI tract and could eventually elucidate the regulatory mechanism(s) involved. 
These approaches could help to unravel the geographical differentiation of L. plantarum 
gene expression along the GI tract, i.e., specific induction in the stomach, small intestine 
or colon. 

Another promising development is the optimization of bacterial RNA isolation 
protocols from fecal samples (67), and Gl-tract samples from conventional mice fed with 
L. plantarum (Marco et al. unpublished data) or human cancer patients who volunteered to 
consume an oatmeal-based drink containing high numbers of L. plantarum prior to surgery 
(de Vries et al. unpublished data). Although it is technically difficult to isolate high quality 
bacterial mRNAs from these samples, such RNA samples originating from an in vivo 
animal tissue, could prove extremely valuable, as they should allow analysis using 
DNA micro-array technology, providing direct information on the in situ expression 
levels of thousands of bacterial genes. Moreover, comparison of bacterial responses 
in samples from the GI tract from animal models and of human origin could provide an 
indication of the overlap in the response of L. plantarum during residence in the GI tract of 
different hosts. 

Studies in gnotobiotic mice have indicated that there is specific signaling between 
the commensal bacterium B. thetaiotaomicron and its host. Synthesis of host epithelial 
glycans is elicited by a B. thetaiotaomicron signal of which the expression is regulated by 
a fucose-binding bacterial transcription factor. This factor senses environmental levels of 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 181 

fucose and coordinates the decision to generate a signal for production of host 
fucosylated glycans when environmental fucose is limited or to induce expression of the 
bacteria's fucose utilization operon when fucose is abundant (68). Additional studies 
have evaluated the global intestinal response to colonization of gnotobiotic mice with 
B. thetaiotaomicron. This colonization dramatically affected the host's gene expression, 
including several important intestinal functions such as nutrient absorption, mucosal 
barrier fortification, and postnatal intestinal maturation (9). From the in situ global 
transcription profiles mentioned above and follow-up experiments it could be established 
that the production of a previously uncharacterized angiogenin is induced when 
gnotobiotic mice are colonized with B. thetaiotaomicron, revealing a mechanism 
whereby intestinal commensal bacteria influence Gl-tract bacterial ecology and shape 
innate immunity (69). In addition, the cellular origin of the angiogenin response was 
investigated when different intestinal cell types were separated by laser-capture 
microdissection and analyzed by qRT-PCR, revealing that angiogenin-3 mRNA is 
specifically induced only in crypt epithelial cells. Hence, these experiments strongly 
suggest an intestinal tissue specific response of the host during colonization (9). 
Interestingly, comparison of the changes in global host gene expression in mice after 
colonization with B. thetaiotaomicron, Bifidobacterium infantis or E. coli led to the 
observation that part of this host response was only induced in mice by colonization with 
B. thetaiotaomicron (9). However, analysis of a broader range of members of the 
intestinal microbiota will reveal what the level of bacterial response specificity within the 
host's tissues actually is. One such study is currently performed for L. plantarum (Peters 
et al. unpublished data). Overall, the aforementioned studies on B. thetaiotaomicron 
colonization of gnotobiotic mice provided valuable information on the influence of one 
particular member of the microbiota on the host. However, the host response during 
colonization by more complex mixtures of microbes and/or the host response in other 
animal systems remained to be investigated at that time. Recently, it was found that 
conventionalization of adult gnotobiotic mice with normal microbiota harvested from the 
distal intestine of conventionally raised mice produced a 60% increase in body fat 
content and insulin resistance despite reduced food intake. Studies of gnotobiotic and 
conventionalized mice revealed that the microbiota promotes absorption of monosac- 
charides from the gut lumen, which results in induction of de novo hepatic lipogenesis. 
Fastin-induced adipocyte factor (Fiaf), a member of the angiopoietin-like family of 
proteins, is selectively suppressed in the intestinal epithelium of normal mice by 
conventionalization. Analysis of gnotobiotic and conventionalized, normal and Fiaf 
knockout mice established that Fiaf is a circulating lipoprotein lipase inhibitor and that its 
suppression is essential for the microbiota-induced deposition of triglycerides in 
adipocytes. These results suggest that the gut microbiota have a major impact on food- 
derived energy harvest and storage in the host (70). Another recent study investigated the 
host response during colonization of a different animal model. DNA micro-array 
comparison of gene expression in the digestive tracts of six days post-fertilization 
gnotobiotic, conventionalized, and conventionally raised zebrafish (Danio Rerio) 
revealed 212 genes regulated by the microbiota. Notably, 59 of these genes were also 
found to be regulated in the mouse intestine during colonization, including genes that 
encode functions involved in stimulation of epithelial proliferation, promotion of nutrient 
metabolism, and innate immune response, indicating a substantial overlap in the genetic 
response of mice and zebrafish towards intestinal colonization (71). Despite these recent 
developments, an important future challenge lies within the translation of these animal 
host response analyses to the human system. 



182 Bronetal. 

CONCLUDING REMARKS AND FUTURE PERSPECTIVES 

Historically, research on the bacterial flora of the GI tract has concentrated on the 
inhabitants that have negative effects on their hosts. More recently, research has expanded 
from these pathogenic to non-pathogenic bacteria, including symbionts and commensals. 
One obvious reason for this is the accumulating evidence that certain bacteria, especially 
strains from the genus Lactobacillus and Bifidobacterium, may have probiotic effects in 
man and animals (72). At present there is a detailed understanding on the distribution of 
specific microbes along the human colon and the variations that can occur between 
different individuals (73-75). Moreover, knowledge on the activity and response of 
specific species to the conditions encountered when they transit through this complex 
niche is starting to accumulate. Several in vitro studies mimicking specific conditions in 
the GI tract have been performed, which allowed the identification of the repertoire of 
genes and their corresponding proteins that respond to the condition applied. More 
recently, in vivo approaches aiming at the identification of bacterial genes that are induced 
during passage of the GI tract have been performed in various microbes, including food- 
grade species. The current knowledge on promoter elements regulating gene expression of 
food-grade bacteria in the GI tract could have application possibilities, as these bacteria 
have been shown to possess great potential to serve as delivery vehicles of health- 
promoting or therapeutic compounds to the human GI tract (76-84). (R-)IVET approaches 
have provided the required promoters that will allow the construction of LAB -based 
dedicated Gl-tract delivery vehicles that only express certain desired functions in situ. 
Moreover, geographically more detailed insight in the exact site of in situ gene activation 
in the GI tract derived from qRT-PCR using specific tissue samples might allow the 
construction of highly site-specific delivery vehicles. Combination of these promoters 
with certain genes, e.g., bacteriophage-derived or other lytic cassettes, might generate 
LAB strains that release their cellular content at a specific location in the GI tract. 

At present, a large part of the consortium of bacteria residing in the GI tract has not 
been cultured in vitro. Since most genetic approaches require the culturability of the 
microbe under investigation, the expansion of our knowledge of this group of bacteria is 
highly challenging and very limiting at this stage. Metagenomic approaches might shed 
light on the genetic complexity of the collective genomic material of the intestinal 
microbiota (85). Moreover, such studies could reveal previously unknown, critical genes 
for intestinal microbiota functioning. However, effective exploration of metagenomic 
functionality will depend on high throughput screening systems that allow function 
identification. Moreover, the development of effective and robust methods to assess 
microbiota activity in situ in a culture independent manner will be critical for our 
functional understanding of the large number of unculturable bacteria in the GI tract. 

A promising prospect from the increasing availability of complete genome 
sequences is the construction of DNA micro-arrays in several laboratories working on 
food-associated microbes. As a consequence, the first publications presenting data from 
these DNA micro-arrays appeared recently (86-88). These genomics-based, global 
investigations of gene expression in food-grade microbes under various conditions will 
further detail our understanding of their behavior. However, the application of these 
transcriptome profiling techniques on microbe-containing GI tract samples will still have 
to overcome some technical hurdles (RNA extraction procedures, response validation, 
etc.), but will eventually lead to a more complete view of the activity of these bacteria in 
this complex niche. Besides the application of DNA micro-array technology to reveal the 
bacterial side of host-microbe interactions in the GI tract, this technology has already been 
used by Hooper and co-workers in several elegant studies aiming at identification of the 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 183 

response of gnotobiotic mice to colonization with the commensal B. thetaiotaomicron 
(9,68,69). In addition, more recent gnotobiote studies have shed light on the differences in 
mouse gene expression upon colonization by a more complex mixture of bacteria (70), and 
have provided the first steps towards the comparative analysis of host responses in 
different animal models (71). Nevertheless, an important question that still remains to be 
answered is to what extent the data obtained on host and bacterial gene expression using 
animal models can be extrapolated to the situation in humans. 

In conclusion, the genome-wide transcript profiling approaches that have been 
performed to date have provided us with clues of the possible role of individual host and 
bacterial genes during host-microbe interactions. Combination of bacterium and host 
transcriptomes should allow the construction of molecular models that describe host- 
microbe interactions, allowing more pinpointed experiments in the future, designed on the 
basis of a molecular interaction hypothesis. As Gl-tract bacteria like L. plantarum and 
B. thetaiotaomicron are genetically accessible, gene deletion and overexpression mutants 
can be constructed and employed to study the effect of a single bacterial gene and its 
corresponding function on host gene expression. After profiling of these host genes, knock- 
out mice and/or antisense RNA approaches might allow gene silencing on the host side of 
the spectrum, thereby enabling us to study the effect of single host gene mutations on the 
colonization of microbes. Ultimately, such studies may provide a molecular knowledge 
base to understand Gl-tract colonization of commensals or symbionts, and could lead to the 
molecular explanation of probiotic effects associated with LAB and related species. 



ACKNOWLEDGMENTS 

The authors gratefully acknowledge Jos Boekhorst and Sergey Konstantinov for the 
construction of Figures 3 and 4, respectively. We cordially thank our WCFS colleagues for 
sharing unpublished data. Part of this work was supported by the EU project LABDEL 
(EU-QLRT-2000-00340). 



REFERENCES 

1 . Savage DC. Microbial ecology of the gastro intestinal tract. Annu Rev Microbiol 1977; 31:133. 

2. Zoetendal EG, Akkermans AD, De Vos WM. Temperature gradient gel electrophoresis 
analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities 
of active bacteria. Appl Environ Microbiol 1998; 64:3854-3859. 

3. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben- Amor K, Akkermans AD, 
de Vos WM. Mucosa-associated bacteria in the human Gl-tract are uniformly distributed 
along the colon and differ from the community recovered from feces. Appl Environ Microbiol 
2002; 68:3401-3407. 

4. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related changes in intestinal bacterial 
populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid 
profiles. Gut 2001; 48:198-205. 

5. Favier CF, Vaughan EE, De Vos WM, Akkermans AD. Molecular monitoring of succession of 
bacterial communities in human neonates. Appl Environ Microbiol 2002; 68:219-226. 

6. Zoetendal EG, Akkermans AD, Akkermans-van Vliet WM, de Visser JAGM, de Vos WM. 
The host genotype affects the bacterial community in the human gastrointestinal tract. Microb 
Ecol Health Dis 2001; 13:129-134. 



184 Bronetal. 

7. Sullivan A, Edlund C, Nord CE. Effect of antimicrobial agents on the ecological balance of 
human microflora. Lancet Infect Dis 2001; 1:101—1 14. 

8. Zoetendal EG, Plugge CM, Akkermans AD, de Vos WM. Victivallis vadensis gen. nov., sp. 
nov., a sugar- fermenting anaerobe from human faeces. Int J Syst Evol Microbiol 2003; 
53:211-215. 

9. Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular analysis of 
commensal host-microbial relationships in the intestine. Science 2001; 291:881-884. 

10. Audia JP, Webb CC, Foster JW. Breaking through the acid barrier: an orchestrated response to 
proton stress by enteric bacteria. Int J Med Microbiol 2001; 291:97-106. 

11. Chou LS, Weimer B. Isolation and characterization of acid- and bile-tolerant isolates from 
strains of Lactobacillus acidophilus. J Dairy Sci 1999; 82:23-31. 

12. Hyronimus B, Le Marrec C, Sassi AH, Deschamps A. Acid and bile tolerance of spore-forming 
lactic acid bacteria. Int J Food Microbiol 2000; 61:193-197. 

13. Jan G, Leverrier P, Pichereau V, Boyaval P. Changes in protein synthesis and morphology 
during acid adaptation of Propionibacterium freudenreichii. Appl Environ Microbiol 2001; 
67:2029-2036. 

14. Gunn JS. Mechanisms of bacterial resistance and response to bile. Microbes Infect 2000; 
2:907-913. 

15. Flahaut S, Frere J, Boutibonnes P, Auffray Y. Comparison of the bile salts and sodium dodecyl 
sulfate stress responses in Enterococcus faecalis. Appl Environ Microbiol 1996; 62:2416-2420. 

16. Leverrier P, Dimova D, Pichereau V, Auffray Y, Boyaval P, Jan G. Susceptibility and adaptive 
response to bile salts in Propionibacterium freudenreichii: physiological and proteomic 
analysis. Appl Environ Microbiol 2003; 69:3809-3818. 

17. Phan-Thanh L, Gormon T. Stress proteins in Listeria monocytogenes. Electrophoresis 1997; 
18:1464-1471. 

18. Rince A, Le Breton Y, Verneuil N, Giard JC, Hartke A, Auffray Y. Physiological and 
molecular aspects of bile salt response in Enterococcus faecalis. Int J Food Microbiol 2003; 
88:207-213. 

19. Begley M, Gahan CG, Hill C. Bile stress response in Listeria monocytogenes L028: adaptation, 
cross-protection, and identification of genetic loci involved in bile resistance. Appl Environ 
Microbiol 2002; 68:6005-6012. 

20. Schmidt G, Zink R. Basic features of the stress response in three species of bifidobacteria: 
B. longum, B. adolescentis, and B. breve. Int J Food Microbiol 2000; 55:41-45. 

21. Krogfelt KA, Hjulgaard M, Sorensen K, Cohen PS, Givskov M. rpoS gene function is 
a disadvantage for Escherichia coli BJ4 during competitive colonization of the mouse large 
intestine. Infect Immun 2000; 68:2518-2524. 

22. Khanduja V, Kang G, Raj an DP, Balasubramanian KA. Oxidative stress response in Shigella & 
nonpathogenic gut bacteria. Indian J Med Res 1998; 108:3-7. 

23. Purdy D, Cawthraw S, Dickinson JH, Newell DG, Park SF. Generation of a superoxide 
dismutase (SOD)-deficient mutant of Campylobacter coli: evidence for the significance of 
SOD in Campylobacter survival and colonization. Appl Environ Microbiol 1999; 
65:2540-2546. 

24. Breton YL, Maze A, Hartke A, Lemarinier S, Auffray Y, Rince A. Isolation and 
characterization of bile salts-sensitive mutants of Enterococcus faecalis. Curr Microbiol 
2002; 45:434-439. 

25. Bron PA, Marco M, Hoffer SM, Van Mullekom E, de Vos WM, Kleerebezem M. Genetic 
characterization of the bile salt response in Lactobacillus plantarum and analysis of responsive 
promoters in vitro and in situ in the gastrointestinal tract. J Bacteriol 2004; 186:7829-7835. 

26. Taranto MP, Fernandez Murga ML, Lorca G, de Valdez GF. Bile salts and cholesterol induce 
changes in the lipid cell membrane of Lactobacillus reuteri. J Appl Microbiol 2003; 95:86-91. 

27. West NP, Sansonetti PJ, Frankel G, Tang CM. Finding your niche: what has been learnt from 
STM studies on GI colonization. Trends Microbiol 2003; 11:338-344. 

28. Mecsas J. Use of signature-tagged mutagenesis in pathogenesis studies. Curr Opin Microbiol 
2002; 5:33-37. 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 185 

29. Mahan MJ, Heithoff DM, Sinsheimer RL, Low DA. Assessment of bacterial pathogenesis by 
analysis of gene expression in the host. Annu Rev Genet 2000; 34:139-164. 

30. Morrow BJ, Graham JE, Curtiss R, III. Genomic subtractive hybridization and selective capture 
of transcribed sequences identify a novel Salmonella typhimurium fimbrial operon and putative 
transcriptional regulator that are absent from the Salmonella typhi genome. Infect Immun 1999; 
67:5106-5116. 

31. Graham JE, Clark-Curtiss JE. Identification of Mycobacterium tuberculosis RNAs synthesized 
in response to phagocytosis by human macrophages by selective capture of transcribed 
sequences (SCOTS). Proc Natl Acad Sci USA 1999; 96:11554-11559. 

32. Daigle F, Graham JE, Curtiss R, III. Identification of Salmonella typhi genes expressed within 
macrophages by selective capture of transcribed sequences (SCOTS). Mol Microbiol 2001; 
41:1211-1222. 

33. Hou JY, Graham JE, Clark-Curtiss JE. Mycobacterium avium genes expressed during growth in 
human macrophages detected by selective capture of transcribed sequences (SCOTS). Infect 
Immun 2002; 70:3714-3726. 

34. Dozois CM, Daigle F, Curtiss R, III. Identification of pathogen-specific and conserved genes 
expressed in vivo by an avian pathogenic Escherichia coli strain. Proc Natl Acad Sci USA 
2003; 100:247-252. 

35. Mahan MJ, Slauch JM, Mekalanos JJ. Selection of bacterial virulence genes that are 
specifically induced in host tissues. Science 1993; 259:686-688. 

36. Gahan CG, Hill C. The use of listeriolysin to identify in vivo induced genes in the 
gram- positive intracellular pathogen Listeria monocytogenes. Mol Microbiol 2000; 
36:498-507. 

37. Angelichio MJ, Camilli A. In vivo expression technology. Infect Immun 2002; 70:6518-6523. 

38. Lowe AM, Beattie DT, Deresiewicz RL. Identification of novel staphylococcal virulence genes 
by in vivo expression technology. Mol Microbiol 1998; 27:967-976. 

39. Gillaspy AF, Hickmon SG, Skinner RA, Thomas JR, Nelson CL, Smeltzer MS. Role of the 
accessory gene regulator (agr) in pathogenesis of staphylococcal osteomyelitis. Infect Immun 
1995; 63:3373-3380. 

40. Smith HE, Buijs H, de Vries RR, Wisselink HJ, Stockhofe-Zurwieden N, Smits MA. 
Environmentally regulated genes of Streptococcus suis: identification by the use of iron- 
restricted conditions in vitro and by experimental infection of piglets. Microbiology 2001; 
147:271-280. 

41. Lai YC, Peng HL, Chang HY. Identification of genes induced in vivo during Klebsiella 
pneumoniae CG43 infection. Infect Immun 2001; 69:7140-7145. 

42. Janakiraman A, Slauch JM. The putative iron transport system SitABCD encoded on SPI1 
is required for full virulence of Salmonella typhimurium. Mol Microbiol 2000; 
35:1146-1155. 

43. Walter J, Heng NC, Hammes WP, Loach DM, Tannock GW, Hertel C. Identification of 
Lactobacillus reuteri genes specifically induced in the mouse gastrointestinal tract. Appl 
Environ Microbiol 2003; 69:2044-2051. 

44. Kilic AO, Herzberg MC, Meyer MW, Zhao X, Tao L. Streptococcal reporter gene-fusion 
vector for identification of in vivo expressed genes. Plasmid 1999; 42:67-72. 

45. Bron PA, Grangette C, Mercenier A, de Vos WM, Kleerebezem M. Identification of 
Lactobacillus plantarum genes that are induced in the gastrointestinal tract of mice. J Bacteriol 
2004; 186:5721-5729. 

46. Reed RR, Grindley ND. Transposon-mediated site-specific recombination in vitro: DNA 
cleavage and protein-DNA linkage at the recombination site. Cell 1981; 25:721-728. 

47. Austin S, Ziese M, Sternberg N. A novel role for site-specific recombination in maintenance 
of bacterial replicons. Cell 1981; 25:729-736. 

48. Campo N, Daveran-Mingot ML, Leenhouts K, Ritzenthaler P, Le Bourgeois P. Cre-loxP 
recombination system for large genome rearrangements in Lactococcus lactis. Appl Environ 
Microbiol 2002; 68:2359-2367. 



186 Bronetal. 

49. Bron PA, Meijer M, Bongers R, de Vos WM, Kleerebezem M. Competitive population 
dynamics of /W-gene mutants of Lactobacillus plantarum in the gastrointestinal tract of mice. 
In Bron PA, the molecular response of Lactobacillus plantarum to intestinal passage and 
conditions. PhD thesis 2004, Wageningen Centre for Food Sciences, Wageningen. 

50. Fleischmann RD, Adams MD, White O, et al. Whole-genome random sequencing and 
assembly of Haemophilus influenzae Rd. Science 1995; 269:496-512. 

5 1 . Ivanova N, Sorokin A, Anderson I, et al. Genome sequence of Bacillus cereus and comparative 
analysis with Bacillus anthracis. Nature 2003; 423:87-91. 

52. Parkhill J, Dougan G, James KD, et al. Complete genome sequence of a multiple drug resistant 
Salmonella enterica serovar Typhi CT18. Nature 2001;848-852. 

53. Glaser P, Frangeul L, Buchrieser C, et al. Comparative genomics of Listeria species. Science 
2001;294:849-852. 

54. Kunst F, Ogasawara N, Moszer I, et al. The complete genome sequence of the gram-positive 
bacterium Bacillus subtilis. Nature 1997;249-256. 

55. Bolotin A, Wincker P, Mauger S, et al. The complete genome sequence of the lactic acid 
bacterium Lactococcus lactis ssp. lactis IL1403. Genome Res 2001; 11:731-753. 

56. Kleerebezem M, Boekhorst J, Van Kranenburg R, et al. Complete genome sequence of 
Lactobacillus plantarum WCFSL Proc Natl Acad Sci USA 2003; 100:1990-1995. 

57. Pridmore RD, Berger B, Desiere F, et al. The genome sequence of the probiotic intestinal 
bacterium Lactobacillus johnsonii NCC 533. Proc Natl Acad Sci USA 2004; 101:2512-2517. 

58. Klaenhammer T, Altermann E, Arigoni F, et al. Discovering lactic acid bacteria by genomics. 
Antonie Van Leeuwenhoek 2002; 82:29-58. 

59. de Vos WM. Advances in genomics for microbial food fermentations and safety. Curr Opin 
Biotechnol2001; 12:493-498. 

60. Xu J, Bjursell MK, Himrod J, et al. A genomic view of the human- Bacteroides 
thetaiotaomicron symbiosis. Science 2003;2074-2076. 

61. Schell MA, Karmirantzou M, Snel B, et al. The genome sequence of Bifidobacterium longum 
reflects its adaptation to the human gastrointestinal tract. Proc Natl Acad Sci USA 2002; 
99:14422-14427. 

62. Paulsen IT, Banerjei L, Myers GS, et al. Role of mobile DNA in the evolution of vancomycin- 
resistant Enterococcus faecalis. Science 2003; 299:2071-2074. 

63. Hooper LV, Midtvedt T, Gordon JI. How host- microbial interactions shape the nutrient 
environment of the mammalian intestine. Annu Rev Nutr 2002; 22:283-307. 

64. Barrangou R, Altermann E, Hutkins R, Cano R, Klaenhammer TR. Functional and comparative 
genomic analyses of an operon involved in fructooligosaccharide utilization by Lactobacillus 
acidophilus. Proc Natl Acad Sci USA 2003; 100:8957-8962. 

65. Boekhorst J, Siezen RJ, Zwahlen MC, et al. The complete genomes of Lactobacillus plantarum 
and Lactobacillus johnsonii reveal extensive differences in chromosome organization and gene 
content. Microbiology 2004; 150:3601-3611. 

66. Rokbi B, Seguin D, Guy B, et al. Assessment of Helicobacter pylori gene expression within 
mouse and human gastric mucosae by real-time reverse transcriptase PCR. Infect Immun 2001; 
69:4759-4766. 

67. Fitzsimons NA, Akkermans AD, de Vos WM, Vaughan EE. Bacterial gene expression detected 
in human faeces by reverse transcription-PCR. J Microbiol Methods 2003; 55:133-140. 

68. Hooper LV, Xu J, Falk PG, Midtvedt T, Gordon JI. A molecular sensor that allows a gut 
commensal to control its nutrient foundation in a competitive ecosystem. Proc Natl Acad Sci 
USA 1999; 96:9833-9838. 

69. Hooper LV, Stappenbeck TS, Hong CV, Gordon JI. Angiogenins: a new class of microbicidal 
proteins involved in innate immunity. Nat Immunol 2003; 4:269-273. 

70. Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates 
fat storage. Proc Natl Acad Sci USA 2004; 101:15718-15723. 

71. Rawls JF, Samuel BS, Gordon JI. Gnotobiotic zebrafish reveal evolutionarily conserved 
responses to the gut microbiota. Proc Natl Acad Sci USA 2004; 101:4596-4601. 



Molecular Analysis of Host-Microbe Interactions in the Gl Tract 187 

72. Ahrne S, Nobaek S, Jeppsson B, Adlerberth I, Wold AE, Molin G. The normal Lactobacillus 
flora of healthy human rectal and oral mucosa. J Appl Microbiol 1998; 85:88-94. 

73. Zoetendal EG, Collier CT, Koike S, Mackie RI, Gaskins HR. Molecular ecological analysis of 
the gastrointestinal microbiota: a review. J Nutr 2004; 134:465-472. 

74. Zoetendal EG, Cheng B, Koike S, Mackie RI. Molecular microbial ecology of the 
gastrointestinal tract: from phylogeny to function. Curr Issues Intest Microbiol 2004; 5:31-47. 

75. Vaughan EE, de Vries MC, Zoetendal EG, Ben-Amor K, Akkermans AD, de Vos WM. The 
intestinal LABs. Antonie Van Leeuwenhoek 2002; 82:341-352. 

76. Grangette C, Muller-Alouf H, Hols P, et al. Enhanced mucosal delivery of antigen with cell 
wall mutants of lactic acid bacteria. Infect Immun 2004; 72:2731-2737. 

77. Hanniffy S, Wiedermann U, Repa A, et al. Potential and opportunities for use of recombinant 
lactic acid bacteria in human health. Adv Appl Microbiol 2004; 56:1-64. 

78. Reuter MA, Hanniffy S, Wells JM. Expression and delivery of heterologous antigens using 
lactic acid bacteria. Methods Mol Med 2003; 87:101-114. 

79. Vandenbroucke K, Hans W, Van Huysse J, et al. Active delivery of trefoil factors by 
genetically modified Lactococcus lactis prevents and heals acute colitis in mice. 
Gastroenterology 2004; 127:502-513. 

80. Steidler L, Neirynck S, Huyghebaert N, et al. Biological containment of genetically modified 
Lactococcus lactis for intestinal delivery of human interleukin 10. Nat Biotechnol 2003; 
21:785-789. 

81. Steidler L, Hans W, Schotte L, et al. Treatment of murine colitis by Lactococcus lactis 
secreting interleukin- 10. Science 2000; 289:1352-1355. 

82. Steidler L. Live genetically modified bacteria as drug delivery tools: at the doorstep of a new 
pharmacology? Expert Opin Biol Ther 2004; 4:439-441. 

83. Steidler L. Genetically engineered probiotics. Best Pract Res Clin Gastroenterol 2003; 
17:861-876. 

84. Grangette C, Muller-Alouf H, Geoffroy M, Goudercourt D, Turneer M, Mercenier A. 
Protection against tetanus toxin after intragastric administration of two recombinant lactic acid 
bacteria: impact of strain viability and in vivo persistence. Vaccine 2002; 20:3304-3309. 

85. Schloss PD, Handelsman J. Biotechnological prospects from metagenomics. Curr Opin 
Biotechnol 2003; 14:303-310. 

86. Bron PA, Molenaar D, de Vos WM, Kleerebezem M. DNA microarray based identification of 
bile-responsive genes in Lactobacillus plantarum. J Appl Microbiol 2006; in press. 

87. Azcarate-Peril MA, McAuliffe O, Altermann E, Lick S, Russell WM, Klaenhammer TR. 
Microarray analysis of a two-component regulatory system involved in acid resistance and 
proteolytic activity Lactobacillus acidophilus. Appl Environ Microbial 2005; 71:5794-5804. 

88. Pieterse B, Leer RJ, Schuren FH, van der Werf MJ. Unravelling the multiple effects of lactic 
acid stress on Lactobacillus plantarum by transcription profiling. Microbiology 2005; 
151:3881-3894. 



10 

The Infant Intestinal Microbiota in Allergy 



Pirkka V. Kirjavainen and Gregor Reid 

Canadian Research and Development Center for Probiotics, The Lawson Health 
Research Institute, London, Ontario, Canada 



INTRODUCTION 

Allergies represent a condition where impaired immunological tolerance to common 
environmental allergens is the fundamental determinant of the disease. The immuno- 
pathological mechanism of the disease development is poorly understood. It is thought to 
involve complex genetic predisposition, which depending on environmental triggers 
and/or protective factors, may lead to allergic sensitization and development of allergic 
disease and the consequent symptoms (1-5). One environmental factor that has received 
particular interest in recent years is the variation in early microbial exposure, which has 
indisputable, although incompletely understood, effects on immunological maturation. 
Wider acknowledgment of the possible association between microbes and allergic 
diseases followed the introduction of what became known as "hygiene hypothesis" by 
Strachan, 1989 (6). Based upon epidemiological findings, he suggested that the rise in 
prevalence of allergic diseases in past decades was due to factors associated with changes 
in life style such as reduced family size and improved hygiene measures. He assumed that 
these epidemiological correlations reflected reduced opportunities for cross-infections in 
families with young children. 

The basic idea linking microbes and allergies is that adequate microbial exposure 
may be able to direct the early immunological development away from allergic type 
responsiveness. In contrast, inadequate exposure does not provide this necessary stimulus 
and may even promote the development of allergic disease. The original hygiene 
hypothesis was based on infections, but what truly constitutes the characteristics and 
source of "adequate" microbial stimulus remains unknown. Intestinal microbiota are at 
least quantitatively the primary source of host-microbe interactions soon after birth. 
Moreover, the early establishment of the microbiota has been shown to be prerequisite for 
the formation of tolerance to mucosally encountered antigens (7-12). Arguably the best 
clinical evidence linking intestinal microbiota and allergies is provided by preliminary 
trials that have had success in preventing or treating allergic conditions by oral 
administration of intestinal bacterial isolates (13-18). Also, early use of antibiotics has 
been implicated to predispose the infant to allergic sensitization and development of 
allergic disease, although this view is controversial (19,20). The aim of this chapter is to 

189 



190 Kirjavainen and Reid 

summarize the current knowledge of the characteristics of gut microbiota in allergic 
infants and discuss their implication in allergic disease development. 



ALLERGIES— AN OVERVIEW 

Allergies are by definition immunological hypersensitivity reactions to substances 
(allergens), usually proteins, tolerated in defined dose by normal individuals (21). Allergic 
reactions are manifested in allergic diseases such as asthma, eczema, and rhinoconjuncti- 
vitis, each defined by a group of symptoms and signs. The life-impairing effect of these 
diseases varies from subtle to dominant. In addition to impairing physical health there may 
be an impact on social and emotional health, especially in childhood (22). Allergic 
symptoms can significantly disturb productivity in school and work where they are among 
the major causes of absenteeism. The personal and social economic burden is considerable 
(22-24). During the second half of the twentieth century the prevalence of allergic 
diseases has increased in epidemic proportions. The highest prevalence is in children and 
teenagers. With, on average, every fourth child affected, allergic diseases represent the 
most common chronic childhood illnesses in many countries (25,26). The reasons for this 
increase are not known (25,27). 

There are many exceptions, but in most cases in established allergic disease the 
inflammatory cascade leading to the symptoms follows allergen contact at mucosal 
membranes in airways or gastrointestinal tract and is initiated through specific recognition 
by Immunoglobulin E (IgE) antibodies (27). Overactive T helper 2 (Th2) cells may be 
considered as the immunopathological cornerstone of these reactions (28). When, for 
example, pollen-derived aeroallergen is inhaled by a non-allergic subject the immune 
system reacts mildly by producing allergen-specific IgG 2 and IgG 4 antibodies. This is 
probably due to specific recognition and action, e.g., production of interferon (IFN)-y by T 
helper 1 cells (Thl) cells (28,29). In contrast, in allergic individuals Th2 cells typically 
infiltrate to the affected tissue and produce cytokines such as interleukins (IL)-4, -5, -9, 
and, -13. These cytokines promote the production of IgE antibodies, development and 
accumulation of mast cells, eosinophils, and basophils (the primary effector cells in allergic 
inflammation) as well as overproduction of mucus and airway hyper-responsiveness in 
asthma. Recognition of allergens by specific IgE antibodies on the surface of mast cells and 
basophils triggers these cells to release pre- and newly formed proinflammatory 
and vasoactive molecules (e.g., histamine) that may cause tissue damage and other detrimental 
effects. Eosinophilic inflammation contributes to the airway hyper-responsiveness (28). 

It is clear that there is a hereditary trait that predisposes to the formation of allergen- 
specific IgE antibodies and development of allergic disease (27). This genetic 
predisposition, known as atopy, affects arguably as many as 30-50% of the world 
population (2,25,27). Although the immunopathological mechanisms in established 
allergic diseases are well characterized, it is poorly understood how and why atopy leads 
or does not lead to allergic sensitization and why only some sensitized individuals develop 
symptomatic allergic disease (30). Intriguingly, the immune responses to common 
environmental allergens are initially dominated by Th2 cells in all newborn infants but 
these responses are not suppressed in atopic infants during the first year of life (31,32). 
This is thought to be due to defects associated with atopy, for example, impaired 
production of IFN-y, which compromise the normal maturation of Th2 antagonistic Thl 
responses. The major driving force for the Thl maturation is considered to be the nature of 
the microbial exposure encountered after birth. Recent studies indicate that another type 
of T helper cells, collectively referred to as regulatory T cells (Tregs), may also be 



The Infant Intestinal Microbiota in Allergy 191 

involved or even be the chief executers in natural suppression of Th2 mediated responses 
to environmental allergens. At least two types of Tregs have been shown to have this 
ability in humans: (1) CD4 + CD25 + Tregs, which probably mediate their action primarily 
via production of immunosuppressive cytokines transforming growth factor (TGF)-(3 (also 
in membrane-bound form) and IL-10 and (2) IL-10 producing Tregs (33-35). Notably, 
there is indication that the numbers of allergen-specific Tregs may be lower and their 
suppressive ability defective in those subjects who become sensitized (36,37). Also, the 
mechanism of successful allergen-injection immunotherapy has been linked with 
induction of IL-10 Tregs that suppress Th2 responses and induce switching from IgE to 
IgG 4 antibody (33). 



ALLERGY-ASSOCIATED COMPOSITIONAL CHARACTERISTICS 
OF INFANT GUT MICROBIOTA 

The predominant site for host-microbe interaction is in the gut. Thus, its compositional 
development has been suggested to be the key determinant in whether or not the atopic 
genotype will be fully expressed and thereby affect the development of allergic diseases. 
The determination of characteristics in compositional development of intestinal 
microbiota in association with the expression of allergies may provide a starting point 
for elucidating which microbial components, if any, may have particular relevance in 
immunopathology of allergic diseases. 

Studies by Traditional Plate Culture Methods 

The first reports associating allergy with characteristic microbial composition in the gut 
appear to be from studies in the former Soviet Union in the early 1980s (38-40). One of these 
studies, reported also in English, involved an assessment of 60 under one-year-old infants 
with food allergy and atopic eczema. It was claimed that the severity of the disease was in 
direct correlation with the stage of aberrancy in the fecal microbiota. This aberrancy was 
characterized as low prevalence of bifidobacteria and lactobacilli and high prevalence of 
Enterobactericeae, pathogenic species of staplylococci and streptococci as well as Candida 
species (39). Indication that such differences may persist beyond infancy was provided a 
few years later by Ionescu and co-workers (1986) who studied 10- to 45-year-old subjects. 
Subjects with atopic eczema (n = 58) were shown to have lower prevalence of lactobacilli, 
bifidobacteria, and enterococci species than the healthy subjects (n = 21) but higher 
prevalence of Klebsiellae, Proteus, Staphylococcus aureus, Clostridium innocuum and 
Candida species (41,42). Supporting findings were later published by this group from a 
comparison of the fecal microbiota of 30 healthy subjects and 110 subjects with atopic 
eczema (43). 

Although these early studies have not received wider acknowledgment in the 
scientific community, they are well in agreement with later studies that began to accumulate 
a decade later. In one study Klebsiellae species were again found more frequently in the 
feces of 6-month-old infants with atopic eczema (n = 27) and the presence of Streptococcus 
species was less frequent than in the healthy controls (n=10) (44). Collectively, the 
predominant anaerobic and facultatively anaerobic microbiota of allergic infants has been 
characterized by significantly lower prevalence of gram-positive species. In a study by 
Bjorksten and co-workers (1999), colonization by lactobacilli was shown to be less common 
in both Estonian and Swedish two-year-old children with food allergies (n = 27) than in the 
age compatible healthy children (n = 36), whilst the opposite was true for coliforms and 



192 Kirjavainen and Reid 

S. aureus (45). In addition, their results indicated that Bacteroides comprised a larger 
proportion of the whole microbiota in healthy compared to allergic infants. They later 
studied the development of microbiota in a prospective follow-up. Surprisingly, lactobacilli 
were significantly more frequently present during the neonatal period in the feces of infants 
who at 2 years had atopic eczema and/or positive skin prick test (n = 18) than in the feces of 
infants who remained symptom free and had negative skin prick test (n = 26) (46). The rest 
of the characteristics that were associated with allergy were in concordance with the 
previous studies with less frequent presence of bifidobacteria and enterococci during 
the neonatal period. Later in the first year of life, a relatively high prevalence of S. aureus 
and numbers of Clostridia and relatively low numbers of Bacteroides were associated with 
allergic eczema (46). The putative differences in the bifidobacterial microbiota were studied 
at species level by Ouwehand and co-workers (2001) and they found that the feces of 2 to 
7-month-old infants with atopic eczema (n = 7) contained more frequently B. adolescentis 
and less frequently B. bifidum than the feces of healthy infants (n = 6) (47). 

Studies by Molecular Methods 

Results obtained by molecular-based culture-independent techniques are largely supportive 
of the findings presented above. In another prospective follow-up, the fecal microbiota in 
Finnish neonates was studied prior to the expression of atopy as detected by a positive skin 
prick test at year one (n= 12). The microbiota of these sensitized children tended to contain 
lower numbers of bifidobacteria and significantly higher numbers of Clostridium 
histolyticum than those in samples from infants with a negative prick test (n= 17) (48). 
The Clostridium species detectable with the oligonucleotide-probe used in that study 
include common infant gut colonizers such as C paraputrificum, C butyricum and 
C perfringens but not C difficile. However, another study indicated that relatively high 
fecal levels of rarely detected i-caproic acid indicative of C. difficile activity was associated 
with presence of IgE mediated allergic condition in Swedish infants at around one year of 
age (49). The association between low numbers of fecal bifidobacteria and subsequent 
allergic sensitization was confirmed in a study showing that neonatal bifidobacteria 
numbers were significantly lower in children who had food allergen- specific IgE antibodies 
in their serum at 2 years (n = 10) than in those who did not have the antibodies (n = 16) (50). 
In addition, the numbers of bifidobacteria present during the neonatal period correlated 
inversely with total IgE concentration at 2 years (n = 25). In accordance with the association 
suggested by the earlier studies between the high prevalence of coliforms and allergy, 
another study showed a direct correlation between the fecal numbers of Escherichia coli and 
total IgE concentration in infants with early onset atopic eczema at mean age of 5 months 
(n=19) (18). Furthermore, at weaning around 1 year of age total bacterial cell counts 
correlated inversely with the severity of eczema as indicated by severity Scoring Atopic 
Dermatitis (SCORAD) scores (44). 

Somewhat contrasting results to those presented by plate culture methods have also 
been reported. In a study of 6-month-old exclusively breast-fed infants the mean 
bifidobacterial numbers were not found to be lower in the feces of infants with early onset 
atopic eczema (n=15) compared to controls (n=10), with the exception of a small 
subgroup of allergic infants (n = 5) that additionally had gastrointestinal symptoms. 
Moreover, as opposed to studies by Bjorksten and co-workers, Bacteroides numbers were 
higher in a subgroup of allergic infants (n = 6) who were later confirmed to have cow milk 
allergy by challenge (44). Bacteroides numbers were also associated with cow milk allergy 
in a later study where the high counts correlated directly with serum total IgE concentration 
in a subgroup of infants intolerant to extensively hydrolyzed whey formula (n = 7) (18). 



The Infant Intestinal Microbiota in Allergy 



193 



During weaning, the numbers of Clostridium histolyticum correlated inversely with the 
severity of atopic eczema as indicated by SCORAD scores whereas lactobacilli/ 
enterococci numbers correlated directly with the serum total IgE levels (44). It is worthy 
of note that although high total IgE concentration represents phenotypic characteristics 
associated with an atopic background, unlike allergen-specific IgE antibodies, the 
immunopathophysiological significance of total IgE is questionable (51). 

Common Trends and Contradictions 

The microbial characteristics of infants presented above are summarized in Figure 1. 
Although some variability exists depending upon the study, there are relatively clear 
trends evident. The most consistent trends associated with allergy are low numbers of 
bifidobacteria and high numbers of S. aureus and certain species of coliforms and 
Clostridia. It should be pointed out that there are several aspects of these studies that 
complicate their interpretation. A fundamental downfall is the evaluation of intestinal 
microbiota by use of the feces, which may only be indicative of the composition of the 
microbial community in the lower bowel (52,53). Notably, it has been shown that 
the proportional quantities of specific strains in the colonic mucosa may differ from those 
in the feces (54). Moreover, the studies on fecal microbiota reveal little with respect to the 
composition of the small intestine, which immunologically may be more relevant than 
the large intestine. Another significant deficiency in these studies is the lack of more 
detailed characterization, especially at the species and strain level. It is well known that 
bacterial properties, including their immunological effects, vary between bacterial species 



Aerobes in total 

Coliforms 

Staph, aureus 

Clostridia 

Bacteroides 

Lactobacilli 

Enterococci 

Bifidobacteria 

Gram + aerobes 

Anaerobes in total 4 













< X X X > 
X X X X 

X X X X > 
X X X X 

X X X X > 


a 




K"^K"^K*^K*^HT^K*^K*^K"^^n X X X X 

^KjIQK^ x x x x 


X X X X > 

X X X X 
X X X X > 

X X X X 

x y y v •> 


a,b 1 ,c\f 








X X X X 
C X X X X 

X X X X 
t X X X X 

X X X X 




x x x x 

X X X X > 

X X X X 
X X X X > 

X X X X 


a,d 




X X X x~ 

X X X X ) 

X X X X 

x x x x > 

X X X X 
X X X X 1 


"X X X X 
X X X X 

X X X X 
X X X X 

X X X X 








b 2 ,d,e 2 




^tf X X X X 

3 x x x x 

VI x x x x 

31 x x x x 

3H X X X x 






a,c 


X X X X > 
X X X X 

X X X X > 
X X X X 

V V V V ^ 








X X X X 

< X X X X 
X X X X 

< X X X X 

V V V V 




a,b 3 ,d,f 






x x x j^ 
x x x/ 

x >-^ 
X \jr 




< X X X X 
X X X X 

< X X X X 

X X X X 

< X X X X 




b 3 ,d 










a,d,e,f,g 














b 














a,b 



AGE 1 st wk 1 



12 



24 REF. 



months 



Potentially allergy 
preventing bacteria 

Potentially allergy 
promoting bacteria 




Low numbers/frequency associated with allergic disease 

Low numbers/frequency associated with allergic disease & allergic sensitisation 

High numbers associated with allergic sensitisation/high total IgE cone. 

High numbers/frequency associated with allergic disease 

High numbers/frequency associated with allergic disease and high total IgE cone. 

No characteristics determined/detected 



Figure 1 Map of bacterial characteristics in infant microbiota during the first 2 years of life relative 
to development and presence of allergic disease and IgE antibodies and total IgE concentrations. 
Klebsiellae in ref. "a", E. coli in ref. "c", Clostridium histolyticum, lactobacilli and enterococci 
enumerated together, 4 Includes results from total microbial cell counts. Abbreviation: IgE, 
Immunoglobulin E. Source: a, From Ref. 45; b, From Ref. 44; c, From Ref. 18; d, From Ref. 46; e, 
From Ref. 48; f, From Ref. 39; g, From Ref. 50. 



194 Kirjavainen and Reid 

and strains of the same species (55,56). Many of the apparent contradictions in the results 
may therefore reflect the fact that different species or strains within the same genera may 
have dramatically different effects on allergies. Having said that, they could be the result of 
different study protocols, methodologies, and particularly differences in the study 
populations and their nutritional and therapeutic management. Clearly defined study 
populations are particularly important in studies of allergies. This applies even within 
allergic diseases such as atopic eczema, which rather than a single disease is an 
aggregation of several conditions which have certain clinical characteristics in common 
(57). It is difficult to state that specific microbial patterns can be generalized to be common 
in all allergic conditions, in part because the microbiota composition remains to be fully 
elucidated in all the mucosal compartments, and as human genomic and environmental 
exposures differ between individuals. In all the studies reported to date, the composition of 
infant intestinal microbiota has been assessed in relation to development of atopic eczema, 
food allergy or signs of allergic sensitization. This is an obvious shortcoming, albeit 
understandable, as these are nearly exclusive manifestations of allergy in childhood. 

INTERPRETING THE GUT MICROBIOTA CHARACTERISTICS 

The reason for the compositional differences in the average microbiota of allergic and 
healthy infants is not yet known. Undisputable conclusions regarding causal relationship 
cannot be drawn based on mere characterization of microbial composition relative to 
clinical sings and symptoms. In a few studies, characteristics of the fecal microbiota have 
been shown to precede the beginning of the expression of atopy, implying that these 
differences are not necessarily secondary to the disease. However, these, and other studies 
have not taken into account changes that occur in the development of the gut mucosa as 
these likely influence which microbes colonize and how these influence clinical signs 
of allergy. 

Theoretically, there are a number of plausible causes for microbial compositional 
differences seen to date; these are listed in Table 1. Many of these factors are intertwined. 
Some plausible ways by which desirable microbes may protect the host from allergic 
sensitization and alleviate symptoms are presented in Figure 2. 

Reflection of Atopic Genotype 

Incomplete knowledge of the genetic characteristics of allergic diseases restricts the full 
understanding of their possible influence on the development of gut microbiota (58). 
Theoretically, microbial colonization could be directly affected for example if the atopic 
genotype was associated with receptor expression on epithelial cells or production of 
intestinal mucus. There is some indication that the atopic genotype is associated with 

Table 1 Possible Causes for Microbial Compositional Differences in Atopic versus 
Healthy Children 

Atopic genotype related defects in the host's ability to interact with bacteria 

The role of microbial stimulus in the normal maturation of the immune system away from allergic 

type responsiveness 
The influence of allergic symptoms and consequent inflammation on microbial colonization 
The effects microbes have on allergen processing and uptake, for example, by inducing gut 

inflammation 
Environmental factors that affect the expression of atopy in parallel with the microbiota or via the 

microbiota 



The Infant Intestinal Microbiota in Allergy 



195 



Gut barrier 




NO 
SENSITIZATION/ 

ALLERGIC 
INFLAMMATION 



SENSITIZATION/ 
ALLERGIC 

INFLAMMATION 



XXXXXXxxXxx 
XXXXXXXXXXX)* 

XXXXXXXXXXX 



Figure 2 Mechanisms by which specific components of intestinal microbiota may protect from 
allergic sensitization and/or alleviate symptoms. "Adequate" microbial composition may reduce 
allergen uptake by providing maturational stimulus for gut barrier function, enhancing allergen 
degradation by production of digestive enzymes (this may also reduce allergen allergenicity), improving 
mucosal integrity by direct exclusion of pathogens that may cause epithelial damage or by enhancing 
secretory IgA (slgA) production (possibly via inducing TGF-fi secretion) and by inducing secretion of 
anti-inflammatory cytokines, which may break a vicious circle where inflammation increases gut 
permeability allowing invasion of pathogens and allergens, which then results in further inflammation. 
Danger signals caused by epithelial damage and inflammation promote the maturation of dendritic cells, 
which influence the differentiation of naive Th cells. Presentation of allergen in absence of danger signals 
may promote formation of regulatory T cells (Treg) and thus formation of tolerance to the allergen. The 
fate of Th cells in the presence of danger signals depends on additional stimulus: presence of TGF-& 
(produced, e.g., by epithelial cells) may promote development of Treg population and again tolerance to 
the allergen, presence of IL-12 andlFN-y (produced, e.g., by macrophages or dendritic cells) promotes 
development of Thl population and non-allergic type immune responses, whereas presence of IL-10 
may promote formation of allergen specific Th2 cells. In the symptomatic phase induction of anti- 
inflammatory cytokines may also directly alleviate the allergic inflammation by active suppression. 
Abbreviations: slgA, secretory IgA; M, M-cell; iDC, immature dendritic cell; mDC, mature dendritic 
cell; IL, interleukin; TGF, transforming growth factor; Th, T-helper; Treg, regulatory T-cell; M<I>, 
macrophage. 



immunological deviancies that could result in impaired recognition of specific bacterial 
groups and thus allow them to flourish. These defects include compromised expression of 
Toll-like receptor (TLR) 4 and its soluble co-receptor CD 14 (sCD14), albeit the results 
regarding sCD14 are conflicting (59-64). However, also low breast-milk levels of sCD14 
have been associated with subsequent development of eczema in children irrespective of 
atopy (65). TLR4 and sCD14 are pattern recognition receptors of innate immune systems 
that are important in detection of components in both Gram-positive and Gram-negative 
bacteria but especially the cell-wall lipopolysaccharides (LPS) in the latter (66,67). 
Notably, CD14-independent recognition of LPS would seem to be defective during the 
neonatal period (68). Compromised recognition may facilitate colonization by bacteria 



196 Kirjavainen and Reid 

which would otherwise be cleared or reduced in numbers due to immune responses mounted 
against them. This could partly explain why relatively a high prevalence and numbers of 
potentially pathogenic Gram-negative bacteria but low numbers of Gram-positive bacteria 
appear to accompany atopic eczema and high levels of IgE (18,39,42-45,50). 

From another perspective, microbial compositional differences may reflect their 
influence on allergic sensitization and disease development. If the recognition of gut 
colonizers is compromised, then so may be the interactions that drive the normal 
immunological maturation (10,32,60,69,70). Recognition of peptidoglycan, a major 
component of Gram-positive cell- wall, is less dependent on CD 14 and TLR4 but rather on 
co-operation between TLRs 2 and 6 (71-73). Thereby, an atopic host, with deficient TLR4 
and CD 14 recognition, may have better chances to interact with Gram-positive than Gram- 
negative bacteria. This interaction may, on one hand, limit the ability of Gram-positive 
bacteria to colonize the gut, but on the other, provide maturational stimulus for the 
developing immune system (44,69). 

Whereas the recognition of one specific bacterial component occurs primarily via one 
or two different pattern recognition receptors, the recognition of whole bacterium is likely to 
involve a set of different receptors such as TLR9 recognizing unmethylated bacterial CpG 
DNA and TLR5 recognizing flagella (74). Accordingly, a quantitatively strong enough 
exposure may compensate the poor recognition of Gram-negative bacteria, especially due to 
ligation of TLR9. This would be in agreement with the observation that postnatal 
administration of exogenous Gram-negative bacteria, namely non-enteropathogenic E. coli 
strain, was associated with reduced risk of developing allergic diseases later in life (14,15). 

Reflection of Effects on Th1, Th2, and Treg Differentiation 

The effects of intestinal bacteria on cytokine production, epithelia-damaging action or 
proinflammatory action may have a major influence on naive T-cell differentiation to Thl, 
Th2 or Treg cells (Fig. 2). A study in mice with compromised Toll-mediated signaling 
capacity indicated that antigen specific Thl responses to food allergens are dependent on 
simultaneously induced Toll-mediated activities, whilst similar dependency was not 
observed in Th2 responses. Re-exposing the mice to the allergen enhanced the production 
of IL-13 by T-cells, a cytokine capable of inducing isotype class-switching of B-cells to 
produce IgE (75). 

Th differentiation is directed by dendritic cells, which monitor the antigenic 
environment and presence of danger signals in the gut. Danger signals may include 
epithelial damage and inflammation. In the absence of maturational/inflammatory stimuli, 
dendritic cells aim to tolerize the immune system to what they assume to be harmless 
antigens. It is noteworthy that the immunological stimulus initiated may vary depending on 
which TLR or combination of TLRs are ligated (76). This may provide a mechanistic basis 
for consistent data from in vitro studies, which indicate that cytokine responses mounted by 
mononuclear cells in response to whole Gram-negative and whole Gram-positive bacteria 
are different. The induction of IL-12 is greater for Gram-positive bacteria and IL-10 for 
Gram-negative bacteria (77-79). IL-12 is produced by dendritic cells and macrophages and 
is a key cytokine promoting the Th cell differentiation into Thl cells. IL-10 may contribute 
in maintaining a Th2 bias, but it may also induce tolerance by promoting the formation of 
Tregs and anergic T-cells (80-82). 

In a study by He and co-workers (2002) bifidobacteria isolated from the feces of 
allergic infants tended to induce murine macrophage-like cells to produce more of IL-12, 
but less IL-10 than bifidobacteria from the feces of healthy infants (83). In their earlier, 
aforementioned, study B. adolescentis was associated with allergic and B. bifidum with 



The Infant Intestinal Microbiota in Allergy 197 

healthy infants (47). Accordingly, in a recent study, Young and co-workers showed that 
B. bifidum enhanced IL-10 production by dendritic cells isolated from cord blood (84). 
However, B. adolescentis, or any other bifidobacteria! strain, did not induce IL-12 
production. Moderate differences were observed in the effects of bifidobacterial strains on 
the expression of dendritic cell activation markers. The basis for speculation on the possible 
significance of these findings is weak until more detailed characterization is performed. 
Arguably, the findings could collectively indicate that bifidobacteria in allergic infants may 
promote formation of tolerogenic responses but this remains to be confirmed (Fig. 2). 

Also Lactobacillus strains have been shown to confer differential effects on cytokine 
production and expression of surface markers on murine dendritic cells (85). Furthermore, 
lactobacilli induced in vitro, in a strain dependent manner, Treg-like low proliferating Th 
population producing TGF-P and IL-10 (86). TGF-P is the key cytokine in induction of 
T-cell differentiation towards Tregs (Fig. 2) (87). In a clinical study, improvement in 
atopic eczema symptoms following oral administration of lactobacilli was accompanied 
by increased serum concentrations of TGF-P (17). Interestingly, oral supplementation of 
lactobacilli in breast-feeding mothers was followed by increased TGF-P concentrations in 
breast-milk (88). This increase may have contributed to subsequently lower prevalence of 
atopic eczema in children. It should be noted, however, that allergic sensitization was not 
affected and allergic rhinitis and asthma may have increased in frequency (89). 
Nevertheless, these studies are not only indicative of the influence of infant microbiota 
on allergy development but also of the possible influence of maternal microbiota during 
pregnancy and via breast-milk. 

Reflection of Effects on Allergen Uptake, Processing, and Presentation 

The original hygiene hypothesis implicated pathogens in an allergy-preventing role. 
However, their role may be two-sided (90). Whereas the host immune system may become 
tolerant towards commensal microbes, this should and will not happen with pathogens 
(91,92). Therefore, pathogens may have a greater potential to stimulate the neonatal 
immunity away from the allergic type responsiveness than the commensal microbes 
towards which tolerance has been formed (90). Conversely, potential pathogens may 
induce and sustain inflammation and compromise the gut barrier (18,93). This may allow 
greater numbers of allergens to pass the barrier and alter their presentation to lymphocytes 
due to the presence of danger signals. Consequently, allergic sensitization may be more 
likely to occur, and may be aggravated in already sensitized subjects with allergic disease 
(94-96). E. coli and Bacteroides bacterial groups colonizing these subjects may include 
strains with such detrimental properties (97-100). Such bacteria were implicated with 
higher serum total IgE concentrations and sensitivity to cow's milk proteins in studies 
referred to above (18,44). Some non-pathogenic bacteria, such as lactobacilli and 
bifidobacteria, may have the opposite effects by reducing gut inflammation either via 
excluding colonization by pathogens or inducing secretion of anti-inflammatory 
cytokines, reducing gut permeability, allergen antigenicity, and fortifying gut defense 
barrier e.g., by stimulating IgA production (101-110). Intestinal microbes are likely to 
affect the allergen uptake also by promoting the maturation and integrity of gut barrier but 
there is little information on how this ability may vary between different bacteria (111). 



Reflection of Allergic Symptoms 

The possibility that allergic symptoms either affect, or are affected by, the microbiota is 
supported by an observation that alleviation in atopic eczema and allergic inflammation 



198 Kirjavainen and Reid 

following oral administration of bifidobacteria was accompanied by modified dynamics in 
the microbiota (i.e., restriction in the growth of E. coli and Bacteroides) (18). Also, earlier 
findings attest to this possibility implicating direct correlation between numbers of 
Enterobacteriaceae family bacteria and severity of atopic eczema symptoms (39). The 
compositional characteristics associated with the severity of symptoms may be caused by 
intestinal inflammation exacerbated in some allergic conditions (95,112-115). 

Reflection of Environmental Factors 

Amongst the best examples of factors which have been clearly shown to influence the 
development of the gut microbiota and have also been implicated in allergic diseases 
include the mode of delivery and breast-feeding (1 16-123). Indeed, it is plausible that the 
characteristics of fecal microbiota associated with atopic eczema and allergic sensitization 
may partly reflect dietary factors. It is well known that changes in diet may dramatically 
affect the microbial composition of the gut. Then again, in allergic infants the diet can reflect 
the child's health status due to food restrictions. In 39-63% of all infants and young 
children, atopic eczema is triggered by one or more challenge-confirmed food allergies 
(124-126). Moreover, the development of manifestations of allergic diseases in children 
correlates with differences in the composition and immunological characteristics of breast- 
milk, which on the other hand are affected by maternal gut microbiota and atopy ( 1 27-1 33). 
For example, the polyunsaturated fatty acid composition in breast-milk has been shown to 
correlate with the development of allergic disease in children (131,132). In vitro these 
compounds have been shown to selectively affect microbial growth and adhesion to 
intestinal cells (134). Recently, lactobacilli in breast-milk were shown to have properties 
in vitro that could promote the development and maintenance of gut barrier in neonates, thus 
warranting further studies on this area (135). Albeit the effect of caesarean delivery in 
promoting allergy is disputable, it is notable that colonization by Lactobacillus- and 
Bifidobacterium-like bacteria, the high numbers of which have mainly been associated with 
non-allergic phenotype, may be delayed for up to 10 days and 1 month, respectively, as 
compared to vaginally delivered infants (136). 

Regarding our earlier discussion on pathogens and E. coli, it is noteworthy that in 
developing countries with low prevalence of allergies, the establishment of intestinal 
microbiota is characterized by rapid initial colonization, formation of enterobacterial 
microbiota predominated by E. coli, and frequent colonization by pathogens such as 
salmonellae. The E. coli population is characterized by a wide spectrum of strains and 
instability (137,138). Whether such rapid colonization and strongly variable exposure has 
special influence on immunological maturation and gut barrier formation and maintenance 
remain to be established. 



CONCLUSION 

It has been well established that allergic sensitization and the development of allergic 
disease are associated, at least in some infants, with characteristic developmental patterns 
in fecal microbiota composition that are atypical to healthy infants. With relative 
consistency these characteristics include low numbers of bifidobacteria and anaerobes in 
total and high numbers of Clostridia, S. aureus and certain coliforms such as Klebsiellae. 
Data on lactobacilli, Bacteroides and E. coli are somewhat variable. How this aberrancy in 
fecal microbiota depicts the situation in the intestine and how it is clinically significant, 
remains to be known. The possibility that the characteristics are secondary to the disease 



The Infant Intestinal Microbiota in Allergy 199 

cannot be excluded, but it is also feasible that they reflect their significance in the aetiology 
of allergy. Extensive experimental data implies that the development of atopic type 
immunoreactivity could be promoted by the establishment of an early gut microbiota that 
(1) is incapable of directing the immune system towards tolerogenic responses to, what 
should be, harmless environmental antigens and/or (2) induces inflammatory responses 
against itself, thereby increasing mucosal permeability to potential allergens. 

It has been convincingly demonstrated that microbial exposure is likely to be the 
primary exogenous stimulus directing the immunological maturation away from allergic 
type immunoresponsiveness early in life. However, it is still not clear what are the 
qualitative or quantitative characteristics of the indigenous microbiota or other sources of 
microbial exposure that could protect from, or conversely promote ("allow"), the 
expression of allergies. Future studies should assess whether specific microbial species 
have particular importance in this respect or whether the "adequate" stimulus is only a 
matter of quantitatively high enough exposure or strongly variable exposure. More efforts 
should be directed to characterizing microbial composition of nasal and oral cavities and 
different compartments in the intestinal tract of children as well as the gut of pregnant 
women and the gut and breast-milk of breast-feeding mothers. 



ACKNOWLEDGMENTS 



Pirkka Kirjavainen gratefully acknowledges financial support from the Academy 
of Finland. 



REFERENCES 

1. Arruda LK, Sole D, Baena-Cagnani CE, Naspitz CK. Risk factors for asthma and atopy. Curr 
Opin Allergy Clin Immunol 2005; 5:153-159. 

2. Steinke JW, Borish L, Rosenwasser LJ 5. Genetics of hypersensitivity. J Allergy Clin 
Immunol 2003; 111:S495-S501. 

3. von Mutius E. Influences in allergy: epidemiology and the environment. J Allergy Clin 
Immunol 2004; 113:373-379; quiz 380. 

4. von Mutius E. The environmental predictors of allergic disease. J Allergy Clin Immunol 2000; 
105:9-19. 

5. O'Connell EJ. Pediatric allergy: a brief review of risk factors associated with developing 
allergic disease in childhood. Ann Allergy Asthma Immunol 2003; 90:53-58. 

6. Strachan DP. Hay fever, hygiene, and household size. BMJ 1989; 299:1259-1260. 

7. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? 
Trends Microbiol 2004; 12:562-568. 

8. Moreau MC, Corthier G. Effect of the gastrointestinal microflora on induction and 
maintenance of oral tolerance to ovalbumin in C3H/HeJ mice. Infect Immun 1988; 
56:2766-2768. 

9. Moreau MC, Gaboriau-Routhiau V. The absence of gut flora, the doses of antigen ingested 
and aging affect the long-term peripheral tolerance induced by ovalbumin feeding in mice. 
Res Immunol 1996; 147:49-59. 

10. Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal 
bacterial flora for the development of an IgE production system fully susceptible to oral 
tolerance induction. J Immunol 1997; 159:1739-1745. 

11. Gaboriau-Routhiau V, Moreau MC. Gut flora allows recovery of oral tolerance to ovalbumin 
in mice after transient breakdown mediated by cholera toxin or Escherichia coli heat-labile 
enterotoxin. Pediatr Res 1996; 39:625-629. 



200 Kirjavainen and Reid 

12. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science 2001; 
292:1115-1118. 

13. Kalliomaki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in 
primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet 2001; 
357:1076-1079. 

14. Lodinova- Zadnikova R, Cukrowska B. Influence of oral colonization of the intestine with a 
non-enteropathogenic E. coli strain after birth on the frequency of infectious and allergic 
diseases after 10 and 20 years. Immunol Lett 1999; 69:64. 

15. Lodinova-Zadnikova R, Cukrowska B, Tlaskalova-Hogenova H. Oral administration of 
probiotic Escherichia coli after birth reduces frequency of allergies and repeated infections 
later in life (after 10 and 20 years). Int Arch Allergy Immunol 2003; 131:209-211. 

16. Majamaa H, Isolauri E. Probiotics: a novel approach in the management of food allergy. 
J Allergy Clin Immunol 1997; 99:179-185. 

17. Isolauri E, Arvola T, Sutas Y, Moilanen E, Salminen S. Probiotics in the management of 
atopic eczema. Clin Exp Allergy 2000; 30:1604-1610. 

18. Kirjavainen PV, Arvola T, Salminen SJ, Isolauri E. Aberrant composition of gut microbiota of 
allergic infants: a target of bifidobacteria! therapy at weaning? Gut 2002; 51:51-55. 

19. Celedon JC, Weiss ST. Use of antibacterials in infancy: clinical implications for childhood 
asthma and allergies. Treat Respir Med 2004; 3:291-294. 

20. Voor T, Julge K, Bottcher MF, Jenmalm MC, Duchen K, Bjorksten B. Atopic sensitization 
and atopic dermatitis in estonian and Swedish infants. Clin Exp Allergy 2005; 35:153-159. 

21 . Johansson SG, Bieber T, Dahl R, et al. Revised nomenclature for allergy for global use: report 
of the nomenclature review committee of the world allergy organization, October 2003. 
J Allergy Clin Immunol 2004; 113:832-836. 

22. O'Connell EJ. The burden of atopy and asthma in children. Allergy 2004; 78:7-11. 

23. Blaiss MS. Important aspects in management of allergic rhinitis: compliance, cost, and quality 
of life. Allergy Asthma Proc 2003; 24:231-238. 

24. Cisternas M, Blanc P, Yen I, et al. A comprehensive study of the direct and indirect costs of 
adult asthma. J Allergy Clin Immunol 2003; 111:1212-1218. 

25. Jarvis D, Burney P. ABC of allergies. The epidemiology of allergic disease. BMJ 1998; 
316:607-610. 

26. Wickman M, Lilja G. Today, one child in four has an ongoing allergic disease in Europe. 
What will the situation be tomorrow? Allergy 2003; 58:570-571. 

27. Johansson SG, Hourihane JO, Bousquet J, et al. A revised nomenclature for allergy. An 
EAACI position statement from the EAACI nomenclature task force. Allergy 2001; 
56:813-824. 

28. Kay AB. Allergy and allergic diseases. First of two parts. N Engl J Med 2001; 344:30-37. 

29. Ebner C, Schenk S, Najafian N, et al. Nonallergic individuals recognize the same T cell 
epitopes of Bet v 1, the major birch pollen allergen, as atopic patients. J Immunol 1995; 
154:1932-1940. 

30. Prescott SL. Allergy: when does it begin and where will it end?. Allergy 2003; 58:864-867. 

3 1 . Prescott SL, Macaubas C, Holt BJ, et al. Transplacental priming of the human immune system 
to environmental allergens: universal skewing of initial T cell responses toward the Th2 
cytokine profile. J Immunol 1998; 160:4730-4737. 

32. Prescott SL, Macaubas C, Smallacombe T, Holt BJ, Sly PD, Holt PG. Development of 
allergen-specific T-cell memory in atopic and normal children. Lancet 1999; 353:196-200. 

33. Robinson DS, Larche M, Durham SR. Tregs and allergic disease. J Clin Invest 2004; 
114:1389-1397. 

34. Nakamura K, Kitani A, Fuss I, et al. TGF-beta 1 plays an important role in the mechanism of 
CD4 + CD25+ regulatory T cell activity in both humans and mice. J Immunol 2004; 
172:834-842. 

35. Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by 
CD4( + )CD25( + ) regulatory T cells is mediated by cell surface-bound transforming 
growth factor beta. J Exp Med 2001; 194:629-644. 



The Infant Intestinal Microbiota in Allergy 201 

36. Ling EM, Smith T, Nguyen XD, et al. Relation of CD4 + CD25 + regulatory T-cell 
suppression of allergen-driven T-cell activation to atopic status and expression of allergic 
disease. Lancet 2004; 363:608-615. 

37. Akdis M, Verhagen J, Taylor A, et al. Immune responses in healthy and allergic individuals 
are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 
cells. J Exp Med 2004; 199:1567-1575. 

38. Kuvaeva IB, Zakharova NV, Orlova NG, Veselova OL. Functional state of the immunological 
system and of the gastrointestinal tract in children with a food allergy. Vopr Pitan 
1980;33-40. 

39. Kuvaeva IB, Orlova NG, Veselova OL, Kuznezova GG, Borovik TE. Microecology of the 
gastrointestinal tract and the immunological status under food allergy. Nahrung 1984; 
28:689-693. 

40. Shaternikov VA, Kuvaeva ID, Ladodo KS, Orlova NG, Veselova OL. General and local 
humoral immunity and intestinal microflora in children with skin manifestations of food 
allergy. Vopr Pitan 1982;51-56. 

41. Ionescu G, Radovicic D, Schuler R, et al. Changes in fecal microflora and malabsorption 
phenomena suggesting a contaminated small bowel syndrome in atopic eczema patients. 
Microecol Ther 1986; 16:273. 

42. Ionescu G, Kiehl R, Ona L, Schuler R. Abnormal fecal microflora and malabsorption 
phenomena in atopic eczema paitents. J Adv Med 1990; 3:71-91. 

43. Ionescu G, Kiehl R, Wichmann-Kunz F, Leimbeck R. Immunobiological significance of 
fungal and bacterial infections in atopic eczema. J Adv Med 1990; 3:47-58. 

44. Kirjavainen PV, Apostolou E, Arvola T, Salminen SJ, Gibson GR, Isolauri E. Characterizing 
the composition of intestinal microflora as a prospective treatment target in infant allergic 
disease. FEMS Immunol Med Microbiol 2001; 32:1-7. 

45. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic estonian 
and Swedish 2-year-old children. Clin Exp Allergy 1999; 29:342-346. 

46. Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal 
microflora during the first year of life. J Allergy Clin Immunol 2001; 108:516-520. 

47. Ouwehand AC, Isolauri E, He F, Hashimoto H, Benno Y, Salminen S. Differences in 
Bifidobacterium flora composition in allergic and healthy infants. J Allergy Clin Immunol 
2001; 108:144-145. 

48. Kalliomaki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of 
neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin 
Immunol 2001; 107:129-134. 

49. Bottcher MF, Nordin EK, Sandin A, Midtvedt T, Bjorksten B. Microflora-associated 
characteristics in faeces from allergic and nonallergic infants. Clin Exp Allergy 2000; 
30:1590-1596. 

50. Kirjavainen P. The Intestinal Microbiota — A Target for Treatment in Infant Atopic 
Eczema, in Department of Biochemistry and Food Chemistry. Turku: University of 
Turku, 2003: 79. 

51. Howard TD, Meyers DA, Bleecker ER. Mapping susceptibility genes for allergic diseases. 
Chest 2003; 123:363S-368S. 

52. Mikelsaar M, Mandar R, Sepp E. Lactic acid microflora in the human microbial ecosystem 
and its development. In: Salminen S, Von Wright A, eds. Lactic Acid Bacteria: Microecology 
and Functional Aspects. New York: Marcel Dekker Inc., 1998:278-342. 

53. Marteau P, Pochart P, Dore J, Bera-Maillet C, Bernalier A, Corthier G. Comparative study of 
bacterial groups within the human cecal and fecal microbiota. Appl Environ Microbiol 2001; 
67:4939-4942. 

54. Zoetendal EG, von Wright A, Vilpponen-Salmela T, Ben-Amor K, Akkermans AD, de 
Vos WM. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly 
distributed along the colon and differ from the community recovered from feces. Appl 
Environ Microbiol 2002; 68:3401-3407. 



202 Kirjavainen and Reid 

55. Kirjavainen PV, EINezami HS, Salminen SJ, Ahokas JT, Wright PF. Effects of orally 
administered viable Lactobacillus rhamnosus GG and Propionibacterium freudenreichii 
subsp. shermanii JS on mouse lymphocyte proliferation. Clin Diagn Lab Immunol 1999; 
6:799-802. 

56. Kirjavainen PV, El-Nezami HS, Salminen SJ, Ahokas JT, Wright PF. The effect of orally 
administered viable probiotic and dairy lactobacilli on mouse lymphocyte proliferation. 
FEMS Immunol Med Microbiol 1999; 26:131-135. 

57. Wollenberg A, Bieber T. Atopic dermatitis: from the genes to skin lesions. Allergy 2000; 
55:205-213. 

58. Feijen M, Gerritsen J, Postma DS. Genetics of allergic disease. Br Med Bull 2000; 
56:894-907. 

59. Koppelman GH, Reijmerink NE, Colin Stine O, et al. Association of a promoter 
polymorphism of the CD14 gene and atopy. Am J Respir Crit Care Med 2001; 163:965-969. 

60. Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, Martinez FD. A Polymorphism* 
in the 5 ; flanking region of the CD14 gene is associated with circulating soluble CD14 levels 
and with total serum immunoglobulin E. Am J Respir Cell Mol Biol 1999; 20:976-983. 

61. Zdolsek HA, Jenmalm MC. Reduced levels of soluble CD 14 in atopic children. Clin Exp 
Allergy 2004; 34:532-539. 

62. Kabesch M, Hasemann K, Schickinger V, et al. A promoter polymorphism in the CD 14 gene 
is associated with elevated levels of soluble CD 14 but not with IgE or atopic diseases. Allergy 
2004; 59:520-525. 

63. Kedda MA, Lose F, Duffy D, Bell E, Thompson PJ, Upham J. The CD14 C-159T 
polymorphism is not associated with asthma or asthma severity in an Australian adult 
population. Thorax 2005; 60:211-214. 

64. Fageras Bottcher M, Hmani-Aifa M, Lindstrom A, et al. A TLR4 polymorphism is associated 
with asthma and reduced lipopolysaccharide-induced interleukin-12(p70) responses in 
Swedish children. J Allergy Clin Immunol 2004; 114:561-567. 

65. Jones CA, Holloway J A, Popplewell EJ, et al. Reduced soluble CD 14 levels in amniotic fluid 
and breast milk are associated with the subsequent development of atopy, eczema, or both. 
J Allergy Clin Immunol 2002; 109:858-866. 

66. Haziot A, Ferrero E, Kontgen F, et al. Resistance to endotoxin shock and reduced 
dissemination of gram-negative bacteria in CD14-deficient mice. Immunity 1996; 4:407-414. 

67. Miller SI, Ernst RK, Bader MW. LPS, TLR4 and infectious disease diversity. Nat Rev 
Microbiol 2005; 3:36^16. 

68. Cohen L, Haziot A, Shen DR, et al. CD14-independent responses to LPS require a serum 
factor that is absent from neonates. J Immunol 1995; 155:5337-5342. 

69. Kirjavainen PV. Exposure to gram-positive bacteria: the key in natural defence against atopic 
sensitisation?. Microecol Ther 2002;109-114. 

70. Sudo N, Yu XN, Aiba Y, et al. An oral introduction of intestinal bacteria prevents the 
development of a long-term Th2-skewed immunological memory induced by neonatal 
antibiotic treatment in mice. Clin Exp Allergy 2002; 32:1112-1116. 

71. Ozinsky A, Underhill DM, Fontenot JD, et al. The repertoire for pattern recognition of 
pathogens by the innate immune system is defined by cooperation between toll-like receptors. 
Proc Natl Acad Sci USA 2000; 97:13766-13771. 

72. Ozinsky A, Smith KD, Hume D, Underhill DM. Co-operative induction of pro-inflammatory 
signaling by Toll-like receptors. J Endotoxin Res 2000; 6:393-396. 

73. Dziarski R, Ulmer AJ, Gupta D. Interactions of CD 14 with components of gram-positive 
bacteria. Chem Immunol 2000; 74:83-107. 

74. Takeda K, Akira S. Toll-like receptors in innate immunity. Int Immunol 2005; 17:1-14. 

75. Schnare M, Barton GM, Holt AC, Takeda K, Akira S, Medzhitov R. Toll-like receptors 
control activation of adaptive immune responses. Nat Immunol 2001; 2:947-950. 

76. Aderem A, Ulevitch RJ. Toll-like receptors in the induction of the innate immune response. 
Nature 2000; 406:782-787. 



The Infant Intestinal Microbiota in Allergy 203 

77. Cross ML, Ganner A, Teilab D, Fray LM. Patterns of cytokine induction by gram- 
positive and gram-negative probiotic bacteria. FEMS Immunol Med Microbiol 2004; 
42:173-180. 

78. Karlsson H, Hessle C, Rudin A. Innate immune responses of human neonatal cells to bacteria 
from the normal gastrointestinal flora. Infect Immun 2002; 70:6688-6696. 

79. Hessle C, Andersson B, Wold AE. Gram-positive bacteria are potent inducers of monocytic 
interleukin-12 (IL-12) while gram-negative bacteria preferentially stimulate IL-10 
production. Infect Immun 2000; 68:3581-3586. 

80. Raghupathy R. Pregnancy: success and failure within the Thl/Th2/Th3 paradigm. Semin 
Immunol 2001; 13:219-227. 

81. Akdis CA, Blaser K. Mechanisms of interleukin-10-mediated immune suppression. 
Immunology 2001; 103:131-136. 

82. Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, Roncarolo MG. Differentiation of 
Trl cells by immature dendritic cells requires IL-10 but not CD25 + CD4+ Tr cells. Blood 
2005; 105:1162-1169. 

83. He F, Morita H, Hashimoto H, et al. Intestinal Bifidobacterium species induce varying 
cytokine production. J Allergy Clin Immunol 2002; 109:1035-1036. 

84. Young SL, Simon MA, Baird MA, et al. Bifidobacteria! species differentially affect 
expression of cell surface markers and cytokines of dendritic cells harvested from cord blood. 
Clin Diagn Lab Immunol 2004; 11:686-690. 

85. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of 
cytokines and maturation surface markers in murine dendritic cells. J Immunol 2002; 
168:171-178. 

86. von der Weid T, Bulliard C, Schiffrin EJ. Induction by a lactic acid bacterium of a population 
of CD4( + ) T cells with low proliferative capacity that produce transforming growth factor 
beta and interleukin-10. Clin Diagn Lab Immunol 2001; 8:695-701. 

87. Huber S, Schramm C, Lehr HA, et al. Cutting edge: TGF-beta signaling is required for the 
in vivo expansion and immunosuppressive capacity of regulatory CD4 + CD25 + T cells. 
J Immunol 2004; 173:6526-6531. 

88. Rautava S, Kalliomaki M, Isolauri E. Probiotics during pregnancy and breast-feeding might 
confer immunomodulatory protection against atopic disease in the infant. J Allergy Clin 
Immunol 2002; 109:119-121. 

89. Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E. Probiotics and prevention of 
atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 2003; 
361:1869-1871. 

90. Kirjavainen PV. In: Mattila-Sandholm T, Saarela M, eds. Probiotics and the management of 
food allergy, in functional dairy products. Cambridge, U.K.: Woodhead Publishing, 
2003:108-131. 

91. Neish AS, Gewirtz AT, Zeng H, et al. Prokaryotic regulation of epithelial responses by 
inhibition of IkappaB -alpha ubiquitination. Science 2000; 289:1560-1563. 

92. Nagler- Anderson C. Man the barrier! Strategic defences in the intestinal mucosa. Nat Rev 
Immunol 2001; 1:59-67. 

93. Kirjavainen PV, Apostolou E, Salminen SJ, Isolauri E. New aspects of probiotics-a novel 
approach in the management of food allergy. Allergy 1999; 54:909-915. 

94. Batt RM, Rutgers HC, Sancak AA. Enteric bacteria: friend or foe?. J Small Anim Pract 1996; 
37:261-267. 

95. Berin MC, Yang PC, Ciok L, Waserman S, Perdue MH. Role for IL-4 in macromolecular 
transport across human intestinal epithelium. Am J Physiol 1999; 276:C1046-C1052. 

96. Gee JM, Wal JM, Miller K, et al. Effect of saponin on the transmucosal passage of beta- 
lactoglobulin across the proximal small intestine of normal and beta-lactoglobulin-sensitised 
rats. Toxicology 1997; 117:219-228. 

97. Dahlgren UI, Wold AE, Hanson LA, Midtvedt T. Expression of a dietary protein in E. coli 
renders it strongly antigenic to gut lymphoid tissue. Immunology 1991; 73:394-397. 



204 Kirjavainen and Reid 

98. Deitch EA, Specian RD, Berg RD. Endotoxin-induced bacterial translocation and mucosal 
permeability: role of xanthine oxidase, complement activation, and macrophage products. Crit 
Care Med 1991; 19:785-791. 

99. Obiso RJ, Jr., Lyerly DM, Van Tassell RL, Wilkins TD. Proteolytic activity of the Bacteroides 
fragilis enterotoxin causes fluid secretion and intestinal damage in vivo. Infect Immun 1995; 
63:3820-3826. 

100. Duchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer zum KH. Buschenfelde, 
Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel 
disease (IBD). Clin Exp Immunol 1995; 102:448-455. 

101 . Moreau MC, Ducluzeau R, Guy-Grand D, Muller MC. Increase in the population of duodenal 
immunoglobulin A plasmocytes in axenic mice associated with different living or dead 
bacterial strains of intestinal origin. Infect Immun 1978; 21:532-539. 

102. De Simone C, Ciardi A, Grassi A, et al. Effect of Bifidobacterium bifidum and Lactobacillus 
acidophilus on gut mucosa and peripheral blood B lymphocytes. Immunopharmacol 
Immunotoxicol 1992; 14:331-340. 

103. Kaila M, Isolauri E, Soppi E, Virtanen E, Laine S, Arvilommi H. Enhancement of the 
circulating antibody secreting cell response in human diarrhea by a human Lactobacillus 
strain. Pediatr Res 1992; 32:141-144. 

104. Yasui H, Nagaoka N, Mike K, Hayakawa K, Ohwaki M. Detection of Bifidobacterium strains 
that induce large quantities of IgA. Microb Ecol Health Dis 1992; 5:155-162. 

105. Majamaa H, Isolauri E, Saxelin M, Vesikari T. Lactic acid bacteria in the treatment of acute 
rotavirus gastroenteritis. J Pediatr Gastroenterol Nutr 1995; 20:333-338. 

106. Isolauri E, Majamaa H, Arvola T, Rantala I, Virtanen E, Arvilommi H. Lactobacillus casei 
strain GG reverses increased intestinal permeability induced by cow milk in suckling rats. 
Gastroenterology 1993; 105:1643-1650. 

107. Matsuzaki T, Yamazaki R, Hashimoto S, Yokokura T. The effect of oral feeding of 
Lactobacillus casei strain Shirota on immunoglobulin E production in mice. J Dairy Sci 1998; 
81:48-53. 

108. Siitas Y, Soppi E, Korhonen H, et al. Suppression of lymphocyte proliferation in vitro by 
bovine caseins hydrolyzed with Lactobacillus casei GG-derived enzymes. J Allergy Clin 
Immunol 1996; 98:216-224. 

109. Siitas Y, Hurme M, Isolauri E. Down-regulation of anti-CD3 antibody-induced IL-4 
production by bovine caseins hydrolysed with Lactobacillus GG-derived enzymes. Scand 
J Immunol 1996; 43:687-689. 

110. Pessi T, Isolauri E, Siitas Y, Kankaanranta H, Moilanen E, Hurme M. Suppression of T-cell 
activation by Lactobacillus rhamnosus GG-degraded bovine casein. Int Immunopharmacol 
2001; 1:211-218. 

111. Hooper LV, Falk PG, Gordon JI. Analyzing the molecular foundations of commensalism in 
the mouse intestine. Curr Opin Microbiol 2000; 3:79-85. 

112. Ogawa H, Yoshiike T. A speculative view of atopic dermatitis: barrier dysfunction in 
pathogenesis. J Dermatol Sci 1993; 5:197-204. 

113. Majamaa H, Laine S, Miettinen A. Eosinophil protein X and eosinophil cationic protein as 
indicators of intestinal inflammation in infants with atopic eczema and food allergy. Clin Exp 
Allergy 1999; 29:1502-1506. 

114. Majamaa H, Miettinen A, Laine S, Isolauri E. Intestinal inflammation in children with atopic 
eczema: faecal eosinophil cationic protein and tumour necrosis factor-alpha as non-invasive 
indicators of food allergy. Clin Exp Allergy 1996; 26:181-187. 

115. Majamaa H, Aittoniemi J, Miettinen A. Increased concentration of fecal alpha 1 -antitrypsin is 
associated with cow's milk allergy in infants with atopic eczema. Clin Exp Allergy 2001; 
31:590-592. 

116. Xu B, Pekkanen J, Hartikainen AL, Jarvelin MR. Caesarean section and risk of asthma and 
allergy in adulthood. J Allergy Clin Immunol 2001; 107:732-733. 

117. Kero J, Gissler M, Gronlund MM, et al. Mode of delivery and asthma — is there a connection?. 
Pediatr Res 2002; 52:6-11. 



The Infant Intestinal Microbiota in Allergy 205 

118. Saarinen UM, Kajosaari M, Backman A, Siimes MA. Prolonged breast-feeding as prophylaxis 
for atopic disease. Lancet 1979; 2:163-166. 

119. Gdalevich M, Mimouni D, Mimouni M. Breast-feeding and the risk of bronchial asthma in 
childhood: a systematic review with meta-analysis of prospective studies. J Pediatr 2001; 
139:261-266. 

120. Gdalevich M, Mimouni D, David M, Mimouni M. Breast-feeding and the onset of atopic 
dermatitis in childhood: a systematic review and meta-analysis of prospective studies. J Am 
Acad Dermatol 2001; 45:520-527. 

121. Schoetzau A, Filipiak-Pittroff B, Franke K, et al. Effect of exclusive breast-feeding and early 
solid food avoidance on the incidence of atopic dermatitis in high-risk infants at 1 year of age. 
Pediatr Allergy Immunol 2002; 13:234-242. 

122. Kull I, Wickman M, Lilja G, Nordvall SL, Pershagen G. Breast feeding and allergic diseases 
in infants-a prospective birth cohort study. Arch Dis Child 2002; 87:478^-81. 

123. Kirjavainen PV, Gibson GR. Healthy gut microflora and allergy: factors influencing 
development of the microbiota. Ann Med 1999; 31:288-292. 

124. Sampson HA. The immunopathogenic role of food hypersensitivity in atopic dermatitis. Acta 
Derm Venereol Suppl (Stockh) 1992; 176:34-37. 

125. Isolauri E, Turjanmaa K. Combined skin prick and patch testing enhances identification of 
food allergy in infants with atopic dermatitis. J Allergy Clin Immunol 1996; 97:9-15. 

126. Burks AW, James JM, Hiegel A, et al. Atopic dermatitis and food hypersensitivity reactions. 
J Pediatr 1998; 132:132-136. 

127. Kalliomaki M, Ouwehand A, Arvilommi H, Kero P, Isolauri E. Transforming growth factor- 
beta in breast milk: a potential regulator of atopic disease at an early age. J Allergy Clin 
Immunol 1999; 104:1251-1257. 

128. Jarvinen KM, Laine S, Suomalainen H. Defective tumour necrosis factor- alpha production in 
mother's milk is related to cow's milk allergy in suckling infants. Clin Exp Allergy 2000; 
30:637-643. 

129. Jarvinen KM, Laine ST, Jarvenpaa AL, Suomalainen HK. Does low IgA in human milk 
predispose the infant to development of cow's milk allergy? Pediatr Res 2000; 48:457-462. 

130. Jarvinen KM, Suomalainen H. Leucocytes in human milk and lymphocyte subsets in cow's 
milk-allergic infants. Pediatr Allergy Immunol 2002; 13:243-254. 

131. Duchen K, Casas R, Fageras-Bottcher M, Yu G, Bjorksten B. Human milk polyunsaturated 
long-chain fatty acids and secretory immunoglobulin A antibodies and early childhood 
allergy. Pediatr Allergy Immunol 2000; 11:29-39. 

132. Duchen K, Bjorksten B. Polyunsaturated n-3 fatty acids and the development of atopic 
disease. Lipids 2001; 36:1033-1042. 

133. Thijs C, Houwelingen A, Poorterman I, Mordant A, van den Brandt P. Essential fatty acids in 
breast milk of atopic mothers: comparison with non-atopic mothers, and effect of borage oil 
supplementation. Eur J Clin Nutr 2000; 54:234-238. 

134. Kankaanpaa P, Nurmela K, Erkkila A, et al. Polyunsaturated fatty acids in maternal diet, 
breast milk, and serum lipid fatty acids of infants in relation to atopy. Allergy 2001; 
56:633-638. 

135. Martin R, Olivares M, Marin ML, Fernandez L, Xaus J, Rodriguez JM. Probiotic potential of 3 
Lactobacilli strains isolated from breast milk. J Hum Lact 2005; 21:8-17 quiz 18-21, 41. 

136. Gronlund MM, Lehtonen OP, Eerola E, Kero P. Fecal microflora in healthy infants born by 
different methods of delivery: permanent changes in intestinal flora after cesarean delivery. 
J Pediatr Gastroenterol Nutr 1999; 28:19-25. 

o 

137. Adlerberth I. In: Hanson LA, Yolken RH, eds. Establishment of Normal Intestinal Microflora 
in the Newborn Infant., in Probiotics, Other Nutritional Factors, and the Intestinal Flora. 
Philadelphia: Vevey/Lippicott-Raven Publishers, 1999. 

138. Mata LJ, Urrutia JJ. Intestinal colonization of breastfed children in a rural area of low socio- 
economic level. Ann NY Acad Sci 1971; 176:93-109. 



11 

Probiotics: A Role in Therapy for 
Inflammatory Bowel Disease 



Barbara Sheil, Jane McCarthy, Liam O'Mahony, and Malik M. Anwar 

Alimentary Pharmabiotic Centre, Departments of Medicine and Surgery, Microbiology 
National Food Biotechnology Centre, National University of Ireland, Cork, Ireland 

Fergus Shanahan 

Alimentary Pharmabiotic Centre, Departments of Medicine and Surgery, National 
University of Ireland, Cork, Ireland 



INTRODUCTION 

Hippocrates is credited with saying: "Let food be thy medicine and medicine be thy food" 
(1). The term "functional food" includes "any food or food ingredient that may provide a 
health benefit beyond the traditional nutrients it contains" (2). Probiotic bacteria are forms 
of functional food that are of particular relevance to gastroenterologists, with evidence for 
their role in the treatment of infectious and antibiotic-associated diarrhea. Their putative 
therapeutic role in inflammatory bowel disease (IBD) is receiving growing interest; 
however, it remains unproven. The Noble laureate, Elie Metchnikoff, suggested that 
bacteria could be of some benefit to the health of man (3). He suggested that the 
consumption of copious amounts of fermented dairy products, which served to introduce 
"beneficial" bacteria to the gastrointestinal tract, was responsible for the longevity of 
Bulgarian peasants. This marked the birth of probiotics, which are live microorganisms 
that, when consumed in an adequate amount, confer a health effect on the host (4). 

The last decade has seen a resurgence of interest in probiotic research. This renewal 
of interest in enteric (intestinal) microbiota and gut host-microbe interactions has been 
generated for a number of reasons. Firstly, the gut contains a complex microbial 
community, the composition of which has remained elusive due to limited bacteriological 
culturing techniques. Molecular techniques have now been applied to accurately profile 
intestinal bacterial groups. Secondly, cross-talk between the gut epithelium and bacteria 
has been demonstrated. The mechanisms underlying this interaction, and the role of the 
microbiota in the development and function of the gastrointestinal tract needs further 
investigation. A breakdown in immune tolerance to enteric microbiota has also been 
implicated in the pathogenesis of inflammatory disorders, such as inflammatory bowel 
disease. While evidence suggests that inflammatory bowel disease is characterized by an 
aggressive immune response to luminal antigens, including members of the commensal 

207 



208 Sheil et al. 

microbiota, the precise role of the luminal microbiota in the pathogenesis of disease has 
yet to be elucidated. Finally, there is evidence suggesting a role for probiotic bacteria in 
ameliorating inflammatory disease. This has led to the suggestion that probiotics may be 
an option in the therapy of inflammatory bowel disease, the rationale being that these 
bacteria without proinflammatory potential might alter the intestinal microbiota balance 
and modulate the immune response (5-8). 

Inflammatory bowel disease encompasses two major diseases, ulcerative colitis 
(UC) and Crohn's disease (CD). These two syndromes, while sharing similar features of 
gut mucosal inflammation, are distinct entities. Their pathogenesis remains incompletely 
understood. Both diseases are commonest in the Western, developed world, with highest 
incidence in northern climates (9,10). 

Genetic factors are known to play a role in the pathogenesis of inflammatory bowel 
disease. This is demonstrated by concordance in monozygous twin studies. Also, 10-25% 
of affected patients have a first-degree relative with the disease. However, the incomplete 
concordance seen in twin studies (concordance rates are 40-50% for CD and < 10% for 
ulcerative colitis) suggests that environmental factors also contribute to the pathogenesis 
of the disease. In addition, there has been a marked rise in the frequency of CD in the 
developed world in the past fifty years, with a prevalence of approximately 100 per 
100,000 population in North America and northern Europe. This rise in incidence in CD 
underscores the importance of environmental factors in its etiology. The increase in the 
incidence of CD has occurred as countries become more developed and industrialized. 
With changes in lifestyle and environment, improving levels of sanitation have altered the 
microbial environment. This means altered patterns of exposure to microbes and 
infections during childhood (11). Inflammatory bowel disease may be a disorder of 
mucosal immune responsiveness due to lack of stimulation and education of the immune 
responses (12). It is interesting that parallel to an increase in CD, other chronic 
inflammatory disorders, including allergies, asthma, multiple sclerosis and insulin- 
dependent diabetes mellitus have also increased in incidence. Environmental changes 
associated with industrialization may alter immune system development and pose a risk 
factor for inflammatory bowel disease in the genetically susceptible individual (12). 



THE ROLE OF THE ENTERIC MICROBIOTA IN THE NORMAL GUT 

Underpinning the probiotic concept is the importance of the normal intestinal microbiota 
in health and disease (12). Establishment of gut microbiota begins within minutes of 
delivery of the newborn (13,14). During delivery the infant is exposed to bacteria in the 
birth canal, the environment, maternal fecal microbiota, and other sources (15). The gut is 
initially colonized by facultative anaerobes such as Escherichia coli and Enterococcus 
species, possibly due to the absence of anaerobic conditions in the intestine (16). 
Colonization with bifidobacteria follows, particularly in breast-fed infants, and as the 
environment becomes more anaerobic, Bacteroides and Clostridia. 

The importance of the intestinal microbiota is suggested by the fact that the healthy 
adult gastrointestinal tract is home to a gut microbiota comprising over 400 different 
species with more bacterial cells in the gut than eucaryotic cells in the human body and with 
the average mass of bacteria being 1-2 kg. Commensal bacteria are present at a number of 
10" per gram of intestinal content in the small bowel, up to 1 per gram of ileal content in 
the distal ileum and up to 10 cells per gram of colonic content (17). 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 209 

The collective metabolic activity of the normal microbiota, of which little is known, 
is estimated to rival that of the liver (18-21). Up to 99% of the microbiota is comprised of 
30 to 40 strains, with the most abundant populations being strict anaerobes (22,23). 

Bacterial members of the genus Bacteroides are amongst the most prominent species 
found in human feces. Other species include bifidobacteria, Clostridia, streptococci, 
enterococci, lactobacilli, ruminococci, and eubacteria (4,22). Information regarding the 
microbiota has been restricted by the limitations of bacteriological culture methodology 
with only 40% of bacterial communities being cultivated on non-selective media in the 
laboratory (24). 



Effects of Enteric Microbiota in the Healthy Intestine 

Experiments with germ-free and re-colonized animals demonstrate beneficial effects of the 
resident microbiota (20). The commensal bacteria act as a defense against infection using 
several mechanisms, including competition for nutrients, the production of antimicrobial 
factors against pathogens, such as lactic acid and bacteriocins, and blockage or antagonism 
of adhesion sites. 

In addition, the integrity of the mucosa requires cell signaling between the 
microbiota, epithelium, and mucosal immune system (7). Without the microbiota, mucosal 
associated lymphoid tissue is underdeveloped and cell mediated immunity is defective. 
The enteric microbiota plays an important role in immune system education by fine-tuning 
T-cell repertoires and Thl/Th2 cytokine profiles (11). Compared with conventional 
animals, germ-free animals have reduced mucosal cell turnover, cytokine production, 
mucosal associated lymphoid tissue and lamina propria cellularity leading to an ineffective 
cell mediated immunity, decreased vascularity and less muscle wall thickness (25-27). 
There are also differences in intraepithelial lymphocytes (28,29). The intestinal microbiota 
primes the mucosal immune response and keeps it in a state of "controlled physiological 
inflammation" (26). Induction and/or maintenance of oral tolerance to ingested antigens 
also require microbial colonization of the gastrointestinal tract in early life. 

Understanding the influence of the gastrointestinal microbiota has prompted interest 
in the therapeutic modification of the enteric microbiota with probiotics or prebiotics. 



THE IMPORTANCE OF THE ENTERIC MICROBIOTA 
IN INFLAMMATORY BOWEL DISEASE 

Considerable evidence implicates the enteric microbiota in the pathogenesis of 
inflammatory bowel disease (Table 1) (7,8,30,31). Firstly, mucosal inflammation occurs 
in areas of the gut with highest bacterial numbers. Secondly, surgical diversion of the fecal 
stream has been associated with clinical improvement in the distal bowel, but relapse is 
predictable following surgical restoration. Thirdly, putative therapeutic efficacy is seen 
with the use of antibiotics in colonic disease. Fourthly, immune reactivity to intestinal 
bacteria is detectable in patients with inflammatory bowel disease suggesting a loss of 
immune tolerance to components of the microbiota (32,33). Fifthly, there are reports of 
increased numbers of bacteria within the mucosa of patients with inflammatory bowel 
disease compared with controls (34,35). The highest bacterial numbers have been seen in 
CD patients and numbers increase with severity of disease. Finally, the description of the 
first susceptibility gene for CD, CARD15/NOD2, has provided a basis for explaining the 
interaction between bacteria and the immune response. CARD15/NOD2 encodes a protein 



210 Sheiletal. 

Table 1 Evidence Implicating the Enteric Microbiota in the Pathogenesis of IBD 

The distribution of the lesions is greatest in areas of highest numbers of luminal bacteria 
Interruption of the fecal stream has been associated with clinical improvement but relapse is 

predictable following surgical restoration 
Evidence for loss of immunological tolerance to components of the commensal microbiota 
Serology and cellular immune reactivity to enteric microbiota that has formed the basis of putative 

diagnostic tests 
Efficacy of antibiotics in patients 

Description of first susceptibility gene for Crohn's disease (CARD15/NOD2) 
Colonization with normal enteric microbiota is required for expression of disease in animal models 

of colitis irrespective of the underlying defect 
Attenuation of inflammation in animal models of enterocolitis 
Efficacy of probiotics in animal models of colitis 
Effect of probiotics in human studies of IBD 

Abbreviation: IBD, inflammatory bowel disease. 



that is involved in the recognition of bacterial products and initiates the inflammatory 
cascade via activation of the transcription factor Nuclear Factor kappaB (NFkB) (36,37). 

Compelling evidence for the interactive role of genes, bacteria, and immunity has 
been derived from experimental animal models of both Crohn' s-like and colitis-like 
disease (38,39). There are now about 30 different spontaneously occurring or genetically 
engineered (knockout or transgenic) animal models for inflammatory bowel disease 
(40-42). Colonization with normal enteric microbiota is required for full expression of 
disease. Thus, the normal microbiota is a common factor driving the inflammatory process 
irrespective of the genetic underlying predisposition and immunological effector 
mechanism (43,44). Several different microorganisms have been demonstrated to induce 
colitis in animal models. These include Enterococcus faecalis, causing colitis in the anti- 
inflammatory interleukin-10 (IL-10) knockout mice, and Bacteroides vulgatus, which 
induced inflammation in the HLA-B27 rat model (45,46). This evidence has prompted the 
therapeutic modification of the enteric microbiota in inflammatory bowel disease. 

In patients with ulcerative colitis, the construction of an ileal pouch following 
a colectomy represents a human "model" showing the contribution of genes, bacteria, 
and immune mechanisms to its pathogenesis. A genetic contribution is consistent with 
the relative frequency of pouchitis in patients undergoing surgery for colitis compared 
with those having a pouch created surgically for familial polyposis coli. The contribution 
of bacteria to the pathogenesis of pouchitis is shown by the efficacy of both antibiotic 
and probiotic therapy in treating the disease (47). The immune system mediates the 
tissue damage and pouchitis appears to be a colitis-like process occurring in the 
colonized ileum. 



Specific Microorganisms in Inflammatory Bowel Disease 

Despite the importance of bacteria in the pathogenesis of colitis and CD, no specific micro- 
organism has been implicated in causing the intestinal inflammation. The roles of 
Mycobacterium paratuberculosis, measles virus, Listeria monocytogenes and adherent 
E. coli in the pathogenesis have been examined. Strains of adherent-invasive E. coli have 
been isolated in the mucosa of patients with CD (48). M. paratuberculosis has been 
cultured from the intestine of patients with CD and detected by molecular methods in the 
granulomas of resected tissue from patients (49). Possible disease modifying mechanisms 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 211 

of transient pathogens include the disruption of the mucosal barrier (allowing increased 
uptake of luminal antigens), mimicry of self-antigens and activation of the mucosal 
immune system via modulation of transcription factors such as NFkB. However, a direct 
cause and effect relationship has not been established for any of these organisms. Indeed, 
conditions favoring transmission of infection (low socio-economic status, overcrowding, 
poor sanitation) appear to protect against inflammatory bowel disease, arguing against an 
infectious aetiology (50). 

Since there is evidence for the role of luminal microbiota in the pathogenesis of 
inflammatory bowel disease, the alteration of the microbiota by the introduction of 
probiotic bacteria may result in clinical improvement of the condition. Conventional drug 
therapy for inflammatory bowel disease involves suppression of the immune system or 
modulation of the inflammatory response. Probiotics offer an alternative without the risk of 
side effects associated with conventional therapy. 



PROBIOTICS 

Probiotic Definition 

Probiotics may be defined as "Live microorganisms which when administered in adequate 
amounts confer a health benefit on the host" (4,51). Probiotics are non-pathogenic 
microbial organisms which survive passage through the gastrointestinal tract and are 
believed to have potential beneficial health effects. The desirable properties of probiotic 
bacteria include having generally regarded as safe status, acid, and bile stability, 
adherence to intestinal cells, persistence for some time in the gut, antagonism against 
pathogenic bacteria and modulation of the immune response (52). Bacteria of human 
origin were originally required for safety reasons and because probiotic efficacy appeared 
to be host-specific. This stipulation may now be unnecessary as potential probiotics are 
fully identified and characterized by phenotypic and genotypic methods and tested for 
safety before use. Probiotic activity has been associated most commonly with lactobacilli 
and bifidobacteria, but other non-pathogenic bacteria including species of streptococci and 
enterococci, non-pathogenic E. coli Nissle 1917, and the yeast Saccharomyces boulardii 
have been used (53). 

However, the current definition of a probiotic may now be too limited. Whilst the 
definition is one of live microorganisms, studies have demonstrated that bacterial DNA or 
bacterial components could themselves be responsible for any observed probiotic effects 
(54). Genetically modified bacteria have also been tested and a genetically engineered 
lactobacillus secreting the anti-inflammatory cytokine IL-10 has attenuated colitis in 
animals (55). Therefore, future use of the functional microbes may be outside the definition 
of probiotics. The definition of probiotics is likely to undergo continuing modification, 
and the term "pharmabiotics" may be more appropriate [(56), www.apc.ucc.ie]. This 
umbrella term includes live and dead organisms and constituents thereof, and 
encompasses genetically engineered microbes. 



How Probiotics May Exert an Effect in Inflammatory Bowel Disease 

The mechanisms of action of probiotic bacteria in the setting of inflammation are not 
completely elucidated and are likely to involve a number of factors and be strain specific. 
Proposed mechanisms focus on how probiotics influence the immune response. Commensal 
microbiota are known to contribute to immune homeostasis (7,26). There are several 



212 Sheiletal. 

molecular pathways which are suggested as candidates for the site of probiotic immune 
effects. In the context of IBD, anti-inflammatory activity may involve signaling with the 
gastrointestinal epithelium and perhaps mucosal regulatory T-cells (7). 

Gut Epithelium and Dendritic Cells 

Within the gut, intestinal epithelial cells are the first point of contact for bacteria and play 
an important role in bacteria-host communication (57). The epithelial cells act as sensors 
of commensal and pathogenic bacteria, with discriminatory capacity to activate signaling 
pathways (8,58,59). Interactions with Toll-like receptors and dendritic cells in the gut are 
believed to be involved in this communication between host and bacteria (8,60). Dendritic 
cells in the gut mucosa are responsible for the stimulation of T cells and seem to have an 
important role in the balance between inducing TH1, TH2, and TH3 cytokine profiles (61). 
Gut dendritic cells are mostly immature and potentially prone to modulation by the 
environment, containing microorganisms. TH1/TH2/TH3 cytokine profiles induced by gut 
dendritic cells have been modulated by the administration of lactobacilli (62). In a further 
study, the probiotic bacteria Bifidobacterium infantis and Lactobacillus salivarius have 
induced dendritic cells to produce the anti-inflammatory cytokine IL-10 rather than pro- 
inflammatory IL-12 (63). In addition, intestinal dendritic cells have been shown to retain 
small numbers of commensal bacteria. This allows induction of protective IgA by the 
dendritic cells, preventing mucosal penetration by bacteria (64). 

Modulation of the Cytokine Response 

The ability of probiotic bacteria to induce an anti-inflammatory or regulatory cytokine 
profile by in vitro immunocompetent cells has been confirmed (65). In vitro studies 
examined the effect of probiotics on cytokine production by human intestinal mucosa. 
Both Lactobacillus casei and Lactobacillus bulgaricus down-regulated the production of 
TNF-a from normal and inflamed mucosa (66,67). The effects of various lactic acid 
bacteria on the cytokine profile produced by peripheral blood mononuclear cells in vitro 
have been studied (57,68-71). Alterations in cytokine production have been observed in 
the IL-10 knockout mouse model which develops colitis similar to human inflammatory 
bowel disease. The anti-inflammatory effects of Lactobacillus salivarius UCC118, and 
Bifidobacterium infantis 35624, when administered both orally and subcutaneously to 
IL-10 knockout mice, were accompanied by a reduction in pro-inflammatory cytokines 
IFN-y, TNF-a and IL-12 from splenocytes, while levels of the regulatory cytokine TGF-P 
were maintained (72,73). 

It is suggested that live bacteria may not be necessary for the immune responses seen 
with probiotics. Indeed bacterial DNA has been shown to have potent immunostimulatory 
effects and has reduced colitis in a number of murine models (54). The DNA sequences 
used are termed immunostimulatory sequences or CpG motifs. CpG DNA can activate 
dendritic cells and its effects are mediated via Toll-like receptors (74,75). 

Nuclear Factor kappaB Pathway 

The NFkB pathway, a nuclear factor involved in the transcriptional regulation of 
inflammatory genes, mediates responses to invasive pathogenic bacteria. Certain non- 
pathogenic organisms have been shown to counterbalance epithelial responses to invasive 
bacteria via an effect on the inhibitor kappaB / NFkB pathway (76). A recent study has 
demonstrated that a commensal bacterium, Bacteroides thetaiotaomicron, also acted on 
NFkB to attenuate pro-inflammatory cytokine expression, but via a unique mechanism. 
The mechanism involved limiting the duration of action of NFkB by promoting its nuclear 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 213 

export through a peroxisome proliferator activated receptor-y-dependent (PPAR-y) 
pathway (77). 

Intestinal Permeability 

Apart from immune mechanisms, it is also suggested that probiotic bacteria may have 
a beneficial effect on permeability of the gut barrier. There is evidence to suggest that the 
epithelial barrier function is reduced in inflammatory bowel disease (78). 

Probiotic strains have demonstrated an ability to enhance the epithelial barrier 
function, based on measurements of intestinal permeability in excised mucosal tissue from 
animal models and humans (79,80). Probiotics given to IL-10 knockout mice normalized 
colonic physiological function and barrier integrity, along with a reduction in severity 
of colitis. 



EFFICACY OF PROBIOTICS IN INFLAMMATORY BOWEL DISEASE 

Probiotics in Animal Models of IBD 

The efficacy of probiotics in attenuating colitis has been demonstrated in experimental 
animal models (Table 2). These models include the interleukin-10 knockout murine model 
(81-84), methotrexate induced colitis (85), HLA-B27 transgenic rats (86), and the 
CD45Rbhi transfer model (87). 

The model of IL-10 knockout mice develop colitis when colonized with normal 
enteric microbiota but remain disease-free if kept in germ-free conditions. In a study of 
IL-10 - ' mice colonization with Lactobacillus plantarum 299 v was performed 2 weeks 
before transferring from a germ-free environment to a specific pathogen-free 
environment (84). This treatment led to a reduction in disease activity and a significant 
decrease in mesenteric lymph node IL-12 and IFN-y production. A role for Lactobacillus 
reuteri in prevention of colitis in IL-10 - mice was also demonstrated (81). In this 
study, the oral administration of the prebiotic lactulose (shown to increase the levels of 
Lactobacillus species) and rectal swabbing with L. reuteri restored Lactobacillus levels 
to normal in neonatal mice, originally found to have low levels of lactobacilli species. 
This effect was associated with the attenuation of colitis. In a placebo controlled trial, 
orally administered Lactobacillus salivarius UCC118 reduced the incidence of colon 
cancer and the severity of mucosal inflammation in IL-10 _/ " mice (82). L. salivarius 
was also shown to modify the gut microbiota in these animals as Clostridium 
perfringens, enterococci and coliform levels were significantly reduced in the probiotic 
group. A further trial confirmed the efficacy of L. salivarius UCC118 and demonstrated 
efficacy for Bifidobacterium infantis 35624 in attenuation of colitis in the IL-10 _/ " 
mouse model (83). The amelioration of disease activity in this study was associated with 
modulation of the gut microbiota as investigated by culture-independent 16S ribosomal 
RNA targeted PCR-direct gradient gel electrophoresis. In addition, mucosal pro- 
inflammatory cytokine production was significantly reduced. Indeed, the oral route of 
administration may not be essential for certain probiotic effects. Reduced inflammatory 
scores and reduced production of pro-inflammatory cytokines have been observed in 
IL-10 - mice which had been injected subcutaneously with L. salivarius UCC1 18 (73). 

Modified Probiotics in Animal Models 

Combinations of probiotic treatment with prebiotic s or antibiotics have been used to 
increase the beneficial effect. The combination of the prebiotic inulin, and the probiotic 



214 



Sheil et al. 



Table 2 Summary of Probiotic Efficacy in Animal Models of Enterocolitis 



Probiotic microorganism 



Type of study 



Trial outcome 



Reference 



Lactobacillus reuteri 



IL-10" /_ mice. N = 4-8 
per group. Placebo 
controlled trial 



Lactobacillus salivarius 
UCC118 



IL-10~ /_ mice. N= 10 
per group. Placebo 
controlled 



Lactobacillus salivarius 
UCC118 and Bifido- 
bacterium infantis 

35624 






IL-10"'" mice. N= 10 
per group. Placebo 
controlled 



Lactobacillus salivarius 
UCC118 






L-10~ /_ mice. CIA 
model N= 10 per 
group. Placebo 
controlled 



.-/- 



Lactobacillus plantar um IL-10 mice. Placebo 
299v controlled 



Lactobacillus rhamnosus HLA-B27 transgenic 

GG rats 

Combination of Lacto- HLA-B27 transgenic 

bacillus acidophilus rats 

La-5, L. delbruckii 

subsp. bulgaricus, 

Bifidobacterium 

Bb-12, and Strepto- 
coccus thermophilus 



Prebiotic lactulose and 
probiotic L. reuteri 
attenuated colitis 
and improved 
mucosal barrier 
function. 

Reduced incidence of 
colon cancer and 
mucosal inflam- 
mation. Modulation 
of fecal microbiota. 

Attenuation of disease. 
Modulation of gut 
microbiota. 
Reduction in in vitro 
production of IFN- 
y,TNF-aandIL-12. 
TGF-(3 levels main- 
tained. 

Attenuation of colitis 
and arthritis 
following subcu- 
taneous adminis- 
tration of probiotic. 
Reduction in proin- 
flammatory cyto- 
kine production. 

Attenuation of colitis. 
Reduction in IL-12 
and IFN-y produced 
by stimulated 
mesenteric lymph 
node cells. 

Prevented recurrence 
of colitis. 

Attenuated colitis 
following treatment 
with the prebiotic 
inulin and a combi- 
nation of probiotic 
organisms. 



Madsen et al. 
1999 (81) 



O'Mahonyetal. 
2001(82) 



McCarthy et al. 
2003 (83) 



Sheil et al. (73) 



Schultz et al. 
2002 (84) 



Dieleman et al. 

2001 (86) 
Schultz et al. 

unpublished 

data 



Abbreviations: HLA, human leukocyte antigen; IFN, interferon; IL, interleukin; N, number of animals; TGF, 
transforming growth factor; TNF, tumor necrosis factor. 



organisms Lactobacillus acidophilus La-5, Lactobacillus delbrueckii subsp. bulgaricus, 
Bifidobacterium lactis Bb-12, and Streptococcus thermophilus significantly decreased 
inflammation in HLA-B27 rats (Schultz, unpublished data). Furthermore, genetically 
modified probiotics have been developed. Lactococcus lactis was engineered to secrete 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 215 

biologically active IL-10 and a significant reduction in inflammation was observed in both 
IL- 10 ~ ' ' and dextran sodium sulfate-induced murine colitis models (55). The investigators 
concluded that genetically engineered bacteria for local administration of a therapeutic 
agent, such as IL-10, may be a useful strategy in the treatment and prevention of IBD. 

Live versus Dead Bacteria 

It may not be necessary to administer live bacteria to achieve benefit. Bacterial DNA has 
been shown to have potent immuno-stimulatory effects. In a trial by Rachmilewitz et al. (54) 
bacterial DNA was used to attenuate colitis in a number of murine models suggesting an 
anti-inflammatory effect for bacterial DNA that warrants further study. A more recent study 
investigated the role of Toll-like receptors in mediating these effects of bacterial DNA (88). 

Human Trials of Probiotics in Patients with Inflammatory Bowel Disease 

Evidence that the enteric microbiota play a role in the pathogenesis of IBD and results 
from models of IBD which have demonstrated beneficial effects for probiotics has 
prompted clinical studies examining the effect of these organisms in patients with 
inflammatory bowel disease. 

Trials in Ulcerative Colitis 

A number of studies have examined the use of a non-pathogenic E. coli strain Nissle 
1917, in the setting of ulcerative colitis. Kruis et al. (89) first performed in 1997 a 
randomized, double-blind clinical trial where 120 patients with inactive ulcerative colitis 
were randomized to receive oral E. coli strain Nissle 1917 or mesalazine. They reported 
that there was no difference in relapse rates in the probiotic treated group compared to 
patients on mesalazine. Relapse rates were 11.3% for the mesalazine treated group and 
16.0% for the E. coli group. Life table analysis showed a relapse free time of 103 + 
4 days for mesalazine and 106 + 5 days for E. coli. From the results of this preliminary 
study, probiotic treatment appeared to offer another option for maintenance therapy of 
ulcerative colitis (89). Further beneficial results were described by Rembacken et al. (90) 
in a study where a total of 116 patients with active ulcerative colitis were recruited. 
Seventy-five percent and 68% of the mesalamine and E. coli groups achieved remission, 
respectively. In the second maintenance part of this study, the relapse rate in both groups 
was markedly higher than the investigators anticipated, 73% for the mesalamine group 
and 67% for the E. coli group. The time to relapse was not significantly different 
between the groups (90). These results suggested that the non-pathogenic E. coli was 
equivalent to mesalazine in maintaining remission, however these relapse rates are 
similar to those of placebo-treated patients. In a larger, 1-year multi-center, randomized, 
double-blind, remission maintenance study of 327 patients, E. coli was shown to be as 
effective as mesalazine in maintaining remission with relapse rates of 45% for the E. coli 
group and 36% in the mesalazine group, therefore offering an alternative to mesalazine 
in maintenance of remission in ulcerative colitis patients (Table 3) (92). 

The probiotic cocktail VSL#3, a mixture of four lactobacilli (Lactobacillus 
plantarum, Lactobacillus casei, Lactobacillus acidophilus, Lactobacillus delbrueckii ssp. 
Bulgaricus), three bifidobacteria strains (Bifidobacterium infantis, Bifidobacterium breve, 
Bifidobacterium longum), and one strain of Streptococcus salivarius ssp. thermophilus, 
has been studied in ulcerative colitis. There is a high concentration of bacteria in this 
mixture with potential synergistic relationships to enhance suppression of potential 
pathogens. The effect of VSL#3 on maintenance of remission in UC patients was 



216 



Sheil et al. 



Table 3 Summary of Human Trials of Probiotic Therapy in Ulcerative Colitis 



Study type 


Organism used 


Trial outcome 


Reference 


Randomized 


E. coli strain Nissle 


Patients with active colitis 


Kruis et al. 


controlled 


1917. N= 120 


demonstrated similar relapse rates 


1997 (89) 


trial 




compared to patients on mesalazine 




Randomized, 


E. coli strain Nissle 


Confirmed result from Kruis 


Rembacken et al. 


controlled 


1917. N=116 


et al. 1997 


1999 (90) 


trial 








Open labeled 


VSL#3. N = 20 


Maintenance of remission in patients 


Venturi et al. 


trial 






1999 (91) 


Randomized 


E. coli strain Nissle 


Remission maintained in patients 


Kruis et al. 


controlled 


1917. N = 327 


receiving probiotic 


2001 (92) 


trial 








Open labeled 


Sac char omyces 


Treatment given in combination with 


Guslandi et al. 


trial 


boulardii. 


mesalamine for relapse of ulcerative 


2003 (93) 




N = 25 


colitis. Remission achieved in 17 
patients 





Abbreviation'. N, number of subjects in trial. 

evaluated using an open label design (91). In this pilot study, 20 patients in remission 
were treated for 12 months. At the end of the trial 15 out of 20 patients (75%) remained 
in remission. 

A recent study has investigated the use of Saccharomyces boulardii in the setting of 
ulcerative colitis. In an open, non-placebo controlled study, 25 patients with a relapse of 
ulcerative colitis were treated with mesalazine in combination with S. boulardii. 
Seventeen patients achieved remission (93). 



Trials in Pouchitis 

Convincing evidence for beneficial probiotic effects in inflammatory bowel disease is 
seen in the treatment of pouchitis. In an open labeled study, patients with pouchitis were 
treated with Lactobacillus GG and fructooligosaccharide (94). The patients reported 
a beneficial effect when the probiotic-prebiotic mix was administered as an adjuvant to 
antibiotic therapy. Remission was documented by suppression of symptom scores and 
reversal of endoscopic findings (94). Gionchetti et al. (95) have studied VSL#3 in the 
setting of pouchitis and have demonstrated the efficacy of this probiotic mix in 
maintenance of remission in patients with chronic pouchitis. In a randomized, double- 
blind, placebo-controlled trial, 40 patients with pouchitis received one month of 
antibiotic treatment and were in clinical and endoscopic remission. Patients were then 
randomized to receive VSL#3 or placebo for 9 months. At the end of the study three 
patients (15%) had relapsed in the VSL#3 group compared to 20 (100%) in the placebo 
group. In a follow-up study, this group has also used VSL#3 as prophylaxis in patients 
after ileo-anal pouch formation surgery to prevent pouchitis. Forty patients were 
randomized to receive VSL#3 or placebo. At 1-year follow-up, 10% of probiotic treated 
patients had developed pouchitis, compared with 40% of the placebo treated group (96). 
A recent study has again examined the role of VSL#3 in maintaining remission 
following treatment of refractory or recurrent pouchitis. Thirty-six patients with 
recurrent pouchitis (at least twice in the past year) or requiring continuous antibiotics, in 
whom remission was induced by 4 weeks of antibiotics, were randomized to receive 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 



217 



Table 4 Summary of Human Trials of Probiotic Therapy in Pouchitis 



Study type 


Organism used 


Trial outcome 


Reference 


Open labeled 


Prebiotic fructooli- 


Effective in inducing remission in 


Friedman et al. 


trial 


gosaccharide and 

probiotic. 

N = 10 


combination with antibiotic 


2000 (94) 


Randomized 


VSL#3. N = 40 


Maintenance of remission in chronic 


Gionchetti et al. 


controlled 




pouchitis after antibiotic induced 


2000 (95) 


trial 




remission. 15% relapse rate 
compared with 100% in control 
group 




Randomized 


VSL#3. N = 40 


Prevention of acute pouchitis in 


Gionchetti et al. 


controlled 




patients after ileo-anal pouch sur- 


2003 (96) 


trial 




gery. 10% pouchitis rate in probiotic 
group compared with 40% in control 
group 




Randomized 


VSL#3 (6 g). 


Maintenance of remission in recurrent 


Mimura et al. 


controlled 


N = 36 


or refractory pouchitis after anti- 


2004 (97) 


trial 




biotic induced remission. 85% 
remained in remission at one year, 
compared with 6% in placebo group 





Abbreviation: N, number of subjects in trial. 



6 gram of VSL#3 or placebo daily for one year or until relapse. Eighty-five percent 
of the VSL#3 treated group remained in remission at one year compared with 6% 
(one patient) in the placebo group (Table 4) (97). 



Trials in Crohn 's Disease 

In CD, an early study involved the use of Sac c char omyces boulardii (98). In a double- 
blind study, 20 patients with moderately active CD were randomized to treatment with this 
organism or placebo for 7 weeks. The probiotic treated patients had a significant decrease 
in CD activity index (CDAI) compared with the control group. More recently, a double- 
blind trial randomized 32 CD patients in clinical remission to receive either mesalamine 
alone or mesalamine plus S. boulardii. Clinical relapse was observed in only 6.25% of 
patients receiving mesalamine plus S. boulardii, while 37.5% relapse rate was observed in 
the group receiving mesalamine alone (Table 5) (103). 

The efficacy of Lactobacillus rhamnosus GG in the treatment of CD has been 
studied (99). Malin et al. (99) reported that in pediatric CD, consumption of Lactobacillus 
GG was associated with increased gut IgA levels which could promote the gut 
immunological barrier. Gupta et al. (101) also reported improved clinical scores and 
improved intestinal permeability in an open labeled pilot study in a small study involving 
four pediatric CD patients. 

A double-blind study investigated the use of the E. coli Nissle 1917 strain in CD (100). 
Malchow et al. randomized 28 patients in remission to receive either E. coli or placebo. At 
1-year follow-up, the relapse rates were significantly reduced in the group that received 
E. coli (30%) compared with 70% in the placebo group. In a large double-blind, randomized 
study the efficacy of VSL#3 combined with antibiotic treatment on the post-operative 
recurrence of CD was compared to treatment with mesalamine alone (102). Forty patients 



218 



Sheil et al. 



Table 5 Summary of Human Trials of Probiotic Therapy in Crohn's Disease 



Study type Organism used 



Trial outcome 



Reference 



Randomized 


Saccharomy- 


controlled 


ces boular- 


trial 


dii. N = 20 


Open labeled 


Lactobacillus 


trial 


rhamnosus 




GG. N=14 


Randomized 


E. coli strain 


controlled 


Nissle 1917 


trial 


N = 28 


Open labeled 


Lactobacillus 


trial 


rhamnosus 




GG in chil- 




dren. N = 4 


Randomized 


VSL#3 with 


controlled 


antibiotic. 


trial 


N = 40 


Randomized 


Saccharomy- 


controlled 


ces boular- 


trial 


dii. N = 32 


Open labeled 


Lactobacillus 


trial 


salivarius 




118. N = 25 


Randomized 


Lactobacillus 


controlled 


rhamnosus 


trial 


GG 



Decrease in CDAI in probiotic group 



Increase in gut IgA response 



Remission achieved in patients on probiotics 
and steroids greater than with steroids alone 

Improved intestinal permeability and CDAI 



Patients with CD had 20% remission when 
given antibiotic and VSL#3 compared to 
40% in mesalamine treated group 

Maintenance of remission in treatment group 
superior as relapse observed in 6.25% of 
patients receiving probiotic plus mesalasine 
compared to 37.5% on mesalamine alone 

Reduction of mean CDAI and induction of 
IgA in patients with relapse 

No difference seen in rate of recurrence 1 year 
after surgery between group given probiotic 
or control 



Plein et al. 
1993 (98) 

Malin et al. 

1996 (99) 

Malchow et al. 

1997 (100) 

Gupta et al. 
2000 (101) 



Campieri et al. 
2000 (102) 

Guslandi et al. 
2000 (103) 



McCarthy et al. 

2001 (104) 

Prantrera et al. 

2002 (105) 



Abbreviations: N, number of subjects in trial; CD, Crohn's disease; CDAI, Crohn's disease activity index. 

were randomized to receive rifaximin for 3 months followed by VSL#3 for 9 months or 
mesalamine for 12 months. At the end of the trial 20% of the patients had recurrent CD in the 
probiotic/antibiotic group while 40% of patients in the mesalamine group relapsed (102). In 
an open study of patients with mildly active CD despite 5-ASA therapy, patients were 
offered either steroids or a trial of Lactobacillus salivarius subsp. salivarius UCC118 for 
6 weeks (104). Of the 25 patients enrolled, 19 successfully completed the study and avoided 
steroids for a 3-month follow-up period. The mean CDAI at enrolment was 217, falling to 
150 at the end of the study period (104). Finally, in a recent study of 45 CD patients who 
underwent curative surgery, the recurrence rate 1 year after surgery in patients treated with 
Lactobacillus rhamnosus GG or placebo was compared. No difference was seen between 
the patients receiving probiotic (16% recurrence rate) and the placebo group (10%) (105). 
In conclusion, while the trials for probiotics in treatment of IBD to date are 
promising, results have been mixed; consequently, better-designed trials are needed. 



DISCUSSION 



Although preliminary studies are promising, large placebo-controlled, randomized, 
double-blinded clinical trials are needed to clarify the role of probiotic bacteria in the 
treatment of inflammatory bowel disease. Studies of probiotics in inflammatory bowel 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 219 

disease in the future will also need to increase our knowledge of how probiotics exert their 
effect. Optimal dosing schedules need to be determined. Detailed comparisons of probiotic 
performance amongst different bacterial strains have not yet been performed, in vivo or 
under clinical trial conditions, and the level of scientific characterization of individual 
organisms has been variable. The route of administration also requires more study, in 
particular to determine whether the oral route is always essential. The issue of live versus 
dead bacteria remains unclear. The beneficial effect of bacterial DNA and other 
metabolites or constituents versus whole organisms needs comprehensive study. 

Irrespective of the mechanism of action, however, there are reasons which might 
favor therapeutic usage of live over dead bacteria. Live bacteria may be more reliable for 
enteric transit and occupation of microbial niche. Secondly, live bacteria offer the 
advantage of elaborating biological molecules other than immunomodulatory DNA. 

Detailed strain characterization is also required for all potential probiotic strains 
before the use of combinations can be recommended. The potential exists for synergistic 
or antagonistic effects amongst bacterial strains and this requires further study. Finally, 
disease-specific probiotic organisms designed to target particular patients, (the "designer 
probiotic"), may become a possibility as we increase our understanding of molecular 
mechanisms behind the anti-inflammatory effects of individual probiotics. What is already 
clear, is that there will be an increasing role for bacteria or bacterial products in a 
therapeutic setting along with conventional treatments for inflammatory bowel disease. 
The concept of a food influencing the health of the gastrointestinal tract is appealing to 
many people. Therapeutic modification of the microbiota with functional foods such as 
probiotics empowers patients with an enhanced sense of control in the management of 
their illness. Microbial therapeutics is an expanding field inviting further investigation, 
and we should not allow ourselves to become captive of the definition of probiotics. 



REFERENCES 

1. Milner JA. Functional foods and health promotion. J Nutr 1999; 129:1395S-1397S. 

2. McCarthy J, Shanahan F. Functional food and probiotics: time for gastroenterologists to 
embrace the concept. Curr Gastroenterol Rep 2000; 2:345-346. 

3. Metchnikoff E. Prolongation of life. London: William Heinmann, 1907. 

4. Fooks LJ, Fuller R, Gibson GR. Prebiotics, probiotics and human gut microbiology. Int Dairy 
J 1999; 9:53-61. 

5. Dunne C. Adaption of bacteria to the intestinal niche: probiotics and gut disorders. Inflamm 
Bowel Dis 2001; 7:136-145. 

6. Schultz M, Sartor RB. Probiotics and inflammatory bowel disease. Am J Gastroenterol 2000; 
95:519-521. 

7. Shanahan F. Probiotics and inflammatory bowel disease: is there a scientific rationale. 
Inflamm Bowel Dis 2000; 6:107-115. 

8. Shanahan F. Therapeutic manipulation of the gut flora. Science 2000; 289:1311-1312. 

9. Shanahan F. Crohn's disease. Lancet 2002; 359:62-69. 

10. Ekbom A. The changing faces of Crohn's disease and ulcerative colitis. In: Targan S, 
Shanahan F, Karp L, eds. Inflammatory Bowel Disease. From Bench to Bedside. 2nd ed. 
Dordrecht: Kluwer Academic Publishers, 2003:5-20. 

11. Rook GAW, Stanford JL. Give us this day our daily germs. Immunol Today 1998; 
19:113-116. 

12. Shanahan F. The host- microbe interface within the gut. Best Pract Res Clin Gastroenterol 
2002; 19:915-931. 

13. Fuller R. Probiotics in man and animals. J Appl Bacteriol 1989; 66:365-378. 



220 Sheil et al. 

14. Collins JK, Thornton G, Sullivan G. Selection of probiotic strains for human applications. 
Int Dairy J 1998; 8:487-490. 

15. Mackie R, Gaskins HR. Gastrointestinal microbial ecology. Sci. Med. 1999; 6:18-27. 

16. Vanderhoof JA, Young RJ. Use of probiotics in childhood gastrointestinal disorders. 
J Paediatr Gastroenterol Nutr 1998; 27:323-332. 

17. Berg RD. The indigenous gastrointestinal microbiota. Trends Microbiol 1996; 434:430-435. 

18. Bengmark S. Ecological control of the gastrointestinal tract. The role of the probiotic flora. 
Gut 1998; 42:2-7. 

19. Mackowiak PA. The normal microbial flora. N Engl J Med 1982; 307:83-93. 

20. Midtvedt T. Microbial functional activities. In: Hanson LA, Yolken RH, eds. Intestinal 
Microflora Nestle Nutrition Workshop Series, Vol. 42. Philadelphia: Lippincott-Raven, 
1999:79-96. 

21. Bocci V. The neglected organ: bacterial flora has a crucial immunostimulatory role. Perspect 
Biol Med 1992; 35:251-260. 

22. Alderberth I, Breslin L, Bjoerkstein B, et al. Digestive flora. In: Gorthier G, Arnaud C, eds. 
Mechanisms of Protection of the Digestive Tract. Paris: John Libby Eurotext, 1998:17-20. 

23. Tannock GW. Probiotic properties of lactic acid bacteria: plenty of scope for fundamental 
R&D. Trends Biotechnol 1997; 15:270-274. 

24. Tannock GW, Munro K, Harmsen HJM, et al. Analysis of the fecal flora of human subjects 
consuming a probiotic containing Lactobacillus rhamnosus DR20. Appl Environ Microbiol 
2000; 66:2578-2588. 

25. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science 2001; 
292:1115-1118. 

26. MacDonald TT, Carter PB. Requirement for a bacterial flora before mice generate cells 
capable of mediating the delayed hypersensitivity reaction in sheep red blood cells. J Immunol 
1979; 122:2426-2429. 

27. Gordon HA, Pesti L. The gnotobiotic animal as a tool in the study of host-microbial 
relationships. Bacterial Rev 1971; 35:390-429. 

28. Kawaguchi M, Nanno M, Umesaki Y, et al. Cytolytic activity of intestinal intraepithelial 
lymphocytes in germ-free mice is strain dependent and determined by T-cells expressing 
T-cell receptors. Proc Natl Acad Sci 1993; 90:8591-8594. 

29. Umesaki Y, Setomaya S, Matsumoto S, et al. Expansion of the alpha beta T-cell receptor- 
baring intestinal intraepithelial lymphocytes after microbial colonisation in germ-free mice, 
and its independence from the thymus. Immunology 1993; 79:32-37. 

30. Guarner F, Casellas F, Borruel N, et al. Role of microecology in chronic inflammatory bowel 
diseases. Eur J Clin Nutr 2002; 56:S34-S38. 

31. Campierei M, Gionchetti P. Probiotics in inflammatory bowel disease: new insight to 
pathogenesis or possible therapeutic alternative. Gastroenterology 1998; 116:1246-1260. 

32. Macpherson A, Khoo UY, Forgacs I, et al. Mucosal antibodies in inflammatory bowel disease 
are directed against intestinal bacteria. Gut 1996; 38:365-375. 

33. Duchmann R, Kaiser I, Hermann E, et al. Tolerance exists towards resident intestinal flora but 
is broken in active inflammatory bowel disease (IBD). Clin Exp Immunol 1995; 102:448-455. 

34. Schultsz C, Van Den Berg F, Ten Kate FW, et al. The intestinal mucus layer from patients 
with inflammatory bowel disease harbors high numbers of bacteria compared with controls. 
Gastroenterology 1999; 117:1089-1097. 

35. Swindinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora in inflammatory bowel disease. 
Gastroenterology 2002; 122:44-54. 

36. Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2 leucine rich repeat variants 
with susceptibility to Crohn's disease. Nature 2001; 411:599-603. 

37. Hugot JP, Zouali H, Leasage S. Lessons to be learned from the NOD2 gene in Crohn's 
disease. Eur J Gastroenterol Hepatol 2003; 15:593-597. 

38. Elson CO, Sartor RB, Tennyson GS, et al. Experimental models of inflammatory bowel 
disease. Gastroenterology 1995; 109:1344-1367. 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 221 

39. Strober W, Fuss IJ, Ehrhardt RO, et al. Mucosal immunoregulation and inflammatory bowel 
disease: new insights from murine models of inflammation. Scand J Immunol 1998; 
48:453-458. 

40. Blumberg RS, Saubermann LJ, Strober W, et al. Animal models of mucosal inflammation and 
their relation to human inflammatory bowel disease. Curr Opin Immunol 1999; 11:648-656. 

41. Wirtz S, Neyrath MF. Animal models of intestinal inflammation: new insights into the 
molecular pathogenesis and immunotherapy of inflammatory bowel disease. Int J Colorectal 
Dis2000; 15:144-160. 

42. Kosiewicz MM, Nast CC, Krishnan A, et al. Thl-type responses mediate spontaneous ileitis in 
a novel murine model of Crohn's disease. J Clin Invest 2001; 107:695-702. 

43. Kuhn R, Lohler J, Rennick D, et al. Interleukin-10-deficient mice develop chronic 
enterocolitis. Cell 1993; 75:263-274. 

44. Shanahan F. Gene-targeted immunologic knockouts: new models of inflammatory bowel 
disease. Gastroenterology 1994; 17:312-314. 

45. Balish E, Warner T. Enterococcus faecalis induces inflammatory bowel disease in 
interleukin-10 knockout mice. Am J Pathol 2002; 160:2253-2257. 

46. Rath HC, Wilson KH, Sartor RB. Differential induction of colitis and gastritis in HLA-B27 
transgenic rats selectively colonized with Bacteroides vulgatus or Escherichia coli. Infect 
Immunol 1999; 67:2969-2974. 

47. Sartor RB. Probiotics in chronic pouchitis: restoring the luminal balance. Gastroenterology 
2000; 119:305-309. 

48. Darfeuille-Michaud A, Neut C, Barnich N, et al. Presence of adherent Escherichia coli strains 
in ileal mucosa of patients with Crohn's disease. Gastroenterology 1998; 115:1405-1413. 

49. Ryan P, Bennett MW, Aarons S, et al. PCR detection of Mycobacterium paratuberculosis in 
Crohn's disease granulomas isolated by laser capture microdissection. Gut 2002; 51:665-670. 

50. Gent AE, Hellier MD, Grace RH, et al. Inflammatory bowel disease and domestic hygiene in 
infancy. Lancet 1994; 343:766-767. 

51. Guarner F, Schaafsma GJ. Probiotics. Int J Food Microbiol 1998; 39:237-238. 

52. Dunne C, O'Mahony L, Murphy L. In vitro selection criteria for probiotic bacteria of human 
origin: correlation with in vivo findings. Am J Clin Nutr 2001; 73:386S-392S. 

53. Shanahan F. Inflammatory bowel disease: immunodiagnostics, immunotherapeutics, and 
ecotherapies. Gastroenterology 2001; 120:622-635. 

54. Rachmilewitz D, Karmeli F, Takabayashi K, et al. Immunostimulatory DNA ameliorates 
experimental and spontaneous murine colitis. Gastroenterology 2002; 122:1428-1441. 

55. Steidler L, Hans W, Schotte L, et al. Treatment of murine colitis by Lactococcus lactis 
secreting interleukin-10. Science 2000; 289:1352-1355. 

56. Shanahan F. Host-flora interactions in inflammatory bowel disease. Inflamm Bowel Dis 2004; 
10:S16-S24. 

57. Haller D, Bode C, Hammes WP, et al. Non-pathogenic bacteria elicit a differential cytokine 
response by intestinal epithelial cell / leukocyte co-cultures. Gut 2000; 47:79-87. 

58. Gordon JI, Hooper LV, McNevin MS, Wong M, Bry L. Epithelial cell growth and 
differentiation, III. Promoting diversity in the intestine: conversations between the microbiota, 
epithelium and diffuse GALT. Am J Physiol 1997; 273:G565-G570. 

59. Hooper LV, Wong MH, Thelin A, et al. Molecular analysis of commensal host/microbial 
relationships in the intestine. Science 2001; 291:881-884. 

60. Marteau P, Seksik P, Jian R. Probiotics and health: new facts and ideas. Curr Opin Biotechnol 
2002; 13:486-489. 

61. Kronin V, Hochrein H, Shortman K, et al. Regulation of T cell cytokine production by 
dendritic cells. Immun Cell Biol 2000; 78:214-223. 

62. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of 
cytokines and maturation surface markers in murine dendritic cells. J Immunol. 2002; 
168:171-178. 

63. O'Mahony L, Zong Y, Sharma S, et al. Probiotic bacteria and pathogenic bacteria elicit diff- 
erential cytokine responses from dendritic cells (abstract). Gastroenterology 2001; 120:1625. 



222 Sheil et al. 

64. Macpherson AJ, Uhr T. Induction of protective IgA by intestinal dendritic cells carrying 
commensal bacteria. Science 2004; 303:1624-1625. 

65. O'Mahony L, Feeney M, MacSharry J, et al. Probiotics, mononuclear cells and epithelial 
cells: an anti-inflammatory network (abstract). Gastroenterology 2000; 118:680. 

66. Borruel N, Carol M, Casellas F, et al. Increased mucosal TNF-a production in Crohn's disease 
can be downregulated ex vivo by probiotic bacteria. Gut 2002; 51:659-664. 

67. Borruel N, Casellas F, Antolin M, et al. Effects of non-pathogenic bacteria on cytokine 
secretion by human intestinal mucosa. Am J Gastroenterol 2003; 98:865-870. 

68. Miettinen M, Matikainen S, Vuopio-Varkila J, et al. Lactobacilli and streptococci induce 
interleukin-12 (IL-12), IL-18, and gamma inteferon production in human peripheral blood 
mononuclear cells. Infect Immun 1998; 66:6058-6062. 

69. Miettinen M, Lehtonen A, Julkunen I, et al. Lactobacilli and streptococci activate NF-kappaB 
and STAT signalling pathways in human macrophages. J Immunol 2000; 164:3733-3740. 

70. Haller D, Serrant P, Granato D, et al. Activation of human NK cells by staphylococci 
and lactobacilli requires cell contact-dependent costimulation by autologous monocytes. 
Clin Diagn Lab Immunol 2002; 9:649-657. 

71. Haller D, Blum S, Bode C, et al. Activation of human peripheral blood mononuclear cells by 
non-pathogenic bacteria in vitro: evidence of NK cells as primary targets. Infect Immun 2000; 
68:752-759. 

72. McCarthy J, O'Mahony L, O'Callaghan L, et al. Double-blind, placebo-controlled trial of two 
probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. 
Gut 2003; 52:975-980. 

73. Sheil B, McCarthy J, O'Mahony L, et al. Is the mucosal route of administration essential for 
probiotic function? Subcutaneous administration is associated with attenuation of murine 
colitis and arthritis Gut 2004; 53:694-700. 

74. Hemmi H, Takeuchi O, Kavai T, et al. A Toll-like receptor recognizes bacterial DNA. Nature 
2000; 408:740-745. 

75. Wagner H. Toll meets bacterial CpG-DNA. Immunity 2001; 14:499-502. 

76. Neish AS, Gerwirtz AT, Zeng H, et al. Prokaryotic regulation of epithelial responses by 
inhibition of IkappaB -alpha ubiquitination. Science 2000; 289:1560-1563. 

77. Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut bacteria attenuate 
inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-y and RelA. Nature 
Immunol 2004; 5:104-112. 

78. Schmitz H, Barmeyer C, Fromm M, et al. Altered tight junction structure contributes to the 
impaired epithelial barrier function in ulcerative colitis. Gastroenterology 1999; 
116:301-309. 

79. Isolauri E, Majamaa H, Arvola T, et al. Lactobacillus casei strain GG reverses increased 
intestinal permeability induced by cow milk in suckling rats. Gastroenterology 1993; 
105:1643-1650. 

80. Madsen K, Cornish A, Soper P, et al. Probiotic bacteria enhance murine and human intestinal 
epithelial barrier function. Gastroenterology 2001; 121:580-591. 

81. Madsen KL, Doyle JS, Jewell LD, et al. Lactobacillus sp prevents development of 
enterocolitis in interleukin- 10 deficient mice. Gastroenterology 1999; 116:1107-1114. 

82. O'Mahony L, Feeney M, O'Halloran S, et al. Probiotic impact on microbial flora, 
inflammation, and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther 
2001; 15:1219-1225. 

83. McCarthy J, O'Mahony L, O'Callaghan L, et al. Double-blind, placebo-controlled trial of two 
probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. 
Gut 2003; 52:975-980. 

84. Shultz M, Veltkamp C, Dieleman LA, et al. Lactobacillus plantarum 299 v in the treatment 
and prevention of spontaneous colitis in Interleukin- 10-deficient mice. Inflamm Bowel Dis 
2002; 8:71-80. 

85. Mao Y, Nobaek S, Kasravi B, et al. The effects of Lactobacillus strains and oat fibre on 
methotrexate induced enterocolitis in rats. Gastroenterology 1996; 111:334-344. 



Probiotics: A Role in Therapy for Inflammatory Bowel Disease 223 

86. Dieleman LA, Goerres MS, Arends A, et al. Lactobacillus GG prevents recurrence of colitis in 
HLA-B27 transgenic rats after antibiotic treatment. Gastroenterology 2001; 118:A4312. 

87. Murphy L, Byrne F, Collins JK, Shanahan F, O' Sullivan GC, Aranda R. Evaluation and 
characterisation of probiotic therapy in the CD45Rbhi transfer model. Gastroenterology 1999; 
116:A3882 (abstract). 

88. Rachmilewitz D, Katakura K, Karmeli F, et al. Toll like receptor 9 signaling mediates the anti- 
inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 2004; 
126:520-528. 

89. Kruis W, Schutz E, Fric P, Fixa B, Judmaier G, Stolte M. Double blind comparison of an oral 
Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. 
Aliment Pharmacol Ther 1997; 11:853-858. 

90. Rembacken BJ, Snelling AM, Hawkey PM, Axon ATR. Non-pathogenic Escherichia coli 
versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet 1999; 
354:635-639. 

91. Venturi A, Gionchetti P, Rizzello F, et al. Impact on the composition of the faecal flora by 
a new probiotic preparation: preliminary data on maintenance treatment of patients with 
ulcerative colitis. Aliment Pharmacol Ther 1999; 13:1103-1108. 

92. Kruis W, Fric P, Stolte M. The Mutflor Study Group. Maintenance of remission in ulcerative 
colitis is equally effective with Escherichia coli Nissle 1917 and with standard mesalazine. 
Gastroenterology 2001; 120:A680 (abstract). 

93. Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative 
colitis. Eur J Gastroenterol Hepatol 2003; 15:697-698. 

94. Friedman G, George J. Treatment of refractory 'pouchitis' with probiotic and prebiotic 
therapy. Gastroenterology 2000; 118:A4167. 

95. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as maintenance treatment in 
patients with chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology 
2000; 119:305-309. 

96. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis onset with probiotic 
therapy: A double-blind, placebo- controlled trial. Gastroenterology 2003; 124:1202-1209. 

97. Mimura T, Rizzello F, Helwig U, et al. Once daily high dose probiotic therapy (VSL#3) for 
maintaining remission in recurrent or refractory pouchitis. Gut 2004; 53:108-114. 

98. Plein K, Holz J. Therapeutic effects of Saccharomyces boulardii on mild residual symptoms 
in a stable phase of Crohn's disease with special respect to chronic diarrhoea-a pilot study. 
Z Gastroenterol 1993; 31:129-134. 

99. Malin M, Suomalainen H, Saxelin M, et al. Promotion of IgA immune response in patients 
with Crohn's disease by oral bacteriotherapy with Lactobacillus GG. Ann Nutr Metab 1996; 
40:137-145. 

100. Malchow HA. Crohn's disease and Escherichia coli. A new approach in therapy to maintain 
remission of colonic Crohn's disease? J Clin Gastroenterol 1997; 25:653-658. 

101 . Gupta P, Andrew H, Kirschner BS, et al. Is Lactobacillus GG helpful in children with Crohn's 
disease? Results of a preliminary open-label study J Pediatr Gastroenterol Nutr 2000; 
31:453-457. 

102. Campieri M, Rizzello F, Venturi A, et al. Combination of antibiotic probiotic treatment is 
efficacious in prophylaxis of post-operative recurrence of Crohn's disease: a randomised 
controlled study vs mesalamine. Gastroenterology 2000; 118:A4179 (abstract). 

103. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in the maintenance of 
Crohn's disease. Dig Dis Sci 2000; 45:1462-1464. 

104. McCarthy J, O'Mahony L, Dunne C, et al. An open trial of a novel probiotic as an alternative 
to steroids in mild/moderately active Crohn's disease (abstract). Gut 2001; 49:2447. 

105. Prantrera C, Scribano ML, Falasco G, Andreoli A, Luzi C. Ineffectiveness of probiotics in 
preventing recurrence after curative resection for Crohn's disease: a randomised controlled 
trial with Lactobacillus GG. Gut 2002; 51:405-409. 



12 



The Gastrointestinal Microbiota in Cancer 



Patricia M. Heavey 

School of Life Sciences, Kingston University, Kingston-upon-Thames, U.K. 

Ian R. Rowland 

Northern Ireland Center for Food and Health, University of Ulster, Coleraine, 
Northern Ireland, U.K. 

Joseph J. Rafter 

Department of Medical Nutrition, Novum, Huddinge University Hospital, 
Karolinska Institutet, Stockholm, Sweden 



INTRODUCTION 

The microbiota of the human gastrointestinal tract and in particular the large intestine, 

1 "y 

comprises a large and diverse range of microorganisms, with over 10 bacteria per gram 
of contents (1). It is therefore not surprising that the activities of this microbial population 
have a significant impact on the health of the host. The microbiota interacts with its host at 
both the local (intestinal mucosa) level, and systemically, resulting in a broad range of 
immunological, physiological, and metabolic effects. From the standpoint of the host, 
these effects have both beneficial and detrimental outcomes for nutrition, infections, 
xenobiotic metabolism, toxicity of ingested chemicals, and cancer. 

The participation of intestinal bacteria in carcinogenesis continues to be 
controversial partly due to the lack of agreement on the molecular mechanisms involved 
in the development of this disease. In normal adult tissues, proliferation, apoptosis, and 
DNA repair are in equilibrium and this ensures a steady state of healthy cells. In the 
progression of changes leading from a normal mucosa to carcinoma, at least five to seven 
major molecular alterations need to occur. Extensive studies on colorectal cancer (CRC) 
have identified specific genetic changes in various proto-oncogenes, tumor suppressor 
genes, and DNA mismatch repair genes, as well as alterations in DNA methlyation status 
and inherited genetic defects. Subsequently, several molecular pathways have been 
identified which can contribute to the development of CRC. In 1990, Fearon and 
Vogelstein (2) proposed a genetic pathway of colorectal tumorigenesis, which is now 
generally accepted as the classical model for the development of CRC. The model 
postulates that at least five to seven major molecular alterations need to occur for a normal 
epithelial cell to proceed to carcinoma. This process is now accepted as central to the 
majority of cancers and has been studied extensively in CRC. 

Bacteria have been linked to cancer by two mechanisms: induction of chronic 
inflammation following bacterial infection and production of toxic bacterial metabolites. 

225 



226 Heavey et al. 

The latter mechanism has a strong link with diet. Carcinogenic agents may be present 
in the diet or formed in vivo during digestion. Many of these mechanisms involve the 
metabolic activities of the microbiota normally resident in the human colon. This paper 
discusses both the detrimental and beneficial consequences of bacterial activity of the 
gastrointestinal tract focusing on the stomach and large intestine. 

THE STOMACH 

The pH of the gastric contents of the fasting normal human is usually less than three, which 
is sufficient to kill most commensal bacteria (3). However, during a meal the gastric acid is 
buffered, allowing bacteria ingested with food to survive at least until the pH falls, and 
thus permitting a transient gastric microbiota. However, where gastric acid secretion is 
impaired, bacteria can survive longer and even proliferate in the elevated pH conditions. 
Reduced gastric acid secretion (hypochlorhydria) occurs naturally with ageing (4) and is 
common after gastric surgery. Certain diseases such as pernicious anemia and 
hypogammaglobulinaemia are associated with achlorhydria, which results in the gastric 
pH rising to seven and above (4). This allows a diverse microbiota with up to 10 9 
organisms per gram to establish, consisting usually of species of salivary bacteria of the 
genera Streptococcus, Neisseria, Staphylococcus, and Veillonella, although Bacteroides, 
Lactobacillus and Escherichia species are also found (4). Hypochlorhydria is also 
common in patients with atrophic gastritis associated with chronic Helicobacter pylori 
H. pylori infection. 

The presence of a gastric microbiota in hypochlorhydric and achlorhydric 
individuals has potential toxicological sequelae since it increases the probability of 
xenobiotic metabolism by the bacteria, particularly since the gastric emptying time of such 
patients may be up to 5 hours (4). It has been suggested that the increased gastric cancer 
risk of achlorhydric patients is linked to increased formation of N-nitroso compounds 
(NOC) by their gastric microbiota (5). 

Helicobacter pylori 

H. pylori is a Gram-negative bacterium found in the human stomach and plays an 
important role in the pathogenesis of chronic gastritis and peptic ulcers (6). Additionally, 
both epidemiological and clinical evidence has indicated that H. pylori is associated with 
an increased risk of gastric carcinoma (7,8) and as such it is the first bacterium to be termed 
a definitive cause of cancer by the International Agency for Research into Cancer (I ARC). 
The cag pathogenicity island appears to play an important role in the aetiology of the 
disease since, in developed countries, strains of H. pylori that carry it are associated with 
an increased risk of peptic ulcer and adenocarcinoma than strains that are negative for the 
cag island (9). 

The precise mechanisms involved in its pathogenesis have yet to be fully elucidated 
although numerous clues can be derived from in vitro models and animal studies. 

The inflammatory effects of H. pylori infection have been related to cancer due to 
increased cell proliferation and production of mutagenic free radicals and NOC (10). In the 
Mongolian gerbil model ofH. pylori infection, it has been shown that H. pylori inoculation 
can induce abnormality in gastric mucosal cell proliferation (11). 

Infection with H. pylori is associated with significant epithelial cell damage as well 
as an increased level of apoptosis. However, the mechanism for H. pylori induced 
apoptosis in gastric epithelial cells remains uncertain. Apoptosis is a genetically 
programmed mode of cell death that is regulated by many genes, including oncogenes 



The Gastrointestinal Microbiota in Cancer 227 

and oncosuppressor genes, which may be mutated, delayed or abnormally expressed in 
neoplasia, thus altering tumor cell susceptibility to apoptosis (12). The role of the p53 
tumor supressor gene in apoptosis is currently of particular interest. Genetic abnormalities 
in this gene have been observed in a wide range of human cancers, and are also closely 
associated with the transition from adenoma to carcinoma (13). The mutational 
inactivation of p53 function allows cells to continue with their cell cycle, meaning 
damaged or mutated DNA is propagated in the next generation of cells. 

Zhang and coworkers (14) examined the effect of H. pylori on gastric epithelial 
cells and the role of p53 and showed that the organism induced a time and dose dependent 
inhibition of cell growth and apoptosis over 72 hours. In agreement with other findings 
(15), at low inoculations of H. pylori, cell DNA synthesis was stimulated compared to the 
controls. They also demonstrated no difference in the induction of gastric cell epithelial 
cell apoptosis and cell proliferation between cells exposed to cagA positive and cagA 
negative strains. In addition, H. pylori infection was associated with changes in oncogene 
and tumor suppressor gene expression as shown by increased ras p21 expression and p53 
mutation in H. pylori positive cases of gastric cancer (16). Cell cycle regulatory proteins 
have also been identified as critical targets during carcinogenesis. It has been shown that 
chronic H. pylori infection is associated with decreased expression of the cyclin 
dependent kinase inhibitor (CDI) p27kipl. Another CDI, pl6Ink4a (pi 6) is over- 
expressed in gastric epithelial cells of H. pylori patients and this is associated with an 
increase in apoptosis (17). 

High dose vitamin C has been shown to inhibit H. pylori growth and colonization 
(18) and at physiological concentrations it induced H. pylori associated apoptosis and 
cell cycle arrest in vitro (19). Such effects may account for the observed negative 
association between dietary vitamin C intake and gastric cancer risk (20) although other 
mechanisms include the ability of vitamin C to scavenge reactive oxygen species and 
inhibit NOC formation. Other studies have implicated cigarette smoking and low levels 
of dietary vitamin C as a contributing factor in those high risk individuals with H. pylori 
infection (21,22). 

Overexpression of cyclooxygenase 2 (COX-2) has also been observed in tissues of 
human gastric cancer. There are two isoforms of COX; COX-1 and COX-2. These are key 
enzymes that convert arachidonic acid to prostaglandins. COX-1 is expressed in most 
human tissues, whereas COX-2 is usually undetectable. Overexpression of COX-2 has 
been implicated in a number of cancers including gastric and colon cancer. It has been 
shown that COX-2 was overexpressed in 84% of gastric cancer specimens and those 
specimens with cagA positive strain expression had a significantly higher expression of 
COX-2 than the specimens with cagA negative strain expression (23). It has therefore been 
suggested that the application of COX-2 selective inhibitors may be an effective 
preventive strategy for gastric cancer and in particular those that would not cause 
gastrointestinal complications. Both nonsteroidal anti-inflammatory drug (NSAID) use 
and H. pylori infection independently and significantly increase the risk of peptic ulcer and 
ulcer bleeding. In a meta-analysis of the data it was interpreted that there was synergism 
for the development of peptic ulcer and ulcer bleeding between H. pylori infection and 
NSAID use (24). 

The prevalence of H. pylori infection is falling in developing countries and this has 
been linked to changes in the epidemiology of gastrointestinal diseases, in particular 
reduced incidence of gastric cancers in western countries (25,26). Improved nutrition, 
water supplies and reduced family sizes have been associated with reduced H. pylori 
colonization (25). Novel treatment of this infection using probiotics is in the initial stages 
and results indicate only a slight improvement (27). 



228 Heavey et al. 

THE LARGE INTESTINE 

It is becoming increasingly evident that the large and complex bacterial population of the 
large intestine and their metabolism has an important role in toxicity of ingested chemicals 
and in cancer (28-31). A number of potential mechanisms have been proposed whereby 
gut bacteria may impact carcinogenesis. They may have a direct effect through the binding 
of potential mutagens and thus reduce exposure to the host (32). The normal microbiota 
present in the gut is known to produce and release toxins, which can bind specific cell 
surface receptors and affect intracellular signal transduction (33). Bacterial involvement in 
CRC has been widely studied with most information being derived from animal work and 
some human studies. Evidence from a wide range of sources supports the view that the 
colonic microbiota is involved in the etiology of cancer (Table 1). 

Gut Bacterial Involvement in Colorectal Cancer 

Comparisons of the fecal microbiota of healthy subjects and colon cancer patients have 
not revealed any consistent patterns, possibly due to the difficulties in culturing and 
identifying gut organisms. Elevated numbers of Bacteroides have been associated with 
increased colon cancer risk in humans (34,35). Similarly, lecithinase-negative Clostridium 
and Lactobacillus were more abundant in colon cancer patients (36) although in another 
study, some Lactobacillus species and Eubacterium aerofaciens have been associated with 
reduced risk (35). 

In animals, the presence of the intestinal microbiota has a major impact on colonic 
tumor formation (37,38). In a study conducted by Reddy and coworkers (38) the rate 
of tumor formation was much more rapid in conventional than in germ-free rats treated 
with the tumor initiator 1,2-dimethylhydrazine (DMH). After 20 weeks, 17% of 
conventional rats had colon carcinomas, whereas there were no tumors (adenomas or 
carcinomas) in the germ-free animals. At 40 weeks, two out of 18 germ-free rats had 
developed benign adenomas (although still none had carcinomas), compared to six out of 
24 conventional rats with tumors (4 cancers, 2 adenomas); thus the gut microbiota had a 
tumor-promoting effect when DHM was the tumor initiator. 

A high incidence of spontaneous CRC has been demonstrated in the T-cell receptor 
(TCR) P chain and p53 double-knockout mice. In one study, 70% of the animals with a 
conventional microbiota developed adenocarcinomas, whereas adenocarcinoma of the 
colon did not occur in germ-free TCR $~'~ p53~'~ mice, thus indicating a major role for 
the intestinal microbiota (39). 

Table 1 Evidence That the Colonic Microbiota Is Involved in the Etiology of Colon Cancer 

Human feces have been shown to be mutagenic, and genotoxic substances of bacterial origin have 

been isolated 
Intestinal bacteria can produce, from dietary components, substances with genotoxic, carcinogenic, 

and tumor-promoting activity 
Gut bacteria can activate procarcinogens to DNA reactive agents 

Germ-free rats fed human diets exhibit lower levels of DNA adducts in tissues than conventional rats 
Germ-free rats treated with the carcinogen 1 ,2-dimethylhydrazine have a lower incidence of colon 

tumors than similarly treated rats having a normal microbiota 
Germ-free T-cell receptor chain and p53 double-knockout (TCR3~ p53~ ") mice did not develop 

adenocarcinoma of the colon at 4 months of age. Adenocarcinomas of the ileocecum and cecum 

were detected in 70% of the conventional TCR(3 -/ ~ p53 _/ ~ mice 



The Gastrointestinal Microbiota in Cancer 229 

Streptococcus bovis has been implicated in colonic neoplasia and supplements of 
this strain of bacteria and antigens extracted from the bacterial cell wall were shown to 
induce formation of hyperproliferative aberrant colonic crypts and increase the expression 
of proliferation markers in carcinogen treated rats (40). The effect of individual bacteria on 
cancer risk varies. Mice mono-associated with Mitsuokella multiacida, Clostridium 
butyricium or Bifidobacterium longum had a higher incidence of colonic adenoma (68% in 
each case) as compared to those associated with Lactobacillus acidophilus (30%) (41). 

Gut Bacterial Metabolism and CRC Risk 

The enormous numbers and diversity of the human gut microbiota is reflected in a large 
and varied metabolic capacity, particularly in relation to xenobiotic biotransformation, 
carcinogen synthesis and activation. The metabolic activities of the gut microbiota can 
have wide-ranging implications for the health of the host (42). To date the vast majority of 
mechanisms whereby bacteria are involved in carcinogenesis involve toxic or protective 
products of bacterial metabolism. Such metabolic activities include numerous enzymatic 
reactions and degradation of undigested dietary residues. Diet can substantially modulate 
these activities by providing a vast array of substrates. A wide range of enzyme activities 
capable of generating potentially carcinogenic metabolites in the colon are associated with 
the gut microbiota, including (3-glucuronidase, (3-glucosidase, nitrate reductase and nitro- 
reductase. These are usually assayed in fecal suspensions and appear to be present in many 
bacterial types (43-52). 

A major role for the intestinal microbiota has been identified in the metabolism of 
the bile acids. The primary bile acids, chenodeoxycholic acid and cholic acid, are subject 
to extensive metabolism by the intestinal microbiota (53), predominantly 7-a- 
dehydroxylation, which converts cholic to deoxycholic acid (DCA) and chenodeoxycholic 
acid to lithocholic acid (LCA). These secondary bile acids exert a range of biological and 
metabolic effects in vitro and in vivo including cell necrosis, hyperplasia, and tumor- 
promoting activity in the colon, induction of DNA damage and apoptosis (54). It has also 
been suggested that secondary bile acids influence CRC by selecting for apoptosis- 
resistant cells or by interacting with various secondary messenger signaling systems. 

A number of human observational studies in patients with adenomas or CRC have 
reported a correlation between fecal bile acid (FBA) concentrations and CRC risk (55,56). 
Some studies have also suggested that high fecal DCA concentrations and DCA to LCA 
ratio are associated with increased CRC risk (57). However, not all studies have confirmed 
this relationship between bile acids and CRC risk (58). 

Formation of Protective Agents During Fermentation 

Both dietary and endogenous carbohydrate substrates (e.g., starch and non-starch 
polysaccharides and intestinal mucins) are hydrolyzed by gut bacterial enzymes to 
produce the short chain fatty acids (SCFAs), acetate, propionate, and butyrate (59). These 
SCFAs provide an energy source for the intestinal cells and are also thought to confer 
beneficial effects on the host. SCFAs decrease colonic and fecal pH and this acidic 
environment is thought to be beneficial to the host (60). Specific oligosaccharides and 
resistant starch that result in SCFAs, and in particular butyrate (61) may have the potential 
to decrease CRC risk. This SCFA is of specific interest since it has been shown to induce 
apoptosis in colon adenoma and colon cell lines. In vitro studies have shown that 
increased butyrate supply to colon cells induces growth of the gut epithelium whereas 
reduced butyrate supply causes gut atrophy and functional impairments (62). Sodium 



230 Heavey et al. 

butyrate has been observed to induce apoptosis and to alter the resistance of colonic tumor 
cells to apoptosis (62). However, the majority of these results have come from 
experiments conducted in vitro and again there have been conflicting views (63). 

It follows from the above that modification of the gut microbiota may exert a 
beneficial effect on the process of carcinogenesis and this opens up the possibility for 
dietary modification of colon cancer risk. Probiotics and prebiotics, which modify the 
microbiota by increasing the numbers of lactobacilli and/or bifidobacteria in the colon, 
have been a particular focus of attention in this regard. In general species of 
Bifidobacterium and Lactobacillus have low activities of those enzymes involved in 
carcinogen formation and metabolism by comparison to other major anaerobes in the gut 
such as Bacteroides, Eubacteria and Clostridia (44). This suggests that increasing the 
proportion of lactic acid bacteria (LAB) in the gut could modify, beneficially, the levels of 
xenobiotic metabolizing enzymes. This manipulation of the gut is discussed in greater 
detail in other chapters within this book. Overall, experimental and animal research show 
encouraging effects of several probiotic strains to decrease colon cancer, leading the way 
to the development of well-designed human intervention trials. 

Effects of Gut Microbiota on Gene Expression 

To date, there are only a few molecular descriptions of how bacteria in the normal 
microbiota regulate gene products with presumed positive functions in the intestine or 
systemically. Dramatic changes in gene expression were noted when germ-free mice 
were mono-colonized with Bacteroides thetaiotaomicron, a component of the normal 
microbiota of adult mice and humans (64). A number of genes involved in general 
mechanisms like nutrient uptake, fortification of the intestinal epithelial barrier, 
postnatal development, and angiogenesis are regulated in response to this commensal 
microbe. In addition, it is becoming clear that metabolic products, produced by the gut 
microbiota, can alter gene expression in the colonocyte [e.g., butyrate, produced by 
bacterial fermentation of dietary fiber, induces p21/Cipl/WAFl mRNA (important in cell 
cycle control)] and secondary bile acids, produced from primary bile acids by the gut 
microbiota, alter AP-1 -dependent and COX-2 gene transcription) (65,66). 



SURROGATE MARKERS FOR DIET-RELATED 
COLON CANCER STUDIES 

As discussed above, the gut microbiota has been implicated in the etiology of CRC by a 
number of studies and these observations form the theoretical basis for the use of several 
gut microbiota biomarkers (fecal biomarkers) in studies on diet and colon cancer. They are 
composed of two main categories; those examining the activity of bacterial enzymes or 
bacterial metabolites and those based on bioassays on fecal water. For a more thorough 
review of this subject, the reader is referred to Rafter and coworkers (67). 

Bacterial Enzymes 

A wide range of enzyme activities capable of generating potentially carcinogenic 
metabolites in the colon are associated with the gut microbiota, including P-glucuronidase 
P-glucosidase, nitrate- and nitro-reductase. These are usually assayed in fecal suspensions 
and appear to be present in many bacterial types. Of these enzymes, P-glucuronidase has 
been the most extensively investigated as a biomarker of CRC risk. It should be noted that 



The Gastrointestinal Microbiota in Cancer 231 

these factors are associated with the generation of carcinogens and promoters and do not 
have a direct link with tumors. 

(5 -Glucuronidase 

Many carcinogenic compounds are metabolized in the liver and then conjugated to 
glucuronic acid before being excreted via the bile into the small intestine. In the colon 
bacterial P-glucuronidase can hydrolyze the conjugates, releasing the parent compound or 
its activated, hepatic metabolite. 

The activity of P-glucuronidase in the colon can alter the likelihood of tumor 
induction in animal models of CRC. The use of a P-glucuronidase inhibitor administered 
in conjunction with the carcinogen azoxy methane (which undergoes activation and 
conjugation in the liver) significantly reduces the number of tumors formed in the rat 
colon, indicating that microbiota P-glucuronidase has a role in tumor induction. Metabolic 
epidemiological studies have shown that populations at high risk of CRC have high levels 
of fecal P-glucuronidase activity. Furthermore, fecal P-glucuronidase activity in colon 
cancer patients is significantly higher than in healthy controls. 

The activity of P-glucuronidase is influenced by diet. High risk diets for CRC have 
consistently been shown to increase P-glucuronidase activity relative to low risk diets. 
Furthermore, various types of fiber decrease the activity of P-glucuronidase in rats. 

Although it represents a simple reproducible marker, evidence for a role for 
P-glucuronidase in human CRC is indirect and is remote from the final endpoint (tumors). 

Metabolites 

A wide range of metabolites with potential genotoxic, tumor-promoting and anti- 
carcinogenic activities have been identified in feces. 

N-Nitroso Compounds 

Nitrate, ingested via diet and drinking water, is reduced by gut bacterial nitrate reductase 
to its more reactive and toxic reduction product, nitrite. Nitrite reacts with nitrogenous 
compounds in the body to produce NOC. The reaction can occur chemically in the acidic 
conditions prevalent in the human stomach and can also be catalyzed at neutral pH by gut 
bacteria in the colon. 

The term NOC covers a wide range of compounds including N-nitrosamines, 
N-nitrosamides, N-nitrosoguanidines, and N-nitrosoureas, the majority of which are 
highly carcinogenic, DNA alkylating agents. However, the genotoxic or carcinogenic 
activity of the NOC produced by the bacterial N-nitrosation process in the large intestine 
has not yet been established. 

Fecal apparent total NOC (ATNC) excretion is increased by red meat consumption. 
In conjunction with high meat intakes, wheat bran, resistant starch and vegetable 
consumption had no effect on fecal ATNC excretion or concentration. 

Secondary Bile Acids 

The primary bile acids, chenodeoxycholic acid and cholic acid, are subject to extensive 
metabolism, predominantly 7-a-dehydroxylation, by the intestinal microbiota, which 
converts cholic to DCA and chenodeoxycholic to LCA. These are termed secondary 
bile acids. 



232 Heavey et al. 

Epidemiological studies indicate that concentrations of secondary bile acids are 
higher in populations at high risk of CRC and in case control studies 7-a-dehydroxylase 
activity is higher in cases than controls. In human studies, high fat intake, which correlates 
with CRC risk, increases FBA concentrations, whereas increased consumption of wheat 
bran (negatively correlated with CRC risk) reduces FBA concentration. 

Short Chain Fatty Acids 

The SCFAs acetate, propionate, and butyrate are the principal end-products of 
carbohydrate fermentation. These are absorbed from the colonic lumen and metabolized 
by various body tissues. Butyrate is preferentially metabolized by colonocytes. 

There is evidence from in vitro studies and animal models (where cecal SCFA 
concentrations can be measured) that the type of carbohydrate has an important influence 
on the amount and proportions of SCFA produced, with starch and wheat bran being 
particularly associated with elevated butyrate production. In human studies, inulin has 
been shown to enhance excretion of total SCFA in human feces, whereas wheat bran 
increased absolute or relative proportions of butyrate in feces. Where the butyrate is 
produced relative to proximal and distal regions of the colon is important and should be a 
methodological consideration. 

Gut bacterial enzymes and fecal metabolites are relatively simple to measure 
routinely and in general may be of use in assessing effects of diet on modulating exposure 
of the colon to potential carcinogens, rather than reflecting cancer risk. 

Fecal Water Activities 

Fecal Water Cytotoxicity 

There is considerable evidence that colon tumors are a result of gut luminal factors 
damaging the mucosa. Furthermore, free reactive and soluble factors are more likely to 
affect the epithelium than substances bound to the insoluble matrix such as fiber. 
Therefore, an alternative approach to assaying enzymes or metabolites in feces is to 
assess toxicological activity of fractions using short-term tests for toxicity, genotoxicity, 
and mutagenicity. Usually the aqueous phase of the human feces (fecal water) is used, 
since this will contain most of the free reactive species. For assessment of fecal water 
cytotoxicity, the effect on proliferation of human colon carcinoma cells in culture 
is used. 

Proliferative zone expansion in the colonic crypts and an increased rate of epithelial 
proliferation are considered to be an early step in carcinogenesis. Stimulation of 
proliferative activity in colonic epithelium may in part be mediated via cytotoxic 
mechanisms, resulting in increased cell loss at the epithelial surface and a compensatory 
rise in mitotic activity of the crypts. Such considerations led to the development of assays 
to assess cytotoxic activity in fecal water towards colon cells in vitro. It is thought that bile 
acids, especially secondary bile acids, make a major contribution to fecal water 
cytotoxicity. In a comparison of fecal water cytotoxicity in patients at low (no colon 
adenomas) medium (small colorectal adenomas) and high (large tubular adenomas) risk of 
CRC, no significant differences between the groups were observed. 

Interventions using dietary regimes associated with increased or decreased CRC risk 
have been shown to modulate appropriately fecal water cytotoxicity. For example, dietary 
calcium has frequently been shown to reduce the cytotoxicity of fecal water presumably by 
precipitating soluble bile acids. Fecal water cytotoxicity was higher in subjects on a high 
fat, low calcium, low fiber diet compared with those on a low fat, high calcium, high fiber 



The Gastrointestinal Microbiota in Cancer 233 

regime. In rats, a high red meat consumption increases the cytotoxicity of fecal water. This 
effect was independent of the fat and bile acid content of the fecal water and may be related 
to dietary haem. 

Fecal Water Genotoxicity 

The presence of DNA damaging activity towards human cultured colon cells has been 
demonstrated in samples of fecal water from healthy human subjects. A wide variation was 
found ranging from negligible to high activity. The presence of genotoxic activity in fecal 
water can be considered to reflect exposure of the colonic mucosa to carcinogens. 

There is now convincing evidence that CRC is induced by a series of mutational 
events in a number of critical genes. Sporadic colorectal tumors have been shown to 
contain mutations and deletions in oncogenes, and tumor suppressor genes such as Ape, 
K-ras, and p53. DNA damage has been detected in biopsies of colon tissue derived 
from laboratory animals and human subjects. Thus, the presence in the colonic lumen 
of DNA damaging agents could represent an important risk factor for CRC. There are 
as yet no reports of validation studies for the endpoint in patients at different risk 
of CRC. 

In healthy subjects, a diet high in fat and meat, but low in dietary fiber (hence 
considered to be of high CRC risk) was associated with a significantly increased fecal 
water genotoxicity by comparison to a diet low in fat and meat. 

Cytotoxicity and particularly genotoxicity of fecal water have a good mechanistic 
link with colon carcinogenesis and hence provide potentially valuable, non-invasive 
methods for assessing CRC risk in human subjects. However, there is a need for more 
extensive validation of these endpoints. 



CONCLUSION 

It is becoming increasingly evident that the microbiota of the gastrointestinal tract and in 
particular that of the large intestine interacts with its host and may exert either harmful or 
protective effects, thus participating in the etiology of cancer. Gastric adenocarcinoma is 
the second leading cause of cancer-related deaths in the world and has been associated 
with the presence of H. pylori in the stomach. Several mechanisms of how this bacterium 
may affect tumorigenesis have been identified as well as dietary and environmental 
agents, which may confer either protective or detrimental effects. Colon cancer is the 
fourth most common cancer worldwide and again environmental factors and in particular 
diet play an important role in this disease. It has been shown that the microbiota of the gut 
interacts with its host both locally and systemically resulting in a broad range of effects, 
which may have both beneficial and detrimental outcomes, for nutrition, infections, 
xenobiotic metabolism, toxicity of ingested chemicals, and cancer. It is important to gain 
more insight into the pathogenesis of these cancers in order to develop more effective 
preventive and treatment strategies. The use of pro- and prebiotics may serve to induce 
beneficial effects on the host. Further research from well-planned intervention trials is 
required to further our understanding of the role of these agents in human carcinogenesis. 
Finally, as our understanding of the role of the gut microbiota in health and disease 
improves, we will be able to develop even better surrogate markers for use in human 
dietary intervention studies. 



234 Heavey et al. 

REFERENCES 

1 . Cummings JH, MacFarlane GT. The control and consequences of bacterial fermentation in the 
human colon. J Appl Bacteriol 1991; 70:443^159. 

2. Fearon ER, Vogelstein B. A genetic model for colorectal tumorgenesis. Cell 1990; 61 :759-767. 

3. Draser BS. The bacterial flora of the intestine. In: Rowland IR, ed. The role of the Gut Flora in 
Toxicity and Cancer. London: Academic Press, 1988:23-38. 

4. Hill MJ. In: Hill MJ, ed. The role of Gut Bacteria in Human Toxicology and Pharmacology. 
London: Taylor and Francis, 1995. 

5. Hill MJ. Gut flora and cancer in humans and laboratory animals. In: Rowland IR, ed. The role 
of the Gut Flora in Toxicity and Cancer. London: Academic Press, 1988:461-502. 

6. Graham DY, Lew GM, Klein PD, et al. Effect of treatment of Helicobacter pylori infection on 
the long-term recurrence of gastric or duodenal ulcer: a randomised, controlled study. Ann Int 
Med 1992; 116:705-708. 

7. Asaka M, Kimura T, Kato M, et al. Possible role of Helicobacter pylori infection in early 
gastric cancer development. Cancer 1994; 73:2691-2694. 

8. Kikuchi S, Wada O, Nakajaima T, et al. Serum anti-He licobacter pylori antibody and gastric 
carcinoma among young adults. Research group on prevention of gastric carcinoma among 
young adults. Cancer 1995; 75:2789-2793. 

9. Atherton JC. H. pylori virulence factors. Br Med Bull 1998; 54:105-120. 

10. Moss SF. The carcinogenic effect ofH. pylori on the gastric epithelial cell. J Physiol Pharmacol 
1999; 50:847-856. 

11. Yao YL, Xu B, Song YG, et al. Effect of Helicobacter pylori infection on gastric mucosal cell 
proliferation in mongolian gerbils. Di Yi Junyi Daxue Xuebao 2002; 22:348-350. 

12. Arends MJ. How do cancer cells die? Apoptosis and its role in neoplastic progression. In: 
Leake R, Gore M, Ward RH, eds. The Biology of Gynaecological Cancer. Royal College of 
Obstetricians and Gynaecological Press, London, 1995:73-91. 

13. Cho KR, Vogelstein B. Suppressor gene alterations in the colorectal adenoma-carcinoma 
sequence. J Cell Biochem 1992; 16G:137-141. 

14. Zhang ZW, Patchett SE, Farthing MJ. Role of Helicobacter pylori and p53 in regulation of 
gastric epithelial cell cycle phase progression. Dig Dis Sci 2002; 47:987-995. 

15. Wagner S, Beil W, Westermann J, et al. Regulation of gastric epithelial cell growth by 
Helicobacter pylori: offdence for a major role of apoptosis. Gastroenterology 1997; 
113:1836-1847. 

16. Wang J, Chi DS, Kalin GB, et al. Helicobacter pylori infection and oncogene expressions in 
gastric carcinoma and its precursor lesions. Dig Dis Sci 2002; 47:107-113. 

17. Shirin H, Hibshoosh H, Kawabata Y, et al. P16Ink4a is overexpressed in H. /?y /on-associated 
gastritis and is correlated with increased epithelial apoptosis. Helicobacter 2003; 8:66-71. 

18. Correa P, Malcolm G, Schmidt B, et al. Antioxidant micronutrients and gastric cancer. Aliment 
Pharmacol Ther 1998; 12:73-82. 

19. Zhang HM, Wakisaka N, Maeda O, Yamamoto T. Vitamin C inhibits the growth of a bacterial 
risk factor for gastric carcinoma: Helicobacter pylori. Cancer 1997; 80:1897-1903. 

20. Zhang ZW, Abdullahi M, Farthing MJ. Effect of physiological concentrations of vitamin C on 
gastric cancer cells and Helicobacter pylori. Gut 2002; 50:165-169. 

21. Kaaks R, Tuyns AJ, Haelterman M, Riboli E. Nutrient intake patterns and gastric cancer risk: a 
case control study in Belgium. Int J Cancer 1998; 78:415-420. 

22. You WC, Zhang L, Gail MH, et al. Gastric dysplasia and gastric cancer: Helicobacter pylori, 
serum vitamin C and other risk factors. J Natl Cancer Inst 2000; 92:1607-1612. 

23. Guo X, Wang L, Yuan Y. Association between Helicobacter pylori cagA strain infection and 
expression of cyclooxygenase 2 in gastric carcinoma. Chin Med J 2002; 82:868-871. 
Breuer NF, Dommes P, Jaekel S, Goebell H. Fecal bile acid excretion pattern in colonic cancer 
patients. Dig Dis Sci 1985; 30:852-859. 

24. Huang JQ, Sridhar S, Hunt RH. Role of Helicobacter pylori infection, and non-steroidal anti- 
inflammatory drugs in peptic-ulcer disease: a meta-analysis. Lancet 2002; 359:14-22. 



The Gastrointestinal Microbiota in Cancer 235 

25. Blaser MJ. Helicobacters are indigenous to the human stomach: duodenal ulceration is due to 
changes in microecology in the modern era. Gut 1998; 43:721-727. 

26. Logan RP, Walker MM. ABC of the upper gastrointestinal tract: epidemiology and diagnosis of 
Helicobacter pylori infection. BMJ 2001; 323:920-922. 

27. Michetti P, Dorta G, Wiesel PH, et al. Effect of whey-based culture supernatant of 
Lactobacillus acidophilus (johnsonii) Lai on Helicobacter pylori infection in humans. 
Digestion 1999; 60:203-209. 

28. Mitsuoka T. Recent trends in research on intestinal flora. Bifidobacteria Microflora 1982; 
3:3-24. 

29. Draser BS. The bacterial flora of the intestine. In: Rowland IR, ed. The Role of the Gut Flora in 
Toxicity and Cancer. London: Academic Press, 1998:23-38. 

30. Rowland IR, ed. Role of the Gut Flora in Toxicity and Cancer. London: Academic Press, 1988. 

31. Rowland IR. Toxicology of the colon: role of the intestinal flora. In: Gibson GR, 
MacFarlane GT, eds. Human Colonic Bacteria: Role in Nutrition, Physiology and Pathology. 
Boca Raton: CRC Press, 1995:155-174. 

32. Orrhage K, Sillerstrom E, Gustafsson JA, et al. Binding of the mutagenic heterocyclic amines 
by intestinal and lactic acid bacteria. Mutat Res 1994; 311:239-248. 

33. Fassano A. Cellular microbiology: can we learn cell physiology from microorganisms? Am 
J Physiol 1999; 276:C765-C776. 

34. Hill MJ, Draser BS, Hawksworth G, et al. Bacteria and aetiology of cancer of the large bowel. 
Lancet 1971; i:95-100. 

35. Moore WE, Moore LH. Intestinal floras of populations that have a high risk of colon cancer. 
Appl Environ Microbiol 1995; 61:3202-3207. 

36. Kanazawa K, Konishi F, Mitsuoka T, et al. Factors influencing the development of sigmoid 
colon cancer. Bacteriologic and biochemical studies. Cancer 1996; 77:1701-1706. 

37. Reddy BS, Narisawa T, Maronpot R, et al. Animal models for the study of dietary factors and 
cancer of the large bowel. Cancer Res 1975; 35:3421-3426. 

38. Reddy BS, Weisburger JH, Narisawa T, Wynder EL. Colon carcinogenesis in germ-free rats 
with dimethylhydrazine, and N-nitrosamines in health and gastroduodenal disease. Lancet 
1974; ii:550-552. 

39. Kado S, Uchida K, Funabashi H, et al. Intestinal microflora are necessary for development of 
spontaneous adenocarcinoma of the large intestine in T-cell receptor 3 chain and p53 double- 
knockout mice. Cancer Res 2001; 61:2395-2398. 

40. Ellmerich S, Djouder N, Scholler M, Klein JP. Production of cytokines by monocytes, 
epithelial and endothelial cells activated by Streptococcus bovis. Cytokine 2000; 12:26-31. 

41. Horie H, Kanawawa K, Okada M, et al. Effects of intestinal bacteria on the development of 
colonic neoplasm: an experimental study. Eur J Cancer Prev 1999; 8:237-245. 

42. Rowland IR, Gangolli SD. Role of gastro-intestinal microflora in the metabolic and 
toxicological activities of xenobiotics. In: Ballentyne B, Marrs, TC, Syveron T eds. General 
and Applied Toxicology 1999; P562-576. 

43. Cole CB, Fuller R, Mallett AK, Rowland IR. The influence of the host on expression of 
intestinal microbial enzyme activities involved in metabolism of foreign compounds. J Appl 
Bacteriol 1985; 58:549-553. 

44. Saito Y, Takano T, Rowland IR. Effects of soybean oligosaccharides on the human gut 
microflora in in vitro culture. Microb Ecol Health Dis 1992; 5:105-110. 

45. Tadaka H, Hirooka T, Hiramatsu Y, Yamamoto M. Effects of beta-glucuronidase inhibitor on 
azoxymethane-induced colonic carcinogenesis in rats. Cancer Res 1982; 42:331-334. 

46. Grasten SM, Juntunen KS, Poutanen KS, et al. Rye bread improves bowel function and 
decreases the concentrations of some compounds that are putative colon cancer risk markers in 
middle-aged women and men. J Nutr 2000; 130:2215-2221. 

47. Kim DH, Jin YH. Intestinal bacterial beta-glucuronidase activity of patients with colon cancer. 
Arch Pharmacol Res 2001; 24:564-567. 

48. Eriyamremu GE, Osagie VE, Alufa OI, et al. Early biochemical events in mice exposed to 
cycas and fed a nigerian-like diet. Ann Nutr Metab 1995; 39:42-51. 



236 Heavey et al. 

49. Rowland IR, Tanaka R. The effects of transgalactosylated oligosaccharides on gut flora 
metabolism in rats associated with a human fecal microflora. J Appl Bacteriol 1993; 
74:667-674. 

50. Mallet AK, Rowland IR. Factors affecting the gut microflora. In: Rowland IR, ed. Role of the 
Gut Flora in Toxicity and Cancer. London: Academic Press, 1988:347-382. 

51. Hughes R, Cross AJ, Pollock JR, Bingham S. Dose-dependent effect of dietary meat on 
endogenous colonic N-nitrosation. Carcinogenesis 2001; 22:199-202. 

52. Silvester KR, Bingham SA, Pollock JR, et al. Effect of meat and resistant starch on fecal 
excretion of apparent N-nitroso compounds and ammonia from the human large bowel. Nutr 
Cancer 1997;29:13-23. 

53. MacDonald IA, Sutherland JD, Cohen BI, Mosbach EH. Effect of bile acid oxazoline 
derivatives on microorganisms participating in 7 alpha-hydroxyl epimenzation of primary bile 
acids. J Lipid Res 1983; 24:1150-1559. 

54. Gill CI, Rowland IR. Diet and cancer: assessing the risk. Br J Nutr 2002; 88:S73-S87. 

55. Imray CHE, Radley S, Davis A, et al. Fecal unconjugated bile acids in patients with colorectal 
cancer or polyps. Gut 1992; 33:1239-1245. 

56. Stadler J, Yeung KS, Furrer R, et al. Proliferative activity of rectal mucosa and soluble fecal 
bile acids in patients with normal colons and in patients with colonic polyps or cancer. Cancer 
Lett 1988; 38:315-320. 

57. Owen RW. Feacal steroids and colorectal carcinogenesis. Scand J Gastroenterol 1997; 
222:76-82. 

58. de Kok TM, van Maanen JM. Evaluation of fecal mutagenicity and colorectal cancer risk. 
Mutat Res 2000; 463:53-101. 

59. Silvi S, Rumney CJ, Cresci A, Rowland IR. Resistant starch modifies gut microflora and 
microbial metabolism in human flora-associated rats inoculated with feces from Italian and 
U.K. donors. J Appl Microbiol 1999; 86:521-530. 

60. Scheppach W. Effects of short chain fatty acids on gut morphology and function. Gut 1994; 
LS35-S38. 

61. Bird AR, Brown IL, Topping DL. Starches, resistant starches, the gut microflora and human 
health. Curr Issues Intest Microbiol 2000; 1:25-37. 

62. Archer S, Meng S, Shei A, Hodin RA. P21WAFI is required for butyrate-mediated growth 
inhibition of human colon cancer cells. Proc Natl Acad Sci USA 1998; 95:6791-6796. 

63. Ishizuka S, Sonoyama K, Kassai T. Changes in the number and apoptosis of epithelial cells in 
the colorectum of wheat bran-fed rats soon after administering 1,2-dimethylhydrazine. Biosci 
Biotechnol Biochem 1997; 61:1337-1341. 

64. Xu J, Bjursell MK, Himrod J, et al. A genomic view of the human- Bacteroides 
thetaiotaomicron symbiosis. Science 2003; 299:2074-2076. 

65. Smith JG, Yokoyama WH, German BG. Butyric acid from the diet: actions at the level of gene 
expression. Clin Rev Food Sci 1998; 38:259-297. 

66. Hodin RA, Meng S, Archer S. Cellular growth state differentially regulates enterocyte gene 
expression in butyrate-treated HT-29 cells. Cell Growth Differ 1996; 7:647-653. 

67. Rafter J, Govers M, Martel P, et al. PASSCLAIM — Diet-related cancer. European Journal of 
Nutrition,. 2004; 43:1147-1184. 



13 



In Vitro Methods to Model 
the Gastrointestinal Tract 



Harri Makivuokko and Paivi Nurminen 

Danisco Innovation, Kantvik, Finland 



INTRODUCTION 

The human intestinal microbiota has never been so intensively studied as in this current 
period. Over the last decade, the use of molecular methods, especially those based on 16S 
ribosomal RNA, have generated much knowledge on the composition of the intestinal 
microbiota of especially humans but also animals. The relatively easy accessible fecal 
sample is the main source of intestinal microbiota used for various analyses. It is uncertain 
how well fecal samples reflect the composition of the microbiota in the proximal parts of 
the colon (1,2) but it is certainly very different from the small intestine. In order to study 
the microbial composition and activity in these sites, one would need in vivo samples from 
a large number of healthy individuals. Invasive sampling from healthy people is ethically 
not acceptable. Animal models can be used for invasive sampling (see chapter by 
Henriksson); however, due to physiological and anatomical differences, animals will have 
a different microbiota. Therefore, in vitro techniques complement animal studies and offer 
means to test specific hypotheses in a controlled, replicable manner without using animal 
models or clinical samplings. With in vitro models, it is possible to simulate the conditions 
in the human oral cavity, stomach, duodenum, jejunum, ileum, and in the ascending, 
transverse, and descending sections of the colon. 



TYPES OF INTESTINAL SIMULATOR MODELS 

In vitro models can be divided into batch cultures, chemostat-type simulators, including 
semi-continuous and continuous cultures, and non-chemostat-type simulators. All models 
of the gastrointestinal tract (GIT) have strictly anaerobic conditions in order to simulate 
the environment that supports the growth of microbiota obtained from the GIT of humans 
or other mammals. In vitro models can be used sequentially, so that in the simulators of 
stomach and small intestine the food matrix can be digested using conditions and enzymes 
representing the physiological conditions in the upper GIT, while the colon simulators 
continue by simulating the microbial metabolism of the nondigestible residue. The 

237 



238 



Makivuokko and Nurminen 



different chemostat- and non-chemostat-type models have major structural differences, 
but the batch fermentors are generally similarly structured, small-scale bottle fermentors. 
The chemostat models can be run using inocula in either an in vitro steady-state (the 
exponential growth of the bacterial has stabilized) achieved with several days of pre- 
fermentation of the fecal inoculum or after a short (16-24 hours) pre-fermentation. 



Batch-Type Simulators 

The simplest and most commonly used in vitro method in microbiological studies is the use 
of batch fermentation with intestinal fluid or fecal slurry to study the effects of different 
added ingredients. These chemostat are typically anaerobically sealed bottles with fecal, 
caecal or rumen material and these models simulate only a certain part of the animal's GIT, 
e.g., mouse cecum or cow's rumen. The transit times of the intestinal fluids through those 
areas are relatively short and therefore the run-times in batch fermenting simulations range 
from 2-24 hours (3-7). The accumulation of fermentation products (e.g., SCFAs) can 
change the conditions in the batch fermentation from the microbially balanced starting 
point to a more competitive environment for the fermentative microbiota, thus affecting the 
in vivo relevance in longer simulations. More complex fermentation models with several 
vessels and fluid transitions between vessels either continuously or semi-continuously 
avoid this accumulation of metabolites and depletion of nutrients. 



Chemostat-Type Simulators 

The in vitro colon simulators were introduced for the first time in 1981 (8), and all models 
functioning today have a lot in common with this model. Rumney and Rowland reviewed 
the first decade of in vitro simulators in their excellent article (3). Of the models reviewed 
by Rumney and Rowland, the Reading model introduced by Gibson and co-workers in 
1988 (9), revised 1998 by Macfarlane and co-workers (10), is still actively being used and 
two new interesting models have been described in the literature. Of these, the SHIME 
(Simulator for Human Intestinal Microbiological Ecosystem) model introduced by Molly 
et al. in 1993 (11) and the EnteroMix® colon simulator introduced by Makivuokko et al. 




N 



Media 



*=<p> = 3r 




Figure 1 The Reading model. This model represents the human colon in three vessels: 
VI proximal, V2 transverse, and V3 distal colon. Media is pumped to system continuously, and at the 
same time there is a continuous overflow from vessel to vessel. Source: From Ref. 9. 



In Vitro Methods to Model the Gastrointestinal Tract 



239 



Table 1 Colon Simulator Models 





Reading 


SHIME 


EnteroMix® 


TIM 1 


TIM 2 


Simulation 


Colon 


Stomach 


Colon 


Stomach 


Colon 


area 




to colon 




to ileum 




Vessel 


220-320 ml 


300-1600 ml 


6-15 ml 


200 ml 


200 ml 


volumes 












pH levels 


5.8-6.8 


5.0-7.0 


5.5-7.0 


1.8-6.5 


5.8 


Running times 


14 days to 
steady state 


30 days 
per cycle 


2 days 


~ 1 day 


~3 days 



Abbreviations: SHIME, Simulator for Human Intestinal Microbiological Ecosystem; TIM, TNO Intestinal Model. 

in 2005 (12), together with the Reading model, are structurally chemostat models having 
3-6 sequentially attached fermenting vessels with computer controlled fluid transition 
systems (Fig. 1) and (Table 1). The Reading model and the EnteroMix model both 
simulate only the human colon, and a similar artificial simulator media described by 
Macfarlane et al. (10) is used in them to simulate the fluid entering the colon from the 
small intestine. The SHIME model simulates the whole human GIT from stomach to colon 
using artificial SHIME media, which has much in common with the medium described by 
Macfarlane and co-workers (10). These three models have three different designs in fluid 
transition. Fluids are either pumped semi-continuously to the subsequent vessels in three- 
hour intervals (EnteroMix® model), there is a continuous overflow of fluids between 
vessels (the Reading model), or the model can be a combination of these two types 
(SHIME). 

Reading Simulator 

The Reading simulator (Fig. 1) simulates the gut using a 3 stage continuous culture with 
three glass vessels (220 ml, 320 ml and 320 ml) and different pH in each vessel (5.8, 6.2, 
and 6.8); mimicking the human proximal, transverse, and distal colon, respectively. 

In the beginning of the simulation, each vessel is inoculated with 100 ml of 20% 
(wt/vol) of human feces. The system is incubated in a batch overnight, after which a 
continuous pumping of fresh simulator fluid to the first vessel is started. At the same time a 
continuous overflow from vessel to vessel begins and the system is run for at least 14 days 
in order achieve a steady- state condition in the vessels. The excess fluid from the third 
vessel is collected to a waste container. The total retention time of the system can vary, 
e.g., between 27 and 67 hours (10). The viability of the microbiota is determined by taking 
samples at regular intervals from the vessels. After the incubation period, the test 
substance is added to the system mixed in the fresh simulation fluid and the system is then 
run to new steady state [e.g., for 22 days (9)]. The last phase is the washout period [e.g., for 
50 days (9)] with the original simulation fluid to determine how long the changes induced 
by the test substance can still be measured in the absence of the substrate itself. 



SHIME Model 

The current SHIME model is a single six-stage system, where the first three glass vessels 
simulate stomach and small intestine and the subsequent three glass vessels the large 
intestine (11a). The original SHIME model (Fig. 2) (11) was a single five-stage system 
without the stomach compartment. Working volumes in these vessels are 300 ml for 
stomach and small intestine, 1000 ml for ceacum and ascending colon, 1600 ml for 



240 



Makivuokko and Nurminen 



acid 



Pancreatic juice 




Vessel 1 



Vessel 2 



Vessel 3 



Vessel 4 



Vessel 5 



Figure 2 The original SHIME model. Vessels 1-5 in the figure mimic the different compartments 
of the human GIT: duodenum + jejunum, ileum, caecum + ascending colon, transverse colon and 
distal colon, respectively. In the revised version of this system, a vessel representing the stomach has 
been added before vessel 1. First five pumps work semi-continuously, and pumps between vessels, 
3-5 and effluent work continuously. Source: From Ref. 11. 



transverse colon, and 1200 ml for descending colon. pH is controlled in vessels 2, 3, 4, 5, 
and 6 in the ranges 5.0-6.5, 6.5-7.0, 5.5-6.0, 6.0-6.5 and 6.5-7.0, respectively. 

The system is inoculated by introducing 10 ml supernatant of a human western diet 
suspension per day to the three first vessels for eight successive days. The remaining three 
vessels 4-6 representing the different compartments of the colon are inoculated with 50 ml 
of fecal suspension for 10 successive days. The contents of these three vessels are pumped 
continuously from vessel to vessel and finally to a discard bottle. The transit time of the 
whole system is 84 hours. 

In the beginning of the simulation, 200 ml of fresh SHIME media (11) is added to 
vessel 1 (stomach) three times per day. Every 2-3 hours, the acidic (pH 2.0) contents of the 
first vessel is pumped to vessel 2 (duodenum + jejunum) along with 100 ml of pancreatic 
juice, supplemented with bile, to neutralize the acidity of the gastric effluent. After four 
hours the contents of vessel 2 is pumped to vessel 3 (ileum). 

After eight days of using SHIME media only, the actual test substrate mixed with the 
SHIME media is introduced to the system. Feeding of the substrate is continued for 12 days, 
followed by another SHIME media-only period for 8-10 days. This cycle of three periods is 
repeated for all the studied substrates and samples are taken after each period. 



The EnteroMix R Colon Simulator 

The EnteroMix® model (Fig. 3) has four parallel units each comprising four glass vessels, 
allowing four simulations to be run simultaneously using the same fecal inoculum (12). 
EnteroMix model vessels 1, 2, 3, and 4 have the smallest working volumes (6, 8, 10, and 
12 ml, respectively) of the three models presented here (Table 1). The pH levels in the 
vessels (5.5, 6.0, 6.5, and 7.0, respectively) are similar to the other models. Because of the 
small volumes of vessels, a 40 ml inoculum of 25% wt/vol human feces and only 4 g of 
test substrate is needed for four parallel 48-hour simulations. 

The simulation begins by filling the vessels of each of the four units with 0.9 mM 
anaerobic NaCl (3, 5, 7, and 9 ml to vessels 1, 2, 3, and 4, respectively) and inoculating the 



In Vitro Methods to Model the Gastrointestinal Tract 



241 



N 2 +NH 3 



+37°C 



fe^ fe^ 




+4°C 

Fresh 

medium 






nT~1 



V1 





V2 




V3 



V4 



6 



+4°C 
Effluent 



3 ml 
5.5 



5 ml 
6.0 



7 ml 
6.5 



9 ml < Volume 

7.0 <— PH 



Figure 3 The EnteroMix® model. The figure represents the initial volumes of the system before 
fresh medium is added to begin the simulation. The vessels VI to V4 are mimicking different 
sections of the human colon: caecum + ascending, transverse, descending, and distal colon, 
respectively. pH controlling and semi-continuous fluid transitions are operated via opening and 
closing of computer controlled valves (S). 



first vessel with 10 ml of fecal inoculum. The inoculum is mixed in the vessel with NaCl 
and 10 ml of the mixed culture is pumped to the next vessel. This procedure continues 
through the vessels and finally the excess inoculum is pumped to waste container from the 
fourth vessel. After three hours of the incubation, 3 ml of fresh simulator media with (three 
test channels) or without (one control channel) test substance is pumped to the first vessel. 
The media is fermented in the first vessel for three hours, after which 3 ml of the fermented 
media is transferred to the second vessel, and 3 ml of fresh media is pumped to the first 
vessel. This procedure of transferring liquid to the next vessel continues through all the 
vessels, so that finally after 15 hours, when 3 ml of fermented fluid has been transferred 
from vessel four to the waste container for the first time, vessels 1, 2, 3, and 4 have 
respective volumes of 6, 8, 10, and 12 ml of fermenting fluid. The fermentation and three- 
hourly fluid transfers continue for 48 hours, after which the system is stopped and samples 
are collected from each vessel. 

Other Simulators 

In addition to simulate different parts of the GIT, chemostat-type simulators have also 
been used to simulate the oral cavity, in particular to investigate plaque formation (13); 
and to simulate the urinary bladder to investigate antibiotic sensitivity of urinary tract 
infection-causing pathogens (14). These simulators usually consist of a single 
chemostat. 



Non-Chemostat Models 

The third type of model is actually comprised of two complementary parts, the TIM (TNO 
Intestinal Model) systems 1 and 2 introduced by Minekus et al. in 1995 (15) and 1999 (16). 
The TIM 1 system (Fig. 4) comprises eight sequentially attached glass modules and 
mimics the stomach and small intestine, while the TIM 2-system consists of four glass 
modules in a loop mimicking the proximal colon of monogastric animals (Fig. 5). These 



242 



Makivuokko and Nurminen 




Figure 4 TIM 1 model. The model is mimicking the different sections of the human small 
intestine: the gastric compartment (1), duodenum (2), jejunum (3) and ileum (4). Gastric (5) and 
intestinal secretions (6), peristaltic valve pumps (7) and dialysis devices (8) are also included in this 
simulator. Source: From Ref. 17. 



dynamic models differ from the three previously presented models in two main aspects: 
fluid transportation from vessel to vessel is executed via peristaltic valve-pumps and there 
is a constant absorption of water and fermentation products through dialysis membranes. 
In both systems the peristaltic movement of the intestinal fluid flowing in a flexible tube in 
the middle of the modules is achieved by changing the pressure of the 37°C heated water 
circulating between the module walls and the flexible tube. The peristaltic pressure around 
the flexible tube is controlled via computer-controlled valves to mimic the gastric 
emptying times. For the simulation of intestinal absorption TIM 1 has two integrated 5 
kDa dialysis membranes, after jejunal and ileal modules, and TIM 2 has one, a hollow- 
fiber membrane (molecular mass cut-off value 50 kDa) in the lumen of the system. The 
TIM 1 dialysis membranes allow real-time collection of absorbable metabolites and water 
that would be absorbable in the human jejunum and ileum. In the tube membrane of TIM 2 
circulates dialysis fluid allowing absorption of e.g., water, and short-chain fatty acids. The 
pH-values are monitored in each compartment. 

In a TIM 1 simulation, a homogenized human meal is introduced into the gastric 
compartment in pre- set times. From the stomach, the fluid is pumped through the 
following six compartments. During the simulation, the secretion of enzymes, bile, and 
pancreatic juice and the pH-controlling of the stomach (a pH gradient from 5.0 to 1.8 in 
80 minutes from the beginning) and duodenum (constant pH 6.5) is regulated 
via computer. 

In a TIM 2 simulation the model is first inoculated with 200 ml of fecal inoculum. 
Microbiota is allowed to adapt to the conditions for 16 hours, after which the actual 
simulation is started by adding ileal medium semi-continuously with or without the tested 
substrate to the system. The pH is constantly maintained constant at 5.8 representing the 
pH-level in the proximal colon. Samples can be taken both from the lumen of the simulator 
and from the dialysis liquid during the simulation. 



In Vitro Methods to Model the Gastrointestinal Tract 



243 




Figure 5 TIM 2 model: The model represents the human proximal colon in one loop-shaped 
system: peristaltic mixing with flexible walls inside (a), pH electrode (b), alkaline pump (c), dialysis 
system (d), fluid level sensor (e), nitrogen inlet (f), peristaltic valves (g), sample port (h), gas 
sampling (i) and ileal medium reservoir. Source: From Ref. 18. 



Comparison of the Models 

The four colon simulation models presented here have structural and functional 
differences (Table 1), but the solutions used to reproduce the critical conditions that 
influence the microbiology of the colon are similar in all four models. Firstly the colonic 
microbiota is simulated in each model using fecal samples from a single donor or several 
donors in a pooled sample, because more realistic samples of gastrointestinal tract bacteria 
from the ileum or cecum of humans are very difficult to obtain both ethically and 
technically. Secondly all the colon simulators use similar growth media that originate from 
media originally published by Gibson et al. in 1988 (9) mimicking the ileal fluids obtained 
from sudden-death victims. Thirdly all the colon models have strictly anaerobic 
conditions, similar pH set-points representing the in vivo situation in the colon of healthy 
humans (19) and all the functions of these systems are computer-controlled. 

The Reading model and the SHIME system are both run until a steady state 
in microbial growth is reached, while TIM 2 and the EnteroMix® model are run for 
a pre-determined time (2 or 5 days). The SHIME system is the only one of the above- 
mentioned four systems having a continuous line from stomach to distal colon, thus 
enabling the simulation of the whole Gl-tract in one run. The simulated ileal fluid 
coming from TIM 1 can also be used indirectly as growth medium in TIM 2. The 
EnteroMix® model has the smallest working volumes (Table 1) in the vessels, enabling 
the simulation of small concentrations of the tested substrate. On the other hand the 



244 Makivuokko and Nurminen 

small volumes do not allow any samplings during the simulation run, which is possible 
in all the other models, because the volume of microbiota would be too heavily 
affected in the vessels. The EnteroMix model is also the only model having parallel 
channels in the same simulator allowing four parallel simulations to be run at the same 
time with the same fecal inoculum. 



SIMULATING THE RUMEN 

Although the simulators described above are mainly aimed at simulating the human GIT, the 
models can also be used to simulate the GIT of other monogastric animals. However for the 
simulation of the ruminant GIT different factors have to be taken into consideration; in 
particular the different functioning of the rumen, retaining and fermenting solid material 
while liquid phase is allowed to pass on into the GIT. 

The anaerobic environment of the rumen is heterogeneous in nature: a large volume 
of free liquid, a complex solid mass of digesta, and a gas phase. Within this mixture, the 
diverse microbial population of bacteria, protozoa, and anaerobic fungi can be described 
as occurring in four different compartments (1) the microbes living free in suspension, (2) 
the microbes loosely associated with the solid material, (3) the microbes that are trapped in 
the solid material, and (4) the microbes close to or attached to the rumen wall (20). The 
complexity is still increased due to the different removal rates of the solid and liquid 
portions of rumen contents, revealing the dynamic nature of the rumen. 

Rumen Simulators 

The artificial rumen techniques developed over the past five decades for investigation of 
rumen physiology as well as evaluation of feed rations, have ranged from batch 
fermentations to more complicated continuous incubations. In addition, the absorption 
function of the rumen wall has been included in some designs, in which a semi-permeable 
membrane is applied for removal of the fermentation end products. 

Batch Culture 

The most simplistic, in vitro fermentations representing the rumen were performed in 
different kinds of tubes (21-23). Another way to conduct a static, batch simulation is to use 
closed glass serum bottles. As an example, in the study of Lopez et al. (24) 0.2 g of diet 
(ground to pass through 1 mm screen) was weighed into the 120 ml serum bottles and the 
fermentation process started by dispensing 50 ml of strained, 1 :4 (v/v) buffered rumen 
fluid under C0 2 flushing. The bottles were sealed with butyl rubber stoppers and 
aluminium caps and incubated in a shaking water bath at +39°C. After 24-hour 
incubation, total gas production and pH were measured and samples for methane, 
hydrogen, and short chain fatty acid analysis taken. 

The durations of the reported batch fermentations employing rumen microbes have 
varied from six (25) to 96 hours (26) or even up to 168 hours (27). The buffer systems 
applied in batch simulations are quite often adopted from by Menke et al. (28), McDougall 
(29), or Goering and van Soest (30). 

Due to the fact that gas production has been used as an indirect measure of 
digestibility and fermentation kinetics of ruminant feeds, a scaled glass syringe (volume 
of 100-150 ml) has also been used as a fermentation vessel (28,37). The piston is allowed 
to move upward without restrain and thus indicates the amount of gas released due to 



In Vitro Methods to Model the Gastrointestinal Tract 



245 



microbial activity. The more sophisticated ways to measure gas production kinetics have 
been reported, for example the syringe/electronic pressure transducer-equipment (32), 
which measured and released the accumulated gas. However more automated systems 
were, both an apparatus which combined electronic pressure transducers and electric 
micro- valves (33) and the automated pressure evaluation system (APES) (34) where the 
overpressure was released by use of pressure sensitive switches and solenoid valves. 

Semi-Continuous Culture (Rusitec) 

The structure of semi-continuous rumen simulation technique Rusitec (Fig. 6), which was 
described by Czerkawski and Breckenridge (35), provides three of the four microbial 
compartments mentioned earlier. A Rusitec reaction vessel with capacity of one liter consisted 
of a Perspex cylinder (254 X 76) with an inlet at the bottom. The cylinder was sealed by flat 
Perspex cover provided with a screw flange for easy access. The cover is provided with two 
outlets, one for sampling and the other for effluent overflow and gas collection. The solids 
(feed or digesta) were placed in nylon bags (pore size 50-100 urn) inside a perforated 
container. This "cage" then slid up and down inside the reaction vessel, allowing the effluent to 
flush the solids. At the bottom of the vessel, the artificial saliva (29) was continuously infused 
and the excess liquid and the gases are forced out through an overflow by a slight positive 




Figure 6 A schematic diagram of semi-continuous Rusitec unit: driving shaft (S), sampling valve 
(V), gas-tight gland (G), flange (F), main reaction vessel (R), rumen fluid (L), perforated food 
container (C), nylon gauze bag (N), rigid tube (T), inlet of artificial saliva (I), outlet through overflow 
(O), line to gas-collection bag (M), vessel for collection of effluent (E). Source: From Ref. 35. 



246 



Makivuokko and Nurminen 



pressure in the gas space. The proper fermentation temperature was maintained by incubating 
the reaction vessel in water bath at 39°C during the experiment. 

The fermentation in Rusitec was started by placing solid rumen digesta in one nylon 
bag and an equal amount of feed to be used in a second nylon bag. The reaction vessel was 
filled up to overflow with strained diluted rumen contents. After 24 hours the inoculum 
bag was removed and replaced with a new bag of food. Removal of the oldest bag 
(48 hours) and adding a new bag was repeated each day. At the beginning of the experiment 
and during feeding, the gas space was flushed with the mixture of C0 2 and N 2 (5:95 v/v). 
The removed bag is drained, placed in a plastic bag and the solids washed twice with the 
artificial saliva. This rumination mimicking process includes gentle pressing of the solids 
and squeezing out excess liquid, which is combined and returned to the reaction vessel. 

The Rusitec technique has been quite widely applied as such. It has been used by a 
number of authors to study, for example, decreased methanogenesis (36,37) and efficiency 
of recovery of particle-associated microbes from ruminal digesta (38). In reported Rusitec 
studies at least up to 16 reaction vessels have been applied simultaneously (39). The 
running times of sample collection periods have exceeded from five (40) to 36 days (36) 
after stabilizing the microbial population for 12 hours (39) to 17 days (40). 

Continuous Culture 

One of the earliest reports of continuous culture apparatus (Fig. 7) is the work of Stewart 
et al. (41). With the device designed by Quinn (42) the incubation time could exceed 
more beyond 24 hours because of the pH control system. In these simulation systems the 



timed periodic impulse ^, 
solenoid inflow valve 



stirring motor /^wvent 
float with 
electrical contacts 



» 
» 



^X 



\ sampling 
-V - " device 



relay 
solenoid outflow value H 



IT 




V, 



thermometer 



water 
bath 



constant 

U/ C °2 

pressure 




ice bath 



magnet 




magnetic 

stirring 

motor 



water bath heater 



» 

/ 

i 
i 

i 



outflow receptacle 



Figure 7 One of the earliest continuous culture systems for studying rumen fermentation. Source: 
FromRef. 41. 



In Vitro Methods to Model the Gastrointestinal Tract 



247 




zzzzzszzzzzzzzzzzzzszzzzzzzzzzzzzzz 



Figure 8 A continuous culture apparatus providing absorption of fermentation products: 
centrifugal water pump (A), gas-sampling port (B), fermentor (C), feeding port (D), water-drainage 
pipe (E), Plexiglas reservoir (F), drainage tube (G), magnetic stirrer (H), water bath (I), dialysis sac 
with cation-exchange resin (J), saliva-inflow ground-glass joint (K), fermentor stirring device (L), 
gas-outlet tube (M), fermentor port (N), sampling glass tube and resin holder (O), liquid-effluent 
collection funnel (P), peristaltic pump (Q), effluent outlet (R), effluent rubber tubing (S), saliva-water 
reservoir (T), gas-collection bladder (U), feed-input apparatus (V). Ports D and N are shown 90° out 
of phase from their actual position to simplify the drawing. Source: From Ref. 44. 



water insoluble substrates were continuously delivered to the vessel in the form of a 
slurry. One of the few devices taking the absorption of fermentation end products into 
account was developed by Rufener et al. (43) and improved by Slyter et al. (44). The 
apparatus (Fig. 8) consisted of six independent fermentation chambers (500 ml) with 
accessories providing anaerobiosis, constant volume, agitation of the fermentation 
mixture and collection of effluents and gases. For controlling the pH, this system 
included a dialysis bag containing a mixture of ion-exchange resins, which absorbed the 
short chain fatty acids. The fermentors were reported to reach the steady state in three to 



248 



Makivuokko and Nurminen 



four days of operation. One criterion for this conclusion was the stabilization of 
protozoal numbers even though their density in the vessels was merely 2% of that found 
in the inoculum. 

The dual flow continuous culture system described by Hoover et al. (45) and modified 
later by Crawford et al. (46) and Hannah et al. (47) simulates the differential flows of liquids 
and solids that occur in the rumen. In the design described by Hannah et al. (Fig. 9) (47), the 
mineral buffer solution (48) supplemented with urea is infused to maintain fixed liquid 
dilution rate, and solids retention is regulated by adjusting the ratio of the filtered to overflow 
effluent volumes using a filtering device. Temperature of the vessel is kept constant at 
+ 39°C and pH is adjusted by infusion of 5N HC1 or 5N NaOH. The vessel is constantly 
purged with N 2 to preserve anaerobic conditions and mixing of the fermentation broth is 
performed with magnetic impeller system. The ground and pelleted diet is semi- 
continuously fed to the vessel in eight equal portions over the 24-hour period by use of 
an automated feeder. 

In typical experiments, durations of stabilization periods have varied from five to 
seven days followed by three-day effluent sampling period. Fermentation gases are neither 
collected nor analyzed from this simulation system. Depending on the experiment, 
systems consisting of four (49) to eight (50) glass vessels with a volume of 1.0 (49) to 
1.26 liters (51) have been reported. 



(A) 




(B) 





Figure 9 (A) General schematic of dual flow continuous culture system. (B) Schematic of 
fermenter flask components. A, Automatic feeding device and feed input port; B, magnetic impeller 
assembly; C, sodium hydroxide infusion port; D, hydrochloric acid infusion port; E, filters; F, buffer 
infusion port; G, nitrogen sparger; H, thermocouple assembly; I, coaxial heat exchanger apparatus; 
J, pH electrode; K, overflow port. Source: From Ref. 47. 



In Vitro Methods to Model the Gastrointestinal Tract 249 

Possibilities and Limitations of Rumen Simulation Methods 

The in vitro environmental conditions (temperature, pH, buffering capacity, osmotic 
pressure, dry matter content and oxidation-reduction potential) should represent as closely 
as possible those of the rumen. Irrespective of the technique applied, the quality of the 
inoculum is one of the most important aspects in rumen simulations. In most studies the 
rumen fluid is strained through two, sometimes even four layers of cheesecloth. As a 
result, the inoculum is likely to represent only the microbes occurring in free liquid and a 
major part of the cellulolytic micro-organisms is lost. 

Efforts that can more effectively reproduce the real conditions within the rumen 
will be very useful. Nevertheless the designs may be too complicated for routine and 
easy use: particle block up in the outlet filter or daily opening of the fermentor for 
feeding the microbes prevents the usability. A continuous culture system of two (52) to 
21 (53) reaction vessels with running times of three to four weeks is not a very rapid 
method for analyzing the effects of feed substances on fermentation patterns of rumen 
microbes. The advantage of a batch simulation over continuous one is not only the 
possibility to have more replicates but also the flexibility to test a greater number of 
different treatments simultaneously. 

The duration of the fermentation in closed batch culture should be adjusted 
carefully according to the substrates and cell density to prevent the deprivation and 
inhibitory effects of accumulating metabolites. As a consequence in either case, the 
most fastidious bacteria and protozoa are at risk of being lost. A shorter incubation 
time should be used with substrates that are rapidly fermented. By using actual feed 
components and compositions, the risk of substrate deprivation during simulations is 
reduced. For example, Leedle and Hespell (54) have reported the selective effects of 
single or purified carbohydrates and nitrogen substrates on microbial population. The 
amount of feed should be not only adequate in relation to the microbial density in 
vitro, but also in relation to the calculated total digestive nutrient requirement of the 
host (44). 

The lack of substrates or excess of accumulated end products are, more rarely, the 
reasons for microbial changes in continuous culture systems. Those fermentors, which 
have a uniform and fast turnover rate for the total contents, quickly lose part or all of the 
protozoa. Stabilization of the system for several days will lead to selection and survival of 
those microbes best adapted to that environment. Irrespective of the artificial rumen 
technique, the longer the simulation is run, the greater the difference that will develop in 
the microbial populations compared to the original inoculum. However, a stable 
fermentation that can be maintained long enough to allow microbial adaptation, is 
considered desirable by continuous culture users (36,55). The use of actual feed 
components and compositions presumably assists the maintenance of a representative 
population also in continuous culture systems. 

Although some of the artificial rumen techniques are more superior in taking into 
account the microbial compartments or the different transfer rates of liquids and solids, 
none of them include the activity of bacteria associated with the rumen wall or the 
interaction with the host immune system. It is both challenging and difficult to mimic 
ruminal fermentation and measure the parameters as they actually happen in the rumen. 
The real long-term effects of a test substance on rumen microbes and animal physiology 
can be evaluated neither with a short batch simulation nor with continuous culture 
simulation run for several weeks. Nevertheless, simulation of the rumen in vitro is a 
valuable technique for evaluating particular feed components and testing new diets before 
undertaking animal experiments. 



250 Makivuokko and Nurminen 

CONCLUSION 

Despite the advanced techniques used in the simulators described here, they will remain 
only limited models of the authentic gastrointestinal tract. In particular, the interaction 
between the microbes and the host is absent including contact with the mucosa and the 
intestinal immune system. Some of these issues may be addressed by the use of intestinal 
cell lines, either in the simulator, as a separate loop in the simulator or by using simulator 
effluent. While the latter would remain approximations of the real situation, they would 
nevertheless be very valuable for providing further insight into the dynamics and activity 
of the gastrointestinal microbiota. 



REFERENCES 

1. Zoetendal EG, von Wright A, Vilpponen-salmela T, Ben- Amor K, Akkermans ADL, 
de Vos WM. Mucosa-associated bacteria in the human gastrointestinal tract are uniformly 
distributed along the colon and differ from the community recovered from feces. Appl 
Environ Microbiol 2002; 68:3401-3407. 

2. Ouwehand AC, Salminen S, Arvola T, Ruuska T, Isolauri E. Microbiota composition of the 
intestinal mucosa: association with fecal microbiota? Microbiol Immunol 2004; 48:497-500. 

3. Rumney CJ, Rowland IR. In vivo and in vitro models of the human colonic flora. Crit Rev 
Food Sci Nutr 1992; 31:299-331. 

4. Edwards CA, Eastwood MA. Comparison of the effects of ispaghula and wheat bran on rat 
caecal and colonic fermentation. Gut 1992; 33:1229-1233. 

5. Barry J-L, Hoebler C, Macfarlane GT, et al. Estimation of fermentability of dietary fibre 
in vitro: a European interlaboratory study. Br J Nutr 1995; 74:303-322. 

6. Deprez S, Brezillon C, Rabot S, et al. Polymeric proanthocyanidins are catabolized by human 
colonic microflora into low-molecular-weight phenolic acids. J Nutr 2000; 130:2733-2738. 

7. Oufir LE, Barry JL, Flourie B, et al. Relationships between transit time in man and in vitro 
fermentation of dietary fiber by fecal bacteria. Eur J Clin Nutr 2000; 54:603-609. 

8. Miller TL, Wolin MJ. Fermentation by the human large intestine microbial community in an 
in vitro semicontinuous culture system. Appl Environ Microbiol 1981; 42:400-407. 

9. Gibson GR, Cummings JH, Macfarlane GT. Use of a three-stage continuous culture system to 
study the effect of mucin on dissimilatory sulfate reduction and methanogenesis by mixed 
populations of human gut bacteria. Appl Environ Microbiol 1988; 54:2750-2755. 

10. Macfarlane GT, Macfarlane S, Gibson GR. Validation of a three-stage compound continuous 
culture system for investigating the effect of retention time on the ecology and metabolism of 
bacteria in the human colon. Microb Ecol 1998; 35:180-187. 

11. Molly K, Vande WM, Verstraete W. Development of a 5-step multi-chamber reactor as 
a simulation of the human intestinal microbial ecosystem. Appl Microbiol Biotechnol 1993; 
39:254-258. 

11a. Alander M, De Smet I, Verstraete W, von Wright A, Mattila-Sandholm T. The effect of 
probiotic strains on the microbiota of the Simulator of the Human Intestinal Microbial 
Ecosystem (SHIME). Int J Food Microbiol 1999; 46:71-79. 

12. Makivuokko H, Nurmi J, Nurminen P, Stowell J, Rautonen N. In vitro effects on polydextrose 
by colonic bacteria and Caco-2 cell cyclooxygenase gene expression. Nutr Cancer 2005; 
52:93-103. 

13. Herles S, Olsen S, Afflitto J, Gaffar A. Chemostat flow cell system: an in vitro model for the 
evaluation of antiplaque agents. J Dent Res 1994; 73:1748-1755. 

14. Takahashi S, Sano M, Nishimura M, et al. Bactericidal effect of levofloxacin on strains with 
equal susceptibility in an in vitro urinary bladder model. Chemotherapy 1998; 44:337-342. 



In Vitro Methods to Model the Gastrointestinal Tract 251 

15. Minekus M, Marteau P, Havenaar R, Huis in't Veld HJ. A multicompartmental dynamic 
computer-controlled model simulating the stomach and small intestine. ATLA 1995; 
23:197-209. 

16. Minekus M, Smeets-Peeters M, Bernalier A, et al. A computer-controlled system to simulate 
conditions of the large intestine with peristaltic mixing, water absorption and absorption of 
fermentation products. Appl Microbiol Biotechnol 1999; 53:108-114. 

17. Marteau P, Minekus M, Havenaar R, Huis In't Veld JHJ. Survival of lactic acid bacteria in a 
dynamic model of the stomach and small intestine: validation and the effects of bile. J Dairy Sci 
1997; 80:1033. 

18. Krul C, Humblot C, Philippe C, et al. Metabolism of sinigrin (2-propenyl glucosinolate) by the 
human colonic microflora in a dynamic in vitro large-intestinal model. Carcinogenesis 2002; 
23:1011. 

19. Macfarlane GT, Gibson GR, Cummings JH. Comparison of fermentation reactions in different 
regions of the human colon. J Appl Bacteriol 1992; 72:57-64. 

20. Czerkawski JW. Compartmentation in the rumen, p. 65-82. An Introduction to Rumen Studies. 
Oxford OX3 OB W: Pergamon Press Ltd, 1986. 

21. Morgavi DP, Newbold CJ, Beever DE, Wallace RJ. Stability and stabilization of potential feed 
additive enzymes in rumen fluid. Enzyme Microb Technol 2000; 26:171-177. 

22. Hatfield RD, Weimer PJ. Degradation characteristics of isolated and in situ cell wall 
Lucerne pectic polysaccharides by mixed ruminal microbes. J Sci Food Agric 1995; 
69:185-196. 

23. Susmel P, Spanghero M, Marchetti S, Moscardini S. Trypsin inhibitory activity of raw soya 
bean after incubation with rumen fluid. J Sci Food Agric 1995; 67:441-445. 

24. Lopez S, Valdes C, Newbold CJ, Wallace RJ. Influence of sodium fumarate addition on rumen 
fermentation in vitro. Br J Nutr 1999; 81:59-64. 

25. Gomez JA, Tejido ML, Carro MD. Influence of disodium malate on microbial growth and 
fermentation in rumen-simulation technique fermenters receiving medium- and high- 
concentrate diets. Br J Nutr 2005; 93:479-484. 

26. Blummel M, Karsli A, Russell JR. Influence of diet on growth yields of rumen micro-organisms 
in vitro and in vivo: influence on growth yield of variable carbon fluxes to fermentation 
products. Br J Nutr 2003; 90:625-634. 

27. Ranilla MJ, Carro MD, Lopez S, Newbold CJ, Wallace RJ. Influence of nitrogen source on the 
fermentation of fibre from barley straw and sugarbeet pulp by ruminal micro-organisms 
in vitro. Br J Nutr 2001; 86:717-724. 

28. Menke KH, Raab L, Salewski A, Steingass H, Fritz D, Schneider W. The estimation of the 
digestibility and metabolizable energy content of ruminant feedingstuffs from the gas production 
when they are incubated with rumen liquor in vitro. J Agric Sci Camb 1979; 93:217-222. 

29. McDougall EO. Studies on ruminant saliva I. The composition and output of sheep's saliva. 
Biochem J 1948; 43:99. 

30. Goering KH, Van Soest PJ. Forage fiber analysis (apparatus, reagents, procedures, and some 
repplication). Agricultural Handbook. Washington D.C.: Agricultural Research Counsil, 
U.S. Department of Agriculture, 1970. 

31. Wallace RJ, Wallace SJ, McKain N, Nsereko VL, Hartnell GF. Influence of supplementary 
fibrolytic enzymes on the fermentation of corn and grass silages by mixed ruminal 
microorganisms in vitro. J Anim Sci 2001; 79:1905-1916. 

32. Theodorou MK, Williams BA, Dhanoa MS, McAllan AB, France J. A simple gas production 
method using a pressure transducer to determine the fermentation kinetics of ruminant feeds. 
Animal 1994;48:185-197. 

33. Cone JW, van Gelder AH, Visscher GJW, Oudshoorn L. Influence of rumen fluid and substrate 
concentration on fermentation kinetics measured with a fully automated time related gas 
production apparatus. Animal 1996; 61:113-128. 

34. Davies ZS, Mason D, Brooks AE, Griffith GW, Merry RJ, Theodorou MK. An automated 
system for measuring gas production from forages inoculated with rumen fluid and its use in 
determining the effect of enzymes on grass silage. Animal 2000; 83:205-221. 



252 Makivuokko and Nurminen 

35. Czerkawski JW, Breckenridge G. Design and development of a long-term rumen simulation 
technique (Rusitec). Br J Nutr 1977; 38:371-384. 

36. Wallace RJ, Czerkawski JW, Breckenridge G. Effect of monensin on the fermentation of basal 
rations in the Rumen Simulation Technique (Rusitec). Br J Nutr 1981; 46:131-148. 

37. Dong Y, Bae HD, McAllister TA, Mathison GW, Cheng K-J. Lipid-induced depression of 
methane production and digestibility in the artificial rumen system (RUSITEC). Canadian 
J Anim Sci 1997; 77:269-278. 

38. Ranilla MJ, Carro MD. Diet and procedures used to detach particle-associated microbes from 
ruminal digesta influence chemical composition of microbes and estimation of microbial 
growth in Rusitec fermenters. J Anim Sci 2003; 81:537-544. 

39. Wang Y, McAllister TA, Rode LM, et al. Effects of an exogenous enzyme preparation on 
microbial protein synthesis, enzyme activity and attachment to feed in the Rumen Simulation 
Technique (Rusitec). Br J Nutr 2001; 85:325-332. 

40. Russi JP, Wallace RJ, Newbold CJ. Influence of the pattern of peptide supply on microbial 
activity in the rumen simulating fermenter (RUSITEC). Br J Nutr 2002; 88:73-80. 

41. Stewart DG, Warner RG, Seeley HW. Continuous culture as a method for studying rumen 
fermentation. Appl Microbiol 1961; 9:150-156. 

42. Quinn LY. Continuous culture of ruminal microorganisms in chemically defined medium I. 
Design of continuous-culture apparatus. Appl Microbiol 1962; 10:580-582. 

43. Rufener WH, Nelson WO, Jr., Wolin MJ. Maintenance of the rumen microbial population in 
continuous culture. Appl Microbiol 1963; 11:196-201. 

44. Slyter LL, Nelson WO, Wolin MJ. Modifications of a device for maintenance of the rumen 
microbial population in continuous culture. Appl Microbiol 1964; 12:374-377. 

45. Hoover WH, Crooker BA, Sniffen CJ. Effects of differential solid-liquid removal rates on 
protozoa numbers in continuous cultures of rumen contents. J Anim Sci 1976; 43:528-534. 

46. Crawford RJ, Jr., Shriver BJ, Varga GA, Hoover WH. Buffer requirements for maintenance of pH 
during fermentation of individual feeds in continuous culture. J Dairy Sci 1983; 66:1881-1890. 

47. Hannah SM, Stern MD, Ehle FR. Evaluation of a dual flow continuous culture system for 
estimating bacterial fermentation in vivo of mixed diets containing various soya bean products. 
Animal 1986; 16:51-62. 

48. Weller RA, Pilgrim AF. Passage of protozoa and volatile fatty acids from the rumen of the 
sheep and from a continuous in vitro fermentation system. Br J Nutr 1974; 32:341-351. 

49. Karunanandaa K, Varga GA. Colonization of rice straw by white-rot fungi (Cyathus 
stercoreus): effect on ruminal fermentation pattern, nitrogen metabolism, and fiber utilization 
during continuous culture. Animal 1996; 61:1-16. 

50. Mansfield HR, Stern MD, Otterby DE. Effects of beet pulp and animal by-products on milk 
yield and in vitro fermentation by rumen microorganisms. J Dairy Sci 1994; 77:205-216. 

5 1 . Hoover WH, Kincaid CR, Varga GA, Thayne W V, Junkins LL, Jr. Effects of solids and liquid flows 
on fermentation in continuous cultures IV. pH and dilution rate. J Anim Science 1984; 58:692-699. 

52. Fellner V, Sauer FD, Kramer JK. Steady-state rates of linoleic acid biohydrogenation by 
ruminal bacteria in continuous culture. J Dairy Sci 1995; 78:1815-1823. 

53. Newbold CJ, Wallace RJ, Chen XB, Mcintosh FM. Different strains of Saccharomyces 
cerevisiae differ in their effects on ruminal bacterial numbers in vitro and in sheep. J Anim Sci 
1995;73:1811-1818. 

54. Leedle JAZ, Hespell RB. Brief incubations of mixed ruminal bacteria: effects of anaerobiosis 
and sources of nitrogen and carbon. J Dairy Sci 1983; 66:1003-1014. 

55. Jayasuriya MCN, Hamilton R, Rogovic B. The use of an artificial rumen to assess low quality 
fibrous feeds. Biol Wastes 1987; 20:241-250. 



14 



Animal Models for the Human 
Gastrointestinal Tract 



Anders Henriksson 

DSM Food Specialties, Sydney, Australia 



INTRODUCTION 

Scientific research is continuously generating new ingredients for food and pharmaceu- 
tical products. In pace with consumer awareness of healthy products, considerable efforts 
are made to find new ingredients with beneficial effects on human health. The health 
benefits of such ingredients need to be assessed in human trials prior to being developed as 
a product for wider human consumption. Animal trials, conducted prior to human trials, 
offer a sound filtering system that provide the opportunity to identify those ingredients that 
are worthy of the relatively costly human studies that may follow. Animal models are 
important tools used in the study of human gastrointestinal (GI) microbiology. 
Specifically, animal models are used when considering the effect of food and 
pharmaceutical ingredients on GI health and disease. These effects include the metabolic 
and immunological activities of microorganisms that colonize the human gastrointestinal 
tract (GIT). 

This chapter deals with issues related to the use of animal models in studies of human 
GI microbiota or specific microorganisms of human origin. It discusses similarities and 
differences between human and animal physiology and microbiota with specific focus 
on categories of animal models. The following discussion focuses predominantly on 
rodents and highlights some limitations and opportunities that relate to categories of 
rodent models such as "germ- free," "human flora associated" and "surgically or chemically 
modified." 



Physiology and Microbiology of the GI Tract 

Physiology 

The human GIT is the most appropriate environment to conduct studies on the human GI 
microbiota but for practical reasons animal models are used extensively for these types of 
studies. The wide range of similarities between the animal and human GIT makes it 
possible to draw reasonable parallels between these two hosts, however, results from 
studies on the human microbiota in animals may not entirely reflect processes occurring in 

253 



254 



Henriksson 



the human GIT. The reason for this is that there are also many differences between human 
and animal gut physiology, diets, and behavior. Rodents are the most extensively used 
animals in the research of human GI microbiota. The differences between the human and 
rodent GIT may be important in interpreting any research findings. 

When considering the differences between the human and rat GITs, the issue of size 
is certainly obvious. This difference has impact on transit time of GI contents. Also, the 
rate of passage can vary between the type of diet, the particle size of digesta and 
morphological characteristics of the GITs. In rats the transit time is 12-35 hours 
depending on the type of diet and transit markers used (1,2). In humans, native Africans, 
consuming a traditional diet, have an average GI transit time of 33 hours, which is 
approximately half of the transit time that has been observed in Europeans or Africans on a 
Western diet. 

Many more subtle and potentially important morphological and physiological 
differences exist between the human and rat GITs. An example of this exists in the fact that 
the adult human appendix, known to be the undeveloped caecum, does not correspond in 
function to the developed, functioning rodent ceacum. The adult human GIT is roughly 
divided into three major regions, namely, the stomach, small intestine and the large 
intestine (colon). In the human fetus the caecum commences deveploment as a conical 
diversion. As the rest of the intestine grows, caecal growth is arrested and a vermiform 
appendix remains. In adult humans, the colon, which is haustred throughout its entire 
length, takes the shape and function of the caecum which is found in many other animals 
(Fig. IB) (3). 

The mouse and rat GIT is divided into four major regions, namely the stomach, 
small intestine, caecum, and colon (Fig. 1A). In contrast to the human stomach, the 




NON-SECRETING REGION 



STOMACH 



SMALL INTESTINE 



-CAECUM 



COLON 



APPENDIX 




(A) 



(B) 



Figure 1 The mouse (A) and human (B) gastrointestinal tracts. 



Animal Models for the Human Gastrointestinal Tract 



255 



stomach of rats and mice have a large area of nonsecreting epithelium that expands 
considerably as the animals are eating. In rodents, microbial fermentation is mainly 
occurring in the caecum. The colon of these animals is not haustred and is less important 
for microbial fermentation, compared to the caecum. The large intestine of these animals 
is important for re-absorption of water and formation of fecal pellets. 



Microbiota 

The physiological properties of the human GIT, with its many unique features provide a 
vast number of microbial niches. Host factors such as enzymes, mucins, proteases, bile 
acids dietary factors and regimes contribute to this diversity. The result is a complex 
microbial community composed of several hundred microbial species (4) that collectively 
form the GI microbiota. The mammalian GI microbiota forms dense microbial 
populations, particularly in the posterior part of the intestine (5). The composition of 
both human and rodent microbiota has been extensively investigated and discussed 
in several comprehensive studies and reviews (6-8). The microbial profiles of rodents 
such as rats and mice are in many ways similar to that of other mammals, including 
humans (5,9). In such rodents, lactobacilli are present in levels of 10 9 colony forming units 
(CFU) per gram of feces, (5) whereas in humans, the average levels of fecal lactobacilli are 
usually 10 4 -10 6 CFU per gram of feces (10). As described by Finegold et al. (10), diet has 
impact on population levels of lactobacillus and other microbial groups in humans. 
Bifidobacteria may be detected in both human (10) and rodent feces (11), however 
commercial rodent feed may not support GIT colonization by bifidobacteria as much as 
some other diets (Fig. 2). This suggests that the type of diet should be considered carefully 
to ensure that the diet used supports the colonization of important microbial groups. The 
effect of feed composition is further discussed in the section "Conventional Animals." 

There are also behavioral differences between various animal species that may 
contribute to the resulting GI microbiology of these animals. Rodents are known as 
coprophages, and unless coprophagy is prevented, it is possible that the GIT of these 
rodents are continuously re-inoculated with their own fecal microorganisms. This 
behavior, which could possibly affect the microbial profile, may be inhibited by fitting a 
tail cup which makes the fecal pellets unavailable to the animals (13). Other techniques 
have been attempted, such as keeping animals on a grid to allow fecal pellets to fall 
through and become inaccessible, however coprophagic animals, including rats, usually 




HAS 



Type of feed 




Normal 



Figure 2 Fecal bifidobacteria of mice (Balb/C) fed high amylomaize starch (HAS) diet, 
containing 40% starch [AIN 76 (12)] and a commercial rodent feed (Normal). Results presented are 
the average + SDV of six animals per group. 



256 Henriksson 

collect fecal pellets as they are extruded from anus (14), making such a grid less efficient in 
preventing coprophagy. 

The relative importance of coprophagy, and specifically the rate of microbal 
re-inoculation, has been investigated in a number of studies. The rat may consume 35-50 
percent of the total output of feces, or an even larger proportion if the rat is on a vitamin 
depleted diet (14). It has been reported that prevention of coprophagy has reduced 
weight gains in rats and also caused major changes in caecal and fecal lactobacilli, 
enterococci, and coliforms (15). In another report, prevention of coprophagy made no 
change in GI microbial profiles, apart from a minor decrease in lactobacilli of the stomach 
and the lactobacilli of the small intestine (16). A study conducted by Smith (5) indicated 
that coprophagy has no, or minor effects on gastric microbial populations. These studies, 
whilst showing dramatically varying conclusions, possibly resulting from varying feed 
and housing conditions, indicate that coprophagic behavior should remain an 
important consideration. 

The Role of Microbiota on GI Health 

The mammalian microbiota has several important functions. It aids in nutrition by 
degrading complex nutrients and by synthesizing vitamins. It protects against infectious 
disease, by either preventing invading pathogenic bacteria from establishing in the GIT, or 
by conditioning of the mucosal immune system. The microbiota may also influence the 
development of cancer, by modulation of carcinogens, pre-carcinogens or by activation of 
immunological responses. 

Many factors influence the progression and severity of GI infectious disease. Some 
examples of this are seen in the interaction between various microorganisms and also in 
their interaction with dietary factors and the host. A pathogen entering the GIT will meet 
resistance by the microbiota. An invading pathogen is also faced with the host' s immune 
system as well as host factors such as stomach acids, bile acid and enzymes. 

The GI microbiota plays an important role in activation of the innate immune system 
(17-19). Mucosal immune responses are activated as a result of microorganisms 
interacting with the gut associated lymph tissue (GALT). Interaction of microbes and 
antigens with GALT leads to a cascade of responses as outlined in the chapter by Moreau. 
The host mucosal immune system is important in preventing a pathogen from invading the 
GIT and the translocation of a pathogen to both the mesenteric lymph nodes (MLN) and 
the internal organs (20-22). The intestinal microbiota and orally administrated probiotics, 
prebiotics, and other nutrients may also affect the balance of Thl/Th2 cell response, and 
the production of pro and anti-inflammatory cytokines (23,24). The oral administration of 
probiotics to rodents may activate macrophages (25) and natural killer (NK) cells (26), in a 
similar fashion to when they are administered to humans (27). There are a number of 
described animal models that make research on human GI microbiota possible and bring to 
light the effects of the human microbiota on nutrition, immunology, and resistance against 
infections and other diseases (Table 1). 



ANIMAL MODELS USED FOR STUDIES 
ON THE HUMAN GI MICROBIOTA 

Administration Feed and Test Material to the Animal GIT 

The effect of specific agents, such as pro and prebiotics or specific chemicals, on the GI 
microbiota and gut health is monitored after administration of these agents to experimental 



Animal Models for the Human Gastrointestinal Tract 



257 



d 

•1-H 

•c 
O 

d 

cd 

a 

d- 

O 
co 

£ 

CO 
'S 

cd 
OX) 

»H 

c 
c 
'— 
o 



o 

Cd 

o 

cj 

a. 

oo 

i- 
c 

cd 



43 
O 

'— 

o 



d 

cd 

s 

CD 

43 



cj 

C 



d 

CD 

o 



d 
_o 

"43 
o 
CD 

13 

< 



.Q 

(0 

H 



o 
o 

d 

CJ 

»H 

CD 



CO 



O 
Oh 

H 



S3 
O 


CD 


d 
cj 


CO 


-*— » 


bu 


0) 


>^ 


cd 


-a 


X) 


fTl 


p 

•4— » 


-d 


O 

• «— « 


CO 


CD 
O 


s 


d 





0) 


•l-H 


T3 


43 


T3 


d 


O 


0) 

CO 

13 


. ■ — i 

CD 

CO 


d 

• i-H 


CO 


CD 


I— -H 


CO 


u 



u 
O 

Ph 1) 

d ^ 

cd £» 

.9 § 

cd CD 



00 



d 

CD 
T3 

O 



00 0\ O h (S 

(N cn <n en cn 



en rj- *o \o t> 

en m m m m 



ooaNOeN^om'sj-invo ^j- r- 
cnen^Tj-eN^Tj-TtTt cn ^f 



co 

•l-H 
•4-H 

•c 

43 

03 
T3 

co CD 

a ° 

O T3 

3 .g 
2 d 

Oh <L> 

>. S> 

43 =3 



O 



CO 



CO 

• i-H 
CO 

o 

-d 



.9 -S 

« o 

a a 

Oh <h-h 

o ° 

> O 

> ,P. 



d 
O 

• i-H 
■4-J 

cd 
O 

o 

> 03 

8 5 

.a *£ 

P CD 

OX) -" 

03 <4-h 
43 O 

Oh 

d 
O 

•i-H 

co .O 

•"H _d 

- d 



d 

o 







ft 

CO 

■*—• 

CO 

d 



2 -9 •§ 



o 

Sh 
Oh 

<H-H 
O 



S-H 

a 

co 
cj 






CJ 

.1) 



43 

O 

*H 

P3 Oh 



03 

bp 

03 

O „ 
O g 

•tf O 
<•> d 

- J 

O ^3 
»-i 

Oh 



< < < < < 

^ z z ^ z 



X ^ 
rn P3 

U Q 



u u u 

j-o ^ i 

o3 o3 o3 

PQ CQ CQ 



<D 1) 1) <D (D 
o o CJ o o 



IT: 

I 

CO 

o 

■d 

o 

•l-H 

2 
o- 

o 

•l-H 

o 

o- 

co 

>> 

d 
o 

•l-H 
-4— < 

o 

<H-H 

d 

•l-H 

13 
d 

o 



d 

. -H 

03 

^H 

-*— ' 

O 



O 



T3 
< 

z 

co Q 

° ^ 

•z3 d 



c3 



c3 



Oh -tn 

5T.a 

O-fi 



03 

oo 

■>—• 
CO 

d 

•l-H 

03 

bp 

03 
d 

•2 o 

O •--• 
(D O 

O ?h 

Ph pj 



o 

d 

13 

43 
H 

43 

H 

d 

c 

co 

"-4— > 

c 
o 

•— 

Oh 



O 

o -t 1 bp 

CX2 >> o3 

o o 5 

^ °- 

^ B 



d 

c 



03 

d 43 O 

o o "S 

•a s -g 

c ^J d 

•a cu d 

>^> 

cu 



Q 



F Oh 
» d 

o 






d 






a o 

d -ih 



c3 



4^ 



c3 

d 

•l-H 

■*-< 

CO 

cu 



d 

c3 



S 3 o 

03 j3 



c O- 43 

O 'H l *H 

•13 O O 

O ■(-» -w 

CD O O 

"^ CD CD 

2 te ^ 

Ph PJ PJ 



< < < < 

z z z z 



^ Si -J % 

£ £ £ U 

13 Id 13 vo 

PP Ph PQ Ph 



P^ 



CD CD CD CD CD 
O O CJ O O 



53 
o 



CJ 
T3 



co Q 



O 
tj 

T3 
CD 

■*— » 
03 



d 

CJ 



d 



T3 

CJ 






Oh 
^O 

13 
> 

CD 
T3 



tq -h 



d 
O 

• i-H 
-4— ' 

c3 



^H 

.o 



co 

d 

•l-H 

03 
W) 

03 

d 
O 

•l-H 

■*— » 

CJ 

CJ 
■*— » 

O 

•— 
O- 

a 

co 

♦«» 

Q 

co 

C3 



co 

CJ 
43 
O 
s- 
CJ 



CJ 

o Q 

cd PQ 

C-H M 

W <4H<4H 

«H-. o O 



d 

. -H 

03 

^H 
-4— ' 

CJ 



43 
O 

«H 

Oh 
O 

• i-H 

•F Oh 
> CO 



CJ 

c3 



43 

d 

O 



5 
— — 

"ft 



CJ 

c3 

CJ 



=3 g 43 -a 



CQ 



CJ 

" CJ 



CD 



ft 



43 

• S 33 fe3 



d 

CJ 



CJ 

d 

o 



0) 

"3 

l-H 



d 

CJ 

B 



d 

CJ 



CJ 


03 


03 


n 


43 


CJ 


CJ 


d 


-*— > 


l-H 


s- 




■*— ' 


■*— ' 


CD 


M-H 








o 


l-H 


fH 


i- 


,o 


,o 


,o 


*—• 


<-i-H 


«+H 


'-I-. 



£ i 

CD 43 
O c3 

S C/3 

03 

■4-J 

co 

d 



03 
43 

-4—' 

d 

•l-H 

03 
H 

CO 

CJ 



.cd 



d 

CJ 



43 



CJ 



CO 



d -^ 

CO Q 

co .y 

CD o 

CO 'q 

^ 2 

< Ph 



CO 
CJ 



43 

O 

•— 

Ph 



03 
03 



co d 

.-- o 

S '^ 

O o 

•53 CD 

4J ■(-> 

O O 

S^H 1-4 

Ph Ph 



O S ° 

-o 2 ^ 

2 2 ° 



d 

o 



d 

I- 2 

03 X) 



Cj 03 

O 



o 



CJ 



O 

l-H 



V 



X 
m 

U 

d 

03 



ex: 



CJ 
CJ 

d 
4h 

13 

43 



co 

• i-H 

o 

43 
03 



CN ^3 

d 

o 



cj 

c^ 
03 

d 



43 

H 

d 

CO co 

° 2 

•-H S-H 

s 



CO 



or 
CJ 



N 

d 

CJ 

•4-J 

CO 

o 

43 

co 


O 

• -H 

l-H 

03 
> 

d 

o 

CJ 
43 



« ^32 



O 

43 

O 

l-H 

Oh 



_o3 

O 
d 



l-H 

03 



r : "H fl) 

^ CO .s 
O 73 



CJ 
CJ 

d 

CJ 

d 

to *5 

PJ P3 



CJ 
.CJ 



CJ 

'bb 



CJ 

PJ 









CJ 




















d 




















CJ 




















l-H 




















43 














u 






-4—* 

d 
03 














< 






'o 














< 






43 










co 










CJ 










CJ 




t 
















• i-H 
-4—> 

o 




z 




4-H 


-4— » 

CD 










• i-H 

43 




a 

HH 


< 


< < °° 

\ < oo 


S 


< 


< 


< 


oo 
oo 


^< 


< 


z 


Z Z Q 


i 

m 


Z 


z 


z 


Q 


< z 


z 



CN 
i 



o o o 



Z Z 

5 5 § y 7 

53 S3 ffi < £ d 

S S S S CN CN K 43 c3 ^ 



^o 



m 



-J 

CP 

>n ._ 

U £ 



53 

43 

co 



m 

53 

43 

co 

• i-H 

Ph 



CJCDCDCDCDCDCDCDCD 
OCJCJOOCJOCJCJ 



SSSSS SSSSS SSSSSSSSS 



CJ 




CJ w 


■4— > 


^ cd 


cd 


^ P< 


Ph 



^3 
S 

a 



258 



Henriksson 



"^3 

+— 
S 

c 

• y—t 

W) 

• y—t 

S-H 

o 

d 

g 



CO 



CO 

. -H 

d 

o 
o 

O 



o 

4d 

• i-H 

o 

cj 

ft 

O0 

S-i 

O 

cd 



O 

S-H 

u 



d 

g 
<D 

s: 

c 
o 

CO 
<D 

• i-H 

-a 

d 
4— i 

oo 

c 

• i-H 

o 

CO 
CO 

13 
O 



C 
<D 

O 

4-H 

o 

c 
o 

•1-H 

4— I 

cj 

13 

OO 



-Q 

re 



o 

CJ 

c 
o 

HH 

o 



CO 



ft 
>^ 

H 



<D d 

-d <U 

X) 



O 

• i-H 

£ 

C O 

a 



T3 

CJ 

o 

=3 



CD 
CO 

cd 

CD 

CO 



O 



s- 

o 

c3 & 

S § 

cd CD 

H &Q 



OO 



c 

O 



on o i— i (N en ^t 
^t m io wo m in 



o 












,o 












4h 












*— i 












a 












>^ 












S-i 












CJ 












4-H 












c 




d 






c 


c3 




o 






O 


b 




•I-H 






• i-H 




4— > 






4-H 


CJ 




cd 






C3 


X 




o 






CJ 


cd 




O 






O 


4-h 




CO 






CO 


O 




d 


S-H 

CJ 


>> 


C 


c 


cd 


K3 


o 


• ^H 


cd 


• rH 


S-H 


» « 


S-H 


o 


s- 


■*— < 


•l-H 


4-1 


• i-H 

4-H 

i 

s 

42 

C 


d 

X 


CJ 
Oh 


13 
• -H 

S-H 

CJ 
4-H 

CJ 

C3 

X) 


c3 
O 

o 

CJ 

d 


CJ 

E 

s- 

CJ 

4-H 


13 

•c 

CJ 

4-J 

CJ 

cd 


o 


xl 


4-H 


• i-H 


3 


4-H 


— ( 


4-h 


O 


>. 


bfi 


O 


CD 


c 


d 




-a 


c 


, 


o 


13 


CJ 


o 


d 


cd 


• i-H 
4— < 


o 


> 


• i-H 

J — 1 


c3 

d 
CJ 

o 

W) 

cd 

S— < 


CO 

S-H 

CJ 

> 

CJ 

s- 


•I-H 

• i-H 

■§ 

• i-H 


• i-H 

B 

CJ 

-d 

CJ 


c 

s- 

e 

•1-H 


• i-H 

X) 

• i-H 

q 

• i-H 


s- 


i-H 


i-H 


u 


S-H 


s 


O 


,o 


o 


o 


,o 


4-H 


4-H 


4-H 


4-H 


4-H 


O 


CO 


CO 


co 


■CO 


co 


4-h 


O 


CJ 


O 


CJ 


CJ 


O 


• I-H 


• i-H 


• i-H 


• I-H 


• i-H 


4-» 


4-H 


4—1 


4-» 


4-H 


4- ' 


o 


o 


o 


o 


o 


CJ 
CJ 


• l-H 


• i-H 


• i-H 


• i-H 


• i-H 


_C 


X 


X 


pO 


X 


ft- 1 


o 


o 


o 


o 


o 


4—1 


S- 


(h 


S-H 


s- 


S-H 


w 


&h 


ft 


Oh 


Qh 


Oh 



CJ 

d 

si 

4-H 

CJ 

E 

o 

N 

< 



CJ 

4—1 

cd 



60 

X 



O Z Q 



oo 

z 



Z 



>^ >. >> 

0) CD CD 



en 

S-H 

o 

si 

CO 



cd 

Q 

i 

CJ 

3 



cd 

Q 

i 

CJ 

3 



Q 

i 

CJ 

d s-H 



SD t)i) bD c3 

c3 cd cd ti 

J-* S-h Vh .22 

Dh Ch ft 



S-H 

cd 

4-H 

co 



Ph oo oo oo 



^^ 



cd cS cd cd cd A 
P< rt 0^ P< C^ P< 



13 

S-i 

o 
o 



13 
o 

S-i 

3 

13 



d 

■a 

o 

SOh 

§■ 

4J 

o 

d 




Animal Models for the Human Gastrointestinal Tract 259 

animals. In this type of studies, the following should be considered: (1) Type of animal 
feed, (2) Administration of microorganisms (e.g., pathogenic bacteria or probiotics), 
carcinogens or inflammatory agents, (3) Assessment of animal health and properties of the 
GI microbiota. 

Feed 

The effect of specific dietary components are most conveniently assessed after feeding 
animals a feed containing these compounds. There are several basic feed formulations that 
may be used for this purpose. The feed which was described by Rickard et al. (12) and 
modifications thereof (55) are suitable for administration of probiotics. The composition 
of animal diets may have a significant impact on the composition and activity of the GI 
microbiota. These effects are further discussed in the section on Human Flora Associated 
Animals. 

Microorganisms 

Administration of microorganisms to the GIT of animals can be performed in several 
ways. Gavage is a method in which microorganisms may be inoculated directly into the 
stomach of an animal using a gastric probe (33,56). This method allows a known volume 
containing pathogen, probiotic cultures or complex microbial mixtures to be injected into 
the stomach. Animals can also be inoculated by administering feed or water containing 
microorganisms. However, administration through water or feed may not allow for a 
known inoculum size to be administered at a specific time. In order to avoid this issue, 
animals can be left for a short time without water or feed to ensure that the contaminated 
substrate is consumed without delays (43). Alternatively, the microorganisms can be given 
to animals in a sucrose solution, to improve the rate of consumption (57). 

Carcinogens and Inflammatory Agents 

Cancer and inflammation may be induced by exposure of animals to specific agents. 
Administration of carcinogens, pre-carcinogens or pro-inflammatory agents may be 
introduced orally, by gavage or by feeding animals feeds containing the specific agents. A 
desired effect may also be induced by intrarectal or systemic inoculation of specific agents. 

Conventional Animals 

Conventional (CV) animals, being those which have a natural occurring microbiota, may 
be used to stimulate the human GIT. However, due to differences between the human and 
animal microbiota, the results from studies in CV animals may be quite different from 
corresponding studies conducted in humans. CV animal models are useful in studies on 
orally administrated human microorganisms in vivo, where the activities of the indigenous 
microbiota are acceptable. CV rodent models have been used for studies of probiotic 
cultures and particularly their effect on infectious disease. CV animals may also be used to 
assess the survival of probiotic cultures in vivo. Although the GI conditions of CV animal 
models are quite different from those of the human, the conditions of the animal GIT are most 
likely closer to human than what can be simulated in vitro. 

There are several examples of studies where CV animals have been used to assess the 
protective effects of probiotics. These include studies in which Salmonella (32,56,58-60), 
E. coli (61,62) and Listeria (63,64) have been used as model pathogens. Specific Pathogen 
Free (SPF) mice have been useful in similar studies where animals were given single or 



260 Henriksson 

mixed cultures that were considered to be probiotic, before being challenged with 
Salmonella. The progress of infection is determined by monitoring (1) translocation of 
pathogen to internal organs (31,43), (2) change in animal body weight, and (3) mortality 
(65) following the challenge. Out of these three general methods, monitoring changes in 
animal body weight is convenient and relevant in most cases. The virulence of a model 
pathogen is relevant in this regard, since the virulence will affect the progress of infection. A 
too virulent strain may induce an unnecessarily severe infection (66). In other cases, human 
pathogens may not colonize, infect or give a demonstrable effect in an animal model (64,67). 
If this is the case, then a human pathogen may be replaced with a strain known to be virulent 
in animals. Examples given so far relate to models used for the monitoring of GI infection 
and translocation to areas such as MLN and intestinal organs. The protective effect of 
probiotic cultures can also be monitored by assessing the clearance rate of a specific 
pathogen from the feces of animals challenged with that pathogen (65). The clearance rate of 
Listeria was measured in the feces of animals that were fed probiotic cultures and meat 
starter cultures in order to identify specific probiotic cultures that eliminated this particular 
pathogen (64). 

CV animals may also be used as a model system to assess the survival and 
colonization of probiotic cultures and other microorganisms of human origin. The 
survival, during passage through the GIT, can be monitored as long as appropriate 
methods of detection are available. Traditional culturing methods have been important 
tools used in monitoring the survival of probiotic cultures during GI transit (68-70). 
Recent years have seen other more efficient detection methods such as molecular probes, 
which have been developed for the accurate assessments of population sizes of particular 
probiotic cultures in feces (71). Probes of this type may be used to confirm the identity of 
particular probiotic strains (72-74) and detect specific strains even at very sparse 
population levels (75). 

GI microbiota is obviously important for the biochemical profile of the GIT. However, 
simply identifying the survival of microorganisms in feces gives little information about the 
details of their activity in the GIT. In vivo investigations into the activity of particular 
microorganisms require methods other than those used for that of detection. Mice or rats 
may be used to characterize the activity of cultures at specific sites throughout the GIT, 
something that is very difficult to assess in humans. Traditionally this type of study has 
been conducted on animals containing microorganisms of interest by describing the 
biochemical profile of the animal's GI contents. This methodology is adequate in instances 
where the sum of all microbial and host activities are investigated at the time of sampling. 
However, it is less appropriate if the activities of specific microorganisms are assessed, 
where these microorganisms are a part of a complex microbial system. In vivo studies on 
the activities of specific cultures in a complex ecosystem require different animal models. 
The development of a lactobacillus free mouse model has provided the opportunity to 
study the effects of lactobacillus colonization on host physiology, including the effects on 
fecal bile acids and enzyme activities (76-78). This type of model may be used in studies 
on the effect of human lactobacillus strains if animals are colonized by strains of 
human origin. 

Germ-Free Animals 

Under normal conditions animals are exposed to microorganisms during birth and 
continue to be exposed to a wide range of microorganisms throughout their lives. These 
microorganisms form the microbiota, characteristic to CV animals. Hysterectomy at birth, 
allows the unborn fetus to be transferred from the womb to a sterile chamber. If this 



Animal Models for the Human Gastrointestinal Tract 261 

process is carried out under sterile conditions, the animal would not be contaminated with 
microorganisms from the environment. High hygiene standards are required to ensure that 
animals are maintained and bred under germ-free conditions. 

Considerable achievements have been made since the 1970s in investigating the role 
of the gut microbiota using germ-free animals. Germ-free animals have enabled investi- 
gation of animal gut physiology in the absence of the gut microbiota. Studies using germ- 
free animals have revealed that the gut microbiota is indeed of tremendous importance for 
the biochemical properties of the GIT, by metabolizing compounds in ingested feed and 
host factors of mucosal and pancreatic origin. Data from these studies revealed that many 
physiological and biochemical features of the GIT are indeed the result of microbial gut 
activity (79,80). The gut of germ-free animals have different physiological and 
biochemical properties to that of CV animals. The biochemical properties of germ-free 
and CV animals are often regarded as either germ-free associated characteristics (GAC) or 
microbiota associated characteristics (MAC). The characteristics of MAC and GAC are 
described in the chapter by Norin and Midtvedt. 

Germ-free animals provide the opportunity to investigate the role of specific 
microorganisms in the GIT. These microorganisms and their impact on host physiology, 
can be monitored in an environment that is unaffected by a preexisting microbiota. 
Ex-germ-free animals have also been used to study the interaction between a controlled 
composition of microbial species in the GIT (81-84). Germ-free animals have also been 
useful in research that focuses on the role of the GI microbiota in metabolism of host 
factors such as mucin and bile acids (85,86). More recently, germ-free technology has 
been extensively used in research on the effect of specific strains on host immunology 
(87,88). mucosal physiology and morphology (89-91). Although the absence of a diverse 
microbiota enables characterization of specific microbes, it cannot be used to characterize 
their activity in a complex microbial environment. Therefore, animals associated with one 
or a limited number of strains, may not truly reflect the microbial activity of those that 
harbor a CV gut ecosystem. 

Human Flora-Associated Animals 

The establishment of human fecal microbes within animals, provides the opportunity for 
the study of a microbiota of human origin within these animals. Human flora associated 
animals (HFA) have proven to be particularly valuable in studies of the metabolic and 
immunological activities of the human microbiota. Athough HFA animals are valuable for 
investigations related to the human microbiota, several differences between animal and 
human physiology may influence colonization by the human microbiota in animal hosts. 
Such differences may promote host-specific colonization by microorganisms in different 
animals (92,93). As a result, microbes of human origin may be disadvantaged in the animal 
GIT, compared to isolates originating from this particular animal species. 

HFA animals are created by inoculating germ-free animals with a human fecal 
homogenate (94). The resulting microbial profile of HFA animals is partly dependent on 
the differing ability of the various microorganisms in the human fecal sample to colonize 
the animal GIT. Previous studies have shown that certain microorganisms of human fecal 
origin were unable to colonize the rodent GIT (95). There may be several reasons for this, 
such as diets or host factors like transit times and physiological conditions. It has been 
demonstrated that mice, fed with a commercially available animal feed, may have a 
reduced, or even undetectable level of bifidobacteria in feces. However, after feeding these 
mice an alternative diet for several weeks, bifidobacteria could be detected in the mice that 
were fed sucrose or amylose, with particularly dense populations of bififodobacteria 



262 Henriksson 

observed in mice that were fed with an amy lose rich diet (Fig. 2). This suggests that diet, 
and specifically dietary ingredients such as certain carbohydrates, are important for the 
composition of the GI microbota and that previously nondetectable microbial groups may 
be stimulated to detectable levels. Consideration may be given to the possibility that the 
growth of microbal populations due to dietary intervention, may be at the expense of less 
competitive microbial groups. 

The colonization of human originated bifidobacteria within germ-free animals is not 
always successful (95). Hiramaya and co-workers (96) demonstrated that in rodents, the 
source of fecal material containing bifodobacteria influences the ability of bifodobacteria 
to colonize the GIT (95). This may be due to the fact that bifodobacteria from different 
sources possess different natural characteristics. The activity of the human source 
microbiota that is contained within HFA animals may also be dependent on the cultural 
origin and dietary habits of the human source (97,98). For instance, fecal material obtained 
from different human donors has been shown to provide a different degree of effectiveness 
in protection against Salmonella (32). Although this type of model provides a good tool for 
studying the effects of the human microbiota, it cannot be assumed that the microbial 
profile of HFA animals is identical to that of the human donor. 

HFA rodents are useful in studies of the metabolic activity of the human microbiota. 
The effects of microbiota on the metabolism of lignans and isoflavones have been 
investigated in studies using germ-free and HFA rats (98). In similar studies, HFA rats 
have been used to assess the metabolism of dietary fats (99,100). The usefulness of HFA 
animals has also been illustrated in studies such as those conducted on the effect of 
complex carbohydrates on the human microbiota, including the effect of resistant starch 
(97). Other studies include those relating to the production of short chain fatty acids 
(85,101) and microbial enzyme activities (102). HFA animals are also valuable for 
toxicological studies. There are several examples of studies in which HFA animals have 
been used to assess the effect of the human microbiota on potentially carcinogenic 
compounds (47,103). Interestingly, both studies indicated that the source of fecal material 
used to create HFA rats influenced the transformation of pre-carcinogens to 
carcinogenic componds. 

Oozeer and colleagues (28) used a genetically modified L. casei strain to assess 
whether the strain was active throughout the passage of the intestinal tract of HFA mice. 
This strain was modified by the introduction of genes coding for erythromycin resistance 
and luciferase. Results from this study indicate that this strain is both metabolically active 
and able to initiate new protein synthesis during its transit through the GIT. Techniques in 
transcriptomics and metabolomics are paving the ways for new studies on microbial 
activity of the gut contents and detailed studies of biochemical properties of host cells 
lining the GI epithelium. 

Surgically Modified Animals 

Surgical modification of the GIT gives new opportunities for the study of the GI 
microbiota. Using surgical procedures, specific parts of the GIT can be removed in order to 
make modifications to basic physiology. Surgery can also provide the opportunity, by 
means of cannulation of the GIT, to give repeated post-surgical access to specific sites of 
the tract. The human GIT lacks some of the areas that may be found in the rodent and 
porcine GIT, such as the areas of non-secreting epithelium that are found in the stomachs 
of rodents and pigs. It has been suggested that these areas are the primary sites for 
Lactobacillus colonization within such animals, and that bacterial populations contained 
at these sites are in fact seeding the intestinal tract with lactobacillus (104,105). If this was 



Animal Models for the Human Gastrointestinal Tract 263 

to be correct, removal of the non-secreting stomach region could result in a gastric 
microbial profile that is more in line with that of the human. However, surgical removal of 
the non-secreting stomach region has no effect on the luminal levels of lactobacillus in 
either the stomach or in the colon of mice (A. Henriksson, unpublished observations). 
Therefore, it can be assumed that in mice and possibly other animals, this region is not 
responsible for the relatively dense lactobacillus populations found in either the intestinal 
contents or the stomach itself. 

The caecum is an important part of the rodent intestine for microbial fermentation. 
This stands in contrast to the human GIT where the colon is the major site for such 
fermentation. It has been suggested that this difference is another factor that contributes to 
various differences between the microbial profiles of rodents and humans (106). However, 
studies indicate that the microbial biochemical profile of caecectomized mice remains 
significantly different from that of normal humans or mice (106,107). Most studies on 
rodents with surgically modified GIT have failed to give microbial profiles that closely 
resemble that of the human GIT. 

Cannulation is performed to provide access to specific sites of the GIT in order to 
facilitate collection of microbiological samples. Cannulated animals are equipped with a 
port from which samples can be taken at one or several sites along the GIT. Cannulation 
has been performed on dogs, pigs, and other larger animals (108-110). This technology 
has been valuable in assessing the microbial and enzymatic properties of specific areas 
within the GIT. 

Gene Deficient Animals 

In recent years, specific mouse strains have been frequently used in studies of colitis. Colitis 
in mice closely resembles human inflammatory bowel disease (IBD). There are several 
specific inbred mouse strains that are most useful in this area as they are more likely to 
develop spontaneous colitis. Strains displaying a disrupted expression of Interleukin (IL)-2, 
IL-10, and TGF-P have proven to be particularly useful in these studies and have 
contributed to a broader understanding of the role of the human GI microbiota in IBD. 
There are a number of different characteristics associated with animals that express 
irregular cytokine profiles. In mice that are deficient in IL-2, usually when 6-15 weeks 
old, inflammation occurs in the colon only (111). However, in IL-10 deficient mice, 
inflammation may also occur in the small intestine as well as the colon (112). TCRa 
deficient animals have developed inflammation in the caecum, colon, and rectum 
(113-115), whereas HLA-B27 rats develop inflammation in the colon, duodenum, and 
caecum (1 16). These "knock out" models may be used to investigate the effect of the human 
microbiota, both in terms of the aggravating, as well as alleviating effects on IBD (117). 
Immune deficient animals have also been useful in studies relating to the effects 
of probiotic cultures on colitis. Probiotic cultures investigated in IL-10 deficient mice 
include L. salivarius, Bifidobacterium lactis (1 17). IL-10 knock out mice with colitis have 
also been used to investigate the effect of a genetically modified (GM) Lactococcus lactis 
that synthesizes IL-10 (42,118). 

Chemically Induced Responses 

Animals that have been intentionally exposed to specific pro-inflammatory or 
carcinogenic chemicals have been used in studies on the role of microbiota in the 
development of both cancer and IBD (Table 1). Cancer, or other malignant abnormalities 
in the gut mucosa, may be induced by the oral administration of carcinogens. Examples of 



264 Henriksson 

such carcinogens are 1, 2 dimethylhydrazine (DMH), and N-methyl-N'-nitro-Nitrosogua- 
din (MNNG). These types of models, which are based on either CV or HFA animals, have 
also been used to assess the effect of both probiotics and prebiotics on the progression of 
cancer in its various stages from DNA damage through to differentiation of tissue and 
formation of tumors. 

A study by Mclntoch and co-workers (52) investigated the effect of L. acidophilus 
on the incidence of tumor formation as well as the mass of tumors found in animals that 
had been challenged with DMH. It was demonstrated that the animals that had been given 
L. acidophilus were associated with less tumors than those animals that were given other 
probiotic cultures. As a result the most effective culture, in terms of protecting animals 
against cancer, could be isolated out of a range of LAB cultures. 

Another way of investigating the effect of microbiota on the formation of cancer is to 
assess the occurrence of aberrant crypt foci in the intestinal epithelium. In this type of 
model, increased occurrence of aberrant crypts indicate increased formation of tumors. 
This model has been used to assess the effect of GI microbiota and specific dietary factors 
on the development of intestinal cancer (48,119). In similar studies, animals given 
azoxymethane were used to assess the effect of L. casei, of human origin, on the formation 
of aberrant cells (120). Other studies using 3-methylcholanthrene to induce tumor 
formation, demonstrated that the same strain delayed the onset of tumor formation. It was 
suggested that this delay was due to an enhancement of cytotoxicity of NK cells (26). 
Finally, mucosal carcinogenesis may be assessed by determination of the DNA adduct 
formation (121). This type of methodology allows assessment of carcinogenesis without 
visual scoring of aberrant crypts. This method has been successfully used to investigate the 
effects of human intestinal flora on the mutagenicity of dietary factors by assessing DNA 
adduct formation (36). Assessment of DNA adduct formation has been used as a tool in 
investigating the protective effect of potentially probiotic cultures against the formation of 
cancer (122,123). This type of model provides a cost-effective tool used in studies on the 
GI microbiota and its role in formation of intestinal cancers. 

Although both animal models have been used to demonstrate protection against 
cancer by probiotic cultures, the difference between how cancer that has developed in the 
chemically modified animal and how it has developed in the diseased human subject raises 
questions as to what extent such observations are relevant for the human host. The 
opportunity to test probiotic cultures in humans that have been intentionally exposed to 
carcinogens does not exist. However, it is known that some of the probiotic cultures that 
reduce the incidence of tumor formation in animals have a similar effect on cancer in 
humans (124,125). 

Apoptosis is a mechanism inherent to healthy mucosal cells, which ultimately leads 
to the death of cancerous cells. The effect of various dietary factors on apoptosis can be 
assessed in animal models. Several studies have investigated the effect of probiotics and 
prebiotics on apoptosis. Some of these studies have revealed that prebiotics such as Fructo- 
Oligosaccharides (FOS) and inulin increase incidence of apoptosis and thereby provide 
increased protection against the formation of intestinal cancers (126). 

A wide range of animal models have been applied to studies on IBD. Naturally 
occurring animal models have been important tools in studies related to human ulcerative 
colitis and Crohn's disease. IBD-like symptoms have also been induced chemically. The 
application of such chemicals may induce ulceration of the intestinal mucosa as well as 
several immunological responses that are typical to IBD in humans. Simple methods for 
T-cell induced onset of IBD may be initiated by di-nitro chlorobenzene (DNCP) as 
described by Glick and Falchuk (127). This method involves both systemic and local 
application of DNCP. Other chemically induced forms of IBD may be induced by intra 



Animal Models for the Human Gastrointestinal Tract 265 

rectal inoculation of trinitrobenzene sulphonic acid (TNBS) which is dissolved in alcohol. 
The latter treatment results in inflammation that lasts for several weeks after exposure to 
these agents (128). Animals not treated with this agent are normally tolerant to sonicates 
derived from the heterologus intestine of syngenic littermates (BsH). However, in animals 
with IBD induced by TNBS, both local and systemic tolerance to BsH is broken (129). 
Interestingly, this study also demonstrated that tolerance to BsH was abrogated by 
treatment with IL-10 or antibodies to IL-12. 

A study using oral therapy with a probiotic culture had no effect on either the 
severity of colitis or gut permeability in this TNBS model (53). Similarly, oral therapy 
with L. rhamnosus and a mixture of probiotic cultures has been shown to reduce the extent 
of colonic damage in TNBS induced colitis (130). However, both L. rhamnosus and the 
culture mixture significantly ameliorated colitis induced by idoacetamide (130). These 
studies indicate that inflammation induced by a sulfhydryl blocker (e.g., idoacetamide), as 
described by Rachmilewitz and co-workers (128), may be a better model for assessing the 
effect of gut microorganisms on colitis. 



CONCLUSION 

Animal models provide opportunities to investigate the effect of food and pharmaceutical 
ingredients on GI health and the human microbiota in vivo. A wide range of methods that 
use animal models have been described, including those based on CV, germ-free, and HFA 
animals. CV animal models are particularly suitable for studies on the effect of orally 
dosed probiotic strains, or other microorganisms of human origin, on resistance against 
infection and aberrant formations in the GI mucosa. Germ-free animals provide 
opportunities to create HFA animals that are suitable for studies on the effect of the 
total human microbiota in vivo. HFA animals have been used extensively in studies on 
the role of the human microbiota in nutrition and metabolism of nutrients. The effect of the 
microbiota on the immune system can be investigated in chemically modified animals, or 
specific immune deficient "knock out" models. These models have been used in studies on 
the effect of the human microbiota and probiotic cultures on the progress of IBD and other 
diseases that may be caused by a dysfunctional immune system. In addition, chemically 
modified animals have been used in studies on the effect of probiotic cultures on the 
development of tumors and other aberrant formations. The usefulness of animals in studies 
on human microbiota and its effect on GI health has a long standing and clear value. In an 
age where virtual in vivo simulations are becoming increasingly important, it remains 
clear that animal models will continue to be highly valuable in research on the functions of 
the human microbiota and activity of specific microbial strains of human origin. 



REFERENCES 

1 . Luick BR, Penner MH. Nominal response of passage rates to fiber particle size in rats. J Nutr 
1991; 121:1940-1947. 

2. Sakaguchi E, Itoh H, Uchida S, Horigome T. Comparison of fibre digestion and digesta 
retention time between rabbits, guinea-pigs, rats and hamsters. Br J Nutr 1987; 58:149-158. 

3. Stevens CE. Comparative Physiology of the Vertebrate Digestive System. New York: 
Cambridge University Press, 1988. 

4. Savage DS. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol 1977; 
31:107-133. 



266 Henriksson 

5. Smith WH. Observations on the flora of the alimentary tract of animals and factors affecting 
its composition. J Pathol Bacteriol 1965; 89:95-122. 

6. Tannock GW. Normal microbiota of the gastrointestinal tract of rodents. In: Mackie RI, 
White BA, Isaacson RE, eds. Gastrointestinal Microbiology. New York: Chapman & Hall, 
1997 pp. 187-215. 

7. Holdeman LV, Good IJ, Moore WEC. Human fecal flora: variation in bacterial composition 
within individuals and a possible effect on emotional stress. Appl Environ Microbiol 1976; 
31:359-375. 

8. Moore WEC, Holdeman LV. Human fecal flora: the normal flora of 20 Japanese-Hawaiians. 
Appl Microbiol 1974; 27:961-979. 

9. Drasar BS, Barrow PA. Intestinal Microbiology. Washington: Van Nostrand Reinhold, 1985. 

10. Finegold SM, Attebery HR, Sutter VL. Effect of diet on human fecal flora: comparison of 
Japanese and American diets. Am J Clin Nutr 1974; 27:1456-1469. 

1 1 . Mitsuoka T. Taxonomy and ecology of the indigenous intestinal bacteria. Recent Advances in 
Microbial Ecology. Tokyo: Japan Scientific Societies Press, 1989 pp. 493-498. 

12. Rickard KL, Folino M, Mclntyre A, Albert V, Muir J, Young GP. Colonic epithelial biology; 
comparison of resistant starch to soluble and insoluble dietary fibres. Processing Nutr Soc 
1994; 18:57. 

13. Barnes RH, Fiala G, McGehee B, Brown A. Prevention of coprophagy in the rat. J Nutr 1957; 
63:489-528. 

14. Barnes RH. Nutritional implications of coprophagy. Nutr Rev 1962; 20:289-291. 

15. Fitzgerald RJ, Gustafsson BE, McDaniel EG. Effects of coprophagy prevention on intestinal 
microflora in rats. J Nutr 1964; 84:155-160. 

16. Smith HW, Jones JET. Observations on the alimentary tract and its bacterial flora in healthy 
and diseased pigs. J Pathol Bacteriol 1963; 86:387-412. 

17. Cross ML. Microbes versus microbes: immune signals generated by probiotic lactobacilli and 
their role in protection against microbial pathogens. FEMS Immunol Med Microbiol 2002; 
34:245-253. 

18. Schiffrin EJ, Blum S. Interactions between the microbiota and the intestinal mucosa. Eur 
J Clin Nutr 2002; 56:S60-S64. 

19. Borchers AT, Keen CL, Gershwin ME. The influence of yogurt/ Lactobacillus on the innate 
and acquired immune response. Clin Rev Allergy Immunol 2002; 22:207-230. 

20. Berg RD, Owens WE. Inhibition of translocation of viable Escherichia coli from the 
gastrointestinal tract to the mesenteric lymph nodes and other organs in a gnotobiotic mouse 
model. Infect Immun 1979; 25:820-827. 

21. Berg RD. Inhibition of Escherichia coli translocation from the gastrointestinal tract by normal 
cecal flora in gnotobiotic or antibiotic-decontaminated mice. Infect Immun 1980; 29: 1073-108 1 . 

22. Berg RD. Promotion of the translocation of enteric bacteria from the gastrointestinal tracts of 
mice by oral treatment with penicillin, clindamycin, or metronidazole. Infect Immun 1981; 
33:854-861. 

23. Perdigon G, Maldonado Galdeano C, Valdez JC, Medici M. Interaction of lactic acid bacteria 
with the gut immune system. Eur J Clin Nutr 2002; 56:S21-S26. 

24. Sudo N, Yu XN, Aiba Y, et al. An oral introduction of intestinal bacteria prevents the 
development of a long-term Th2-skewed immunological memory induced by neonatal 
antibiotic treatment in mice. Clin Exp Allergy 2002; 32:1112-1116. 

25. Perdigon G, Nader ME, Alvarez MS, Oliver G, Holgado AAPD. Effect of perorally 
administered lactobacilli on macrophage activation in mice. Infect Immun 1986; 53:404-410. 

26. Takagi A, Matsuzaki T, Sato M, Nomoto K, Morotomi M, Yokokura T. Enhancement of 
natural killer cytotoxicity delayed murine carcinogenesis by a probiotic microorganism. 
Carcinogenesis 2001; 22:599-605. 

27. Gill HS, Rutherfurd KJ, Cross ML. Dietary probiotic supplementation enhances natural killer 
cell activity in the elderly: an investigation of age-related immunological changes. J Clin 
Immunol 2001; 21:264-271. 



Animal Models for the Human Gastrointestinal Tract 267 

28. Oozeer R, Goupil-Feuillerat N, Alpert CA, et al. Lactobacillus casei is able to survive and 
initiate protein synthesis during its transit in the digestive tract of human flora-associated 
mice. Appl Environ Microbiol 2002; 68:3570-3574. 

29. Kato I, Endo-Tanaka K, Yokokura T. Suppressive effects of the oral administration of 
Lactobacillus casei on the type II collagen-induced arthritis in DBA/1 mice. Life Sci 1998; 
63:635-644. 

30. Gill HS, Rutherfurd KJ. Viability and dose-response studies on the effects of the 
immunoenhancing lactic acid bacterium Lactobacillus rhamnosus in mice. Br J Nutr 2001; 
86:285-289. 

31. Gill HS, Shu Q, Lin H, Rutherfurd KJ, Cross ML. Protection against translocating Salmonella 
typhimurium infection in mice by feeding the immuno-enhancing probiotic Lactobacillus 
rhamnosus strain HN001. Med Microbiol Immunol 2001; 190:97-104. 

32. Henriksson A, Conway PL. Colonization resistance induced in mice orogastrically dosed with 
human fecal homogenates from different donors. Microbiol Ecol Health Dis 2001; 13:96-99. 

33. Henriksson A, Conway PL. Isolation of the human fecal bifidobacteria which reduce signs of 
Salmonella infection when orogastrically dosed to mice. J Appl Microbiol 2001; 90:223-228. 

34. Cross ML, Mortensen RR, Kudsk J, Gill HS. Dietary intake of Lactobacillus rhamnosus 
HNOOl enhances production of both Thl and Th2 cytokines in antigen-primed mice. Med 
Microbiol Immunol 2002; 191:49-53. 

35. Mahoney M, Henriksson A. The effect of processed meat and meat starter cultures on 
gastrointestinal colonization and virulence of Listeria monocytogenes in mice. Int J Food 
Microbiol 2003; 84:255-261. 

36. Pestka JJ, Ha C-L, Warner RW, Lee JH, Ustunol Z. Effects of ingestion of yoghurts containing 
Bifidobacterium and Lactobacillus on spleen and peyer' s patch lymphocyte populations in the 
mouse. J Food Prot 2001; 64:392-395. 

37. Hirayama K, Baranczewski P, Akerlund JE, Midtvedt T, Moller L, Rafter J. Effects of human 
intestinal flora on mutagenicity of and DNA adduct formation from food and environmental 
mutagens. Carcinogenesis 2000; 21:2105-2111. 

38. Waidmann M, Bechtold O, Frick JS, et al. Bacteroides vulgatus protects against Escherichia 
co//-induced colitis in gnotobiotic interleukin-2-deficient mice. Gastroenterology 2003; 
125:162-177. 

39. Balish E, Warner T. Enterococcus fecalis induces inflammatory bowel disease in interleukin- 
10 knockout mice. Am J Pathol 2002; 160:2253-2257. 

40. Schultz M, Veltkamp C, Dieleman LA, et al. Lactobacillus plantarum 299V in the treatment 
and prevention of spontaneous colitis in interleukin-10-deficient mice. Inflamm Bowel Dis 
2002; 8:71-80. 

41. McCarthy J, O'Mahony L, O'Callaghan L, et al. Double blind, placebo controlled trial of two 
probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. 
Gut 2003; 52:975-980. 

42. Steidler L, Hans W, Schotte L, et al. Treatment of murine colitis by Lactococcus lactis 
secreting interleukin- 10, [comment]. Science 2000; 289:1352-1355. 

43. Hudault S, Lievin V, Bernet-Camard MF, Servin AL. Antagonistic activity exerted in vitro 
and in vivo by Lactobacillus casei (strain GG) against Salmonella typhimurium C5 infection. 
Appl Environ Microbiol 1997; 63:513-518. 

44. Silva SH, Vieira EC, Dias RS, Nicoli JR. Antagonism against Vibrio cholera by diffusible 
substances produced by bacterial components of the human fecal microbiota. J Med Microbiol 
2001;50:161-164. 

45. Li Z, Yang S, Lin H, et al. Probiotics and antibodies to TNF inhibit inflammatory activity and 
improve nonalcoholic fatty liver disease, [comment]. Hepatology 2003; 37:343-350. 

46. Konrad A, Mahler M, Flogerzi B, et al. Amelioration of murine colitis by feeding a solution of 
lysed Escherichia coli. Scand J Gastroenterol 2003; 38:172-179. 

47. Rouzaud G, Rabot S, Ratcliffe B, Duncan AJ. Influence of plant and bacterial myrosinase 
activity on the metabolic fate of glucosinolates in gnotobiotic rats. Br J Nutr 2003; 
90:395-404. 



268 Henriksson 

48. Roland N, Nugon-Bandon L, Flinois P, Beaune P. Hepatic and intestinal cytochrome P-450, 
glutathione-transferase and UDP-glucuronosyl transferase are affected by six types of dietary 
fiber in rats inoculated with human whole fecal flora. J Nutr 1994; 124:1581-1587. 

49. Tache S, Peiffer G, Millet AS, Corpet DE. Carrageenan gel and aberrant crypt foci in the colon 
of conventional and human flora-associated rats. Nutr Cancer 2000; 37:193-198. 

50. Sidhu H, Allison MJ, Chow JM, Clark A, Peck AB. Rapid reversal of hyperoxaluria in a rat 
model after probiotic administration of Oxalobacter formigenes. J Urol 2001 ; 166: 1487-1491 . 

51. Adawi D, Ahrne S, Molin G. Effects of different probiotic strains of Lactobacillus and 
Bifidobacterium on bacterial translocation and liver injury in an acute liver injury model. Int 
J Food Microbiol 2001; 70:213-220. 

52. Mcintosh GH, Royle PJ, Playne MJ. A probiotic strain of L. acidophilus reduces DMH- 
induced large intestinal tumors in male Sprague-Dawley rats. Nutr Cancer 1999; 35:153-159. 

53. Kennedy RJ, Hoper M, Deodhar K, Kirk SJ, Gardiner KR. Probiotic therapy fails to improve 
gut permeability in a hapten model of colitis. Scand J Gastroenterol 2000; 35:1266-1271. 

54. Eizaguirre I, Urkia NG, Asensio AB, et al. Probiotic supplementation reduces the risk of bacterial 
translocation in experimental short bowel syndrome. J Pediatr Surg 2002; 37:699-702. 

55. Wang X, Brown IL, Evans AJ, Conway PL. The protective effects of high amylose maize 
(amylomaize) starch granules on the survival of Bifidobacterium spp. in the mouse 
gastrointestinal tract. J Appl Microbiol 1999; 87:631-639. 

56. Hudault S, Lievin V, Bernet-Camard MF, Servin AL. Antagonistic activity exerted in vivo 
and in vitro by Lactobacillus casei (strain GG) against Salmonella typhimurium C5 infection. 
Appl Environ Microbiol 1997; 63:513-518. 

57. Cohen PS, Rossoll R, Cabelli VJ, Yang S-L, Laux DC. Relationship between the mouse 
colonizing ability of a human fecal Esherichia coli strain and its ability to bind a specific 
mouse colonic mucous gel protein. Infect Immun 1983; 40:62-69. 

58. Paubert-Braquet M, Xiao-Hu G, Gaudichon C, et al. Enhancement of host resistance against 
Salmonella typhimurium in mice fed a diet supplemented with yogurt or milks fermented with 
warious Lactobacillus casei strains. Int J Immunother 1995; XI: 153-161. 

59. Shu Q, Lin H, Rutherfurd KJ, et al. Dietary Bifidobacterium lactis (HN019) ennhances 
resistance to oral Salmonella typhimurium infection in mice. Microbiol Immunol 2000; 
44:213-222. 

60. Bovee-Oudenhoven IM. Increasing the intestinal resistance of rats to the invasive pathogen 
Salmonella enteritidis: additive effects of dietary lactulose and calcium. Gut 1997; 40:497-504. 

61. Shu Q, Gill HS. Immune protection mediated by the probiotic Lactobacillus rhamnosus 
HN001 (DR20) against Escherichia coli 0157:H7 infection in mice. FEMS Immunol Med 
Microbiol 2002; 34:59-64. 

62. Shu Q, Gill HS. A dietary probiotic (Bifidobacterium lactis HN019) reduces the severity of 
Escherichia coli 0157:H7 infection in mice. Med Microbiol Immunol 2001; 189:147-152. 

63. DeWaard R, Garssen J, Bokken GCAM, Vos JG. Antagonistic activity of Lactobacillus casei 
strain Shirota against gastrointestinal Listaria monocytogenes infection in rats. Int J Food 
Microbiol 2002; 73:93-100. 

64. Mahoney M, Henriksson A. The effect of processed meat and meat starter cultures on 
gastrointestinal colonization and virulence of Listeria monocytogenes in mice. Int J Food 
Microbiol 2001; 84:255-261. 

65. Bernet-Camard MF, Lievin V, Bassart D, Neeser JR, Servin AL, Hudault S. The human 
Lactobacillus acidophilus strain LAI secrets a nonbacteriocin antibacterial substance(s) 
active in vitro and in vivo. Appl Environ Microbiol 1997; 63:2747-2753. 

66. Voravuthikunchai SP. Investigation of Caecectomy as a Perturbation of the Mouse Intestinal 
Ecosystem. Sydney: The University of New South Wales, 1988. 

67. Babour AH, Rampling A, Hormaeche CE. Variation in the infectivity of Listeria 
monocytogenes isolates following intregastric inoculation of mice. Infect Immun 2001. 

68. Yuki N, Watanabe K, Mike A, et al. Survival of a probiotic, Lactobacillus casei strain Shirota, 
in the gastrointestinal tract: selective isolation from feces and identification using monoclonal 
antibodies. Int J Food Microbiol 1999; 48:51-57. 



Animal Models for the Human Gastrointestinal Tract 269 

69. Saxelin M, Elo S, Salminen S, Vapaatalo H. Dose response colonization of feces after oral 
administration of Lactobacillus easel strain GG. Microb Ecol Health Dis 1991; 4:209-214. 

70. Goldin BR, Gorbach SL, Saxelin M, Barakat S, Gualtieri L, Salminen S. Survival of 
Lactobacillus species (strain GG) in human gastrointestinal tract. Dig Dis Sci 1992; 
37:121-128. 

71. Charteris WP, Kelly PM, Morelli L, Collins JK. Selective detection, enumeration and 
identification of potentialy probiotic Lactobacillus and Bifidobacterium species in mixed 
bacterial populations. Int J Food Microbiol 1997; 35:1-27. 

72. Lynch PA, Gilpin BJ, Sinton LW, Savill MG. The detection of Bifidobacterium adolescentis 
by colony hybridization as an indicator of human fecal pollution. J Appl Microbiol 2002; 
92:526-533. 

73. Lucchini F, Kmet V, Cesena C, Coppi L, Bottazzi V, Morelli L. Specific detection of a 
probiotic Lactobacillus strain in fecal samples by using multiplex PCR. FEMS Microbiol Lett 
1998; 158:273-278. 

74. Kaneko T, Kurihara H. Digoxigenin-labeled deoxyribonucleic acid probes for the 
enumeration of bifidobacteria in fecal samples. J Dairy Sci 1997; 80:1254-1259. 

75. Su P, Tandianus JE, Park JH, Henriksson A, Dunn NW. A colony hybridization method for 
Bifidobacterium lactis LAFTI B94. Aust Dairy Technol 2003; 58:213. 

76. Tannock GW, Crichton C, Welling GW, Koopman JP, Midtvedt T. Reconstitution of the 
gastrointestinal microflora of lactobacillus-free mice. Appl Environ Microbiol 1988; 
54:2971-2975. 

77. Tannock GW, Dashkevicz MP, Feighner SD. Lactobacilli and bile salt hydrolase in the 
murine intestinal tract. Appl Environ Microbiol 1989; 55:1848-1851. 

78. McConell MA, Tannock GW. Lactobacilli do not influence enzyme activities of duodenal 
enterocytes of mice. Microb Ecol Health Dis 1993; 6:315-318. 

79. Midtvedt T, Gustafsson BE. Microbial convesion of bilirubin to urobilin in vitro and in vivo. 
Acta Pathol Microbiol Scand Sect B 1981; 89:57-60. 

80. Norin KE, Midtvedt T, Gustafsson BE. Influence of intestinal microflora on the tryptic activity 
during lactation in rats. Lab Anim Sci 1986; 20:234-237. 

81. Corthier C, Dubos F, Ducluzeau R. Prevention of Clostridium difficile induced mortality in 
gnotobiotic mice by Saccharomyces boulardii. Can J Microbiol 1986; 32:894-896. 

82. Rodrigues ACP, Nardi RM, Bambirra EA, Vieira EC, Nicoli JR. Effects of Saccharomyces 
boulardii against oral infection with Salmonella typhimurium and Shigella flexneri in 
conventional and gnotobiotic mice. J Appl Bacteriol 1996; 81:251-256. 

83. Wilson KH, Sheagren JN, Freter R, Weatherbee L, Lyerly D. Gnotobiotic models for study of 
the microbial ecology of Clostridium difficile and Escherchia coli. J Infect Dis 1986; 
153:547-551. 

84. Kabir AM, Aiba Y, Takagi A, Kamiya S, Miwa T, Koga Y. Prevention of helicobacter pylori 
infection by lactobacilli in a gnotobiotic murine model. Gut 1997; 41:49-55. 

85. Kleessen B, Hartmann L, Blaut M. Oligofructose and long-chain inulin: influence on the gut 
microbial ecology of rats associated with a human fecal flora. Br J Nutr 2001; 86:291-300. 

86. Chikai T, Nakao H, Uchida K. Deconjugation of bile acids by human intestinal bacteria 
implanted in germ-free rats. Lipids 1987; 22:669-671. 

o 

87. Herias MV, Hessle C, Telemo E, Midtvedt T, Hanson LA, Wold AE. Immunomodulatory 
effects of Lactobacillus plantarum colonizing the intestine of gnotobiotic rats. Clin Exp 
Immunol 1999; 116:283-290. 

88. Scharek L, Hartmann L, Heinevetter L, Blaut M. Bifidobacterium adolescentis modulates the 
specific immune response to another human gut bacterium, Bacteroides thetaiotaomicron, in 
gnotobiotic rats. Immunobiology 2000; 202:429-441. 

89. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Annu Rev Nutr 2002; 22:283-307. 

90. Husebye E, Hellstrom PM, Sundler F, Chen J, Midtvedt T. Influence of microbial species on 
small intestinal myoelectric activity and transit in germ-free rats. Am J Physiol Gastrointest 
Liver Physiol 2001; 280:G368-G380. 



270 Henriksson 

91 . Kleessen B, Hartmann L, Blaut M. Fructans in the diet cause alterations of intestinal mucosal 
architecture, released mucins and mucosa-associated bifidobacteria in gnotobiotic rats. Br 
J Nutr 2003; 89:597-606. 

92. Lin JH-C, Savage DC. Host specificity of the colonization of murine gastric epithelium by 
lactobacilli. FEMS Microbiol Lett 1984; 24:67-71. 

93. Conway PL, Henriksson A. Strategies for the isolation and characterization of functional 
probiotics. Human Health: The Contribution of Microorganisms. London: Springer- Verlag, 
1993 pp. 75-94. 

94. Mallett AK, Bearne CA, Rowland IR, Farthing MJ, Cole CB, Fuller R. The use of rats 
associated with a human fecal flora as a model for studying the effects of diet on the human gut 
microflora. J Appl Bacteriol 1987; 63:39-45. 

95. Hirayama K. Ex-germfree mice harboring intestinal microbiota derived from other animal 
species as an experimental model for ecology and metabolism of intestinal bacteria. Exp 
Anim 1999; 48:219-227. 

96. Hirayama K, Miyaji K, Kawamura S, Itoh K, Takahashi E, Mitsuoka T. Development of 
intestinal flora of human-flora-associated (HFA) mice in the intestine of their offspring. Exp 
Anim 1995; 44:219-222. 

97. Silvi S, Rumney CJ, Cresci A, Rowland IR. Resistant starch modifies gut microflora and 
microbial metabolism in human flora-associated rats inoculated with feces from Italian and 
U.K. donors. J Appl Microbiol 1999; 86:521-530. 

98. Bowey E, Adlercreutz H, Rowland I. Metabolism of isoflavones and lignans by the gut microflora: 
a study in germ-free and human flora associated rats. Food Chem Toxicol 2003; 41:631-636. 

99. Ward FW, Coates ME, Cole CB, Fuller R. Effect of dietary fats on endogenous formation of 
N-nitrosamines from nitrate in germ-free and conventional rats and rats harbouring a human 
flora. Food Addit Contam 1990; 7:597-604. 

100. Kamlage B, Hartmann L, Gruhl B, Blaut M. Linoleic acid conjugation by human intestinal 
microorganisms is inhibited by glucose and other substrates in vitro and in gnotobiotic rats. 
J Nutr 2000; 130:2036-2039. 

101. Roland N, Nugon-Baudon L, Andrieux C, Szylit O. Comparative study of the fermentative 
characteristics of inulin and different types of fibre in rats inoculated with a human whole fecal 
flora. Br J Nutr 1995; 74:239-249. 

102. Djouzi Z, Andrieux C. Compared effects of three oligosaccharides on metabolism of intestinal 
microflora in rats inoculated with a human fecal flora. Br J Nutr 1997; 78:313-324. 

103. Lhoste EF, Ouriet V, Bruel S, et al. The human colonic microflora influences the alterations of 
xenobiotic-metabolizing enzymes by catechins in male F344 rats. Food Chem Toxicol 2003; 
41:695-702. 

104. Barrow PA, Brooker BE, Fuller BE, Newport MJ. The attachment of bacteria to the gastric 
epithelium of the pig and its importance in the microbiology of the intestine. J Appl Bacteriol 
1980;48:147-154. 

105. Jonsson E, Conway PL. Probiotics for pigs. In: Fuller R, ed. Probiotics, the Scientific Basis. 
London: Chapman & Hall, 1992:87-110. 

106. Voravuthikunchai SP, Lee A. Cecectomy causes long-term reduction of colonization 
resistance in the mouse gastrointestinal tract. Infect Immun 1987; 55:995-999. 

107. Henriksson A, Khaled AKD, Conway PL. The effect of caecectomy on the fecal 
concentrations of urobilinogen and active trypsin in mice. Microb Ecol Health Dis 1996; 
9:61-65. 

108. Simmons HA, Ford EJ. Multiple cannulation of the large intestine of the horse. Br Vet J 1988; 
144:449-454. 

109. Walker J A, Harmon DL, Gross KL, Collings GF. Evaluation of nutrient utilization in the 
canine using the ileal cannulation technique. J Nutr 1994;2672S-2676S. 

110. Hagberg L, Bruce AW, Reid G, Eden CS, Lincoln K, Lidin-Janson G. Colonization of the 
urinary tract with bacteria from the normal fecal and urethral flora in patients with recurrent 
urinary tract infections. In: Kass HE, Eden CS, eds. Host-Parasite Interactions in Urinary 
Tract Infections. Chicago: University of Chicago Press, 1989:194-197. 



Animal Models for the Human Gastrointestinal Tract 271 

111. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, Horak I. Ulcerative colitis-like disease 
in mice with a disrupted interleukin-2 gene, [comment]. Cell 1993; 75:253-261. 

1 12. Kuhn RJ, Lohler J, Rennick D, Rajewsky K, Muller W. Interleukin 10-deficient micedevelop 
chronic enterocolitis. Cell 1993; 75:263-274. 

113. Gaskins HR, Vondrak-Juergens GL, McCracken BA, Woolsey JH. Specific-pathogen-free 
conditions enhance inflammatory bowel disease in T-cell receptor knockout, but not 
C3H/HeJBir mice, [comment]. Lab Anim Sci 1997; 47:650-655. 

1 14. Dianda L, Hanby AM, Wright NA, Sebesteny A, Hayday AC, Owen MJ. T cell receptor-alpha 
beta-deficient mice fail to develop colitis in the absence of a microbial environment. Am 
J Pathol 1997; 150:91-97. 

115. Mombaerts P, Mizoguchi E, Grusby MJ, Glimcher LH, Bhan AK, Tonegawa S. Spontaneous 
development of inflammatory bowel disease in T cell receptor mutant mice, [comment]. Cell 
1993; 75:274-282. 

116. Taurog JD, Richardson JA, Croft JT, et al. The germfree state prevents development of gut 
and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med 1994; 180:2359-2364. 

117. Desreumaux P, Colombel JF. Intestinal flora and Crohn's disease. Ann Pharm Fr 2003; 
61:276-281. McCarthy J, O'Mahony L, O'Callaghan L, et al. Double blind, placebo 
controlled trial of two probiotic strains in interleukin 10 knockout mice and mechanistic link 
with cytokine balance. Gut 2003; 52:957-980. 

118. Steidler L. In situ delivery of cytokines by genetically engineered Lactococcus lactis. Antonie 
van Leeuwenhoek 2002; 82:323-331. 

119. Hambly RJ, Rumney CJ, Fletcher JM, Rijken PJ, Rowland IR. Effects of high- and low-risk 
diets on gut microflora-associated biomarkers of colon cancer in human flora-associated rats. 
Nutr Cancer 1997; 27:250-255. 

120. Yamazaki K, Tsunoda A, Sibusawa M, et al. The effect of an oral administration of 
Lactobacillus casei strain shirota on azoxymethane-induced colonic aberrant crypt foci and 
colon cancer in the rat. Oncol Rep 2000; 7:977-982. 

121. Rumney CJ, Rowland IR, Coutts TM, et al. Effects of risk-associated human dietary 
macrocomponents on processes related to carcinogenesis in human-flora-associated (HFA) 
rats. Carcinogenesis 1993; 14:79-84. 

122. Knasmuller S, Steinkellner H, Hirschl AM, Rabot S, Nobis EC, Kassie F. Impact of bacteria in 
dairy products and of the intestinal microflora on the genotoxic and carcinogenic effects of 
heterocyclic aromatic amines. Mutat Res 2001; 480-481:129-138. 

123. Horie H, Zeisig M, Hirayama K, Midtvedt T, Moller L, Rafter J. Probiotic mixture decreases 
DNA adduct formation in colonic epithelium induced by the food mutagen 2-amino-9H- 
pyrido[2,3-b]indole in a human-flora associated mouse model. Eur J Cancer Prev 2003; 
12:101-107. 

124. Aso Y, Akaza H, Kotake T, Tsukamoto T, Imai K, Naito S. Preventive effect of a 
Lactobacillus casei preparation on the recurrence of superficial bladder cancer in a double- 
blind trial. The BLP Study Group. Eur Urol 1995; 27:104-109. 

125. Aso Y, Akazan H. Prophylactic effect of a Lactobacillus casei preparation on the recurrence 
of superficial bladder cancer. BLP Study Group. Urol Int 1992; 49:125-129. 

126. Hughes R, Rowland IR. Stimulation of apoptosis by two prebiotic chicory fructans in the rat 
colon. Carcinogenesis 2001; 22:43-47. 

127. Glick ME, Falchuk ZM. Dinitrochlorobenzene-induced colitis in the guinea-pig: studies of 
colonic lamina propria lymphocytes. Gut 1981; 22:120-125. 

128. Rachmilewitz D, Simon PL, Schwartz LW, Griswold DE, Fondacaro JD, Wasserman MA. 
Inflammatory mediators of experimental colitis in rats. Gastroenterology 1989; 97:326-337. 

129. Duchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer zum Buschenfelde KH. 
Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel 
disease (IBD) [comment]. Clin Exp Immunol 1995; 102:448-455. 

130. Shibolet O, Karmeli F, Eliakim R, et al. Variable response to probiotics in two models of 
experimental colitis in rats. Inflamm Bowel Dis 2002; 8:399-406. 



15 

Born Germ-Free — Microbial Dependent 



Elisabeth Norin and Tore Midtvedt 

Microbiology and Tumor Biology Center, Karolinska Instituted Stockholm, Sweden 



INTRODUCTION 

The essence of research on germ-free life is isolation. Any isolation must be attained 
mechanically, proven scientifically, and understood philosophically. As early as 1885, 
Louis Pasteur declared that the concept of a multicellular life free of all demonstrable 
living microbes could be looked upon as "mission impossible." Germ-free animal research 
began when Nuttal and Thierfelder in 1895/96 (1) succeeded in keeping a small number of 
Caesarean-derived guinea pigs alive and germ-free for more than a week. From their work, 
one can see that the major elements of germ-free research are similar today. They 
described how to determine the time for partition; developed anesthetic procedures that 
would not too adversely affect the offspring; and worked out procedures of aseptic 
Cesarean section and transfer of the offspring from the uterus into a sterile environment 
and sterilization procedures for food, water, and air, as well as proper methods for testing 
the sterility of the isolator. 

In the decades to follow, several scientists did some work on germ-free multi- 
cellular organisms, but they all had to work with the first generation. A real breakthrough 
in germ-free animal research came in 1945, when the second generation of germ-free rats 
were born at the Lobund Laboratory, Notre Dame, USA. In the following decades, units 
for germ-free animal research were established in several countries all around the world. 
Bengt E. Gustafssson's lightweight stainless steel isolators (2) represented a major 
technical improvement, and so did Trexler's plastic isolators. In the 1980s and 1990s, 
there has been a temporary decline in germ-free animal research since much resources 
from bioscience research were allocated to HIV and AIDS. However, in the last 
5-7 years, there has been an increased interest in germ-free animals as well as in animals 
with a specific, known microbiota, i.e., gnotobiotic animals. This increased interest is 
partly based on progress in molecular methods for studying prokaryot-eukaryote cross- 
talk in health and disease, partly on the mere fact that investigators, when working with 
transgenic or knock-out laboratory animals have realized the tremendous influence of the 
microbiota on the physiological and pathophysiological consequences of the new genetic 
construct. Therefore, it is easy to forecast that germ-free animals and gnotobiotic 
technology will be of increasing interest in the years to come. In the following, we will 
focus on the role of the microbiota on some anatomical structures, physiological, and 

273 



274 Norin and Midtvedt 

biochemical functions in the host. Additionally, the immunological impact of the 
microbiota will briefly be commented on. 



TERMINOLOGY 

With a slight travesty of the well-known terminology introduced by Claude Bernhard, the 
mammalian organism itself — a mouse, a rat or a human — can be characterized as a Milieu 
interieur (MI), a normal microbiota as a Milieu exterieur (ME) and the macroorganism and 
its microbiota as a Milieu total (MT) (3). In studies on the interplay between MI and ME, 
two terms, i.e., Microflora Associated Characteristic (MAC) and Germ-free Animal 
Characteristic (GAC) — have been found to be of considerable value (4). A MAC is defined 
as the recording of any anatomical structure, physiological, biochemical or immunological 
function in a macroorganism, which has been influenced by the microbiota. When 
microorganism(s) influencing the parameter(s) under study are absent — as in germ-free 
animals, newborns, or in relation to ingestion of antibiotics — the recording of a MAC can 
be defined as a GAC. Consequently, a germ-free organism is a sum of all GACs, and a 
normal macroorganism is a sum of MACs. Studies in germ-free animals and healthy 
newborns have given us the values of GACs, i.e., the MI. When we are investigating 
conventional organisms — MT — the question "what have the microbes done, " can be 
answered by the equation MT minus MI = ME. A gnotobiotic animal harboring a known 
microbiota, may present a set-up of some MACs and some GACs, depending on the 
specific activity of its microbiota. 

Over the years, the MAC/GAC concept has been applied in several studies (5). So 
far, most studies have been related to a phenotypic expression of what the microbes have 
done. However, the concept is applicable also when studying host-microbe cross-talk on a 
molecular, genotypic level (6-8). In the following, some major discrepancies between 
germ-free and conventional animals will be highlighted (Table 1). 



GERM-FREE ANIMALS AND DIETARY REQUIREMENTS 

Contrary to what is generally believed, germ-free animals require a higher dietary caloric 
intake than their conventional counterparts. The main reason is very simple. A normal 
microbiota will break down indigestible dietary substances to compounds that can be 
absorbed by the host. That is most prominent in ruminants, i.e., the microbiota digest 
cellulose into short chain fatty acids (SCFAs). 

Also contrary to what is generally believed, germ-free animals require a higher 
intake of nitrogen than their conventional counterparts. The main reason for this is most 
probably the great loss of non-degraded material from expelled enterocytes that are found 
in germ-free animals. In conventional animals, the microbiota converts the expelled 
material into absorbable compounds. 

In many germ-free macroorganisms, there might be a demand for an increased 
dietary intake of some vitamins. Broadly speaking, the gastrointestinal microbiota, placed 
between the ingesta and the host, may utilize dietary vitamins or produce vitamins 
themselves. 

Among the earliest evidence that the vitamin synthesis is connected to functions by 
the intestinal microbes was the demonstration that germ-free rats reared without a dietary 
source of vitamin K developed hemorrhages and hypoprotothrombinemia soon, whereas 
their conventional controls had normal prothrombin levels and no bleeding tendencies (9). 



Born Germ-Free — Microbial Dependent 



275 



Table 1 Influences of the Microbiota on Some Intestinal Anatomic, Physiological, and Biochemical 
Parameters 



Parameter 


MAC 


GAC 


Microbes 


Anatomical/physiological 








Intestinal wall 


Thicker 


Thinner 


Unknown 


Cell kinetics 


Fast 


Slower 


Unknown 


Migration motor complexes 


Normal 


Fewer 


Unknown 


Production of peptides 


Normal 


Altered 


Unknown 


Sensitivity to peptides 


Normal 


Reduced 


Unknown 


Caecum size (rodents) 


Normal 


Enlarged 


Partly known 


Osmolality 


Normal 


Reduced 


Unknown 


Colloid osmotic pressure 


Normal 


Increased 


Unknown 


Oxygen tension 


Low 


High (as in tissue) 


Several species 


Electropotential Eh, mv 


Low (under 100) 


High (above 100) 


Unknown 


Biochemical 








(3-aspartylglycine 


Absent 


Present 


Unknown 


Bile acid metabolism 


Deconjugation 


No deconjugation 


Many species 




Dehydrogenation 


No dehydrogenation 


Many species 




Dehydroxylation 


No dehydroxylation 


Few species 


Bilirubin metabolism 


Much deconjugation 


Little deconjugation 


Many species 




Urobilin 


No urobilin 


One species 


Cholesterol 


Coprostanol 


No coprostanol 


Few species 


Intestinal gases 


Carbon dioxide 


Some carbon 
dioxide 


Many species 




Methane 


No methane 


Few species 




Hydrogene 


No hydrogene 


Few species 


Mucin 


Degraded 


No degradation 


Some species 


SCFAs 


Large amounts 


Far less 


Many species 


Tryptic activity 


Little or absent 


High activity 


Few species 



Abbreviations: MAC, microflora associated characteristic; GAC, germ-free animal characteristic; SCFAs, short 
chain fatty acids. 
Source: From Ref. 8. 

Administration of vitamin Ki restored prothrombin levels to normal values within a few 
hours, but e.g., vitamin K 3 was less effective. If the germ-free animals were inoculated 
with an intestinal microbiota from conventional animals, the prothrombin levels were 
normalized quickly. The vitamin K dependent plasmaprotein factors II, VII, IX, and X are 
taking part in the blood coagulation cascade. It has been shown that some bacterial strains 
were effective in reversing vitamin K deficiency (10). It has also been shown, by hindrance 
of coprophagy in rodents, that the intestinal microbiota supplies the host with parts of the 
vitamin B complex. 



INTESTINAL MICROBIOTA, GROSS ANATOMY, HISTOLOGY, 
AND MOTILITY 



An enlargement of the cecum in the Caesarean-derived guinea pigs was the first 
anatomical difference observed when the epoch of germ-free research started (1), and 
similar differences have been observed in all rodents so far investigated. This enlargement 
might partly be explained by an absence of mucin breakdown in the germ-free animals, 
partly by a reduced degradation of dietary compounds, such as fiber, and partly by 



276 Norin and Midtvedt 

a reduced sensitivity to biogenic amines in germ-free animals (11). Interestingly, it has 
been shown that a mono-association of germ-free animals with Clostridium difficile 
markedly reduced the cecum size (12). 

For years, it was generally accepted that the villi were more slender and uniform in 
shape and that the crypts were shallower, containing less cells in germ-free animals as 
compared to their conventional counterparts. Moreover, the lamina propria was supposed 
to be thinner, and the turn-over rate of epithelial loss was slower. However, most recently 
it was shown — in germ-free, and conventional rats and mice — that age, gender, and the 
intestinal compartment actually under study have to be taken into proper consideration 
before stating significant differences (13-15). 

Another striking difference than an enlarged cecum, is a reduction in spontaneous 
muscular activity in germ-free animals. This may in part be due to a reduced sensitivity to 
biogenic amines (11), partly also to a reduction in motor migrating complexes (16). 
Interestingly, it was found that mono-association of germ-free animals with some bacterial 
species, including a probiotic strain, switches the function from a GAC to an MAC pattern 
within a few days. Furthermore, the area of endocrine cells in the GI tract is enlarged in 
germ-free animals (17). 

Most recently it has been found that experimental post-surgical intestinal adhesion 
formation is markedly reduced in germ- free rats ( 1 8). After mono-associated with lactobacilli, 
i.e., a probiotic strain, the animals reacted similar to the germ-free control, whereas they 
switched to a conventional pattern after being mono-associated with Escherichia coli. 
Obviously, germ-free animals should be used for solving this important question in surgery. 

Additionally, germ-free animals may express a compartmentalized reduced 
osmolarlity in intestinal content, an increased colloid osmotic pressure, a higher oxygen 
tension, and a higher redox potential than their conventional counterparts. As a 
consequence of this, strictly anaerobes are often difficult to establish as a monoculture 
in germ-free animals (this is often a dose-dependency). 



BIOCHEMICAL FUNCTIONS AND THE 
GASTROINTESTINAL MICROBIOTA 

Microbial Conversion of Bilirubin to Urobilinogen 

Bile pigments, consisting almost exclusively of bilirubin, are the end products of the 
catabolism of hemoglobin and some other heme-containing enzymes. Bilirubin, taken up 
by the liver, is conjugated to glucuronate in the liver and excreted with the bile to the 
intestine, where the bilirubin conjugates are de-conjugated, and transformed to a series of 
urobilinogens, usually collectively termed urobilins. Some intestinal (3-glucuronidases are 
derived from endogenous sources (19), but most of them are of microbial origin (20). The 
capacity to alter deconjugated bilirubin to urobilins seems to be a rare property among 
intestinal microorganisms. So far, only one bacterium, a strain of Clostridium ramosum, 
has been found capable of performing this transformation (21,22). 

Studies in children as well as adults, in rats and mice, and in pigs and horses show 
that this is a function normally present in any organism with a normal acting micro- 
biota (8). In infants, this function is established within the first month of life (23). In adults, 
fecal levels of urobilins are significantly higher in men than in women (p<0.05). 
Furthermore, in 36 to 50-year-old men the mean level of urobilins is significantly lower 
than for younger men ( < 36 years). In the case of women, the highest fecal values are 
found in women younger than 35 years of age (24). 



Born Germ-Free — Microbial Dependent 277 

Other studies have shown that intake of different antimicrobial drugs used in clinical 
practice significantly suppressed this MAC (25). 

Microbial Bile Acid Metabolism 

In all mammals, bile acids are derived from cholesterol in the liver. Cholic acid and 
chenodeoxycholic acid are common, but many other primary bile acids may be found. The 
primary bile acids are conjugated, usually with taurine or glycine, sometimes also with 
sulphate or glucuronate, and excreted into the bile. In the intestinal tract, the conjugated 
primary bile acids are attacked by microbial enzymes and converted into a variety of 
metabolites. The so-called secondary bile acids thus formed may then either be excreted 
with the feces, or reabsorbed, and sometimes further metabolized by hepatic enzymes to 
so-called tertiary bile acids before re-excretion in the bile. When present in the intestine, 
the bile acids (primary, secondary or tertiary) are subject to a number of microbial 
transformations such as deconjugation, desulfatation, deglucuronidation, dehydroxylation, 
and other oxidation-reduction reactions at the hydroxy 1 groups (8). In general, the 
metabolites formed are less water-soluble, less active in forming micelles, and sometimes 
more toxic to the host. 

Over the years, many hypotheses have been brought forward regarding the 
influence(s) of various bile acids on several host-related signs and symptoms (intestinal 
motility, cell-turnover, bacterial over-growth, effects similar as pheromones, development 
of cancer etc). Obviously, further works on these areas are needed. 

Microbial Conversion of Cholesterol to Coprostanol 

Cholesterol is a component in all mammalian cellular membranes and a precursor of 
steroid hormones, vitamin D, and bile acids. Pathophysiological^, it is thought to be an 
important factor in the pathogenesis of atheromatous arterial disease, hypertension, cancer 
of the large bowel, and other disorders (8). The intestinal cholesterol is derived mainly 
from two sources — partly from synthesis occurring in the liver and the small intestine and 
partly from foods of animal origin. The main elimination routes for the plasma cholesterol 
are biliary excretion of cholesterol into the intestine as well as hepatic conversion of 
cholesterol to bile acids. The intestinal cholesterol can be absorbed to the entero-hepatic 
circulation or undergo microbial conversion. The major microbial metabolite is 
(unabsorbable) coprostanol which is excreted with the feces. The organisms responsible 
for the conversion are all strictly anaerobic, Gram-positive, nonspore-forming coccoid 
rods, probably belonging to the genus Eubacterium. 

By definition, any germ-free organism lacks the intestinal microbial excretion route 
for cholesterol. From a functional point of view, conversion of cholesterol to coprostanol 
can be looked upon as a sharp "microbial intestinal knife," influencing the normal entero- 
hepatic circulation of cholesterol (26). As early as 1959, higher serum cholesterol 
concentrations were found in germ-free than in conventional rats fed the same diet (27). 

Studies in many mammalian species show that this function is present in all 
animals soon after birth (5). However, data from infants indicate that this function is 
established — when established — in the second part of their first year. Comparative data 
from several countries show that one of five healthy adults might be a "non-excretor" or 
"low-excretor" of coprostanol. We have hypothesized that a genetically determined 
receptor determines whether an environmental receptor modulation determines if a 
cholesterol converting microbiota will be established. So far, however, the nature of 
the(se) receptor(s) is still unknown (8). 



278 Norin and Midtvedt 

It has been claimed by some probiotic-producing companies that their microbial 
products decrease the level of plasma cholesterol, by mechanisms(s) still under discussion. 
Gnotobiotic animal studies seem very applicable for further mechanistic investigations. 

Microbial Degradation of Mucin 

Mucin in the GI tract is produced by goblet cells in the mucosa and glandular mucous cells 
in the submucosa. Mucin consists of a peptide core with oligosaccharide side chains 
O-glycosidically bound, and it has several important physiological and patho- 
physiological roles. It acts as lubricant, as a barrier and stabilizer for the intestinal 
microclimate as well as a source of energy for the microbiota. There is growing evidence 
that the mucin pattern may be a relevant issue to take into account in the pathophysiology 
of some intestinal diseases, such as ulcerative colitis, Crohn's disease, gastric and 
duodenal ulceration, and colon adenocarcinoma. 

In contrast to conventional rats and healthy adult humans, organisms without any 
intestinal microbiota excrete large amounts of mucin with their feces (28). The complete 
degradation of mucin requires various glycosidases and peptidases, and the degradation is a 
sequential action of several bacterial strains (28,29). However, one Peptostreptococcus 
strain can degrade mucin in vitro and in vivo (30). Additionally, some strains belonging to 
other species can act upon mucin (31) e.g., Bifidobacterium and Ruminococcus genera (31) 
have been isolated and are related to degrading of mucin. 

In all mammalian species so far studied, the intestinal microbiota is capable of 
breaking down mucin (8). In healthy children the function is successively established 
within the first year of life. It has also been shown that the microbiota might act upon the 
glycosylation pattern of mucin (32). In fact, alteration in glycosylation was the first 
observation of a molecular, quorum sensing dependent cross-talk between a host and a 
single microbial strain present in the GI tract (33). Also regarding this intestinal function, 
it has been demonstrated that different antibiotics cause disturbance of this microbial 
function in animals and man (34,35). 

Microbial Degradation of Intestinal Enzymes 

In the following section, trypsin is used as a model substance for endogenously derived 
enzymes. It is excreted as a precursor, trypsinogen, from the pancreas, and activated in the 
small intestine, mainly by brush border enzymes (36). Fecal tryptic activity represents the 
net sum of processes involving the secretion of trypsinogen, its activation to trypsin, 
trypsin inactivators, and the presence in the intestine of microbial- and diet-derived 
compounds and enzymes that inactivate or degrade trypsin and trypsin inactivators. Feces 
of germ-free rats contain large amounts of tryptic activity, whereas far less is found in their 
conventional counterparts (37,38). Obviously, intestinal microorganisms are responsible 
for the inactivation of trypsin, and at least one strain of Bacteroides distasonis capable of 
performing this inactivation, has been described (39). 

In most mammals, except man, the intestinal microbiota is breaking down trypsin, 
yielding fecal tryptic activity to be absent or very low. In man, most adults express tryptic 
activity in their feces, although the levels are influenced upon by age and gender (24). 

Microbial Degradation of p-Aspartylglycine 

The biochemical background for the presence of (3-aspartylglycine in feces is probably as 
follows: host-derived intestinal proteolytic enzymes break down some dietary proteins to 



Born Germ-Free — Microbial Dependent 279 

the P-carboxyl dipeptide P-aspartylglycine. The P-carboxyl dipeptide bonds are then 
cleaved by proteases derived from microbes (40). This is substantiated by findings in germ- 
free animals: lambs, piglets, rats, and mice. In feces from germ-free lambs and piglets 
(Welling, personal communication), and adult germ-free rats and mice (41) P-aspartyl- 
glycine is always found in germ-free rats and mice, whereas never in samples from their 
conventional counterparts. Thus, the presence or absence of p-aspartylglycine represents a 
functional parameter, depending on the presence of dietary precursors, the presence of host- 
derived proteolytic enzymes, and the presence/absence of microbial derived proteolytic 
enzymes. Previously it has been shown that the amount of P-aspartylglycine gradually 
diminishes in feces from ex-germ-free mice, as the number of microbes in their GI 
microbiota gradually increases (8). This dipeptide has been suggested as an indicator for 
colonization resistance i.e., a barrier against opportunistic pathogens and other 
microbes (42). Thus, presence of the dipeptide P-aspartylglycine in feces indicates that 
the normal intestinal microbial ecosystem is seriously altered. 

Microbial Production of Short-Chain Fatty Acids 

All Short-Chain Fatty Acids (SCFAs) but acetic acid are microbial anabolic and catabolic 
products following microbial degradation of many exogenously and endogenously derived 
compounds in the GI tract of all mammalian species. Endogenous production of acetate 
may occur in the liver or in the peripheral tissue. However, in intestinal contents nearly all 
acetate present derives from microbial metabolism. The microbial origin of GI SCFAs has 
been substantiated by comparative studies in germ-free and conventional animals (43). In 
conventional animals, the total GI production will partly be influenced by anatomical 
factors (far more in ruminants than in monogastric animals), partly by dietary habits (more 
in herbivores than in carnivores). Fecal SCFAs represent the net sum of production, 
absorption, and possible secretion of SCFAs throughout the GI tract. In short, each 
mammalian species can be expected to have its own "excretion profile" (8). 

The mere fact that so many physiological and clinical roles, ranging from sodium 
absorption (5,23) to cancer pathogenesis are attributed to SCFAs, make them an extremely 
interesting parameter. Significant alteration in fecal SCFAs profiles have been found in 
atopic children (44). Intake of antibiotics (45,46) and dietary changes (47,48) may also 
cause alterations in fecal SCFAs. Therefore, when studying this parameter in gnotobiotic 
animals — or in patients — a consequence analysis, as outlined in Table 2, might give an 
extra incitement for a proper evaluation. 



IMMUNOLOGY AND GERM-FREE LIFE 

In general, the major difference between germ-free and conventional animals is on a 
quantitative rather than a qualitative level. This seems to hold true for innate as well as for 
acquired immunity. 

Serum from germ-free animals contains complement in similar amounts as in 
conventional animals, whereas the levels of specific antibodies are reduced. On a cellular 
level, polymorphonuclear neutrophiles (PMNs) from germ-free animals are equal to their 
conventional counterparts with regard to phagocytic capacity (49) and chemotaxis 
(50), and an apparent reduction in phagocytosis is due to humoral factors, i.e., reduced 
antibodies (52). 

The most striking difference between germ-free and conventional animals is found 
with regard to the lymphoid immune system. In most — if not all — conventional 



280 Norin and Midtvedt 

Table 2 A Consequence Analysis of One Microbiota Associated Characteristic 

Statement: SCFAs are normally produced in high amounts by the intestinal microbiota; they are 

partly absorbed, and partly excreted in feces 
Mechanism behind possible consequences 

Biochemical: SCFAs are involved in several metabolic pathways 
Immunological: Uncertain consequences 
Place 
Locally 

In the intestinal lumen 
At the mucosa surface 
Within the mucosa cells 
Distant 

In the liver, pancreas, brain etc. 
Form 
Direct 
Locally 

Main anions in intestinal content 
Growth promotion of some microbes 
Growth suppression of others 
Growth regulation of mucosa cells 
Distant 

Energy supply to the general metabolism 
Indirect 
Locally 

"Promoted" microbes produce suppressive bacteriocins 
Direct suppression provides niches for other microbes to grow 
Distant 

Metabolic alternations act on production of insulin, etc. 
Consequence 
Physiological 

Locally the SCFAs are parts of direct/indirect regulatory 

Mechanisms for water and electrolyte absorption; the net effect is antidiarrheic, involved in 
regulation of carbohydrate metabolism, etc. 
Pathophysiological 

Involved in hepatic coma. 
Probably involved in colonic cancer, ulcerative, and pseudomembraneous colitis, etc. 

Abbreviation: SCFA, short chain fatty acid. 



mammalian species, there are more lymphoid cells associated with the GI tract than with 
the spleen, peripheral lymph nodes, and blood taken together, and gut-associated B cells 
account for more than 80% of all B cells in the human body (52). The total daily output of 
dimeric IgA is 0.8 g per m of intestine, an amount equivalent to the output of a lactating 
mammary gland (52). 

The gut-associated lymphoid tissue (GALT) has to be considered both from the 
perspective of its composition and spatial complexity, and an extensive evaluation is 
beyond the scope of this review. Interested readers may search in Medline for names such 
as Bengt Bjorksten, Per Brandtzaeg, John Cebra, and Agnes Wold, among others. In the 
future, it is reasonable to assume that germ-free animals will be used in several settings, as 
(1) germ-free inbred animals, with and without genetic manipulation of defined 
components of their immune systems or their epithelium, (2) isogenic strains of a given 
bacterial species expressing defined endogenous or foreign epitopes, and (3) prior or 



Born Germ-Free — Microbial Dependent 281 

simultaneous administration of other competing organisms. In all these future 
experiments, it might be wise to keep in mind that it is a constant "trialogue" of 
interactions between intestinal microbes, epithelium, and GALT. As pointed out 
elsewhere (6) these interactions are probably dynamic, reciprocal, and combinatorial, 
making it difficult to separate out a single tune in this cacophony of noise. Utilization of 
gnotobiotic animals might represent a suitable reductionistic "noise filter, " allowing us to 
study host-microbe cross-talks in greater details. For more information on the role of the 
intestinal microbiota on the immune system, see the chapter by Moreau elsewhere in 
this book. 



CONCLUSION 

For more than a century, germ-free and gnotobiotic animals have been used to investigate 
the influence of the intestinal microbiota and specific members of the intestinal microbiota 
on the functioning and health of the host. This has provided much insight into the intricate 
relation between the host and its microbes. However, as outlined above, much still remains 
to be studied, and germ-free animals will remain an important tool in the study of the 
interactions between the intestinal microbiota and the host. 



REFERENCES 

1. Nuttal GHF, Thierfelder H. Tierisches leben ohne Bakterien im Verdauungskanal. Hoppe 
Seyler's Zeitsch Physiol Chemie 1896-1897; 33:62-73. 

2. Gustafsson BE. Lightweight stainless steel systems for rearing germfree animals. Ann N Y 
AcadSci 1959;78:17-28. 

3. Midtvedt T. Influence of antibiotics on biochemical intestinal microflora-associated 
characteristics in man and animals. In: Gillessen G, Opferkuch W, Peters G, Pulverer G, 
eds. The Influence of Antibiotics on the Host-Parasite Relationship III. Berlin, Heidelberg: 
Springer Verlag, 1989:209-215. 

4. Midtvedt T, Bjorneklett A, Carlstedt-Duke B, et al. Germfree research; microflora control and 
its application to the biomedical sciences. In: Wostmann BS, Pleasants JR, Pollard M, 
Teah BA, Wagner M, eds. The Influence of Antibiotics Upon Microflora- Associated 
Characteristics in Man and Animals. New York: Alan R Liss Inc, 1985:241-244. 

5. Collinder E, Bjornhag G, Cardona M, Norin E, Rehbinder C, Midtvedt T. Gastrointestinal host- 
microbial interactions in mammals and fish. Comparative studies in man, mice, rats, pigs, 
horses, cows, elks, reindeers, salmon, and cod. Microb Ecol Health Dis 2003; 15:66-78. 

6. Falk GP, Hooper LV, Midtvedt T, Gordon JI. Creating and maintaining the gastrointestinal 
ecosystem; what we know and need to know from gnotobiology. Microbiol Molecul Biol Rev 
1998;1 157-1 170. 

7. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Ann Rev Nutr 2002; 22:283-307. 

8. Midtvedt T. Probiotics, other nutritional factors, and intestinal microflora: microbial functional 

o 

activities. In: Hansson LA, Yolken RH, eds. In: Nestle Nutrition Workshop Series. 
Philadelphia: Lippincott-Raven Publishers, 1999:79-96. 

9. Gustafsson BE. Vitamin K deficiency in germfree rats. Ann N Y Acad Sci 1959; 
78:166-174. 

10. Gustafsson BE, Draft FS, McDaniel EG, Smith JC, Fitzgerald RJ. Effects of vitamin K-active 
compounds and intestinal s in vitamin K deficient germfree rats. J Nutr 1962; 78:461-468. 

11. Strandberg K, Sedvall G, Midtvedt T, Gustafsson BE. Effect of some biologically active 
amines on the cecum wall of germ-free rats. Proc Soc Exp Biol Med 1966; 121:699-702. 



282 Norin and Midtvedt 

12. Gustafsson BE, Midtvedt T, Strandberg K. Effects of microbial contamination on the cecum 
enlargement of germ-free rats. Scand J Gastroent 1970; 5:309-314. 

13. Banasaz M, Alam M, Norin E, Midtvedt T. Gender, age, and microbial status influence upon 
cell kinetics in a compartmentalised manner. An experimental study in germ-free and 
conventional rats. Microb Ecol Health Dis 2000; 12:208-218. 

14. Banasaz M, Norin E, Midtvedt T. The role of gender, age, and microbial status on cell kinetics 
in the gastrointestinal tract of mice. An experimental study in germ-free and conventional mice. 
Microb Ecol Health Dis 2001; 13:135-142. 

o 

15. Banasaz M, Akerlund T, Norin E, Burman L, Midtvedt T. Reduced mitotic activity of intestinal 
mucosal cells in germ-free rats mono-associated with toxin producing Clostridium difficile. 
Microb Ecol Health Dis 2003; 4:146-152. 

16. Husebye E, Hellstrom PM, Midtvedt T. Intestinal microflora stimulates myoelectric activity of 
rat wall intestine by promoting cyclic initiation and abnormal propagation of migrating 
myoelectric complexes. Dig Dis Sci 1992;946-956. 

17. Alam M, Midtvedt T. In: Hashimoto K, ed. Microflora and Gastrointestinal Peptides. 
Shiozawa, Japan: XII IGS Publishing Committee, 1996:409-412. 

18. Bothin C, Okada M, Midtvedt T, Perbeck L. The intestinal flora influences adhesion formation 
around surgical anastomoses. Br J Surg 2001; 88:143-145. 

19. Rod TO, Midtvedt T. The origin of intestinal ^-glucuronidase in germfree, monocontaminated, 
and conventional rats. Acta Path Microbiol Scand Sect B 1977; 85:271-276. 

20. Gadelle D, Raibaud P, Sacquet E. (3-glucuronidase activities of intestinal bacteria 
determined both in vitro and in vivo in gnotobiotic rats. Appl Environ Microbiol 1985; 
49:682-685. 

21. Gustafsson BE, Swenander-Lanke L. Bilirubin, and urobilins in germ-free, ex-germ-free, and 
conventional rats. J Exp Med 1960; 112:975-981. 

22. Midtvedt T, Gustafsson BE. Microbial conversion of bilirubin to urobilins in vitro and in vivo. 
Acta Pathol Microbiol Scand Sect B 1981; 89:57-60. 

23. Midtvedt AC, Carlstedt-Duke B, Norin KE, Saxerholt H, Midtvedt T. Development of five 
metabolic activities associated with the intestinal microflora of healthy infants. J Pediatr 
Gastroenterol Nutr 1988; 7:559-567. 

24. Benno P, Alam M, Collinder E, Norin E, Midtvedt T. Fecal tryptic activity and excretion of 
urobilinogens in 573 healthy subjects living in Sweden, Norway, and Scotland. Microb Ecol 
Health Dis 2003; 15:169-175. 

25. Saxerholt H, Carlstedt-Duke B, Hoverstad T, et al. Influence of antibiotics on the fecal 
excretion of bile pigments in healthy subjects. J Gastroenterol 1986; 2:991-996. 

26. Midtvedt T. Metabolism of endogenous substances. In: Clercq De, ed. Frontiers in 
Microbiology. From Antibiotics to AIDS. Dordrecht/Boston/Lancaster: Martinus Nijhoff 
Publishers, 1987:79-87. 

27. Danielsson H, Gustafsson BE. On serum-cholesterol levels and neutral fecal sterols in germfree 
rats. Bile acids and steroids 59. Arch Biochem Biophys 1959; 83:482-485. 

28. Gustafsson B, Carlstedt-Duke B. Intestinal water-soluble mucins in germfree, ex-germfree, and 
conventional animals. Acta Path Microbiol Immunol Scand Sect B 1984; 92:247-252. 

29. Hoskins LC, Agustines M, McKee WB, Boulding ET, Kriaris M, Niedermeyer G. Mucin 
degradation in human colon ecosystems. Isolation and properties of fecal strains that degrade 
ABH blood group antigens and oligosacharrides from mucin glycoproteins. J Clin Invest 1985; 
75:944-953. 

30. Carlstedt-Duke B, Midtvedt T, Nord CE, Gustafsson BE. Isolation and characterization of a 
mucindegrading strain of Peptostreptococcus from rat intestinal tract. Acta Path Microbiol 
Immunol Scand Sec B 1986; 94:292-300. 

3 1 . Karlsson KA. Animal glucosphingolipids as membrane attachment sites for bacteria. Ann Rev 
Biochem 1989; 58:309-350. 

32. Freitas M, Axelsson LG, Cayuela C, Midtvedt T, Trugnan G. Microbial-host interactions 
specifically control the glycosylation pattern in intestinal mouse mucosa. Histochem Cell Biol 
2002; 118:149-161. 



Born Germ-Free — Microbial Dependent 283 

33. Bry L, Falk PG, Midtvedt T, Gordon JI. A model of host-microbial interactions in an open 
mammalian ecosystem. Science 1996; 6:1380-1383. 

34. Carlstedt-Duke B, H0verstad T, Lingaas E, et al. Influence of antibiotics on intestinal mucin in 
healthy subjects. Eur J Clin Microbiol 1986; 5:634-638. 

35. Carlstedt-Duke B, Aim L, H0verstad T, et al. Influence of clindamycin, administered together 
with or without lactobacilli, upon intestinal ecology in rats. FEMS Microbiol Ecol 1987; 
45:251-259. 

36. Norin KE, Midtvedt T, Gustafsson BE. Influence of the intestinal microflora on the tryptic 
activity during lactation. Lab Anim 1986; 20:234-237. 

37. Norin KE. The regulatory and protective role of the normal microflora. In: Grubb R, 
Midtvedt T, Norin KE, eds. The Normal Microflora and Intestinal Enzymes. New York: 
Stockton Press, 1988:219-237. 

38. Norin KE, Gustafsson BE, Midtvedt T. Strain differences in fecal tryptic activity of germfree 
and conventional rats. Lab Anim 1986; 20:67-69. 

39. Ramare F, Hautefort I, Verhe F, Raibaud P, Iovanna J. Inactivation of tryptic activity by a 
human-derived strain of Bacteroides distasonis in the large intestines of gnotobiotic rats and 
mice. Appl Environ Microbiol 1996; 62:1434-1436. 

40. Welling GW, Helmus G, de Vries-Hospers HG, et al. Germ-free research; microflora control, 
and its application to the biomedical sciences. In: Wostmann BS, Pleasants JR, Pollard M, 
Teah BA, Wagner M, eds. Rationale for Use of (3-Aspartylglycine as Indicator of Colonization 
Resistance. New York: Alan R Liss Inc., 1985:155-158. 

41. Welling GW, Groen G, Tuinte HM, et al. Biochemical effects on germ-free mice of association 
with several strains of anaerobic bacteria. J Gen Microbiol 1980; 117:57-63. 

42. van der Waaij D, van der Waaij BD. The colonization resistance of the digestive tract in 
different animal species and in man: a comparative study. Epidemiology and Infections 1990; 
105:237-243. 

43. H0verstad T, Midtvedt T. Short-chain fatty acids in germ-free mice and rats. J Nutr 1986; 
116:1772-1776. 

44. Bottcher M, Norin EK, Sandin A, Midtvedt T, Bjorksten B. Microflora associated 
characteristics in faeces from allergig and nonallergic infants. Clin Exp Allergy 2000; 
30:1590-1596. 

45. H0verstad T, Carlstedt-Duke B, Lingaas E, et al. Influence of ampicillin, clindamycin, and 
metronidazole on fecal excretion of short-chain fatty acids in healthy subjects. Scand 
J Gastroenterol 1986; 21:621-626. 

46. H0verstad T, Carlstedt-Duke B, Lingaas E, et al. Influence of oral intake of seven different 
antibiotics on fecal of short-chain fatty acid excretion in healthy subjects. Scand J Gastroent 
1986;21:997-1003. 

47. Siigur U, Norin KE, Allgood G, Schlagheck T, Midtvedt T. Effects of olestra on fecal water and 
short-chain fatty acids. Microb Ecol Health Dis 1996; 9:9-17. 

48. Siigur U, Norin KE, Allgood G, Schlagheck T, Midtvedt T. Effect of olestra upon intestinal 
microecology as reflected by five microflora associated characteristics in man. Microb Ecol 
Health Dis 1996; 9:297-303. 

49. Trippestad A, Midtvedt T. The phagocytic activity of polynuclear leucocytes from germfree 
and conventional rats. Acta Path Microbiol Scand Sect B 1971; 79:519-522. 

50. Trippestad A, Midtvedt T. Chemotaxis of polymorphonuclear leucocytes from germfree rats 
and generation of chemotactic activity in germfree rat sera. Clin Exp Immunol 1971; 
8:639-646. 

51. Midtvedt T, Trippestad A. Specificity of opsonic and bactericidal response of gnotobiotic rat 
sera. Acta Path Microbiol Scand Sect B 1971; 79:291-296. 

52. Brandtzaeg P, Halstensen TS, Kett K, et al. Immunobiology and immunopathology of human gut 
mucosa; humoral immun intraepithelial lymphocytes. Gastroenterology 1989; 97:1562-1584. 



16 



Modifying the Human Intestinal 
Microbiota with Prebiotics 



Ross Crittenden 

The Preventative Health Flagship, Food Science Australia, Werribee, Victoria, Australia 

Martin J. Playne 

Melbourne Biotechnology, Hampton, Victoria, Australia 



INTRODUCTION 

The aim of both prebiotic and probiotic functional food ingredients is to improve the 
health of consumers by selectively altering the composition and/or activity of microbial 
populations within the gastrointestinal tract. While the probiotic approach endeavors to 
directly deliver supplemental beneficial bacteria to the gut, prebiotics offer an alternative 
strategy. Rather than supplying an exogenous source of live bacteria, prebiotics aim to 
selectively stimulate the proliferation and/or activity of desirable bacterial populations 
already resident in the consumer's intestinal tract. 

The prebiotic strategy offers a number of practical and theoretical advantages over 
modifying the intestinal microbiota using probiotics or antibiotics. This chapter aims to 
provide an overview of the prebiotic approach, modes of action, and an evaluation of their 
effectiveness in modulating intestinal microbial populations and providing health benefits 
to consumers. The production, properties and applications of prebiotics are outlined and 
likely future developments in prebiotics are discussed. However, before exploring the 
concept of modifying the intestinal microbiota using prebiotics, it is perhaps pertinent to 
first reflect briefly on why we might want to alter the composition and activity of the 
intestinal microbiota in the first place. 



WHY MODIFY THE INTESTINAL MICROBIOTA? 

Far from being inconsequential to our lives, the bacteria residing within our 
gastrointestinal tracts are highly important to our health and well-being. They provide 
us with a barrier to infection by intestinal pathogens (1), much of the metabolic fuel for our 
colonic epithelial cells (2), and contribute to normal immune development and func- 
tion (3,4). Intestinal bacteria have also been implicated in the etiology of some chronic 
diseases of the gut such as inflammatory bowel disease (IBD) (5,6). As we age, changes 

285 



286 Crittenden and Playne 

occur in the composition of the intestinal microbiota that may contribute to an increased 
level of undesirable microbial metabolic activity and subsequent degenerative diseases of 
the intestinal tract (7,8). 

Modifying the composition of the intestinal microbiota to restore or maintain a 
beneficial population of micro-organisms would appear to be a reasonable approach in 
cases where a deleterious or sub-optimal population of micro-organisms has colonized the 
gut. The difficulty facing intestinal microbiologists is trying to determine what constitutes 
a "normal," healthy intestinal microbiota. A switch in recent years from culture-based, 
phenotypic examination of microbial ecosystems to the application of culture- 
independent, molecular techniques has helped speed progress. It has also provided new 
insights into the great diversity of bacteria within the human intestinal tract. Historical 
estimates based on culture methods did recognize the complexity of the ecosystem, 
placing the number of bacterial species within the gastrointestinal microbiota at around 
400, dominated by perhaps 30-40 (9). However, it is now believed to be far richer, with 
the number of identified taxa expected to eventually exceed 1000 (10). 

It is clear that we are only at the very beginning of understanding the role of 
individual bacterial populations in health and disease and their interactions with each 
other, the host, and the diet. Addressing these fundamental questions is an essential 
prerequisite to targeted disease intervention strategies involving modification of the 
intestinal microbiota. While acknowledging that the science of manipulating the intestinal 
microbiota to achieve improved health is still very much in its infancy, progress is being 
made, and strategies that may lead to tangible health benefits in specific populations 
are emerging. 



THE PREBIOTIC STRATEGY TO MODIFYING 
THE INTESTINAL MICROBIOTA 

For a variety of reasons, the two bacterial genera most often advocated as beneficial 
organisms with which to augment the intestinal microbiota are lactobacilli and 
bifidobacteria, both of which are common members of the human intestinal microbiota 
(11,12). These bacteria are numerically common, non-pathogenic, non-putrefactive, non- 
toxigenic, saccharolytic organisms that appear from available knowledge to provide little 
opportunity for deleterious activity in the intestinal tract. As such, they are reasonable 
candidates to target in terms of restoring a favorable balance of intestinal species. 

While the probiotic strategy aims to supplement the intestinal microbiota via the 
ingestion of live bacteria, the prebiotic strategy aims to stimulate the proliferation and/or 
activity of beneficial microbial populations already resident in the intestine. The 
characteristics shared by all successful prebiotics is that they remain largely undigested 
during passage through the stomach and small intestine and selectively stimulate only 
beneficial populations of bacteria in the colon. That is not to say that prebiotics cannot be 
theoretically designed to target bacteria within the stomach and small intestine, but rather 
those currently developed tend to target bifidobacteria, which predominantly reside in the 
colon. Importantly, prebiotics should not stimulate the proliferation or pathogenicity of 
potentially deleterious micro-organisms within the intestinal microbiota. To date, most 
prebiotics have been non-digestible carbohydrates, particularly oligosaccharides. Since 
the prebiotics identified to date promote the proliferation of bifidobacteria in particular, 
they are often referred to as bifidogenic factors or bifidus factors. Historically, lactobacilli 
and bifidobacteria have been targeted as beneficial organisms with which to augment the 
intestinal tract. However, as discussed later in this chapter, the manipulation more broadly 



Modifying the Human Intestinal Microbiota with Prebiotics 



287 



of the metabolic activity of the microbiota is of increasing interest for improving intestinal 
health (13). 

A number of largely prophylactic health targets have been proposed for prebiotics 
that, as might be expected, overlap considerably with the targets of probiotic interventions. 
The mechanisms of action remain largely theoretical, but rational hypotheses have been 
developed as our understanding of the intestinal microbiota has advanced. Proposed 
benefits in the gut include protection against enteric infections, increased mineral 
absorption, immunomodulation, trophic and anti-neoplastic effects of short chain fatty 
acids (SCFA), fecal bulking, and reduced toxigenic microbial metabolism (Figs. 1-4). 



A BRIEF HISTORY OF THE DEVELOPMENT OF BIFIDUS FACTORS 
AND PREBIOTICS 

Bifidogenic or bifidus factors were recognized as early as 1954 with Gyorgy et al. 
(14,15) describing such components in milk and colostrum, including a range of amino 
sugars and non-glycosylated casein peptides. Glycoproteins from whey were also 
shown to have bifidogenic potential (16) along with lactoferrin (17,18). Bifidogenic 
effects have been reported for pantethine from carrot extracts (19,20) and for 2-amino- 
3-carboxy-l,4-naphthoquinone (ACNQ), a compound isolated from Propionibacterium 
freudenreichii (21,22). 

Interest in bifidogenic compounds accelerated with the identification of non- 
digestible oligosaccharides (NDOs) in human milk as major factors responsible for 
maintaining an intestinal microbiota numerically dominated by bifidobacteria in breast- 
feeding infants. In contrast, infants fed cow's milk-based formula developed a mixed 
microbiota, including higher levels of potentially deleterious organisms (23,24). Human 



prebiotics 

OOOO 00 

ooo 



intestinal lumen 



stimulate growth 
of bifidobacteria 



colonocytes 




lamina propria 




immunomodulation IgE-mediated allergy 



IL-12 



suppression 

► 



allergy prevention 




IL-4 



Figure 1 Proposed mechanisms of immunomodulation by prebiotics for the prevention of IgE- 
mediated food allergies that are mediated by a skewing of the immune response at the T helper (Th) 
cell level towards a Th2 response. Prebiotics stimulate the growth of bifidobacteria that are sampled 
by the gut-associated lymphoid tissue via M-cells or dendritic cells (DC). The commensal bacteria 
drive a counterbalancing Thl response producing interferon-y (IFN-y), and/or a tolerogenic 
response by regulatory T-cells (Tr) producing the anti-inflammatory cytokines interleukin-10 
(IL-10) and transorming growth factor-(3 (TGF-(3) that quell the allergenic Th2 response. 



288 



Crittenden and Playne 



00 
prebiotics 

oooo 



ooo 



intestinal lumen + . , * 

stimulate growth 
and fermentation 



eliminate pro-IBD 
microbial antigens 



SCFA + 
I 




*^> 



trophic effects fj Jju Sj 



colonization 
resistance 



on mucosa 



rYY^fYWYYWYl f\nT\ 



colonocyte > 



lamina propria 




Mcell 

1 




nnn 

o 



fYY^nnn 



o 











nnn 

o 



-t/ 



immunomodulation 



IL-10 
TGF-P 



anti-inflammatory 



suppress 

IBD 

inflammation 



Figure 2 Proposed mechanisms by which prebiotics may ameliorate inflammatory bowel disease 
(IBD). Abbreviations'. DC, dendritic cell; IL, interleukin; SCFA, short chain fatty acids; Tr, 
regulatory T cell; TGF, transforming growth factor. 

milk oligosaccharides (HMOs) (discussed later in this chapter) were then, and remain 
today, too complex to be synthesized commercially. However, other NDOs were shown 
to replicate the bifidogenic effect of milk oligosaccharides. The Japanese research 
community in particular studied the ability to modify the intestinal microbiota using 
lactulose and fructo- and galacto-oligosaccharides. Although often lacking rigorous 
design, early studies (25-30) at least provided the impetus for later, randomized controlled 
studies that have demonstrated the notion that some NDOs selectively promote the 
proliferation of bifidobacteria in the intestinal tract. 

Concurrently in the late 1980s and early 1990s, interest was rising in the use of 
probiotics to modify the intestinal microbial balance. The term "prebiotic" was coined by 



selective growth 
and fermentation 



prebiotics 

0000 



oo 



ooo 




block ligand 
adhesion to 
mucosa 



nnnnnn 



o 



S £/ 




nutrient and 

niche 

competition 




► SCFA 



antimicrobials 



> lower intestinal pH 



antagonism 




nnnnnn 



o 



-ZS 







rinn 

o 



-a> 



^nnr^nr\ 



-ZS 



o 



o 



nnnnnn 



-£/ 







-<^ 







nnn 

o 



Figure 3 Proposed mechanisms by which prebiotics may enhance colonization resistance against 
bacterial pathogens in the gastrointestinal tract. Abbreviation: SCFA, short chain fatty acids. 



Modifying the Human Intestinal Microbiota with Prebiotics 



289 



alleviate constipation 



de novo 
lipo genesis 



controlled 
p. serum lipids 
and cholesterol 



ooooo 

prebiotics 

0000 




JUL JUL 

increased 

mineral 

absorption 



selective 
fermentation 



► lower intestinal pH 



antagonism of 



■► fewer toxic 





trophic and 
anti-neoplastic 
effects ^ 


putrefactive bacteria 


microbial 
metabolites 


nnn 


nnn 


r v^r 


^nr 


^nr 


fYY^fYY^fYY^fYY^r ^innnnn 

















c 

















. i) 


*■ 6 


e) 


■ o 


^ o 


o 


k £/ 


\ £/ 


■ o 


« £/ 


. £/ 



reduced cancer risk 



reduced cancer risk 



Figure 4 Proposed mechanisms by which the selective fermentation of prebiotics and subsequent 
production of short chain fatty acids (SCFA) improve bowel habit, increase dietary mineral 
absorption, and may reduce the risk of colon cancer. 



Gibson and Roberfroid in 1995 (31) and effectively linked these two concepts for promoting 
beneficial populations of intestinal bacteria. Gibson and Roberfroid (31) broadened the 
narrow bifidogenic target to include the specific stimulation of any potentially beneficial 
microbial genera. There is an obvious potential for synergy between prebiotic and probiotic 
ingredients, and hence, foods containing both prebiotic and probiotics ingredients were 
termed "synbiotics." 



CURRENTLY AVAILABLE PREBIOTIC CARBOHYDRATES 



The prebiotics most commonly used as functional food ingredients are non-digestible 
oligosaccharides (NDOs), of which a variety of types are commercially available (32). 
Most of these NDOs are natural components of many common foods including honey, 
milk, and various fruits and vegetables (32-34). Commercially, they are produced as food 
ingredients by four main processes: 

1. Extraction and purification from plants, e.g., soybean oligosaccharides and 
inulin from chicory 

2. Controlled enzymatic degradation of polysaccharides, e.g., xylo-oligosacchar- 
ides, isomalto-oligosaccharides, and some fructo-oligosaccharides 

3. Enzymatic synthesis from disaccharides, e.g., some fructo-oligosaccharides, 
galacto-oligosaccharides and lactosucrose (32,33) 

4. Chemical isomerization, e.g., lactulose. 



290 Crittenden and Playne 

In nearly all cases, the commercial oligosaccharide products contain a range of 
oligosaccharide structures of differing molecular weights and often with a variety of 
glycosidic linkages between sugar moieties. To date, the largest number of reported 
studies and the most consistent evidence accumulated for prebiotic effects have been for 
fructo-oligosaccharides and the polyfructan inulin (34-39). Good evidence from human 
studies also exists for the prebiotic activities of galacto-oligosaccharides (40-43) and 
lactulose (44-47). Boehm and Stahl (48) have summarized 28 of the human studies 
conducted on the physiological effects of galacto-oligosaccharides and fructans (fructo- 
oligosaccharides and inulin). Most of these studies were between one and three weeks in 
duration. Commercial food-grade oligosaccharide was fed at between 8 and 15 g/day in 
most experiments. Higher levels (40 g/day) were fed when inulin was used. They list 14 
trials on galacto-oligosaccharides involving 298 adults and 27 infants, and another 14 with 
fructans involving 238 adults and 34 infants. In nearly all cases, only healthy volunteers 
were tested. 

A number of other NDOs, to which less rigorous study has been so far applied, have 
at least indications of prebiotic potential. These include lactosucrose (49-52), gluco- (53), 
xylo- (54,55), isomalto- (56-59), and soybean oligosaccharides (60-63). Additionally, 
bifidogenic effects have been reported for lactitol (45), polydextrose (64) and glucono-5- 
lactone (65) in small human feeding studies. 

Evidence that some dietary fibers, such as resistant starches (66-72), arabinoxylan 
(73,74) and plant gums (75) have prebiotic potential is accumulating, but to date remains 
limited largely to in vitro and animal studies. These large carbohydrates may have some 
advantages in the intestinal tract over rapidly fermented oligosaccharides. They minimize 
rapid gas formation and osmotic effects in the gut, which can lead to intestinal discomfort, 
flatulence and diarrhea at high doses of NDOs (typically above 15-20 g per day). 
Additionally, they persist as substrates for saccharolytic fermentation more distally in the 
colon where carbohydrate limitation is believed to promote toxigenic microbial reactions 
leading to an increased risk of colorectal cancer (76-79). 

The molecular structure of the prebiotic can be expected to determine its 
physiological effects as well as which microbial species are able to utilize it as a carbon 
and energy source in the bowel. However, it appears that despite the diversity in molecular 
sizes, sugar compositions, and structural linkages within the range of prebiotic carbo- 
hydrates, it is the bifidobacteria that are almost universally observed to respond. Some 
established and emerging prebiotics, including lactulose (46), galacto-oligosaccharides 
(40,80,81) and resistant starches (69,71) have been sporadically reported to stimulate 
intestinal Lactobacillus populations. Indeed, some lactobacilli have been shown to possess 
the metabolic machinery to use fructo-oligosaccharides (82,83). Despite this, bifidobacteria 
remain the major beneficiaries of these substrates in the gut. Given the benefits attributed to 
probiotic lactobacilli, the development of novel prebiotics directly targeting Lactobacillus 
species remains an opportunity. The rise in these beneficial bacterial populations during 
prebiotic feeding has often been shown to be accompanied by concomitant reductions in the 
numbers of putrefactive organisms such as Clostridia and Bacteroides spp. and 
Enterobacteriaceae (31,44-46,60,84), possibly due to antagonism by SCFA production, 
acidification of the colonic environment, or direct antagonism (Figs. 3-4). 



MODIFYING THE INTESTINAL BIFIDOBACTERIUM POPULATION 

The composition of the human intestinal microbiota changes naturally with age, and 
prebiotic strategies need to be targeted to reflect the desired outcome for specific 



Modifying the Human Intestinal Microbiota with Prebiotics 



291 



demographics. This section describes how prebiotics might provide benefits for specific 
human populations in relation to the characteristics of their own particular intestinal 
microbiota, and outlines some of the evidence for health effects accumulated so far. A 
brief summary of the main physiological effects of prebiotics is listed in Table 1 . 



Infants 

Bifidobacteria colonize the human intestinal tract during or soon after birth and in breast-fed 
infants they eventually dominate the microbiota (85). The numerical dominance of 
bifidobacteria is induced by bifidogenic components in breast milk, including 
oligosaccharides (85,86). Indeed, human milk oligosaccharides (HMOs) are the original 
prebiotics. The concentration of oligosaccharides found in human milk (5 to 10 g/L) is 
about 100 times that found in cow's milk (0.03 to 0.06 g/L). HMOs are complex with more 
than 130 identified structures (87). Each individual oligosaccharide is based on a variable 



Table 1 Summary of Physiological Effects of Prebiotics 



Level of 
substantiation 



Effect 



Comments 



Strong 



Moderate 



Weak 



Increase intestinal 
numbers of 
bifidobacteria 



Improved bowel 
habit 

Alleviate hepatic 
encephalopathy 

Increase calcium 
absorption 



Control of serum 
lipid levels 

Prevention of 

colorectal 

cancer 
Improved 

colonization 

resistance 



Immune 
modulation 



Magnitude of bifidogenic effect is inversely 
proportional to the size of the initial intestinal 
Bifidobacterium population. Best evidence for FOS, 
inulin, lactulose, and GOS, with emerging evidence 
for a range of NDOs and some dietary fibers. 

Improved frequency of defecation and stool consistency 
demonstrated with many prebitoics. Lactulose has a 
long history of pharmaceutical use as a laxative. 

Lactulose has a long history of use as a pharmaceutical. 

Positive results in animal studies and now more 
consistently in human trials. Prebiotics appear to 
enhance colonic Ca + + uptake. Indications that larger 
prebiotics with sustained colonic fermentation may 
be the more effective. 

Consistently positive results in animal studies, but 
mixed results in human trials. Mechanism appears to 
be control of de novo lipogenesis via SCFA. 

Demonstrations of anti-cancer effects in rodent models 
for a range of prebiotics. Reduced intestinal 
genotoxicity in human studies. 

Lactulose effective against chronic Salmonella 
infection. Some evidence from animal studies for 
other prebiotics against intestinal and systemic 
infections. Possible deleterious effects of rapid 
acidification on gut mucosa require investigation. 

Limited evidence from animal studies for anti-allergy 
effects. Suggestions of immunomodulation from 
antiviral effects and enhanced immune responses to 
vaccination. 



Abbreviations: NDOs, nondigestible oligosaccharides; SCFA, short chain fatty acids; FOS, fructo- 
oligosaccharides; GOS, galacto-oligosaccharides. 



292 Crittenden and Playne 

combination of glucose, galactose, sialic acid, fucose and/or TV-acetylglucosamine, with 
varied sizes and linkages accounting for the considerable variety (88). 

In contrast to breast-fed infants, infants fed cow's milk-based formulae develop a 
more mixed intestinal microbiota, with lower counts of bifidobacteria and higher counts of 
Clostridia and enterococci (89). Formula-fed infants have also been observed to have 
higher fecal ammonia and other potentially harmful bacterial products (90,91). The 
bifidogenic effect of HMOs can be emulated using FOS and GOS (40,41,92). However, 
there is increasing evidence for roles of HMO outside their bifidogenic impact in the gut. 
These include blocking adhesion of pathogens to the intestinal mucosa (93-95) and roles 
in developing cognition (96). Hence, Af-acetylneuraminic acid derivatives or sialyl-lactose 
are also commonly added to infant milk formulae. The complexity of HMOs has thwarted 
attempts to synthesize their full range of structures commercially, although specific 
oligosaccharides have been synthesized using chemical and biotechnological approaches 
(97-100). There is a ready market in infant milk formulas for oligosaccharides that more 
closely replicate all of the properties of HMOs and research to synthesize them will no 
doubt continue. 



Effects on Immune System Maturation 

There is a growing recognition of the importance of the intestinal microbiota to the healthy 
maturation of the host's immune system, including appropriate programming of oral 
tolerance to dietary antigens (101). Differences have been observed between the intestinal 
bacterial populations of healthy infants and those suffering from atopic eczema. These 
included differences within the genus Bifidobacterium, which are found in lower numbers 
in the feces of allergic infants (102-108) and with a more adult-like species composition 
dominated by Bif. adolescentis (109,1 10) rather than the usual species associated with the 
infant intestine such as Bif. bifidum, Bif. breve, and Bif longum ( = Bif infantis) (111,112). 
Recent indications that probiotics may reduce the severity of atopic eczema in 
infants (113,114) has led to interest in understanding if similar effects can be achieved with 
prebiotics. The proposed mechanisms are outlined in Figure 1 and involve stimulation of 
Thl cells and/or regulatory T cells. Nagura et al. (115) tested the ability of raffinose 
consumption to re-balance a Th2-biased immune response in a controlled study using an 
engineered murine model of IgE-mediated allergy to ovalbumin. Feeding a relatively high 
dose of raffinose stimulated a counterbalancing Thl -type immune response, reduced Th2 
cell activity and suppressed the synthesis of serum IgE to ovalbumin in response to long- 
term allergen challenge. Using a similar model, Yoshida et al. (116) recently reported 
similar positive results for bifidogenic alginate-oligosaccharides, indicating that prebiotics 
may be able to replicate the benefits seen for probiotics in allergy prevention. 



Adults 

The proportion of bifidobacteria in the colonic microbiota drops following weaning and 
the introduction of solid food. In adults, they account for 1-5% of the total bacteria in 
feces. Although they form a slightly higher proportion of total bacteria in the caecum 
(117-121), the total numbers of bifidobacteria per gram of intestinal contents increases 
approximately 100-fold with passage from the caecum to the colon. In the feces of healthy 

o i r\ 

adults bifidobacteria are found in numbers generally in the order of 10 -10 cells per 
gram. (10,122-125). While these figures represent the typical Bifidobacterium cell density, 
a proportion of healthy adults harbor considerably lower numbers of Bifidobacteria in their 
gut (by several orders of magnitude) without any discernable adverse effects (125-128). 



Modifying the Human Intestinal Microbiota with Prebiotics 293 

It is yet to be determined how the total number of bifidobacteria within a stable microbiota 
influences the long-term health of the human host. In individuals with naturally low levels 
of bifidobacteria, other micro-organisms with similar functionalities may occupy a similar 
niche and fulfill a similar role in the intestinal tract. 

It is clear from the number of human feeding studies reported to date that 
consumption of prebiotics can increase the numbers of bifidobacteria in the colon of 
adults. For NDOs, consumption of typically 10-15 g/day can induce 10- to 100-fold 
increases in Bifidobacterium numbers (129,130). However, a range of factors may 
influence the size of any increase in Bifidobacterium numbers, the most important being 
the initial size of the population within the intestinal tract. In comparing different trials 
conducted using fructo-oligosaccharides, Rao (130) observed that the size of the 
bifidogenic response was inversely proportional to the size of the initial Bifidobacterium 
population rather than showing a strong dose response. In individuals colonized with an 
already large population of bifidobacteria, prebiotic consumption appears not to increase 
the total Bifidobacterium population size further. 

Bif. adolescentis, Bif.catenulatum/pseudocatenulatum, Bif. bifidum, and Bif. longum 
are the most frequently reported Bifidobacterium species in the intestines of adults, with 
considerable variation between individuals (121,125,126,131,132). To date, no clear 
rationale for promoting one species of Bifidobacterium over others has emerged. Indeed, it 
may be quite difficult to achieve major shifts within the population dynamic of 
bifidobacteria at the species level even if this was desirable. In one study to investigate 
this, feeding 8 g/day of galacto-oligosaccharides to healthy adult volunteers did not result 
in marked changes in the composition of their intestinal bifidobacterial populations at the 
species level (133-135). Similarly, despite observing increases in total Bifidobacterium 
numbers, Harmsen et al. (136) also saw no changes in the species composition of 
bifidobacteria in a study where adult volunteers were fed 9 g/day of inulin. The species 
composition within intestinal bifidobacteria has been shown to remain fairly stable over 
many months in adults (10,121,125,137,138) suggesting that day-to-day fluctuations in 
diet have little impact on the species dynamic. 

Even if they do not significantly alter the bacterial population dynamics in all 
individuals, prebiotics may still be effective in providing benefits to the consumer if they 
beneficially modulate the metabolic activity of the microbiota. Hypothetical examples 
might be increased production of SCFA or vitamins that benefit the health of the colonic 
epithelium, or synthesis of antagonistic metabolites that augment colonization resistance 
against pathogens. Tannock et al. (139) used molecular techniques to investigate both 
phylogenetic (DNA-DGGE) and metabolic (RNA-DGGE) changes in the intestinal 
microbiota induced by galacto- or fructo-oligosaccharides. While no discernable changes 
were observed in bacterial communities using DNA-DGGE (nor increases in total 
Bifidobacterium numbers by traditional culturing), RNA-DGGE analysis revealed that the 
prebiotics increased the activity of some bacterial groups including bifidobacteria. A 
current research need is to identify metabolic activities of the microbiota that affect the 
health of the host (positively or negatively) and to demonstrate that these can be 
specifically modulated with prebiotics in situ. 



Prebiotics in the Treatment of Inflammatory Bowel Disease 

A genetic predisposition to develop an over-zealous inflammatory immune response to 
components of the intestinal microbiota has been implicated in the etiology of IBD (140). 
Elimination of specific bacterial antigens, immunomodulation, and trophic effects of 
SCFA on the intestinal epithelium have all been proposed as mechanisms by which 



294 Crittenden and Playne 

prebiotics could alleviate IBD (Fig. 2). The size of the intestinal Bifidobacterium 
population has been shown to be relatively small (141,142) in subjects afflicted with IBD, 
although cause and effect links between disease and a diminished intestinal 
Bifidobacterium population remain to be established. Interventions with prebiotics have 
shown some benefit in ameliorating inflammation in both animal and human feeding trials. 
Using differing rodent models of IBD, a number of research groups have demonstrated 
amelioration of inflammation using prebiotic interventions. These include studies with 
lactulose (143), inulin (140) and fructo-oligosaccharides (144). In contrast, Holma et al. 
(145) observed no reduction in inflammation by intervention with galacto-oligosacchar- 
ides despite an increase in Bifidobacterium numbers. 

In addition to NDOs, larger polysaccharides with prebiotic potential have also been 
shown to have promise in the treatment of IBD. Resistant starch was demonstrated to 
ameliorate IBD in rodent models of disease (146,147), and in one study (147) out- 
performed a diet with an equivalent dose of fructo-oligosaccharides. Additionally, an 
arabinoxylan-rich germinated barley product has been reported to have benefits in the 
treatment of active IBD. This ingredient was shown to induce the proliferation of 
bifidobacteria in the human intestine (148), consistent with other in vitro and animal 
studies of the fermentation of arabinoxylans by intestinal bacteria (73,74,149). In rodent 
models of IBD, and in two small, non-blinded human studies of subjects with ulcerative 
colitis, consumption of the germinated barley product reduced inflammation (150-153). 

These results suggest that prebiotics have at least some potential to benefit IBD 
sufferers. However, convincing evidence of a consistent clinical benefit in the treatment of 
IBD remains to be demonstrated in large, randomized, double-blind, placebo-controlled 
trials. 



Elderly 

The proposed benefits of prebiotics for the elderly have been based on early studies using 
culture methods that showed Bifidobacterium levels substantially decreased as a 
proportion of the total fecal microbiota in elderly Japanese, while the numbers of 
putrefactive bacteria such as Clostridia increased (154). These findings have only recently 
been re-addressed using modern bacteriological and molecular techniques, with mixed 
results. While a study from the United Kingdom (155,156) supported the earlier 
observations of a drop in Bifidobacterium numbers, other studies of elderly Italians and 
Dutch did not show any reduction in the size of the Bifidobacterium population (157,158). 
Still, prebiotics may be of benefit in elderly subjects with a low level of bifidobacteria and 
high levels of deleterious bacteria. One such group is elderly people with Clostridium 
difficile -associated diarrhea (CDAC) who have been shown to have a diminished 
bifidobacterial population (159). Prebiotic intervention may eventually prove to be 
beneficial in the prevention of such conditions in the elderly. 

Prebiotics are also hypothesized to have potential to provide protection for 
degenerative diseases in the elderly such as colon cancer and osteoporosis, and 
experimental evidence for benefits in these conditions are discussed in a later section of 
this chapter. 



SYNBIOTICS 

Products containing both prebiotic and probiotic ingredients are termed "synbiotics" due 
to the obvious potential for synergy between these ingredients. Although the prebiotic may 



Modifying the Human Intestinal Microbiota with Prebiotics 295 

not necessarily be utilized by the included probiotic bacteria in a synbiotic food, attempts 
have been made to maximize potential synergies by using complementary prebiotics that 
may aid the colonization and in situ functionality of the included probiotic strains. In a 
study in pigs, Brown et al. (160) showed that the inclusion of resistant starch or 
oligosaccharides in a synbiotic combination with a probiotic Bifidobacterium resulted in 
significantly higher numbers of bifidobacteria in the intestinal tract than with feeding of 
the probiotic alone. Continuing prebiotic feeding after the cessation of probiotic feeding 
also significantly extended the intestinal persistence of the probiotic. 

In terms of potential health benefits, synbiotic combinations have shown enhanced 
impact over feeding solely probiotics or prebiotics in rodent models investigating anti- 
cancer effects (161-164) and colonization resistance (165). To increase the specificity of 
synbiotics for the added probiotic strains, bifidobacteria have themselves been exploited as 
a source of enzymes to synthesize NDOs (166-168) including synthesis of galacto- 
oligosaccharides in yoghurt during fermentation (166). 



MECHANISMS OF THE BIFIDOGENIC EFFECT 

The mechanism(s) by which prebiotics promote the relatively specific proliferation of 
bifidobacteria remain speculative. It is probably due to the efficient utilization of these 
carbohydrates as carbon and energy sources by bifidobacteria relative to other intestinal 
bacteria, and their tolerance to the SCFA and acidification of the microenvironment 
resulting from fermentation. Additionally, many bifidobacteria adhere to large granular 
substrates such as resistant starch and these may provide a site for colonization as well as 
a substrate (13,169). The ability of bifidobacteria to use a wide variety of oligosaccharides 
and other complex carbohydrates reflects their evolution in the hind-gut of humans and 
animals where the ability to metabolize a diverse range of food and host-derived complex 
carbohydrates and glycoproteins provides a competitive advantage. Analysis of the 
Bif. longum genome has revealed a large number of proteins specialized for the catabolism 
of carbohydrates (170). 

Interestingly, while many bifidobacteria grow well when cultured with prebiotic 
oligosaccharides as their sole carbon and energy source, they often do not grow when 
supplied only with the monosaccharides from which these oligosaccharides are 
composed (74,171,172). This physiology may be another consequence of their evolution 
in an environment with a limited availability of simple sugars. It suggests that 
bifidobacteria lack transport mechanisms for many monosaccharides and import 
prebiotic oligosaccharides before hydrolyzing and metabolizing them. This presumably 
minimizes the availability of released simple sugars for cross-feeding by other intestinal 
bacteria and may be another factor contributing to the specific bifidogenic effect 
of NDOs. 



ADVANTAGES OF THE PREBIOTIC STRATEGY 

While both antibiotics (see chapter by Sullivan and Nord) and probiotics (see chapter by 
Khedkar and Ouwehand) can modify the intestinal microecology, the prebiotic strategy 
offers a number of advantages over these two approaches. 



296 Crittenden and Playne 

Advantages over Antibiotics 

Eliminating pathogenic groups with antibiotics is an obvious approach to beneficially 
modifying the intestinal microbiota. However, perturbation of indigenous microbial 
ecosystems caused by the collateral damage to desirable populations can lead to 
potentially serious side effects. These include antibiotic-associated diarrhea and 
pseudomembranous colitis involving overgrowth of Clostridium difficile as well as oral 
or vaginal candidiasis (173-175). Prebiotics and probiotics can ameliorate the potential of 
opportunistic infections caused by disturbances to the microbiota by restoring populations 
of beneficial bacteria (176-179). No long-term side effects have been reported for either 
prebiotic or probiotic ingredients, enabling their safe long-term use in prophylactic 
strategies to minimize disease. In contrast, long-term use of antibiotics may elicit a range 
of side-effects including liver damage, hypersensitivity, sensitivity to sunlight, and 
increasing the risk of developing antibiotic-resistant bacterial strains (180,181). This latter 
risk is particularly serious, and applies also to the sub-therapeutic use of antibiotics in 
intensive livestock farming in order to minimize infections and maximize yields, 
particularly for poultry and pork. Alternatives to antibiotics are urgently sought, and there 
has been considerable interest in the use of both prebiotics and probiotics in animal feeds 
to aid production. Although they have shown some promise (182,183), further research is 
needed into their application within an overall management strategy in order to match the 
performance of antibiotics. 



Advantages over Probiotics 

Storage Stability 

With the exception of some mechanisms of immunomodulation, the theoretical basis for 
many of the anticipated probiotic effects of bifidobacteria rely on the bacteria being viable 
in the intestinal tract. Currently, probiotics are limited by their stability largely to fresh 
food products such as fermented dairy products and juices, and nutraceutical products 
where they are formulated as dried powders. In contrast, prebiotics are stable, can be heat- 
processed, and can therefore be incorporated into a wider range of processed foods and 
beverages with longer shelf lives than probiotics. 

Host-Microbiota Compatibility 

It is clear that selected probiotic bifidobacteria do survive transit through the stomach and 
small intestine and can be recovered in feces. However, in most cases, ingested probiotic 
strains persist only transiently in the intestine (134,184-188). An introduced probiotic 
strain must compete with an already established microbiota. The application of molecular 
techniques to profile the complex microbial communities has revealed that each person has 
a unique intestinal microbiota at the community, genus, and species level (137-139). This 
has been demonstrated in the case of bifidobacteria using PCR-DGGE analysis of 
Bifidobacterium species in feces, where each individual has their own particular 
combination of species (121,125). This uniqueness appears to extend to the strain level 
too, with molecular fingerprinting techniques showing that each person generally harbors 
multiple and unique Bifidobacterium strains (138,189-191). This host-microbiota stability 
and individuality suggests that certain host-microbiota compatibilities exist, and using 
prebiotics that augment an individual's own bacteria may prove more successful than 
introducing an exogenous strain for some applications. 



Modifying the Human Intestinal Microbiota with Prebiotics 297 

The importance of host species/probiotic species specificity remains a contentious 
question. It is often recommended that probiotics be selected from bacteria indigenous to 
the intestinal tract of the targeted host species (192). However, the predominant probiotic 
Bifidobacterium species currently used in human probiotics is Bifi animalis ( = Bif. lactis) 
(11), which is not an autochthonous member of the human intestinal microbiota. This 
species is taxonomically distant from human intestinal species (193), but is used because 
of its superior technological stability compared with human intestinal isolates. The 
prebiotic strategy overcomes any potential host/probiotic strain compatibility issues by 
targeting those strains already resident in the intestinal tract of an individual. 



Inhibition of Pathogen Adhesion 

One mechanism by which oligosaccharides may provide protection against infection by 
pathogenic micro-organisms has been hypothesized to be that of blocking adhesion to 
intestinal mucosa by acting as soluble receptor analogues (Fig. 3) (194-196). Microbial 
virulence factors, such as fimbriae and other membrane-based adhesins, control mucosal 
attachment and colonization of tissues. The recognition domains of fimbriae are similar to 
lectins that bind to carbohydrate epitopes on membrane glycocojugates of epithelial cells. 
Kunz and Rudloff (197) have listed the receptor specificities of glyco- and lactose-derived 
oligosaccharides and various pathogenic bacteria and viruses. Carbohydrate-mediated cell 
interactions affect cell-cell interactions, as well as bacterium, viral and toxin interactions 
with epithelial cells. The specificity of attachment provides potential for control of gastro- 
intestinal infections through the use of specific oligosaccharide structures. 

Stimulation of Fermentative Activity in the Gut 

In addition to modifying population dynamics, prebiotics also modify the activity of the 
microbiota by providing a source of readily fermentable carbohydrate. Indeed, it may be 
this dietary fiber-like characteristic of modifying the fermentative activity of the existing 
microbiota that is the important factor in providing a number of health benefits to 
consumers (Figs. 2-4). Proposed health effects of prebiotics that are speculated to be 
largely contingent on modifications to metabolic activity of the microbiota include 
reductions in risk factors for colon cancer, increased mineral absorption, improved lipid 
metabolism, and increased resistance to intestinal pathogens. 

Reduced Risk Factors for Colon Cancer. The intestinal microbiota has a number 
of biochemical activities relevant to colon cancer risk that relate to the composition and 
activity of different bacterial populations. Hence, prebiotics may have a role in reducing 
risk factors for colon cancer. Since they supply a source of fermentable carbohydrate to the 
colon, dietary fiber-like anti-carcinogenic effects have been proposed for prebiotics 
(Fig. 4). Proposed mechanisms include supplying the colonic epithelium with SCFA 
(particularly butyrate); suppression of microbial protein metabolism, bile acid conversion 
and other mutagenic and toxigenic bacterial reactions; and immunomodulation. Butyrate 
production in the distal colon is suspected to be beneficial in preventing the development 
of colorectal cancers (198-200). While Lactobacillus and Bifidobacterium probiotics do 
not produce butyrate as major fermentation end products, prebiotics can stimulate butyrate 
production by the colonic microbiota, which provides a potential advantage of this 
approach (37,201). To date, the capacity of prebiotics to significantly contribute to a 
reduced incidence of colorectal cancer remains unproven. However, the results of 
preliminary human and animal experiments have provided sufficient encouragement to 
maintain the impetus for continued research into the protective effects of prebiotics. 



298 Crittenden and Playne 

Numerous studies in humans and animals have shown that consumption of 
prebiotics can produce an improved colonic environment in terms of reducing the levels 
of mutagenic enzyme activities (e.g., (3-glucuronidase and azoreductase) and bacterial 
metabolites (e.g., secondary bile acids, phenols and indoles) that are purportedly 
associated with colon cancer risk. Examples include studies with lactulose (44,45,202), 
galacto-oligosaccharides (203), resistant starch (69,204-206) and lactosucrose (51). 
However, not all prebiotic feeding studies have shown improvements in these parameters 
(46,47,66,207), and in any case, the quantitative importance of these markers to eventual 
cancer development remains to be established. 

A growing number of studies report protection by prebiotics against the 
development of pre-neoplastic lesions and/or tumors in rodent models of colon 
carcinogenesis. Again, these have used a variety of prebiotics including fructo- 
oligosaccharides and inulin (summarized by Pool-Zobel et al. (37)), lactulose (161,208) 
and resistant starch (209,210). Dose effects have been observed (37), but in general, very 
high doses of NDOs have been used in the animal studies. An important question that is 
beginning to be addressed is the significance of the sustainability of fermentation provided 
by different prebiotics during passage through the colon on their effectiveness in 
preventing colon cancer. The distal colon and rectum are the major sites of disease 
in humans, but SCFA produced by bacterial fermentation in the colon are rapidly absorbed 
by the colonic mucosa near the site of their production. Hence, prebiotics that can supply a 
persistent source of fermentable carbohydrate that sustains SCFA synthesis through to the 
distal colon may prove to be the most effective. Indeed, studies with different molecular 
sized fructan prebiotics have reported increased protection with the larger, more slowly 
fermented prebiotics (37). 

Improving Mineral Absorption. As seen for dietary fibres, a number of prebiotics 
have been shown to increase mineral absorption in animal models (21 1-214). The precise 
mechanisms of prebiotic-mediated improvements in mineral uptake remain unclear, but 
fermentative activities of the microbiota including SCFA production and reductions in 
luminal pH are believed to be involved (Fig. 4) (213). Calcium and magnesium are the 
main minerals for which uptake is improved. Under normal circumstances dietary calcium 
is predominately absorbed in the small intestine with little calcium absorbed in the colon 
(215). However, prebiotic fermentation is believed to extend calcium uptake into 
the colon (34). In rats, increased calcium uptake has led to improved bone mineralization 
for animals fed galacto-oligosaccharides (216), lactulose (217) and fructo-oligosac- 
charides (218). 

Although two human studies have shown little impact on mineral uptake (219,220), a 
number have reported beneficial effects on calcium (221-225) and magnesium absorption 
(226) using fructo-oligosaccharides, inulin and galacto-oligosaccharides. Differences in 
results have been attributed to differences in study designs and treatment populations 
(212,225). Griffin et al. (225) saw no effect with short chain fructo-oligosaccharides in a 
population of pubertal girls, but a significant increase in the calcium absorption and balance 
was observed when the girls consumed a mixture of fructo-oligosaccharides and inulin, 
perhaps reflecting a more sustained colonic fermentation. Overall, results so far are 
encouraging of a role for prebiotics in improving calcium uptake. Further research is 
warranted to investigate links between long-term prebiotic consumption and improved bone 
density in humans at risk of developing osteoporosis. 

Effects on Serum Lipids and Cholesterol. A role for prebiotics in controlling 
hyperlipidemia has been proposed and a relatively large number of animal and human 
studies have focused on the effects of oligosaccharide and inulin intake on lipid 
metabolism. These include eight human trials summarized by van Loo et al. (34), and 



Modifying the Human Intestinal Microbiota with Prebiotics 299 

more recent trials (227-231). The mechanism by which lowering of serum lipids and 
cholesterol may occur has been speculated to be regulation of host de novo lipogenesis via 
SCFA absorbed from the gut (Fig. 4) (232). While convincing positive effects on lowering 
serum triglycerols and cholesterol have often been reported in animal studies (233) the 
results from human studies have tended to be contradictory, although no deleterious effects 
have been reported (232). The trials conducted to date indicate that while there is certainly 
potential for prebiotics to control serum lipids, more research is needed to identify the 
most appropriate target populations, the impact of background diet, and the mechanisms 
of action. 

Improving Colonization Resistance in the Gut. The ability of prebiotics to 
improve colonization resistance and prevent bacterial infections from the gut has been 
only scantly explored, but results so far indicate a potential application for lactulose and 
NDOs in this capacity. Lactulose has the most accumulated evidence. Ozaslan et al. (234) 
observed lower caecal overgrowth and translocation of Escherichia coli in rats with 
obstructive jaundice when they were fed lactulose, while Bovee-Oudenhoven et al. (235) 
reported that consumption of lactulose increased colonization resistance against the 
invasive pathogen Salmonella enteritidis in rats. Indeed, lactulose consumption at high 
doses (up to 60 g per day) is effective in eliminating salmonella from the intestinal tract of 
chronic human carriers and is used as a pharmaceutical for this purpose in some countries 
(236). The mode of action is speculated to be acidification of the gut that prevents growth 
of this acid-sensitive pathogen. 

The anti-infective effects of fructo-oligosaccharides and inulin have been examined 
in mice challenged with the enteric pathogen Candida albicans and with systemic 
infections of Salmonella and Listeria monocytogenes (237). Prebiotic feeding significantly 
reduced intestinal colonization by Candida and the mortality of the mice with the systemic 
infections, the latter effect hypothesized as being due to gut microbiota-induced 
immunomodulation. However, two randomized, blinded, and controlled trials in which 
Peruvian infants living in environments with a high burden of gastrointestinal and other 
infections were fed oligofructose failed to show any significant benefit in terms of 
preventing diarrhea or the use of health care resources (238), although a high level of 
breast feeding amongst these infants may have limited the opportunity for effect. Prebiotic 
intervention may prove effective in rapidly restoring colonization resistance and 
preventing infections in cases where the intestinal microbiota has been perturbed. 



Other Physiological and Technological Benefits of Prebiotics 

In addition to the effects elicited by prebiotics discussed thus far, prebiotics have a 
number of other functional properties that make them attractive pharmaceuticals and 
food ingredients. Through their action in fecal bulking and water retention in the 
bowel, prebiotics are effective in relieving constipation and maintaining normal stool 
frequency (34). Additionally, by stimulating bacterial protein synthesis and reducing 
production of ammonia by the microbiota, lactulose is effective in the treatment of 
hepatic encephalopathy (236). NDOs are sweet and can be used as low-cariogenic and 
low-calorific sugar substitutes, while polysaccharides such as inulin are used as fat 
replacers. Their indigestibility and subsequent impact on glucose and insulin responses 
also make them suitable for diabetics. In terms of food technology, NDOs supply a 
number of valuable physicochemical functionalities. They can be used to increase 
viscosity, reduce Malliard reactions, alter water retention, depress freezing points, and 
suppress crystal formation. Hence, they are used commercially in a wide variety of 
foods and beverages. 



300 Crittenden and Playne 

DISADVANTAGES OF THE PREBIOTIC APPROACH 

While there are many advantages of the prebiotic approach, the use of this strategy to 
modify the intestinal microbiota is not without its disadvantages. First among these is the 
potential for intestinal side-effects if excessive doses of prebiotic oligosaccharides are 
consumed (discussed in more detail in the following section). Secondly, there are 
instances where probiotics may be more applicable to restoring colonization resistance in 
the gut. One example is during episodes of diarrhea when mucosal damage may lead to 
reduced capacity for sugar digestion. Ingestion of prebiotic oligosaccharides under these 
conditions may exacerbate symptoms associated with sugar malabsorption even at usually 
tolerable doses. Thirdly, there may be mechanisms, such as immunomodulation, where the 
introduction of an exogenous probiotic strain could theoretically provide a superior 
stimulus. Finally, some effects of probiotics are known to be strain specific and prebiotics 
cannot at this stage emulate that specificity. 



SAFE DOSAGE LEVELS 

Safety of use must always be a dominant issue in the development of new food products. 
Fortunately, it is well established that lactulose, short-chain oligosaccharides, inulin, 
resistant starch and dietary fiber are not toxic, even in high doses. Non-digestible 
carbohydrates are consumed as part of the normal daily diet, as they are natural 
components of most plants (239). Estimates of resulting intakes of fructo-oligosaccharides 
and inulin are between 1 and 10 g/day from normal diets in Europe and the United States 
of America (239,240). It is likely that intakes of around 8 g/day by adults are normal. 
Thus, any recommended dosages of non-digestible carbohydrates will be additional to the 
natural basal dose consumed. Recommended effective doses of prebiotic oligosaccharides 
in adults usually range from 10 to 15 g/day. With the shorter chain oligosaccharides, such 
as fructo- and galacto-oligosaccharides, intakes exceeding 15 g/day in adults can lead to 
flatulence, abdominal discomfort and cramping (241-243). With adaptation, larger doses 
of up to of 25-30 g/day can be tolerated with few ill effects. Excessive consumption of 
lactulose and NDOs can result in diarrhea due to osmotic water retention in the colon, with 
the offending dose depending on the weight of the individual, rate of consumption (single 
dose or frequent smaller doses spread over the day), and the composition and activity of 
the intestinal microbiota. 

A possible side-effect from the consumption of rapidly fermented, acidogenic 
prebiotic sugars was recently identified by Dutch researchers (244,245). While 
investigating the effects of lactulose and fructo-oligosaccharides on the translocation of 
Salmonella in rats, the researchers noted that feeding the prebiotics left the animals more 
susceptible to pathogen translocation from the gut. Intestinal acidification was observed 
due to the rapid prebiotic fermentation, and while this inhibited the acid-sensitive 
pathogen in the intestinal lumen, it possibly also damaged the mucosa leading to an 
impaired barrier effect. Further research is needed to investigate possible negative impacts 
of high doses of rapidly fermented sugars on the intestinal mucosa. 



CONCLUSION AND FUTURE DIRECTIONS 

There is little doubt from the volume of accumulated evidence from human and animal 
studies that prebiotics can modify the dynamics of the colonic microbiota. Bifidobacteria 



Modifying the Human Intestinal Microbiota with Prebiotics 301 

are the dominant group of bacteria stimulated by all prebiotics developed so far. That such 
a range of diverse carbohydrate structures can promote the selective proliferation of 
bifidobacteria is testament to the remarkable metabolic agility of these organisms. The 
magnitude of the bifidogenic effect is largely affected by the size of the intestinal 
Bifidobacterium population, and little impact on Bifidobacterium numbers is observed in 
individuals who already harbor high numbers of these bacteria. 

Beyond Bifidobacteria 

Although traditional microbiology culture methods have enabled some assessment of the 
selectivity of prebiotics, new molecular techniques that enable analysis of non-cultivable 
bacteria are starting to be applied in studies investigating the impact of prebiotics on the 
colonic microbiota. Almost certainly, other bacterial populations that are affected by 
the intake of current prebiotics will emerge. While evidence to date supports the beneficial 
role of bifidobacteria and lactobacilli in the intestinal tract (11,12), they are but two of a 
multitude of bacterial genera within the intestinal microbiota that potentially confer 
benefits to the host. As we gradually shed light on the activities of newly identified 
intestinal bacteria and their interactions with the host in health and disease new beneficial 
and detrimental organisms will be undoubtedly be identified. The challenge will be to find 
or design selective prebiotics to modulate populations and activities of these 
particular organisms. 

Phylogenetic vs. Physiological Modulation of the Microbiota 

It should be emphasized that altering the microbial population dynamic is only one aspect 
of prebiotic action. While stimulating the proliferation of particular groups of bacteria 
might be important for some health effects (e.g., immunomodulation), this may be 
secondary to specifically altering the metabolic activity of the microbiota for other effects 
(e.g., anti-cancer). Marked differences between the phylogenetic and physiologic effects of 
prebiotics on particular groups of organisms have been observed (139). Because of its 
trophic and anti-neoplasic effects on the colonic epithelium, stimulating specific 
populations of butyrigenic bacteria in the colon may well be the next important target 
for prebiotics. In situ measurement of specific bacterial activities remains problematic, but 
advances in functional genomics may provide a new avenue to explore the interactions 
between prebiotics, the intestinal microbiota and the host in health and disease. 

Blurring the Distinctions Between Prebiotics, Dietary Fibers, and Other 
Fermentable Dietary Carbohydrates in the Colon 

The greatest volume of research and evidence for prebiotic effects has been accrued for 
fructo-oligosaccharides and inulin, but there is accumulating evidence of prebiotic actions 
by a number of non-digestible carbohydrates. Lactulose and galacto-oligosaccharides 
have strong claims to be classified as prebiotics, while there is promising evidence for 
prebiotic activity by isomalto-, xylo-, and soybean-oligosaccharides. There is growing 
interest in the impact of dietary fibers on the composition as well as the activity of the 
intestinal microbiota, and resistant starches and arabinoxylans in particular warrant further 
study for bifidogenic and other prebiotic effects. 

It has been hypothesized that synergies might exist between NDOs that stimulate a 
bifidogenic response and SCFA production in the proximal colon and larger 
polysaccharides that sustain a source of fermentable carbohydrate through to the distal 



302 Crittenden and Playne 

colon. ORAFTI (Belgium) market a prebiotic (Synergy 1) that includes both short chain 
fructo-oligosaccharides and the longer chain fructan inulin and have reported synergistic 
effects in this combination for a range of physiological effects (246). Similarly, 
complementary effects have been noted for FOS/inulin and resistant starches (72,247). 
Development of synergistic prebiotic combinations to optimize the composition and 
activity of the microbiota throughout the length of the intestinal tract, or to target specific 
intestinal regions (e.g., for treatment of IBD) is set to provide continuing avenues for 
future research. 



Effects on Human Health 

A growing understanding of the intestinal microbiota and its contribution to health and 
disease has enabled rational hypotheses to be developed for prebiotic interventions 
targeted to specific human populations. Testing of these hypotheses is still mostly centered 
at the animal model or pilot human trial stage. Prebiotic oligosaccharides are already used 
in some infant formulas and efforts to replicate the activities of HMOs are likely to 
continue. Although the effects of prebiotics overlap somewhat with probiotics, the 
prebiotic strategy does provide some potential advantages. Despite these physical and 
potentially physiological advantages, research into the clinical effects of prebiotics still 
lags that devoted to probiotics. 

There is good evidence that prebiotics can relieve constipation and control hepatic 
encephalopathy, and lactulose is currently used pharmaceutically for these purposes. 
Additionally, a number of other health targets proposed for prebiotics have accumulating 
evidence of benefits. The most promising targets have been discussed in this chapter and 
include increasing calcium uptake, boosting colonization resistance against intestinal 
pathogens, and ameliorating IBD. Evidence for these benefits is still largely preliminary, 
but is sufficiently encouraging to warrant continuing investigation. While research efforts 
have naturally focused on the health benefits of prebiotics, and to date few reports of 
deleterious effects have surfaced, further quantification of the potential risks of prebiotics 
at different doses, in combination with different diets, and for different demographics, both 
healthy and diseased should be conducted. It is also important that prebiotics be trialed in 
the context of total diets, since other dietary components, for example the presence of 
dietary fibers that influence intestinal transit rates, can be expected to affect the 
clinical outcomes. 

Recent years have seen marked progress in our understanding of the microecology 
of the gastrointestinal tract. However, we are still only at the very beginning of developing 
an appreciation of the functional relationships between the microbiota and the host, in 
health and disease. A more profound understanding of what constitutes a "healthy" 
intestinal microbiota composition, and which microbial groups and activities are involved 
in health and disease, is a prerequisite to the future development of prebiotics with 
specifically targeted health effects. The challenge remains to demonstrate clinically 
relevant benefits to health by prebiotic interventions in well-designed and controlled 
human trials. 



REFERENCES 

1. Bourlioux P, Koletzko B, Guarner F, Braesco V. The intestine and its microflora are partners 
for the protection of the host: report on the Danone Symposium " The Intelligent Intestine," 
held in Paris, June 14, 2002. Am J Clin Nutr 2003; 78:675-683. 



Modifying the Human Intestinal Microbiota with Prebiotics 303 

2. Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of 
resistant starch and nonstarch polysaccharides. Physiol Rev 2001; 81:1031-1064. 

3. Blum S, Schiffrin EJ. Intestinal Microflora and Homeostasis of the Mucosal Immune 
Response: Implications for Probiotic Bacteria? Curr Issues Intest Microbiol 2003; 4:53-60. 

4. Tlaskalova-Hogenova H, Stepankova R, Hudcovic T, et al. Commensal bacteria (normal 
microflora), mucosal immunity and chronic inflammatory and autoimmune diseases. 
Immunol Lett 2004; 93:97-108. 

5. Cummings JH, Macfarlane GT, Macfarlane S. Intestinal bacteria and ulcerative colitis. Curr 
Issues Intest Microbiol 2003; 4:9-20. 

6. Marteau P, Seksik P, Shanahan F. Manipulation of the bacterial flora in inflammatory bowel 
disease. Best practice res:. Clin Gastroenterol 2003; 17:47-61. 

7. Saunier K, Dore J. Gastrointestinal tract and the elderly: functional foods, gut microflora and 
healthy ageing. Dig Liver Dis 2002; 34:S19-S24. 

8. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003; 361:512-519. 

9. Tannock GW. Molecular assessment of intestinal microflora. Am J Clin Nutr 2001; 
73:410S^14S. 

10. Vaughan EE, de Vries MC, Zoetendal EG, Ben-Amor K, Akkermans ACL, de Vos WM. The 
intestinal LABs. Antonie van Leeuwenhoek 2002; 82:341-352. 

11. Crittenden R. An update on probiotic bifidobacteria. In: Salminen S, von Wright A, 
Ouwerhand A, eds. Lactic Acid Bacteria: Microbiological and Functional Aspects. New 
York: Marcel Dekker, 2004:125-157. 

12. Salminen S, Gorbach S, Lee Y-K, Benno Y. Human studies on probiotics: what 
is scientifically proven today? In: Salminen S, von Wright A, Ouwerhand A, eds. Lactic 
Acid Bacteria: Microbiological and Functional Aspects. New York: Marcel Dekker, 
2004:515-530. 

13. Topping DL, Fukushima M, Bird AR. Resistant starch as a prebiotic and synbiotic: state of the 
art. Proc Nutr Soc 2003; 62:171-176. 

14. Gyorgy P, Norris RF, Rose CS. Bifidus factor I. A variant of Lactobacillus bifidus requiring a 
special growth factor. Arch Biochem Biophys 1954; 48:193-201. 

15. Gyorgy P, Kuhn R, Rose CS, Zilliken F. Bifidus factor II. Its occurrence in milk from different 
species and in other natural products. Arch Biochem Biophys 1954; 48:202-208. 

16. Rasic JL, Kurmann JA. Bifidobacteria and their role. Boston, MA, U.S.A.: Birkhauser Verlag, 
1983. 

17. Modler HW. Bifidogenic factors — sources, metabolism and applications. Int Dairy J 1994; 
4:383-407. 

18. Saito H, Miyakawa H, Ishibashi N, Tamura Y, Hayasawa H, Shimamura S. Effect of iron-free 
and metal-bound forms of lactoferrin on the growth of bifidobacteria E.coli and S. aureus. 
Biosci Microflora 1996; 15:1-7. 

19. Samejima K, Yoshioka M, Tamura Z. Bifidus factors in carrot I. Chem Pharm Bull (Tokyo) 
1971; 19:166-177. 

20. Yoshioka M, Tamura Z. Bifidus factors in carrot II. Chem Pharm Bull (Tokyo) 1971; 
19:178-185. 

21. Kaneko T. A novel bifidogenic growth stimulator produced by Propionibacterium 
freudenreichii. Biosci Microflora 1999; 18:73-80. 

22. Mori H, Sato Y, Taketomo N, et al. Isolation and structural identification of bifidogenic 
growth stimulator produced by Propionibacterium freudenreichii. J Dairy Sci 1997; 
80:1959-1964. 

23. Benno K, Sawada T. The intestinal microflora of infants: composition of fecal flora in breast- 
fed and bottle-fed infants. Microbiol Immunol 1984; 28:975-986. 

24. Balmer SE, Wharton BA. Diet and fecal flora in the newborn: breast milk and infant formula. 
Arch Dis Child 1989; 64:1672-1677. 

25. Petuely F. Bifidusflora bei flaschenkindern durch bifidogene substanzen (bifidusfaktor). Z 
Kinderheilkd 1957; 79:174-179 (In German). 



304 Crittenden and Playne 

26. Ruttloff H, Taufel A, Krause W, Haenel H, Taufel K. Die intestinal-enzymatische spaltung 
von galakto-oligosacchariden im darm von tier und menschen mit besonderer berucksichti- 
gung von Lactobacillus bifidus. II. Mitt, zum intestinalen verhalten der lactulose. Nahrung 
1967; 11:39-46 (In German). 

27. Yazawa K, Imai K, Tamura Z. Oligosaccharides and polysaccharides specifically utilizable by 
bifidobacteria. Chem Pharm Bull (Tokyo) 1978; 26:3306-3311. 

28. Minami Y, Yazawa K, Tamura Z, Tanaka T, Yamamoto T. Selectivity of utilisation of 
galactosyl-oligosaccharides by bifidobacteria. Chem Pharm Bull 1983; 31: 1688—1691. 

29. Hidaka H, Eida T, Takizawa T, Tokunaga T, Tashiro Y. Effects of fructooligosaccharides on 
intestinal flora and human health. Bifidobacteria Microflora 1986; 5:37-50. 

30. Mitsuoka T, Hidaka H, Eida T. Effect of fructooligosaccharides on intestinal microflora. 
Nahrung 1987; 31:427-436. 

31. Gibson GR, Roberfroid MB. Dietary modulation of the human colonic microbiota — 
Introducing the concept of prebiotics. J Nutr 1995; 125:1401-1412. 

32. Playne MJ, Crittenden R. Commercially available oligosaccharides. Bull Int Dairy Fed 1996; 
313:10-22. 

33. Crittenden RG, Playne MJ. Production, properties and applications of food-grade 
oligosaccharides. Trends Food Sci Technol 1996; 7:353-361. 

34. van Loo J, Cummings J, Delzenne N, et al. Functional food properties of non-digestible 
oligosaccharides: a consensus report from the ENDO project (DGXII AIRII-CT94-1095). Br J 
Nutr 1999; 81:121-132. 

35. Kolida S, Tuohy K, Gibson GR. Prebiotic effects of inulin and oligofructose. Br J Nutr 2002; 
87:S193-S197. 

36. Taper HS, Roberfroid MB. Inulin/oligofructose and anticancer therapy. Br J Nutr 2002; 
87:S283-S286. 

37. Pool-Zobel B, van Loo J, Rowland I, Roberfroid MB. Experimental evidences on the potential 
of prebiotic fructans to reduce the risk of colon cancer. Br J Nutr 2002; 87:S273-S281. 

38. Buddington KK, Donahoo JB, Buddington RK. Dietary oligofructose and inulin protect mice 
from enteric and systemic pathogens and tumor inducers. J Nutr 2002; 132:472-477. 

39. Harmsen HJM, Raangs GC, Franks AH, Wildeboer-Veloo ACM, Welling GW. The effect of 
the prebiotic inulin and the probiotic Bifidobacterium longum on the fecal microflora of 
healthy volunteers measured by FISH and DGGE. Microb Ecol Health Dis 2002; 14:21 1-219. 

40. Ben XM, Zhou XY, Zhao WH, et al. Supplementation of milk formula with galacto- 
oligosaccharides improves intestinal micro-flora and fermentation in term infants. Chinese 
Med J 2004; 117:927-931. 

41. Boehm G, Lidestri M, Casetta P, et al. Supplementation of a bovine milk formula with an 
oligosaccharide mixture increases counts of fecal bifidobacteria in preterm infants. Arch Dis 
Childhood 2002; 86:F178-F181. 

42. Bouhnik Y, Flourie B, D'Agay-Abensour L, et al. Administration of transgalacto- 
oligosaccharides increases fecal bifidobacteria and modifies colonic fermentation metabolism 
in healthy humans. J Nutr 1997; 127:444-448. 

43. Ito M, Deguchi Y, Matsumoto K, Kimura M, Onodera N, Yajima T. Influence of galacto- 
oligosaccharides on human fecal microflora. J Nutr Sci Vitaminol 1993; 39:635-640. 

44. Terada A, Hara H, Kataoka M, Mitsuoka T. Effect of lactulose on the composition and 
metabolic activity of the human fecal flora. Microbial Ecol Health Dis 1992; 5:43-50. 

45. Ballongue J, Schumann C, Quignon P. Effects of lactulose and lactitol on colonic microflora 
and enzymatic activity. Scand J Gastroenterol 1997; 32:41-44. 

46. Tuohy KM, Ziemer CJ, Klinder A, Knobel Y, Pool-Zobel BL, Gibson GR. A human volunteer 
study to determine the prebiotic effects of lactulose powder on human colonic microbiota. 
Microb Ecol Health Dis 2002; 14:165-173. 

47. Bouhnik Y, Attar A, Joly FA, Riottot M, Dyard F, Flourie B. Lactulose ingestion increases 
fecal bifidobacterial counts: a randomised double-blind study in healthy humans. Eur J Clin 
Nutr 2004; 58:462-466. 



Modifying the Human Intestinal Microbiota with Prebiotics 305 

48. Boehm G, Stahl B. In: Mattila-Sandholm T, Saarela M, eds. Oligosaccharides. In: Functional 
Dairy Products. Cambridge: Woodhead Publishing/CRC Press, 2003:203-243. 

49. Fujita K, Hara K, Sakai S, et al. Effects of 4-(3-D-galactosylsucrose (lactosucrose) on 
intestinal flora and its digestibility in humans. J Jpn Soc Starch Sci 1991; 38:249-255. 

50. Yoneyama M, Mandai T, Aga H, Fujii K, Sakai S, Katayama Y. Effects of 4-(3-D- 
galactosylsucrose (lactosucrose) intake on intestinal flora in healthy humans. J Jpn Soc Nutr 
Food Sci 1992;45:101-107. 

51. Hara H, Li S-T, Sasaki M, et al. Effective dose of lactosucrose on fecal flora and fecal 
metabolites of humans. Bifidobacteria Microflora 1994; 13:51-63. 

52. Ohkusa T, Ozaki Y, Sato C, Mikuni K, Ikeda H. Long-term ingestion of lactosucrose increases 
bifidobacterium sp. in human fecal flora. Digestion 1995; 56:415-420. 

53. Djouzi Z, Andrieux C, Pelenc V, et al. Degradation and fermentation of alpha-gluco- 
oligosaccharides by bacterial strains from human colon: in vitro and in vivo studies in 
gnotobiotic rats. J Appl Bacteriol 1995; 79:117-127. 

54. Okazaki M, Fujikawa S, Matsumoto N. Effect of xylo-oligosaccharide on the growth of 
bifidobacteria. Bifidobacteria Microflora 1990; 9:77-86. 

55. Campbell JM, Jr., Fahey GC, Wolf BW. Selected indigestible oligosaccharides affect large 
bowel mass, cecal and fecal short-chain fatty acids, pH and microflora in rats. J Nutr 1997; 
127:130-136. 

56. Kohmoto T, Fukui F, Takaku H, Machida Y, Arai M, Mitsuoka T. Effect of isomalto- 
oligosaccharides on human facal flora. Bifidobacteria Microflora 1988; 7:61-69. 

57. Kohmoto T, Fukui F, Takaku H, Mitsuoka T. Dose-response test of isomaltooligosaccharides 
for increasing fecal bifidobacteria. Agric Biol Chem 1991; 55:2157-2159. 

58. Kaneko T, Kohmoto T, Kikuchi H, et al. Effects of isomaltoologosaccharides intake on 
defecation and intestinal environment in healthy volunteers. J Home Econ Jpn 1993; 
44:245-254 (in Japanese). 

59. Kaneko T, Kohmoto T, Kikuchi H, Shiota M, lino H, Mitsuoka T. Effects of 
isomaltooligosaccharides with different degrees of polymerisation on human fecal 
bifidobacteria. Biosci Biotech Biochem 1994; 58:2288-2290. 

60. Benno Y, Endo K, Shiragani N, Sayama K, Mitsuoka T. Effects of raffinose intake on human 
fecal microflora. Bifidobacteria Microflora 1987; 6:59-63. 

61 . Hara T, Ikeda N, Hatsumi K, Watabe J, lino H, Mitsuoka T. Effects of small amount ingestion 
of soybean oligosaccharides on bowel habits and fecal flora of volunteers. Jpn J Nutr 1997; 
55:79-84. 

62. Hayakawa K, Mitzutani J, Wada K, Masai T, Yoshihara I, Mitsuoka T. Effects of soybean 
oligosaccharides on human fecal microflora. Microb Ecol Health Dis 1990; 3:293-303. 

63. Wada K, Watabe J, Mizutani J, Tomoda M, Suzuki H, Saitoh Y. Effects of soybean 
oligosaccharides in a beverage on human fecal flora and metabolites. J Agric Chem Soc Jpn 
1992;66:127-135. 

64. Jie Z, Bang-Yao L, Ming-Jie X, et al. Studies on the effects of polydextrose intake on 
physiologic functions in Chinese people. Am J Clin Nutr 2000; 72:1503-1509. 

65. Asano T, Yuasa K, Kunugita K, Teraja T, Mitsuoka T. Effects of gluconic acid on human fecal 
bacteria. Microbial Ecol Health Dis 1994; 7:247-256. 

66. Kleessen B, Stoof G, Proll J, Schmiedl D, Noack J, Blaut M. Feeding resistant starch affects 
fecal and cecal microflora and short-chain fatty acids in rats. J Anim Sci 1997; 75:2453-2462. 

67. Brown IL, Wang X, Topping DL, Playne MJ, Conway PL. High amylose maize starch as a 
versatile prebiotic for use with probiotic bacteria. Food Aust 1998; 50:603-610. 

68. Wang X, Brown IL, Evans AJ, Conway PL. The protective effects of high amylose maize 
(amylomaize) starch granules on the survival of Bifidobacterium spp. in the mouse intestinal 
tract. J Appl Microbiol 1999; 87:631-639. 

69. Silvi S, Rumney CJ, Cresci A, Rowland IR. Resistant starch modifies gut microflora and 
microbial metabolism in human flora-associated rats inoculated with feces from Italian and 
U.K. donors. J Appl Microbiol 1999; 86:521-530. 



306 Crittenden and Playne 

70. Bielecka M, Biedrzycka E, Majkowska A, Juskiewicz J, Wroblewska M. Effect of non- 
digestible oligosaccharides on gut microecosystem in rats. Food Res Int 2002; 35:139-144. 

7 1 . Wang X, Brown IL, Khaled D, Mahoney MC, Evans AJ, Conway PL. Manipulation of colonic 
bacteria and volatile fatty acid production by dietary high amylose maize (amylomaize) starch 
granules. J Appl Microbiol 2002; 93:390-397. 

72. Le Blay G, Michel C, Blottiere H, Cherbut C. Raw potato starch and short-chain fructo- 
oligosaccharides affect the composition and metabolic activity of rat intestinal microbiota 
differently depending on the caecocolonic segment involved. J Appl Microbiol 2003; 
94:312-320. 

73. van Laere KM, Hartemink R, Bosveld M, Schols HA, Voragen AG. Fermentation of plant cell 
wall derived polysaccharides and their corresponding oligosaccharides by intestinal bacteria. 
J Agric Food Chem 2000; 48:1644-1652. 

74. Crittenden R, Karppinen S, Ojanen S, et al. In vitro fermentation of cereal dietary fiber 
carbohydrates by probiotic and intestinal bacteria. J Sci Food Agric 2002; 82:1-9. 

75. Okubo T, Ishihara N, Takahashi H, et al. Effects of partially hydrolyzed guar gum intake on 
human intestinal microflora and its metabolism. Biosci Biotech Biochem 1994; 58:364-1369. 

76. Muir JG, Yeow EG, Keogh J, et al. Combining wheat bran with resistant starch has more 
beneficial effects on fecal indexes than does wheat bran alone. Am J Clin Nutr 2004; 
79:1020-1028. 

77. Champ MM. Physiological aspects of resistant starch and in vivo measurements. J AOAC Int 
2004; 87:749-755. 

78. Andoh A, Tsujikawa T, Fujiyama Y. Role of dietary fiber and short-chain fatty acids in the 
colon. Curr Pharm Design 2003; 9:347-358. 

79. Muir JG, Yeow EGW. Importance of combining indigestible carbohydrate with protein 
sources in the diet: implications for reducing colorectal cancer risk. Proc Nutr Soc Aust 2000; 
24:196-204. 

80. Smiricky-Tjardes MR, Grieshop CM, Flickinger EA, Bauer LL, Fahey GC, Jr. Dietary 
galactooligosaccharides affect ileal and total-tract nutrient digestibility, ileal and fecal 
bacterial concentrations, and ileal fermentative characteristics of growing pigs. J Animal Sci 
2003; 81:2535-2545. 

81. Ito M, Deguchi Y, Miyamori A, et al. Effects of administration of galacto-oligosaccharides on 
the human fecal microflora, stool weight and abdominal sensation. Microb Ecol Health Dis 
1990; 3:285-292. 

82. Barrangou R, Altermann E, Hutkins R, Cano R, Klaenhammer TR. Functional and 
comparative genomic analyses of an operon involved in fructooligosaccharide utilization by 
Lactobacillus acidophilus. Proc Nat Acad Sci USA 2003; 100:8957-8962. 

83. Kaplan H, Hutkins RW. Metabolism of fructooligosaccharides by Lactobacillus paracasei 
1195. Appl Environ Microbiol 2003; 69:2217-2222. 

84. Roberfroid MB, Van Loo JAE, Gibson GR. The bifidogenic nature of chicory inulin and its 
hydrolysis products. J Nutr 1998; 128:11-19. 

85. Harmsen HJM, Wildeboer-Veloo ACM, Raangs GC, et al. Analysis of intestinal flora 
development in breast-fed and formula-fed infants by using molecular identification and 
detection methods. J Pediatr Gastroenterol Nutr 2000; 30:61-67. 

86. Mountzouris KC, McCartney AL, Gibson GR. Intestinal microflora of human infants and 
current trends for its nutritional modulation. Br J Nutr 2002; 87:405-420. 

87. Brand-Miller J, McVeagh P. Human milk oligosaccharides: 130 reasons to breast-feed. Br 
J Nutr 1999; 82:333-335. 

88. Kunz C, Rudloff S, Baier W, Klein N, Strobel S. Oligosaccharides in human milk: structural, 
functional, and metabolic aspects. Ann Rev Nutr 2000; 20:699-722. 

89. Adlerberth I. Establishment of a normal intestinal microflora in the newborn infant. In: 
Hansen LA, Yolken RH, eds. Probiotics,Other Nutritional Factors and Intestinal Microflora 
(Nestle Nutrition Workshop Series, Vol 42, Nestec Ltd, Vevey). Philadelphia: Lippincott- 
Raven, 1999:63-78. 



Modifying the Human Intestinal Microbiota with Prebiotics 307 

90. Heavey PM, Savage SA, Parrett A, Cecchini C, Edwards CA, Rowland IR. Protein- 
degradation products and bacterial enzyme activities in feces of breast-fed and formula-fed 
infants. Br J Nutr 2003; 89:509-515. 

91. Edwards CA, Parrett AM. Intestinal flora during the first months of life: new perspectives. Br 
J Nutr 2002; 88:S11-S18. 

92. Moro G, Minoli I, Mosca M, et al. Dosage-related bifidogenic effects of galacto- and 
fructooligosaccharides in formula-fed term infants. J Pediatr Gastroenterol Nutr 2002; 
34:291-295. 

93. Newburg DS. Human milk glycoconjugates that inhibit pathogens. Curr Med Chem 1999; 
6:117-127. 

94. Martin-Sosa S, Martin MJ, Hueso P. The sialylated fraction of milk oligosaccharides is 
partially responsible for binding to enterotoxigenic and uropathogenic Escherichia coli 
human strains. J Nutr 2002; 132:3067-3072. 

95. Morrow AL, Ruiz-Palacios GM, Altaye M, et al. Human milk oligosaccharides are associated 
with protection against diarrhea in breast-fed infants. J Pediatr 2004; 145:297-303. 

96. Wang B, Brand-Miller J. The role and potential of sialic acid in human nutrition. Eur J Clin 
Nutr 2003; 57:1351-1369. 

97. Vincent SJ, Faber EJ, Neeser JR, Stingele F, Kamerling JP. Structure and properties of the 
exopolysaccharide produced by Streptococcus macedonicus Scl36. Glycobiology 2001; 
11:131-139. 

98. Rencurosi A, Poletti L, Guerrini M, Russo G, Lay L. Human milk oligosaccharides: an 
enzymatic protection step simplifies the synthesis of 3'- and 6 / -0-sialyllactose and their 
analogues. Carbohydr Res 2002; 337:473-483. 

99. La Ferla B, Prosperi D, Lay L, Russo G, Panza L. Synthesis of building blocks of human milk 
oligosaccharides. Fucosylated derivatives of the lacto- and neolacto-series. Carbohydr Res 
2002; 337:1333-1342. 

100. Dumon C, Samain E, Priem B. Assessment of the two Helicobacter pylori alpha- 
1,3-fucosyltransferase ortholog genes for the large-scale synthesis of LewisX human milk 
oligosaccharides by metabolically engineered Escherichia coli. Biotechnol Prog 2004; 
20:412-419. 

101. Ouwehand A, Isolauri E, Salminen S . The role of the intestinal microflora for the development 
of the immune system in early childhood. Eur J Nutr 2002; 41:132-137. 

102. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic Estonian 
and Swedish 2-year old children. Clin Exp Allergy 1999; 29:342-346. 

103. Bjorksten B, Sepp E, Julge K, Voor T, Mikelsaar M. Allergy development and the intestinal 
microflora during the first year of life. J Allergy Clin Immunol 2001; 108:516-520. 

104. Bottcher M, Sandin A, Norin E, Midtvedt T, Bjorksten B. Microflora associated 
characteristics in feces from allergic and non-allergic children. Clin Exp Allergy 2000; 
30:590-596. 

105. Gronlund MM, Arvilommi H, Kero P, Lehtonen OP, Isolauri E. Importance of intestinal 
colonization in the maturation of humoral immunity in early infancy: a prospective follow up 
study of healthy infants aged 0-6 months. Arch Dis Child 2000; 83:F186-F192. 

106. Kalliomaki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of 
neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin 
Immunol 2001; 107:129-134. 

107. Kirjavainen PV, Apostolou E, Arvola T, Salminen SJ, Gibson GR, Isolauri E. Characterizing 
the composition of intestinal microflora as a prospective treatment target in infant allergic 
disease. FEMS Immunol Med Micrbiol 2001; 32:1-7. 

108. Kirjavainen PV, Arvola T, Salminen SJ, Isolauri E. Aberrant composition of gut microbiota of 
allergic infants: a target of bifidobacterial therapy at weaning? Gut 2002; 51:51-55. 

109. He F, Ouwehand AC, Isolauri E, Hashimoto H, Benno Y, Salminen S. Comparison of mucosal 
adhesion and species identification of bifidobacteria isolated from healthy and allergic infants. 
FEMS Immunol Med Microbiol 2001; 30:43-47. 



308 Crittenden and Playne 

110. Ouwehand AC, Isolauri E, He F, Hashimoto H, Benno Y, Salminen S. Differences in 
Bifidobacterium flora composition in allergic and healthy infants. J Allergy Clin Immunol 
2001; 108:144-145. 

111. Mackie RI, Sghir A, Gaskins HR. Developmental microbial ecology of the neonatal 
gastrointestinal tract. Am J Clin Nutr 1999; 69:1035S-1045S. 

112. Matsuki T, Watanabe K, Tanaka R, Fukuda M, Oyaizu H. Distribution of bifidobacterial 
species in human intestinal microflora examined with 16S rRNA-gene-targeted species- 
specific primers. Appl Environ Microbiol 1999; 65:4506-4512. 

113. Kalliomaki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E. Probiotics in 
primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet 2001; 
357:1076-1079. 

1 14. Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E E. Probiotics and prevention of 
atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 2003; 
361:1869-1871. 

115. Nagura T, Hachimura S, Hashiguchi M, et al. Suppressive effect of dietary raffinose on 
T-helper 2 cell-mediated immunity. Br J Nutr 2002; 88:421-427. 

116. Yoshida T, Hirano A, Wada H, Takahashi K, Hattori M. Alginic acid oligosaccharide 
suppresses Th2 development and IgE production by inducing IL-12 production. Int Arch 
Allergy Immunol 2004; 133:239-247. 

117. Marteau P, Pochart P, Dore J, Bera-Maillet C, Bernalier A, Corthier G. Comparative study of 
bacterial groups within the human cecal and fecal microbiota. Appl Environ Microbiol 2001; 
67:4939-4942. 

118. Franks AH, Harmsen HJ, Raangs GC, Jansen F, Schut GW. Variations of bacterial 
populations in human feces measured by fluorescent in situ hybridization with group-specific 
16S rRNA-targeted oligonucleotide probes. Appl Environ Microbiol 1998; 64:3336-3345. 

119. Sghir A, Gramet G, Suau A, Rochet V, Pochart P, Dore J. Quantification of bacterial groups 
within human fecal flora by oligonucleotide probe hybridization. Appl Environ Microbiol 
2000; 66:2263-2266. 

120. Harmsen HJ, Wildeboer-Veloo AC, Grijpstra J, Knol J, Degener JE, Welling GW. 
Development of 16S rRNA-based probes for the Coriobacterium group and the Atopobium 
cluster and their application for enumeration of Coriobacteriaceae in human feces from 
volunteers of different age groups. Appl Environ Microbiol 2000; 66:4523-4527. 

121. Requena T, Burton J, Matsuki T, et al. Identification, detection, and enumeration of human 
Bifidobacterium species by PCR targeting the transaldolase gene. Appl Environ Microbiol 
2002; 68:2420-2427. 

122. Tannock GW. Analysis of the intestinal microflora using molecular methods. Eur J Clin Nutr 
2002; 56:S44-S49. 

123. Mangin I, Bouhnik Y, Bisetti N, Decaris B. Molecular monitoring of human intestinal 
Bifidobacterium strain diversity. Res Mincrobiol 1999; 150:343-350. 

124. Tannock GW. The bifidobacterial and Lactobacillus microflora of humans. Clin Rev Allergy 
Immunol 2002; 22:231-253. 

125. Satokari RM, Vaughan EE, Akkermans ADL, Saarela M, de Vos WM. Bifidobacterial 
diversity in human feces detected by genus-specific PCR and denaturing gradient gel 
electrophoresis. Appl Environ Microbiol 2001; 67:504-513. 

126. Matsuki T, Watanabe K, Tanaka R, Fukuda M, Oyaizu H. Distribution of bifidobacterial 
species in human intestinal microflora examined with 16S rRNA-gene-targeted species- 
specific primers. Appl Environ Microbiol 1999; 65:4506-4512. 

127. Hayashi H, Sakamoto M, Benno Y. Phylogenetic analysis of the human gut microbiota using 
16S rDNA clone libraries and strictly anaerobic culture-based methods. Microbiol Immunol 
2002; 46:535-548. 

128. Matsuki T, Watanabe K, Fujimoto J, et al. Development of 16S rRNA-gene-targeted group- 
specific primers for the detection and identification of predominant bacteria in human feces. 
Appl Environ Micrbiol 2002; 68:5445-5451. 



Modifying the Human Intestinal Microbiota with Prebiotics 309 

129. Crittenden RG. Prebiotics. In: Tannock GW, ed. Probitoics: A Critical Review. Wymondham 
U.K.: Horizon Scientific Press, 1999:141-156. 

130. Rao AV. Dose-response effects of inulin and oligofructose on intestinal bifidogenesis effects. 
JNutr 1999; 129:1442S-1445S. 

131. Saito Y, Hamanaka Y, Saito K, Takizawa S, Benno Y. Stability of species composition of 
fecal bifidobacteria in human subjects during fermented milk administration. Curr Microbiol 
2002; 44:368-373. 

132. Ventura M, Elli M, Reniero R, Zink R. Molecular microbial analysis of Bifidobacterium 
isolates from different environments by the species-specific amplified ribosomal DNA 
restriction analysis (ARDRA). FEMS Microbiol Ecol 2001; 36:113-121. 

133. Alander M, Matto J, Kneifel W, et al. Effect of galacto-oligosaccharide supplementation on 
human fecal microflora and on survival and persistence of Bifidobacterium lactis Bb-12 in the 
gastrointestinal tract. Int Dairy J 2001; 11:817-825. 

134. Satokari RM, Vaughan EE, Akkermans ADL, Saarela M, de Vos WM. Polymerase chain 
reaction and denaturing gradient gel electrophoresis monitoring of fecal Bifidobacterium 
populations in a prebiotic and probiotic feeding trial. Syst Appl Microbiol 2001; 24:227-231. 

135. Malinen E, Matto J, Salmitie M, Alander M, Saarela M, Palva A. PCR-ELISA— II: Analysis 
of Bifidobacterium populations in human fecal samples from a consumption trial with 
Bifidobacterium lactis Bb-12 and a galacto-oligosaccharide preparation. Syst Appl Microbiol 
2002; 25:249-258. 

136. Harmsen HJM, Raangs GC, Franks AH, Wildeboer-Veloo ACM, Welling GW. The effect of 
the prebiotic inulin and the probiotic Bifidobacterium longum on the fecal microflora of 
healthy volunteers measured by FISH and DGGE. Microb Ecol Health Dis 2002; 
14:211-219. 

137. Zoetendal EG, Akkermans AD, de Vos WM. Temperature gradient gel electrophoresis 
analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities 
of active bacteria. Appl Environ Microbiol 1998; 64:3854-3859. 

138. Tannock GW. A fresh look at feces. Microbiol Aust 2003; 24:34-35. 

139. Tannock GW, Munro K, Bibiloni R, et al. Impact of consumption of oligosaccharide- 
containing biscuits on the fecal microbiota of humans. Appl Environ Microbiol 2004; 
70:2129-2136. 

140. Schultz M, Timmer A, Herfarth HH, Sartor RB, Vanderhoof JA, Rath HC. Lactobacillus GG 
in inducing and maintaining remission of Crohn's disease. BMC Gastroenterol 2004; 15:5. 

141. Linskens RK, Huijsdens XW, Savelkoul PHM, Vandenbroucke-Grauls CMJE, 
Meuwissen SMG. The bacterial flora in inflammatory bowel disease: Current insights in 
pathogenesis and the influence of antibiotics and probiotics. Scand J Gastroenterol 2001; 
36:S29-S40. 

142. Favier C, Neut C, Mizon C, Cortot A, Colombel JF, Mizon J. Fecal beta-D-galactosidase 
production and Bifidobacteria are decreased in Crohn's disease. Dig Dis Sci 1997; 
42:817-822. 

143. Rumi G, Tsubouchi R, Okayama M, Kato S, Mozsik G, Takeuchi K. Protective effect of 
lactulose on dextran sulfate sodium-induced colonic inflammation in rats. Dig Dis Sci 2004; 
49:1466-1472. 

144. Cherbut C, Michel C, Lecannu G. The prebiotic characteristics of fructooligosaccharides are 
necessary for reduction of TNBS-induced colitis in rats. J Nutr 2003; 133:21-27. 

145. Holma R, Juvonen P, Asmawi MZ, Vapaatalo H, Korpela R. Galacto-oligosaccharides 
stimulate the growth of bifidobacteria but fail to attenuate inflammation in experimental 
colitis in rats. Scand J Gastroenterol 2002; 37:1042-1047. 

146. Jacobasch G, Schmiedl D, Kruschewski M, Schmehl K. Dietary resistant starch and chronic 
inflammatory bowel diseases. Int J Colorectal Dis 1999; 14:201-211. 

147. Moreau NM, Martin LJ, Toquet CS, et al. Restoration of the integrity of rat caeco-colonic 
mucosa by resistant starch, but not by fructo-oligosaccharides, in dextran sulfate sodium- 
induced experimental colitis. Br J Nutr 2003; 90:75-85. 



310 Crittenden and Playne 

148. Kanauchi Y, Fujiyama K, Mitsuyama Y, et al. Increased growth of Bifidobacterium and 
Eubacterium by germinated barley foodstuff, accompanied by enhanced butyrate production 
in healthy volunteers. Int J Mol Med 1999; 3:175-179. 

149. Oikarinen S, Heinonen S, Karppinen S, et al. Plasma enterolactone or intestinal 
Bifidobacterium levels do not explain adenoma formation in multiple intestinal neoplasia 
(Min) mice fed with two different types of rye-bran fractions. Br J Nutr 2003; 90:1 19-125. 

150. Bamba T, Kanauchi O, Andoh A, Fujiyama Y. A new prebiotic from germinated barley for 
nutraceutical treatment of ulcerative colitis. J Gastroenterol Hepatol 2002; 17:818-824. 

151. Fukuda M, Kanauchi O, Araki Y, et al. Prebiotic treatment of experimental colitis with 
germinated barley foodstuff: a comparison with probiotic or antibiotic treatment. Int J Mol 
Med 2002; 9:65-70. 

152. Kanauchi K, Mitsuyama Y, Araki A, Andoh A. Modification of intestinal flora in the 
treatment of inflammatory bowel disease. Curr Pharm Des 2003; 9:333-346. 

153. Kanauchi T, Suga M, Tochihara T, et al. Treatment of ulcerative colitis by feeding with 
germinated barley foodstuff: first report of a multicenter open control trial. J Gastroenterol 
2002; 37:67-72. 

154. Mitsuoka T. Recent trends in research on intestinal flora. Bifidobacteria Microflora 1982; 
1:3-24. 

155. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related changes in intestinal bacterial 
populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid 
profiles. Gut 2001; 48:198-205. 

156. Hopkins MJ, Sharp R, Macfarlane GT. Variation in human intestinal microbiota with age. Dig 
Liver Dis 2002; 34:S12-S18. 

157. Canzi E, Casiraghi MC, Zanchi R, et al. Yogurt in the diet of the elderly: a preliminary 
investigation into its effect on the gut ecosystem and lipid metabolism. Lait 2002; 
82:713-723. 

158. Silvi S, Verdenelli MC, Orpianesi C, Cresci A. EU project Crownalife: functional foods, gut 
microflora and healthy ageing - Isolation and identification of Lactobacillus and 
Bifidobacterium strains from fecal samples of elderly subjects for a possible probiotic use 
in functional foods. J Food Eng 2003; 56:195-200. 

159. Hopkins MJ, MacFarlane GT. Changes in predominant bacterial populations in human feces 
with age and with Clostridium difficile infection. J Med Micrbiol 2002; 51:448-454. 

160. Brown I, Warhurst M, Arcot J, et al. Fecal numbers of bifidobacteria are higher in pigs fed 
Bifidobacterium longum with a high amylose cornstarch than with a low amylose cornstarch. 
J Nutr 1997; 127:1822-1827. 

161. Challa A, Rao DR, Chawan CB, Shackelford L. Bifidobacterium longum and lactulose 
suppress azoxymethane-induced colonic aberrant crypt foci in rats. Carcinogenesis 1997; 
18:517-521. 

162. Rowland IR, Rumney CJ, Coutts JT, Lievense LC. Effect of Bifidobacterium longum and 
inulin on gut bacterial metabolism and carcinogen-induced aberrant crypt foci in rats. 
Carcinogenesis 1998; 19:281-285. 

163. Gallaher DD, Khil J. The effect of synbiotics on colon carcinogenesis in rats. J Nutr 1999; 
129:1483S-1487S. 

164. Femia AP, Luceri C, Dolara P, et al. Antitumorigenic activity of the prebiotic inulin enriched 
with oligofructose in combination with the probiotics Lactobacillus rhamnosus and 
Bifidobacterium lactis on azoxymethane-induced colon carcinogenesis in rats Carcinogenesis 
2002;23:1953-1960. 

165. Asahara T, Nomoto K, Shimizu K, Watanuki M, Tanaka R. Increased resistance of mice 
to Salmonella enterica serovar Typhimurium infection by synbiotic administration of 
Bifidobacteria and transgalactosylated oligosaccharides. J Appl Microbiol 2001; 
91:985-996. 

166. Lamoureux L, Roy D, Gauthier SF. Production of oligosaccharides in yogurt containing 
bifidobacteria and yogurt cultures. J Dairy Sci 2002; 85:1058-1069. 



Modifying the Human Intestinal Microbiota with Prebiotics 311 

167. Hung MN, Lee BH. Purification and characterization of a recombinant beta-galactosidase 
with transgalactosylation activity from Bifidobacterium infantis HL96. Appl Micrbiol 
Biotechnol 2002; 58:439-445. 

168. Jorgensen F, Hansen OC, Stougaard P. High-efficiency synthesis of oligosaccharides with a 
truncated beta-galactosidase from Bifidobacterium bifidum. Appl Microbiol Biotechnol 2001; 
57:647-652. 

169. Crittenden R, Laitila A, Forssell P, et al. Adhesion of bifidobacteria to granular starch and its 
implications in probiotic technologies. Appl Environ Microbiol 2001; 67:3469-3475. 

170. Schell MA, Karmirantzou M, Snel B, et al. The genome sequence of Bifidobacterium longum 
reflects its adaptation to the human gastrointestinal tract. Proc Natl Acad Sci USA 2002; 
99:14422-14427. 

171. Hopkins MJ, Cummings JH, Macfarlane GT. Inter-species differences in maximum specific 
growth rates and cell yields of bifidobacteria cultured on oligosaccharides and other simple 
carbohydrate sources. J Appl Microbiol 1998; 85:381-386. 

172. Crittenden RG, Morris LF, Harvey ML, Tran LT, Mitchell HL, Playne MJ. Selection of a 
Bifidobacterium strain to complement resistant starch in a synbiotic yoghurt. J Appl Microbiol 
2001;90:268-278. 

173. Beaugerie L, Petit JC. Microbial-gut interactions in health and disease. Antibiotic-associated. 
Best Pract Res Clin Gastroenterol 2004; 18:337-352. 

174. Sherman RG, Prusinski L, Ravenel MC, Joralmon RA. Oral candidosis. Quintessence Int 
2002; 33:521-532. 

175. Dan M, Kaneti N, Levin D, Poch F, Samra Z. Vaginitis in a gynecologic practice in Israel: 
causes and risk factors. Isr Med Assoc J 2003; 5:629-632. 

176. Plummer S, Weaver MA, Harris JC, Dee P, Hunter J. Clostridium difficile pilot study: 
effects of probiotic supplementation on the incidence of C. difficile. Int Microbiol 2004; 
7:59-62. 

177. Gill HS. Probiotics to enhance anti-infective defences in the gastrointestinal tract. Best Pract 
Res Clin Gastroenterol 2003; 17:755-773. 

178. Marteau P, Seksik P, Jian R. Probiotics and intestinal health effects: a clinical perspective. Br 
JNutr2002; 88:S51-S57. 

179. Kanamori Y, Hashizume K, Kitano Y, et al. Anaerobic dominant flora was reconstructed by 
synbiotics in an infant with MRSA enteritis. Pediatr Int 2003; 45:359-362. 

180. Stern RS. Photocarcinogenicity of drugs. Toxicol Lett 1998; 102-103:389-392. 

181. Dancer SJ. The problem with cephalosporins. J Antimicrob Chemother 2001; 48:463-478. 

182. Patterson JA, Burkholder KM. Application of prebiotics and probiotics in poultry production. 
Poultry Sci 2003; 82:627-631. 

183. Flickinger EA, van Loo J, Jr., Fahey GC. Nutritional responses to the presence of inulin and 
oligofructose in the diets of domesticated animals: a review. Crit Rev Food Sci Nutr 2003; 
43:19-60. 

184. Mattila-Sandholm T, Blum S, Collins JK, et al. Probiotics: towards demonstrating efficacy. 
Trends Food Sci Technol 1999; 10:393-399. 

185. von Wright A, Vilpponen-Salmela T, Llopis MP, et al. The survival and colonic adhesion of 
Bifidobacterium infantis in patients with ulcerative colitis. Int Dairy J 2002; 12:197-200. 

186. Shimakawa Y, Matsubara S, Yuki N, Ikeda M, Ishikawa F. Evaluation of Bifidobacterium 
breve strain Yakult-fermented soymilk as a probiotic food. Int J Food Microbiol 2003; 
81:131-136. 

187. Fujiwara S, Seto Y, Kimura A, Hashiba H. Intestinal transit of an orally administered 
streptomycin-rifampicin-resistant variant of Bifidobacterium longum SBT2928: its long-term 
survival and effect on the intestinal microflora and metabolism. J Appl Micrbiol 2001; 
90:43-52. 

188. Brigidi P, Swennen E, Vitali B, Rossi M, Matteuzzi D. PCR detection of Bifidobacterium 
strains and Streptococcus thermophilus in feces of human subjects after oral bacteriotherapy 
and yogurt consumption. Int J Food Microbiol 2003; 81:203-209. 



312 Crittenden and Playne 

189. McCartney AL, Wenzhi W, Tannock GW. Molecular analysis of the composition of the 
bifidobacteria! and Lactobacillus microflora of humans. Appl Environ Microbiol 1996; 
62:4608-4613. 

190. Kimura K, McCartney AL, McConnell MA, Tannock GW. Analysis of fecal populations of 
bifidobacteria and lactobacilli and investigation of the immunological responses of their 
human hosts to the predominant strains. Appl Environ Microbiol 1997; 63:3394-3398. 

191. Mangin I, Bouhnik Y, Bisetti N, Decaris B. Molecular monitoring of human intestinal 
Bifidobacterium strain diversity. Res Mincrobiol 1999; 150:343-350. 

192. Dunne C, O'Mahony L, Murphy L, et al. In vitro selection criteria for probiotic bacteria of 
human origin: correlation with in vivo findings. Am J Clin Nutr 2001; 73:386S-392S. 

193. Germond JE, Mamin O, Mollet B. Species specific identification of nine human 
Bifidobacterium spp. in feces. Syst Appl Micrbiol 2002; 25:536-543. 

194. Zopf D, Roth S. Oligosaccharide anti-infective agents. Lancet 1996; 347:1017-1021. 

195. Kunz C. Microbial receptor analogs in human milk: structural and functional aspects. In: 
Hansen LA, Yolken RH, eds. Probiotics, Other Nutritional Factors and Intestinal Microflora. 
(Nestle Nutrition Workshop Series, Vol. 42, Nestec Ltd, Vevey). Philadelphia: Lippincott- 
Raven, 1999:157-173. 

196. Kunz C, Rudloff S. Health benefits of milk-derived carbohydrates. Bull Int Dairy Fed 2002; 
375:72-79. 

197. Kunz C, Rudloff S. Biological functions of oligosaccharides in human milk. Acta Paediatr 
1993; 82:903-912. 

198. Orchel A, Dzierzewicz Z, Parfiniewicz B, Weglarz L, Wilczok T. Butyrate-induced 
differentiation of colon cancer cells is PKC and JNK dependent. Dig Dis Sci 2005; 
50:490-498. 

199. Miller SJ. Cellular and physiological effects of short-chain fatty acids. Mini Rev Med Chem 
2004; 4:839-845. 

200. Lupton JR. Microbial degradation products influence colon cancer risk: the butyrate 
controversy. J Nutr 2004; 134:479-482. 

201. Young GP, Le Leu RK. Resistant starch and colorectal neoplasia. J AOAC Int 2004; 
87:775-786. 

202. De Preter V, Geboes K, Verbrugghe K, et al. The in vivo use of the stable isotope-labelled 
biomarkers lactose- [N-15]ureide and [H-2(4)]tyrosine to assess the effects of pro- and 
prebiotics on the intestinal flora of healthy human volunteers. Br J Nutr 2004; 92:439-446. 

203. Rowland IR, Tanaka R. The effects of transgalactosylated oligosaccharides on gut flora 
metabolism in rats associated with a human fecal microflora. J Appl Bacteriol 1993; 
74:667-674. 

204. Phillips J, Muir JG, Birkett A, et al. Effect of resistant starch on fecal bulk and fermentation- 
dependent events in humans. Am J Clin Nutr 1995; 62:121-130. 

205. Hylla S, Gostner A, Dusel G, et al. Effects of resistant starch on the colon in healthy 
volunteers: possible implications for cancer prevention. Am J Clin Nutr 1998; 67:136-142. 

206. Grubben MJ, van den Braak CC, Essenberg M, et al. Effect of resistant starch on potential 
biomarkers for colonic cancer risk in patients with colonic adenomas: a controlled trial. Dig 
Dis Sci 2001; 46:750-756. 

207. Bouhnik Y, Flourie B, Ouarne F, et al. Effects of prolonged ingestion of fructo- 
oligosaccharides (FOS) on colonic bifidobacteria, fecal enzymes and bile acids in humans. 
Gastroenterol 1994; 106:A598. 

208. Rowland IR, Bearne CA, Fischer R, Pool-Zobel BL. The effect of lactulose on DNA damage 
induced by DMH in the colon of human flora-associated rats. Nutr Cancer 1996; 26:37-47. 

209. Perrin P, Pierre F, Patry Y, et al. Only fibers promoting a stable butyrate producing colonic 
ecosystem decrease the rate of aberrant crypt foci in rats. Gut 2001; 48:53-61. 

210. Nakanishi S, Kataoka K, Kuwahara T, Ohnishi Y. Effects of high amy lose maize starch and 
Clostridium butyricum on metabolism in colonic microbiota and formation of azoxymethane- 
induced aberrant crypt foci in the rat colon. Microbiol Immunol 2003; 47:951-958. 



Modifying the Human Intestinal Microbiota with Prebiotics 313 

211. Scholz-Ahrens KE, Schaafsma G, van den Heuvel EG, Schrezenmeir J. Effects of prebiotics 
on mineral metabolism. Am J Clin Nutr 2001; 73:459S-464S. 

212. Scholz-Ahrens KE, Schrezenmeir J. Inulin, oligofructose and mineral metabolism — 
experimental data and mechanism. Br J Nutr 2002; 87:S179-S186. 

213. Morohashi T. The effect on bone of stimulated intestinal mineral absorption following 
fructooligosaccharide consumption in rats. Biosci Microflora 2002; 21:21-25. 

214. Sakuma K. Molecular mechanism of the effect of fructooligosaccharides on calcium 
absorption. Biosci Microflora 2002; 21:13-20. 

215. Hillman LS, Tack E, Covell DG, Vieira NE, Yergey AL. Measurement of true calcium 
absorption in premature infants using intravenous 46 Ca and oral 44 Ca. Pediatr Res 1988; 
23:589-594. 

216. Chonan K, Matsumoto M, Watanuki. Effects of galactooligosaccharides on calcium 
absorption and preventing bone loss in ovariectomized rats. Biosci Biotechnol Biochem 
1995; 59:236-239. 

217. Mizota T. Lactulose as a growth- promoting factor for Bifidobacterium and its physiological 
aspects. Bull Int Dairy Fed 1996; 313:43-48. 

218. Ohta A, Osakabe N, Yamada K, Saito Y, Hidaka H. Effects of fructooligosaccharideson Ca, 
Mg and P absorption in rats. J Jap Soc Nutr Food Sci 1993; 46:123-129. 

219. van den Heuvel EG, Schaafsma G, Muys T, van Dokkum W. Nondigestible oligosaccharides 
do not interfere with calcium and nonheme-iron absorption in young, healthy men. Am J Clin 
Nutr 1998; 67:445-451. 

220. Teuri U, Korpela R, Saxelin M, Montonen L, Salminen S. Increased fecal frequency and 
gastrointestinal symptoms following ingestion of galacto-oligosaccharide-containing yogurt. 
J Nutr Sci Vitaminol 1998; 44:465-471. 

221. Coudray C, Bellanger J, Castiglia-Delavaud C, Remesy C, Vermorel M, Rayssignuier Y. 
Effect of soluble or partly soluble dietary fibers supplementation on absorption and balance of 
calcium, magnesium, iron and zinc in healthy young men. Eur J Clin Nutr 1997; 51:375-380. 

222. van den Heuvel EG, Muijs T, van Dokkum W, Schaafsma G. Lactulose stimulates calcium 
absorption in postmenopausal women. J Bone Miner Res 1999; 14:1211-1216. 

223. van den Heuvel EG, Muys T, van Dokkum W, Schaafsma G. Oligofructose stimulates calcium 
absorption in adolescents. Am J Clin Nutr 1999; 69:544-548. 

224. van den Heuvel EG, Schoterman MHC, Muijs T. Transgalactooligosaccharides stimulate 
calcium absorption in postmenopausal women. J Nutr 2000; 130:2938-2942. 

225. Griffin IJ, Davila PM, Abrams SA. Non-digestible oligosaccharides and calcium absorption in 
girls with adequate calcium intakes. Br J Nutr 2002; 87:S187-S191. 

226. Coudray C, Demigne C, Rayssiguier Y. Effects of dietary fibers on magnesium absorption in 
animals and humans. J Nutr 2003; 133:1-4. 

227. van Dokkum W, Wezendonk B, Srikumar TS, van den Heuvel EG. Effect of nondigestible 
oligosaccharides on large-bowel functions, blood lipid concentrations and glucose absorption 
in young healthy male subjects. Eur J Clin Nutr 1999; 53:1-7. 

228. Jackson KG, Taylor GR, Clohessy AM, Williams CM. The effect of the daily intake of inulin 
on fasting lipid, insulin and glucose concentrations in middle-aged men and women. Br J Nutr 
1999; 82:23-30. 

229. Kruse HP, Kleessen B, Blaut M. Effects of inulin on fecal bifidobacteria in human subjects. Br 
J Nutr 1999; 82:375-382. 

230. Brighenti F, Casiraghi MC, Canzi F, Ferrari A. Effect of consumption of a ready-to-eat 
breakfast cereal containing inulin on the intestinal milieu and blood lipids in healthy male 
volunteers. Eur J Clin Nutr 1999; 53:726-733. 

23 1 . Alles MS, de Roos MN, Bakx JC, van de Lisdonk E, Zock PL, Hautvast JGAC. Consumption 
of fructo-oligosaccharides does not favourably affect blood glucose and serum lipids in non- 
insulin dependent diabetic patients. Am J Clin Nutr 1999; 69:64-69. 

232. Williams CM, Jackson KG. Inulin and oligofructose: effects on lipid metabolism from human 
studies. Br J Nutr 2002; 87:S261-S264. 



314 Crittenden and Playne 

233. Delzenne NM, Daubioul C, Neyrinck A, Lasa M, Taper HS. Inulin and oligofructose 
modulate lipid metabolism in animals: review of biochemical events and future prospects. Br 
J Nutr 2002; 87:S255-S259. 

234. Ozaslan C, Tiirkcapar AG, Kesenci M, et al. Effect of Lactulose on Bacterial Translocation. 
Eur J Surg 1997; 163:463-467. 

235. Bovee-Oudenhoven IMJ, Termont DMSL, Heidt PJ, Van der Meer R. Increasing the intestinal 
resistance of rats to the invasive pathogen Salmonella enteritidis: additive effects of dietary 
lactulose and calcium. Gut 1997; 40:497-504. 

236. Schumann C. Medical, nutritional and technological properties of lactulose. An update. Eur 
J Nutr 2002; 41:17-25. 

237. Buddington KK, Donahoo JB, Buddington RK. Dietary oligofructose and inulin protect mice 
from enteric and systemic pathogens and tumor inducers. J Nutr 2002; 132:472-477. 

238. Duggan C, Penny ME, Hibberd P, et al. Oligofructose-supplemented infant cereal: 
2 randomized, blinded, community-based trials in Peruvian infants. Am J Clin Nutr 2003; 
77:937-942. 

239. Moshfegh AJ, Friday JE, Goldman JP, Chug Ahuja JK. Presence of inulin and oligofructose in 
the diets of Americans. J Nutr 1999; 129:1407S-1411S. 

240. van Loo J, Coussement P, De Leenheer L, Hoebregs H, Smits G. On the presence of inulin and 
oligofructose as natural ingredients in the Western diet. Crit Rev Food Sci Nutr 1995; 
35:525-552. 

241. Smith PB. Safety of short-chain fructo-oligosaccharides and GRAS affirmation by the 
U.S.FDA. Biosci Microflora 2002; 21:27-29. 

242. Deguchi Y, Matsumoto K, Ito A, Watanuki M. Effects of 3 1-4 galacto-oligosaccharides 
administration on defaecation of healthy volunteers with a tendency to constipation. Jap J 
Nutr 1997;55:13-22. 

243. Saavedra JM, Tschernia A. Human studies with probiotics and prebiotics: clinical 
implications. Br J Nutr 2002; 87:S241-S246. 

244. Bovee-Oudenhoven IMJ, ten Bruggencate SJM, Lettink-Wissink MLG, van der Meer R. 
Dietary fructo-oligosaccharides and lactulose inhibit intestinal colonization but stimulate 
translocation of Salmonella in rats. Gut 2003; 52:1572-1578. 

245. ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML, van der Meer R. Dietary 
fructo-oligosaccharides dose-dependently increase translocation of Salmonella in rats. J Nutr 
2003; 133:2313-2318. 

246. van Loo JA. Prebiotics promote good health: the basis, the potential, and the emerging 
evidence. J Clin Gastroenterol 2004; 38:S70-S75. 

247. Younes H, Coudray C, Bellanger J, Demigne C, Rayssiguier Y, Remesy C. Effects of two 
fermentable carbohydrates (inulin and resistant starch) and their combination on calcium and 
magnesium balance in rats. Br J Nutr 2001; 86:479-485. 



17 



Modifying the Gastrointestinal Microbiota 
with Probiotics 



Chandraprakash D. Khedkar 

Department of Dairy Microbiology and Biotechnology (Maharashtra Animal 
and Fishery Sciences University Nagpur), College of Dairy Technology 
Warud (Pusad), India 

Arthur C. Ouwehand 

Danisco Innovation, Kantvik, and Functional Foods Forum, University 
of Turku, Turku, Finland 



INTRODUCTION 

The origin of fermentations involving the production of lactic acid are lost in the ancient 
times, but it is not difficult to imagine how nomadic communities gradually acquired the 
art of preserving their meager supplies of milk by storing them in animal skins or crude 
earthenware pots. Initially, the intention could well have been simply to keep the milk cool 
through the evaporation of whey from the porous surface, but the chance transformation of 
the raw milk into a refreshing, slightly viscous foodstuff would soon have been recognized 
as a desirable innovation resulting in yogurt-like products. 

At the beginning of last century, Eli Metchnikoff proposed the, now classic, theory 
that the apparent longevity of Bulgarian tribesmen was a direct result of their lifelong 
consumption of yogurt-like fermented milk products, probably mostly fermented by 
lactobacilli (1). This inspired an interest in the nutritional and therapeutic characteristics of 
these products. The validity of these hypotheses was debated for many years but one 
undeniable effect of his work was a marked increase in the popularity of yogurt throughout 
Europe. At about the same time, Henri Tissier suggested that bifidobacteria could be 
administered to children with diarrhea to help restore their gut microbiota balance (2). 

Fermented milk products like yogurt and other products containing beneficial or 
"probiotic" cultures, such as lactobacilli, bifidobacteria, lactococci, and propionibacteria 
are currently among the best-known examples of functional foods in many countries 
around the world. These products are associated with a range of health claims, some more 
documented then others, including alleviation of symptoms of lactose intolerance (3), 
treatment of diarrhea (4), cancer risk reduction (5) and restoration of gastrointestinal (6) 
and urogenital microbiota (7), and constipation (8). Milk is an ideal food system to act as a 
carrier of these versatile bacteria to the human gastrointestinal tract (GIT) and support 

315 



316 



Khedkar and Ouwehand 



their viability. From these beginnings, the probiotic concept has progressed considerably 
and is now the focus of much research attention worldwide. Significant advances have 
been made in the selection and characterization of specific cultures and substantiation of 
health claims relating to their consumption. Subsequently, the area of probiotics has 
advanced from anecdotal reports, with scientific evidence now accumulating to back up 
health claim properties of specific strains. Nowadays the majority of scientific and 
commercial attention is concentrated on probiotic microorganisms like Lactobacillus and 
Bifidobacterium, with the result that an expanding range of probiotic dairy products 
containing these species are now available to the consumer. 

This paper will critically examine the health claims and evidence for beneficial 
effects of probiotic organisms in relation to modifying the gastrointestinal microflora and 
its functioning. 



PROBIOTICS 

Definition 

The term probiotic is derived from Greek, meaning "for life" and originated to describe 
substances produced by one microorganism which stimulate the growth of others (9). The 
Food and Agriculture Organization of the United Nations (FAO) and the World Health 
Organization (WHO) have stated that there is adequate scientific evidence to indicate that 
there is potential for probiotic foods to provide health benefits and that specific strains are 
safe for human consumption (10). An expert panel commissioned by FAO and WHO 
defined probiotics as "Live microorganisms which when administered in adequate 
amounts confer a health benefit on the host." This definition will be used in the current 
chapter instead of the term biotherapeutic agents (11) which is sometimes used as well to 
indicate probiotics. 



Probiotic Microorganisms 

Lactobacillus and Bifidobacterium are the principal bacterial genera central to both 
probiotic and prebiotic approaches to dietary modulation of the intestinal microflora. In 



Table 1 Commonly Used Probiotic Microorganisms 







Lacto- 










Bifido- 


coccus 


Strepto- 


Entero- 




Lactobacillus 


bacterium 


lactis subsp 


coccus 


coccus 


Saccharomyces 


acidophilus, brevis, 


adolescentis, 


cremoris, 


thermophilus 


faecium 


cerevisiae 


delbruekii, 


animalis/ 


lactis 






(boulardii) 


fermentum, gasseri, 


lactis? 










johnsonii, lactis, 


bifidum, breve, 










paracasei, 


infantis, lactis, 










plantarum, rham- 


longum, 










nosus, reuteri 


thermophilum 











a The current taxonomic status of B. animalis and B. lactis is unclear. 

b Saccharomyces boulardii is likely to be identical to Saccharomyces cerevisiae. 

Source: From Refs. 12-16. 



Modifying the Gastrointestinal Microbiota with Probiotics 317 

addition, there are many different microorganisms currently used as probiotics. A list of 
microbes commonly used as probiotics is given in Table 1. Some of these organisms have 
been studied much more extensively than others. It is therefore important that probiotics 
are referred to by their strain designation as well as their species. Although other members 
of the same species share most characteristics, different probiotic strains may differ in 
some essential properties (17). 



Desirable Characteristics of Probiotic Microorganisms 

Many desirable characteristics have been proposed by various researchers for probiotic 
lactobacilli and bifidobacteria (and other microbes) to be used as dietary adjuncts for 
gastrointestinal and related health benefits. These organisms should have the ability to 
survive in sufficient numbers, the acidity of the gastric juices and to pass in a viable state to 
the small intestinal region (18-23). Ability of these organisms to proliferate and/or 
colonize the gut is also an important desirable, although appears not so common, property. 
In practice the desired properties of these microorganisms are dependent on the host for 
which probiotic administration is intended, the anatomical site within the host toward 
which the probiotic is directed (most often the GIT) and the desired effect at that site are 
the principal focus of probiotic applications (19,21,23). 

A general set of desirable properties of probiotic microorganisms, regardless of the 
intended host or site of application is presented in Table 2. In vitro tests based on these 
selection criteria, although not a definite means of strain selection, may provide useful initial 
information. In addition, well-characterized, and validated model systems such as the TNO 
Intestinal Models (TIM-I and II) and the Simulator of the Human Intestinal Microbial 
Ecosystem (SHIME), which aim to mimic complex physiological and physicochemical 
in vivo reactions, may also be of value in strain selection (for a description of 
intestinal models, see the chapter by Makivuokko and Nurminen). Several tests for gastric 
passage and gastric digestion of the candidature organisms, as well as pH resistance and 
ability to pass through the stomach are presented in Table 3. Such types of tests are less 
expensive than human or animal trials and do not have the associated ethical drawbacks 
(29). However, ultimate proof of probiotic effects requires validation in well-designed, 
randomized, double-blind, placebo controlled, statistically sound clinical trials (30). 

Administration of a large number of these organisms will increase the number of 
surviving microbes, but various strains of these organisms may differ in acid tolerance and 
survival. However, as the transit time of fermented milk products through the stomach is 
shorter than many other foods (31), and fermented dairy products provide a buffer towards 
gastric juice (32), this has been shown to lead to the appearance in high numbers of the 
administered strains in the feces (33). 



PROPOSED HEALTH BENEFITS OF PROBIOTICS 

The health benefits of probiotics can be direct or indirect through modulation of the 
composition and/or activity of the endogenous microbiota or of the immune system. Many 
health claims have been made concerning probiotics, especially concerning their potential 
to prevent or help cure gastrointestinal and related ailments. These include improved lactose 
digestion and other direct enzymatic effects, prevention, and curative treatment of 
gastroenteritis, antibiotic-associated diarrhea, traveler's diarrhea, constipation, intestinal 



318 



Khedkar and Ouwehand 



Table 2 Desirable Properties of Probiotics 



Probiotic characteristics 



Technological/Functional properties 



Stability: bile salts and 

gastric acidity 
Adherence: ability to 

adhere to the intestinal 

mucosa 
Transient colonization 
Safety 



Antagonism: against 
pathogenic and putre- 
factive organisms 

Proven health effects 



Stability: stability/viabil- 
ity during processing 
and storage 

Technological suitability 



Survival in human gastrointestinal tract 

Immune cell modulation and competitive inhibition of the 
pathogenic organisms 

Growth and multiplication in the human gastrointestinal tract 

Well-documented clinical safety, organism must be accurately 
identified to strain level before recommending its use. It should 
be non-toxic, non-pathogenic, non-allergenic, non-mutagenic, 
non-carcinogenic and have no transferable antibiotic resistance 

Prevention of pathogen colonization through competition for 
nutrients and binding sites and through production of 
antimicrobial substances 

Clinically documented and validated therapeutic effects. Dose- 
response data for minimum effective dosage of the probiotic 
organism in different formulations 

All of the aforementioned desirable characteristics should be 
maintained during processing and storage of these products 
organism should be genetically stable, no plasmid transfer 

Culture should be suitable for production of acceptable quality 
finished products with desirable viable counts 



infections and to suppress colonization of the gut by pathogenic organisms colonized in gut, 
irritable bowel syndrome (IBS) and various conditions of diarrhea, hypocholesterolaemea, 
urogenital tract infection, atopic diseases, skin diseases, gastrointestinal well-being, 
inflammatory bowel disease (IBD) and colon cancer (16,31,34,35). 



Table 3 Experiments Demonstrating Resistance Tests for Survival of an Organism in the Upper 
Digestive Tract for Selected Probiotic Strains 



Resistance test method 



Organisms tested 



Reference 



Gastric digestion in vivo (mixture 
of HC1 + pepsin + rennet) 



PH 

Human gastric juices conditions of 
the stomach: cultured milk 
mixed with gastric juice (70:30) 

Artificial gastric juices (at pH 3.0 
incubation by 37°C) 



Lb. acidophilus (survival) 

Propionibacterium freudenreichii (survival 

without loss of vitality) 
Yogurt, buttermilk and sour milk cultures 

(survival with different digestion times) 
Bif. bifidum (4 strains) 2 hours at pH 2.4 and 

6.5 (strong action at pH 2) 
Lb. acidophilus (survival) yogurt and sour 

milk cultures (addition of gastric juices 

with pH 3.48-6.75, no bacteriocidal or 

bacteriostatic effect observed) 
Lb. acidophilus and Lb. plantarum survive 

3 hours; Lb. bulgaricus less resistant, 

survives only for 1 hour 



(24) 
(25) 



(26) 
(27) 



(28) 



Modifying the Gastrointestinal Microbiota with Probiotics 319 

These microorganisms possess various immunological functions viz., mitogenic 
activity (36), adjuvant activity (21), macrophage activation (37), enhancement of antibody 
production (38), induction of interferon-y production (39) and antitumor effects (40), 
amongst others. It has further been indicated by a number of studies that both the cell wall 
and cytoplasm of specific probiotic bacteria induced mitogenic responses of spleen 
cells (37,41,42). 

Health benefits of probiotic organisms related that may impact on the gut microbiota 
are summarized in the following paragraphs. 



Use of Probiotics to Combat Gastrointestinal Infections 

Probiotics have been shown to be useful in the treatment of a variety of gastrointestinal 
disorders, and the details are presented in Table 4. A number of these disorders have a 
significant inflammatory component in the small and/or large intestine and there is a 
growing body of research to suggest that probiotic bacteria may be useful particularly in 
many of these pediatric gastrointestinal conditions. Specific strains of Lactobacillus 
rhamnosus, Lb. reuteri, Lb. plantarum, Bifidobacterium lactis, and Saccharomyces 
cerevisiae (boulardii) have all been extensively studied. Probiotics can reduce the duration 
and severity of rotaviral enteritis, as well as decrease the risk of antibiotic-associated 
diarrhea in children and Clostridium difficile diarrhea in adults. Prevention of viral 
diarrhea in day-care centers as well as traveler's diarrhea has been demonstrated with 
some probiotics, although not all are equally effective (67). Small bowel bacterial 
overgrowth conditions may respond to probiotic use. How the probiotic bacteria 
counteract the inflammatory process by enhancing the degradation of external antigens, 
reducing the secretion of inflammatory mediators and maintaining the healthy gut 
microbiota by exclusion of pathogens is schematically shown in Figure 1 . 



Possible Mode of Action of Probiotics in Reducing the Duration of Diarrhea 

Several potential mechanisms have been proposed for how probiotics reduce the duration 
of rotavirus diarrhea, but none have been proven and each theory has its limitations. The 
first is competitive blockage of receptor sites (69) in which probiotics bind to receptors, 
thereby preventing adhesion and invasion of the virus. This concept might be plausible if 
there was evidence for specific receptor competition. In most cases, by the time a probiotic 
is ingested, the patient will already have had diarrhea for possibly 12 hours. By this time, 
the virus has infected mature enterocytes in the mid- and upper region of the small 
intestinal villi. The virus and/or its entero toxin, NSP4, will then have disturbed fluid and 
electrolyte transport, thereby lowering fluid and glucose absorption. The toxin could have 
then potentially activated secretory reflexes, causing loss of fluids from secretory epithelia, 
resulting in diarrhea (70). At best, subsequent competitive exclusion of viruses would only 
be effective for attachment of progeny, and it is not known whether such inhibition would 
reduce diarrhea. If probiotic organisms somehow competed with the toxin or peptides 
released from villous endocrine cells, it is feasible that the cascade that leads to diarrhea 
could be prevented. 

The second potential mechanism may be that the immune response is enhanced by 
probiotics, leading to the observed clinical effect (45). This is supported by the protective 
effect which local immunoglobulin A (IgA) antibodies appear to confer against rotavirus 
(71). However, a problem with this theory is given that diarrhea appears to cease within 1 
to 3 days in patients who would otherwise suffer for 4 to 6 days; the probiotics would need 



320 



Khedkar and Ouwehand 



Table 4 Examples of the Effects of Probiotics on Microbial Infections 



Disorder 


Subject 


Probiotics 


Effect 


Reference 


Infantile 


Human 


Lactobacillus 


Reduced duration of 


(43-47) 


diarrhea 




GG 


diarrhea etc. 






Human 


Lb. reuteri 


Reduced duration 
of diarrhea 


(49) 




Human 


Bif. Bifidum-\- 
Str. thermo- 
philus 


Prevention of diarrhea 


(23) 




Human 


Bif. breve 


Prevention of diarrhea 


(51) 


Antibiotic- 


Human 


Bif. longum 


Decreased course of erythromy- 


(52) 


associated 






cin-induced diarrhea 




diarrhea 












Human 


Lactobacillus 
GG 


Decreased course of erythromy- 
cin-induced diarrhea, and other 
side effects of erythromycin 


(53) 




Human 


Str. faecium 


Decreased diarrhea associated 
with anti-tubercular drugs 
administered for pulmonary TB 


(54) 




Human 


Sc. boulardii 


Reduce incidence of diarrhea 


(55) 
(56) 
(56a) 


Relapsing C. 


Human 


Lactobacillus 


Improves/terminates colitis 


(57) 


difficile 




GG 




(58) 


colitis 












Human 


Lactobacillus 


Eradicated associated 


(59) 






GG 


diarrhea 


(60) 


Travelers' 


Human 


Lb. acidophilus 


Decrease frequency, not 


(61) 


diarrhea 




-\-Bif. bifidum 


duration of diarrhea 


(61a) 




Human 


Lactobacillus 
GG 




(62) 


Foodborne 


Male 


Lb. casei Shirota 


Increased resistance to lethal 


(62a) 


pathogen 


BALB/c 




infection with Salmonella, E. 




exclusion 


Mice 




coli, and L. monocytogenes 






Male rat 




Increased resistance to 
salmonellosis infection 


(63) 




Mice 


Bif. lactis HN019 


Increased survival of 
Salmonella infection 


(49a) 




Mice 


Lb. rhamnosus 
HN001 


Increased survival of E. coli 
0157:H7 infection 


(50) 




In vitro 


Yogurt bacteria 


Inhibit growth of Salmonella 


(64,65) 




Human 


Lb. acidophilus 
+ Lactobacil- 
lus GG 


Decreased shigellosis-associated 
diarrhea 


(66) 



Abbreviations: Bif, Bifidobacterium; C, Clostridium; E, Escherichia; L, Listeria; Lb, Lactobacillus; 
Sc, Saccharomyces; Str, Streptococcus. 



to trigger the antibody response rapidly so that it interfered with further viral activity. 
Animal studies do indicate that secretory IgA can be triggered by probiotic ingestion (72), 
but the rate was not determined, nor was the influence on cessation of fluid loss across the 
secretory cell membranes. Modification of the cytokine profile to one that enhances 



Modifying the Gastrointestinal Microbiota with Probiotics 



321 



anti-inflammatory cytokines (73) or attenuation of the virus' and/or toxin's effect on the 
enteric nervous system might provide rapid cessation of epithelial secretion and diarrhea. 
Alternatively, stimulation of T cells to produce gamma interferon, leading to potential 
inhibition of chloride secretion, might also inhibit diarrhea. One aspect of the immunity 
theory that needs to be clarified is why lactobacilli, which we assume are present in the 
child intestine, appear unable to prevent infection; yet those administered orally thereafter 
help to clear the diarrhea. 

A third mechanism could involve a signal(s) from probiotics to the host that down- 
regulates the secretory and motility defenses designed to remove perceived noxious 
substances. Glycosylated intestinal mucins inhibit rotaviruses (74), and MUC2 and MUC3 
mRNA expression is increased in response to probiotics signaling, protecting cells against 
pathogenic bacterial adhesion (13). However, direct host cell signaling between probiotic 
organism and secretory cells has not yet been investigated. Attachment of the virus causes 
cytokine prostaglandin and nitric oxide to be released from the enterocytes, both of which 
could affect motility. The possibility exists that lactobacilli could alter this release (75). 
The intestinal host defense mechanisms comprise complex systems involving the innate 
and adaptive immune responses, and protective effects of the indigenous microbiota. The 
commensal microorganisms colonizing the intestinal mucosa provide a barrier effect 
against pathogens by using a variety of mechanisms, such as occupation of habitats, 
competition for nutrients, and production of antimicrobials. It is also established that the 
probiotic organisms can modulate the homeostasis of the host's defense mechanisms, both 
innate and adaptive immune functions (4). 

A final theory is that the probiotics produce substances that inactivate the viral 
particles. This has been shown in vitro (76), with supernatants from Lactobacillus 
rhamnosus GR-1 and L. fermentum RC-14 inactivating 10 9 particles of the double-stranded 
DNA adenovirus and the negative-stranded RNA vesicular stomatitis virus within 
10 minutes. The effect was likely due to acid, but more specific antiviral properties have 
not been ruled out. Whether or not viral inactivation can inhibit diarrhea remains to 
be confirmed. 



Inflammation 




Immune 
reponse 



Unbalanced 
microbiota 



Figure 1 Schematic representation of the possible ways by which probiotics may counteract ( => ) 
the intestinal inflammatory process. Source: From Ref. 68. 



322 Khedkar and Ouwehand 

More detailed investigation is needed to understand how probiotic strains reduce the 
duration of diarrhea in conjunction with rehydration therapy. Such studies could lead to a 
better understanding of the dynamics within the intestinal microbiota that is being 
disrupted and depleted by rapid fecal loss. In doing so, new intervention therapies should 
be generated to quickly and effectively trigger the cessation of not only rotavirus 
infections but also other gastrointestinal infections that debilitate patients for 2 to 3 days. 

The possible mode of action for diarrhea and other gastrointestinal diseases, such as 
IBS and IBD, are the subject of intense investigation in many labs, using genomics, 
knockout mice models, etc., (77,78). 



MODIFYING INTESTINAL MICROBIOTA COMPOSITION 
THROUGH INTAKE OF PROBIOTICS 

In the human GIT, variability exists in bacterial numbers and composition between the 
stomach, small intestine and colon. The total bacterial count in gastric contents is usually 
below 10 3 per gram contents with numbers in the small intestine ranging from about 10 4 per 
ml of contents to about 10-10 at the terminal ileum (79). In comparison to other regions of 
the GIT, the human large intestine is a complex, heavily populated and diverse microbial 

11 1 

ecosystem. Bacterial numbers in the human large intestine are in the range of 10 -10 for 
every gram of the gut contents (80). The colonic microbiota is capable of responding to 
anatomical and physicochemical variations that are present. The right or proximal colon is 
characterized by a relatively high substrate availability (due to dietary input), a pH of around 
5.5-6.0 (from acids produced during microbial fermentation) and a more rapid transit than 
the distal region. The left or distal area of colon has a lower concentration of available 
substrate, in particular carbohydrates, the pH is approximately 6.5-7.0, and the flow of the 
digesta is slower. The proximal region tends to be a more saccharolytic environment than 
the distal gut, the latter having higher bacterial proteolysis. Several hundred different 
species of bacteria are known to be present in the large intestine (see also the chapter by Ben 
Amor and Vaughan). Gram-negative rods belonging to the Bacteroides fragilis group are 
the numerically predominant culturable bacteria in the colon. The other main groups 
consist of different (Gram-positive) rods and cocci, such as bifidobacteria, Clostridia, 
peptococci, streptococci, eubacteria, lactobacilli, peptostreptococci, ruminococci, enter- 
ococci, coliforms, methanogens, dissimilattory sulfate-reducing bacteria, and acetogens. 
The microbiota includes saccharolytic organisms, proteolytic species and bacteria that can 
metabolize gases. Despite the huge diversity of bacteria present in the large gut (estimated 
over 1000 species), it is certain that the vast majority has hitherto not been identified or 
cultured [(81), see also the chapter by Ben Amor and Vaughan]. 



Increasing Numbers of Beneficial Microbes 

One of the properties thought to be important for the health benefits of consumed probiotic 
organisms is their ability to adhere to the intestinal mucosa. As such they can resist 
peristalsis and occupy a habitat at the expense of potentially harmful organisms. The 
probiotic applications to the human gut are already widespread, and evidence is mounting 
that these organisms have a beneficial effect on the host. It is now well established that the 
probiotic organisms can transiently establish themselves in the GIT and inhibit the 



Modifying the Gastrointestinal Microbiota with Probiotics 



323 



adhesion and growth of enteropathogens. Table 5 delineates the effect of feeding selected 
probiotic preparations on the human gut microbiota. 



Suppressing Numbers of Potentially Harmful Microbes 

The artificial manipulation of the human intestinal microbiota by consumption of large 
numbers of probiotic microorganisms may lead to the presence of large numbers of lactic 
acid-producing microorganisms in the small intestine. Any available sugars will be quickly 
fermented to various organic acids and/or ethanol. This leads to a change in the environment 
where the production of various low-molecular toxic metabolites and antigenic 
macromolecules by various intestinal, potentially pathogenic microbes and the effects of 
endotoxins may be strongly reduced (Table 5). The intestinal growth of all other types of 
nonintestinal pathogens is strongly inhibited by abundant probiotic fermentation in the 
small intestine. Reduction of viral infectivity was attributed to ethanol or acid-mediated 
denaturation of viral envelope proteins. In addition to organic acids, bacteriocins, such as 
e.g., Lactacin F (88), and some unidentified compounds synthesized by probiotic organisms 



Table 5 Effect of Feeding Selected Probiotic Preparations on Human Gut Microbiota 



Type of probiotic organisms 



Effect on gut microbiota 



Reference 



Lb. rhamnosus GG 



Lb. rhamnosus GG 



Lb. plantarum (VTTE-79098) 



Lb. paracasei ssp. paracasei 

(VTTE-94506) 
Lb. paracasei ssp. paracasei 

(VTTE-94510) 
L. rhamnosus (VTTE-94510) 
Bifidobacterium sp. (VTTE- 

94508) 
L. casei Shirota 
Bif. bifidum 
Lb. acidophilus-hBK}/3 



Lb. acidophilus-LBKV3 suppli- 
mented with Propionibacter- 
ium freundenrichii ssp. 

Shermanii 

Bif. lactis HN019 



Attachment of probiotic organism to CaCo-2 
intestinal cell line and in vivo to human 
colonic mucosa 

Increased the number of fecal bifidobacteria and 
lactobacilli 

Concomitant decrease in Clostridia counts 

Reduction in enterobacteriaceae counts of 4 log 
cycles, Clostridia 1 log cycle, and slight 
decreases in enterococci counts in a SHIME 
reactor 



Balancing of intestinal microbiota 

Balancing of intestinal microbiota 

Highly significant increases in fecal lactobacilli, 
bifidobacteria, propionibacteria and lacto- 
cocci counts and concomitant decreases in 
coliforms, Clostridia, staphylococci and 
enterococci in tribal kids of 2-5 years 

Increases in vivo antimicrobial activity of the 
microflora against putrefactive organisms in 
the gut of tribal kids of 2-5 years 

Increase in fecal lactobacilli and bifidobacteria 



(82) 



(83) 



(82) 



(84) 

(84a) 

(85) 



(86) 



(87) 



Abbreviation'. SHIME, Simulator for Human Intestinal Microbiological Ecosystem. 



324 Khedkar and Ouwehand 

may confer an additional growth-inhibiting effect (89). However, it is still uncertain whether 
such substances are produced in situ in the intestine and are effective. 



MODIFYING THE MICROBIAL METABOLIC ACTIVITY 

Due to its numbers and taxonomic diversity, the intestinal microbiota has an enormous 
metabolic potential. The microbiota' s metabolic activity is comparable to that of the 
liver, our metabolically most active organ. This metabolism has a pronounced influence 
on the health and well being of the host, as described in more detail in the chapter by 
Goldin. Probiotics have been shown to be able to change the metabolic activity of the 
intestinal microbiota. In part, this may relate to a direct change in its composition, but it 
may also relate to a change in metabolism of some members of the microbiota in 
response to a shift in the intestinal environment. The main metabolic markers that are 
potentially influenced by probiotics are the production of short chain fatty acids (SCFA) 
and fecal enzyme activity. 



Short Chain Fatty Acid Production 

Principal end products of bacterial fermentation in the colon are SCFA, i.e., acetate, 
propionate, and butyrate. Other fermentation products include ethanol, lactate, succinate, 
formate, valerate, and caproate. Branched chain fatty acids such as isobutyrate, 2-methyl- 
butyrate, and iso valerate may also be formed from the fermentation of amino acids. 



Short Chain Fatty Acids 

The production of SCFA by the intestinal microbiota serves to salvage energy from the 
digesta that would otherwise be lost for the host (90). Butyrate provides an important 
energy source for the intestinal epithelium. Propionate is metabolized in the liver where it 
possibly serves as a precursor for gluconeogenesis. Acetate is mainly taken up by muscle 
tissue but is also used by adipocytes for lipogenesis. Lactate is also metabolized by 
muscle tissue. However, despite the fact that enterocytes only slowly absorb lactate, it is 
usually found only at low concentrations in the digesta as it is used to a large extent by 
members of the intestinal microbiota (91) and only accumulates in disease (92). 



Probiotics and Short Chain Fatty Acids 

Probiotics will, when they are metabolically active, produce organic acids in the intestine; 
these will mainly be lactate and acetate. Furthermore, the metabolic activity will influence 
the metabolism of other microbes present in the intestine, through competition for 
nutrients and through the production of metabolites. It is, however, not really known to 
what extent probiotics are metabolically active in the human intestine, in particular in the 
colon, and whether probiotics produce antimicrobials such as bacteriocins in situ. Studies 
in mice, colonized with a human microbiota, do however indicate metabolic activity (93). 
Assessment of the data presented in Table 6 indicates that most probiotics tested do 
not affect the composition of the fecal short chain fatty acid composition. This may be 
explained by the lack of metabolic activity of the probiotics in the colon, but it is more 
likely to reflect the efficient absorption of fatty acids by the colon (2). Therefore, to assess 
the influence of probiotics, and for that matter also prebiotics, on the availability of SCFA, 



Modifying the Gastrointestinal Microbiota with Probiotics 



325 



Table 6 Influence of Probiotics on Fecal Short Chain Fatty Acids (SCFA) and Fecal Enzyme 
Activity in Humans, Selected References 





Dose 






SCFA 


Fecal enzyme 




Probiotic 


(CFU/day) 


Duration 


Subjects 


change 


activity change 


Reference 


B. lactis 


2.8X10 10 


6 hours 


Ileostomists 


No change 


— 


(94) 


Bb-12 














S. cerevi- 


lg 


6 days 


Healthy 


No change 


— 


(95) 


siae bou- 






adults 








lardii 














S. cerevi- 


lg 


6 days 


Patients 


Increase 


— 


(95) 


siae bou- 






with total 








lardii 






enteral 
nutrition 








Yogurt +L 


3X10 8 L. 


6 months 


Healthy 


No change 


— 


(13) 


acidophi- 


acidophi- 




adults 








lus 


lus 












145+5. 


3X10 7 B. 












longum 


longum 












913 














Kefir 






Healthy 
adults 


Increase, 
though not 
different 
from con- 
trol (milk) 




(96) 


L. plan- 




4 weeks 


Healthy 


No change 


No change 


(97) 


tar um 






adults 








299v 














L. casei 


3X10 11 


4 weeks 


Healthy 


Decrease 


Decrease 


(98) 


Shirota 






adults 








L. rhamno- 


1.6 X10 9 


6 months 


Healthy 


No change 


No change 


(99) 


sus 






adults 








HN019 














L. casei 


1.3X10 10 


1 month 


Healthy 


No change 


Decrease 


(100) 


DN-114 






infants 








001 














L. gasseri 


10 9 , 10 10 , 


41 days 


Healthy 


No change 


Decrease/ 


(100a) 


SBT2055 


10 11 




adults 




no change 




L. gasseri 


2X10 10 


11 days 


Healthy 


— 


Decrease 


(101) 


ADH 






elderly 
Elderly 
with 
atrophic 
gastritis 








L. rhamno- 


1.4X10 10 


4 weeks 


Healthy 


— 


No change 


(86) 


sus GG 






adults 








L. rhamno- 


2X10 10 


2 weeks 


Healthy 


— 


Decrease 


(102) 


sus GG 






elderly 








L. rhamno- 


1-2X10 10 


2 weeks 


Healthy 


— 


Decrease 


(103) 


sus GG 






adults 








L. rhamno- 


4X10 10 


4 weeks 


Healthy 


— 


Decrease 


(103a) 


sus GG 






adults 









(Continued) 



326 



Khedkar and Ouwehand 



Table 6 Influence of Probiotics on Fecal Short Chain Fatty Acids (SCFA) and Fecal Enzyme 
Activity in Humans, Selected References (Continued) 



Probiotic 



Dose 
(CFU/day) Duration Subjects 



SCFA Fecal enzyme 

change activity change Reference 



L. rham 


1-2X10 10 


4 weeks 


Healthy 


nosus 


L. rham 




elderly 


LC-705 


nosus 






P. freuden- 


2-4X10 10 






reichii JS 


P. freu- 
denrei- 
chii 






L. reuterii 


7.2X10 8 


4 weeks 


Healthy 
elderly 


B. longum 


1.3 X10 10 


3-6 weeks 


Healthy 
adults 


L. acido- 


4X10 10 


4 weeks 


Healthy 


philus 






adults 


NCFM 








VSL#3 


9X10 11 


20 days 


Irritable 


(bifido- 






bowel 


bacteria 






syn- 


+ lacto- 






drome 


bacilli 






patients 



+ strep- 
tococci) 



Decrease 



(104) 



No change 


(104) 


Decrease 


(105) 


Decrease 


(106) 


Decrease/ 


(107) 


increase 





sampling should preferably take place in the proximal colon where substrates are more 
abundant and the microbes more active. 



Fecal Enzyme Activity 

Fecal Enzymes 

One of the detrimental effects the human intestinal microbiota may have on host health is 
the production of tumor promoters, mutagens, and carcinogens from undigested dietary 
substrates and endogenous residues. Bacterial enzymes involved in the formation of such 
substances are (3-glucoronidase, azoreductase, nitroreductase, and nitrate reductase (108); 
see also the chapters by Rafter and Rowland, and Goldin. A reduction in the activity of 
these enzymes can be expected to lead to a reduced exposure to carcinogenic substances. 
Animal models have suggested this also leads to a reduced incidence in colorectal 
cancers (106). However, it is not clear if this also holds true for humans. 

Probiotics and Fecal Enzyme Activity 

Most of the probiotics (listed in Table 6) tend to induce a reduction in fecal enzyme 
activity. This appears to be therefore one of the more general and reproducible properties 
of probiotics. However, since fecal enzyme activity is not a definite biomarker for cancer 
risk, one should be cautious when drawing conclusions and extrapolating from animal 



Modifying the Gastrointestinal Microbiota with Probiotics 327 

experiments to humans. As with SCFA production, the mechanism behind this is probably 
competition for nutrients and production of inhibitory metabolites. 

CONCLUSION 

The area of modulation of gastrointestinal microbiota through intake of probiotics seems 
to hold much promise for the prophylactic management and/or treatment of gut disorders, 
as mediated by pathogens. The growing realization by consumers that our food profoundly 
influences our health has fueled the introduction of food products with health claims such 
as probiotics into the market. It seems that the use of probiotics in general clinical practice 
is not far away, given that products such as VSL#3, containing a mixture of lactic acid 
bacteria probiotics, are already being used. However, it is relevant to note that studies on 
particular strains may not necessarily be extrapolated to all probiotic microorganisms. 
Molecular tools will continue to be used to understand and manipulate probiotic bacteria 
with a view to produce vaccines and new and improved products. The critical step in wider 
application will be to make products available that are safe and clinically proven in a 
specific formulation easily accessible to physicians and consumers. Systematically 
randomized, double-blind and placebo-controlled studies including large numbers of 
human volunteers are needed to advance the scientific knowledge of probiotics and 
gastrointestinal microbiota. Technological advances like protective coating(s), micro- 
encapsulation, or addition of prebiotic compounds that can serve as growth factors for 
probiotic organisms will improve the survival of strains in the gut of consumers. It is 
necessary to clearly understand the functionality of these organisms in the intestinal 
ecosystem. 



REFERENCES 

1. Metchnikoff E. The Prolongation of Life. New York: G. Putnam's Sons, 1908. 

2. Tissier H. Traitement des infections intestinales par la methode de la flore bacterienne de 
l'intestin. Crit Rev Soc Biol 1906; 60:359-361. 

3. Kim HS, Gilliland SE. Lactobacillus acidophilus as a dietary adjunct for milk to aid lactose 
digestion in humans. J Dairy Sci 1983; 66:959-966. 

4. van Niel CW, Freudtner C, Garrison MM. Christakis, D.A. Lactobacillus therapy for acute 
infectious diarrhoea in children: a meta-analysis. Pediatrics 2002; 109:678-684. 

5. Ohashi Y, Nakai S, Tsukamoto T, et al. Habitual intake of lactic acid bacteria and risk 
reduction of bladder cancer. Urol Int 2002; 68:273-280. 

6. Orrhage K, Sjostedt S, Nord CE. Effect of supplements with lactic acid bacteria and 
oligofructose on the intestinal microflora during administration of cefpodoxime proxetil. 
J Antimicrob Chemother 2000; 46:603-611. 

7. Gardiner GE, Heinemann C, Bruce AW, Beuerman D, Reid G. Persistence of Lactobacillus 
fermentrum RC-14 and Lactobacillus rhamnosus GR-1 but not L. rhamnosus GG in the 
human vagina as demonstrated by randomly amplified polymorphic DNA. Clin Diag Lab 
Immunol 2002; 9:92-96. 

8. Marteau P, Cuillerier E, Meance S, et al. Bifidobacterium animalis strain DN-173 010 
shortens the colonic transit time in healthy women: a double-blind, randomized, controlled 
study. Aliment Pharmacol Ther 2002; 16:587-593. 

9. Lilly DM, Stillwell RH. Therapeutic properties of substances produced by lactic acid 
bacteria. Science 1965; 147:747-748. 

10. Food and Agriculture Organization of United Nations and World Health Organization 
2001. Regulatory and clinical aspects of dairy probiotics. Food and Agriculture 



328 Khedkar and Ouwehand 

Organization of United Nations and World Health Organization Expert Consultant Report. 
Food and Agriculture Organization of United Nations and World Health Organization 
Working Group Report (Online). 

11. Food and Agriculture Organization of United Nations and World Health Organization 
2002. Guidelines for the evaluation of probiotics in food. Food and Agriculture 
Organization of United Nations and World Health Organization Working Group Report 
(Online). 

12. Dunne C, O'Mahony L, Murphy L. In vitro selection criteria for probiotic bacteria of 
human originxorrelation with in vivo findings. Am J Clin Nutr 2001; 73:386S-392S. 

13. Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA. Probiotics inhibit 
enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression. 
Am J Physiol 1999; 276:G941-G950. 

14. Mattila-Sandholm T, Salminen S. Up-to-date on probiotics in Europe. Gastroenterol Intern 
1998; 1:8-12. 

15. Ventura M, Zink R. Rapid identification, differentiation, and proposed new taxonomic 
classification of Bifidobacterium lactis. Appl Environ Microbiol 2002; 68:6429-6434. 

16. Gardiner GE, Heinemann C, Baroja ML, et al. Oral administration of the probiotic 
combination Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 for human intestinal 
applications. Int Dairy J 2002; 12:191-196. 

17. Ouwehand AC, Vesterlund S. Health aspects of probiotics. IDrugs 2003; 6:573-580. 

18. Collins JK, Thornton G, O' Sullivan G. Selection of probiotic strains for human 
applications. Int Dairy J 1998; 8:487-490. 

19. Gardiner GE, Ross RP, Kelly PM, Stanton C, Collins JK, Fitzgerald G. Microbiology of 
therapeutic milks. In: Robinson RK, ed. Dairy Microbiology Handbook: The Microbiology 
of Milk and Milk Products. New York: Wiley Interscience Publishers, 2002:431-468. 

20. Havenaar R, Huis in't Veld JHJ. In: Wood BJB, ed. Lactic Acid Bacteria in Health and 
Disease. London: Elsevier Applied Science Publishers, 1992:151-170. 

21. Kohwi Y, Hashimoto Y, Tamura Z. Antitumor and immunological adjuvant effect of 
Bifidobacterium adolescentis M 101-104. Biosci Biotech Biochem 1982; 57:2127-2132. 

22. Lee YK, Salminen S. Probiotics in gastrointestinal disorders. Trends Food Sci Technol 
1995; 6:241-245. 

23. Saavedra JM, Bauman NA, Oung I, Perman JA, Yolken RH. Feeding of Bifidobacterium 
bifidum and Streptococcus thermophilus to infants in hospital for prevention of diarrhoea 
and shedding of rotavirus. Lancet 1994; 344:1046-1049. 

24. Breslaw ES, Kleyn DH. Factors affecting growth of Bifidobacteria. Food Sci 1973; 
38:1016-1021. 

25. Mantere-Alhonen S. Fermented milks and human health. Meijeritieteellinen Aikakauskirja 
1983;41:217-237. 

26. Pattersson L, Graf W, Sewelin U. Symposium of the Swedish nutrition foundation. XV. In: 
Hallgren Bo, ed. Nutrition and the Intestinal Flora. Stockholm, Sweden: Almquist & 
Wiksell International, 1983. 

27. Lindwall S, Fonden R. Fermented Milks and Health, FIL/IDF Doc. 1984; 179:21-33 

28. Yakult Honsha Co. Ltd. 1971. The summary of Reports Yakult. Yakult Honsha Co. Ltd. 1- 
1-19, Higshi Shinbashi, Minato-ku, Tokyo, Japan and Intestinal flora of microorganisms 
and health. Prospectus. 

29. Huis in't Veld JHJ, Shortt C. Gut flora and health- past, present and future. In: Leeds RA, 
Rowland IR, eds. International Congress and Symposium Series No. 219. London: Royal 
Society of Medicine Press, 1996:27-39. 

30. Reid G, Jass J, Sebulsky MT, McCormick JK. Potential uses of probiotics in clinical 
practice. Clin Microbiol Rev 2003; 16:658-672. 

31. Fooks LJ, Gibson GR. Probiotics as modulators of the gut flora. Br J Nutr 2002; 
88:S39-S49. 

32. Conway PL, Gorbach SL, Goldin BR. Survival of lactic acid bacteria in the human stomach 
and adhesion to intestinal cells. J Dairy Sci 1987; 70:1-12. 



Modifying the Gastrointestinal Microbiota with Probiotics 329 

33. Aim L, Leijenmark CE, Persson AK, Midtvedt T. The regulatory and protective role of the 
normal microflora. In: WennerGren International Symposium Series. 1988;52: 293-297. 

34. Mountzouris KC, McCartney AL, Gibson GR. Intestinal microflora of human and infants 
and current trends for its nutritional modulation. Br J Nutr 2002; 87:405-420. 

35. Salminen SJ, Gueimonde M, Isolauri E. Probiotics that modify disease risk. J Nutr 2005; 
135:1294-1298. 

36. Kado-oka Y, Fujiwara S, Hirota T. Effects of bifidobacteria cells on mitogenic response of 
splenocytes and several functions of phagocytes. Milchwissenschaft 1991; 46:626-630. 

37. Sekine K, Ohta J, Onishi M, et al. Analysis of antitumor properties of effector cells 
stimulated with a cell wall preparation (WPG) of Bifidobacterium infantis. Biol Pharm Bull 
1995; 18:148-152. 

38. Yasui H, Nagaoka N, Mike A, Hayakawa K, Ohwaki M. Detection of Bifidobacterium 
strains that induce large quantities of IgA. Microb Ecol Health Dis 1992; 5:155-162. 

39. Kishi A, Uno K, Matsubara Y, Okuda C, Kishida T. Effect of the oral administration of 
Lactobacillus brevis subsp. coagulans on interferon-a producing capacity in humans. J Am 
Coll Nutr 1996; 15:408-412. 

40. Sekine K, Kawashima T, Hashimoto Y. Comparison of the TNF-a levels induced by 
human-derived Bifidobacterium longum and rat-derived Bifidobacterium animalis in mouse 
peritoneal cells. Bifidobact Microflora 1994; 13:79-89. 

41. Hosono A, Lee J, Ametani A, et al. Characterization of a water-soluble polysaccharide 
fraction with immunopotentiating activity from Bifidobacterium adolescentis M 101-4. 
Biosci Biotech Biochem 1997; 61:312-316. 

42. Tone-Shimokawa Y, Toida T, Kawashima T. Isolation and structural analysis of 
polysaccharide containing galactofuranose from the cell walls of Bifidobacterium infantis. 
JBacteriol 1996; 178:317-320. 

43. Isolauri E, Juntunen M, Rautanen T, Sillanaukee P, Koivula T. A human Lactobacillus 
strain {Lactobacillus GG) promotes recovery from acute diarrhoea in children. Paediatrics 
1991;88:90-97. 

44. Isolauri E, Kaila M, Mykkanen H, Ling WH, Salminen S. Oral bacteriotherapy for 
gastroenteritis. Digest Dis Sci 1994; 39:2595-2600. 

45. Kalia M, Isolauri E, Soppi E, Virtanen E, Laine S, Arvilommi H. Enhancement of the 
circulating antibody secreting cell response in human diarrhoea by human Lactobacillus 
strain. Paed Res 1992; 32:141-144. 

46. Majaama H, Isolauri E, Saxelin M, Vesikari T. Lactic acid bacteria in the treatment of acute 
rotavirus gastroenteritis. J Paed Gastroenterol Nutr 1995; 20:333-339. 

47. Raza S, Graham SM, Allen SJ, Sultana S, Cuevas L, Hart CA. Lactobacillus GG 
promotes recovery form acute non-bloody diarrhoea in Pakistan. Pediatr Infect Dis J 
1995; 14:107-111. 

48. Salminen S, Ouwehand AG, Benno Y, Lee YK. Probiotics: how should they be defined? 
Trends Food Sci Technol 1999; 10:107-110. 

49. Shornikova AV, Casas I A, Isolauri E, Mykkanan H, Vesikari T. Lactobacillus re uteri as a 
therapeutic agent in acute diarrhoea in young children. J Pediatr Gastroenterol Nutr 1997; 
24:399-404. 

49a. Shu Q, Hai L, Rutherfurd KJ, Fenwick SG, Gopal PK, Gill HS. Dietary Bifidobacterium 
lactis (HN019) enhances resistance to oral Salmonella typhimurium in mice. Microbiol 
Immunol 2000; 44:213-222. 

50. Shu Q, Gill HS. Imune protection mediated by the probiotic Lactobacillus rhamnosus 
HN001 (DR10™) against Escherichia coli 0157:H7 infection in mice. FEMS Immunol 
Med Microbiol 2002; 34:59-64. 

51. Hotta M, Sato SIN. Clinical effects of Bifidobacterium preparation on pediatric intractable 
diarrhoea. Keio J Med 1987; 36:298-314. 

52. Colombel JF, Corot A, Neut C, Romond C. Yogurt with Bifidobacterium longum reduces 
erythromycin-induced gastro-intestinal effects. Lancet 1987; 2:43-44. 



330 Khedkar and Ouwehand 

53. Sittonen S, Vapaatalo H, Salminen S, et al. Effect of Lactobacillus GG yogurt in prevention 
of antibiotic associated diarrhoea. Ann Med 1990; 22:57-59. 

54. Borgia M, Sepe N, Brancato V, Borgia RA. A controlled clinical study on Streptococcus 
faecium preparation for the prevention of side reactions during long-term antibiotic 
therapy. Curr Therapeutic Res 1982; 31:265-271. 

55. Surawicz CM, Elmer LW, Speelman P, McFarland LV, Chinn J, vanBelle G. Prevention of 
antibiotic-associated diarrhoea by Saccharomyces boulardii: a prospective study. 
Gastroenterology 1989; 96:981-988. 

56. Macfarlane GT, Cummings JH. Probiotics and prebiotics: can regulating the activities of 
intestinal bacteria benefit health. BMJ 1999; 318:999-1003. 

56a. Elmer GW, Surawicz C, McFarland L. Biotherapeutic agents: a neglected modality for the 
treatment and prevention of selected intestinal and vaginal infections. J. Am. Med. Asso. 
1996; 275:870-876. 

57. Gorbach SL, Chang TW, Goldin B. Successful treatment of relapsing Clostridium difficile 
Colitis with Lactobacillus GG. Lancet 1987; 26:1519-1522. 

58. Bartlett JG, Chang TW, Gurwith M. Antibiotic-associated pseudomembraneous colitis due 
to toxin-producing Clostridia. N Engl J Med 1982; 57:141-145. 

59. Bennet RG, Laughon B, Lindsay J. 1990. Lactobacillus GG treatment of Clostridium 
difficile infection in nursing home patients. Abstract, 3rd International Conference on 
Nosocomial Infections. Atlanta. 

60. Biller JA, Katz AJ, Flores AF, Buie TM, Gorbach SL. Treatment of recurrent Clostridium 
difficile colitis with Lactobacillus GG. J Pediatr Gastroenterol Nutr 1995; 21:224-226. 

61. Black FT, Andersen PL, Orskov F, Gaarslev K, Laulund S. Prophylactic efficacy of 
lactobacilli on traveller's diarrhoea. Travellers Med 1989; 8:1750-1753. 

61a. Oksanen PJ, Salminen S, Saxelin M, Hamalmein P, Arja IV, Leena MI, Seppo N, 
Oksanen T, Posti I, Salminen E, Siitonen S, Struckey H, Topilla A, Vapaatalo H. 
Prevention of traveller's diarrhoea by Lactobacillus GG. Ann Med 1990; 22:53-56. 

62. Hilton E, Kolakowski P, Smith M, Singer C. Efficacy of Lactobacillus GG as a diarrhoea 
preventive measure. J Travellers Med 1996; 4:3-7. 

62a. Nomoto K, Nagaoka M, Yokokura T, Mutai M. Augmentation of resistance of mice to 
bacterial infection by a polysaccharide peptidoglycan complex (PSPG) extracted from 
Lactobacillus casei. Biotherapy 1989; 1:169-171. 

63. Hitchins AD, Wells P, McDonough FE, Wong NP. Amelioration of the adverse effect of 
gastrointestinal challenge with Salmonella enteriditis on weaning rats by a yogurt diet. Am 
J Clin Nutr 1986;41:91-100. 

64. Bovee-Oudenhoven I, Termont D, Dekker R, Van der Meer R. Calcium in milk and 
Fermentation by yogurt bacteria increase the resistance of rats to Salmonella infection. Gut 
1996; 38:59-65. 

65. Brassart D, Neeser JR, Michetti P, Sevin A. The selection of dairy bacterial strains with 
probiotic properties based on their adhesion to human intestinal epithelial cells. Les 
Bacteries Lactiques/Lact ic Acid Bacteria. Adria Normandie: Presses Unversitaires de Caen, 
1995 pp. 201-202. 

66. Sepp E, Tamm E, Torm S, Lutsar I, Mikelsaar M, Salminem S. Impact of a Lactobacillus 
probiotics on the faecal microflora in children with shigellosis. Microecol Ther 1995; 
23:74-80. 

67. Majaama H, Isolauri E, Saxelin M, Vesikari T. Lactic acid bacteria in the treatment of acute 
rotavirus gastroenteritis. J Pediatr Gastroenterol Nutr 1995; 20:333-338. 

68. Isolauri E, Kirjavainen PV, Salminen S. Probiotics: a role in the treatment of intestinal 
infection and inflammation? Gut 2002; 50:iii54-iii59. 

69. Bernet MF, Brassart D, Neeser JR, Servin AL. Lactobacillus acidophilus LA 1 binds to 
cultured human intestinal cell lines and inhibits cell attachment and cell invasion by 
enterovirulent bacteria. Gut 1994; 35:483-489. 

70. Lundgren O, Svensson L. Pathogenesis of rotavirus diarrhea. Microbes Infect 2001; 
3:1145-1156. 



Modifying the Gastrointestinal Microbiota with Probiotics 331 

7 1 . Ward LA, Rosen BI, Yuan L, Saito LJ. Pathogenesis of an attenuated and a virulent strain of 
group A human rotavirus in neonatal gnotobiotic pigs. J Gen Virol 1996; 77:1431-1441. 

72. Reid G, Charbonneau D, Gonzalez S, Gardiner G, Erb J, Bruce AW. Ability of 
Lactobacillus GR-1 and RC-14 to stimulate host defences and reduce gut translocation and 
infectivity of Salmonella typhlmurium. Nutraceut Food 2002; 7:168-173. 

73. Christensen HR, Frokiaer H, Pestka JJ. Lactobacilli differentially modulate expression of 
cytokines and maturation surface markers in murine dendritic cells. J Immunol 2002; 
168:171-178. 

74. Yolken RH, Ojeh C, Khatri IA, Sajjan U, Forstner JF. Intestinal mucins inhibit rotavirus 
replication in an oligosaccharide-dependent manner. J Infect Dis 1994; 169:1002-1006. 

75. Xu J, Verstraete W. Evaluation of nitric oxide production by lactobacilli. Appl Microbiol 
Biotechnol 2001; 56:504-507. 

76. Cadieux P, Burton J, Gardiner G, et al. Lactobacillus strains and vaginal ecology. JAMA 
2002;287:1940-1941. 

77. Rachmilewitz D, Katakura K, Karmeli F, et al. Toll-like receptor 9 signaling mediates the 
anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 
2004; 126:520-528. 

78. Mohamadzadeh M, Olson S, Kalina WV, et al. Lactobacilli activate human dendritic cells that 
skew T cells toward T helper 1 polarization. Proc Natl Acad Sci USA 2005; 102:2880-2885. 

79. Gorbach SL, Nahas L, Lerner PI. Studies on intestinal microflora I: Effects of diet, age and 
periodic sampling on numbers of faecal microorganisms in man. Gastroenterology 1967; 
53:845-855. 

80. Cummings JH, Macfarlane GT. A review: the control and consequences of bacterial 
fermentation in the human colon. J Appl Bacteriol 1991; 70:443-459. 

81. Blaut M, Collins MD, Welling GW, Dore J, van Loo J, de Vos W. Molecular biological 
methods for studying the gut microbiota: the EU human gut flora project. Br J Nutr May 
2002;87:S203-S211. 

82. Alander M, De Smet I, Nollet L, Verstraete W, Mattila-Sandholm T. The effect of probiotic 
strains on the microbiota of the simulator of the human intestinal microbial ecosystem 
(SHIME). Int J Food Microbiol 1999; 46:71-79. 

83. Benno Y, Hosono M, Hashimoto H, Kojima T, Yamazaki K, lino H. Effects of 
Lactobacillus GG yoghurt on human intestinal microecology in Japanese subjects. Nutr 
Today 1996; 3L9S-12S. 

84. Aso Y, Akazan H. Prophylactic effect of Lactobacillus casei preparation on the recurrence 
of superficial bladder cancer. Urol Int 1992; 49:125-129. 

84a. Marteau P, Flourie B, Pochart P. Effect of chronic ingestion of a fermented dairy product 
containing lactobacillus and Bifidobacterium bifidum on metabolic activities of the colonic 
flora in humans. Am J Clin Nutr 1990; 52:685-688. 

85. Khedkar CD, Patil MR, Gyananath G, et al. Studies on implantationability of a probiotic 
culture of Lactobacillus acidophilus in gastrointestinal tract of tribal children. In 
Proceedings of International Seminar and Workshop on Fermented Foods, health status 
and social well-being, held at Anand(India) 2003:62-63. 

86. Khedkar CD, Gyananath G, Patil MR, Bajad DN, Khojare AS, Sarode AR. 2006. Studies on 
effect of feeding probiotic Lactobacillus supplemented with Propionibacterium culture on 
gastrointestinal microflora of tribal children. Lait (In press). 

87. Gopla PK, Prasad J, Gill HS. Effects of consumption of Bifidobacterium lactis DR10 and 
galactooligosaccharides on the microecology of the gastrointestinal tract in human subjects. 
Nutr Res 2001; 23:1313-1318. 

88. Altermann E, Russell WM, Azcarate-Peril MA, et al. Complete genome sequence of the 
probiotic lactic acid bacterium Lactobacillus acidophilus NCFM. Proc Natl Acad Sci USA 
2005; 102:3906-3912. 

89. Bongaerts GPA, Severijnen RSVM. The beneficial, antimicrobial effect of probiotics. Med 
Hypotheses 2001; 56:174-177. 



332 Khedkar and Ouwehand 

90. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient 
environment of the mammalian intestine. Annu Rev Nutr 2002; 22:283-307. 

91. van Nuenen MHMC, Meyer PD, Venema K. The effect of various inulins and Clostridium 
difficile on the metabolic activity of the human colonic microbiota in vitro. Microb Ecol 
Health Dis 2003; 15:137-144. 

92. Tsukahara T, Iwasaki Y, Nakayama K, Ushida K. Microscopic structure of the large 
intestinal mucosa in piglets during an antibiotic associated diarrhea. J Vet Med Sci 2003; 
65:301-306. 

93. Oozeer R, Mater DD, Goupil-Feuillerat N, Corthier G. Initiation of protein synthesis by a 
labeled derivative of the Lactobacillus casei DN-114 001 strain during transit from the 
stomach to the cecum in mice harboring human microbiota. Appl Environ Microbiol 2004; 
70:6992-6997. 

94. Hove H, Nordgaard-andersen I, Brobech Mortensen P. Effect of lactic acid bacteria on the 
intestinal production of lactate and short-chain fatty acids, and the absorption of lactose. Am 
J Clin Nutr 1994; 59:74-79. 

95. Girard-Pipau F, Pompei A, Schneider S, et al. Intestinal microflora, short chain and cellular 
fatty acids, influence of a probiotic Saccharomyces boulardii. Microb Ecol Health Dis 2002; 
14:220-227. 

96. St-Onge M-P, Farnworth ER, Savard T, Chabot D, Mafu A, Jones PJH. Kefir consumption 
does not alter plasma lipid levels or cholesterol fractional synthesis rates relatiev to milk in 
hyperlipidemic men: a randomized controlled trial. BMC Complement. Altern Med 2002; 
2:1-789. 

97. Goossens D, Jonkers D, Russel M, Stobberingh E, van den Bogaard A, Stockbriigger R. The 
effect of Lactobacillus plantarum 299v on the bacterial composition and metabolic activity 
in faeces of healthy volunteers: a placebo-controlled study on the onset and duration of 
effects. Aliment Pharmacol Ther 2003; 18:495-505. 

98. Spanhaak S, Havenaar R, Schaafsma G. The effect of consumption of milk fermented by 
Lactobacillus casei strain Shirota on the intestinal microflora and immune parameters in 
humans. Eur J Clin Nutr 1998; 52:899-907. 

99. Tannock GW, Munro K, Harmsen HJM, Welling GW, Smart J, Gopal PK. Analysis of the 
fecal microflora of human subjects consuming a probiotic product containing Lactobacillus 
rhamnosus DR20. Appl Environ Microbiol 2000; 66:2578-2588. 

100. Guerin-Danan C, Chabanet C, Pedone C, et al. Milk fermented with yogurt cultures and 
Lactobacillus casei compared with yogurt and gelled milk: influence on intestinal microflora 
in healthy infants. Am J Clin Nutr 1998; 67:111-117. 

100a. Fujiwara S, Seto Y, Kimura A, Hashiba H. Establishment of orally administered 
Lactobacillus gasseri SBT 2055 SR in the gastrointestinal tract of humans and its influence 
on intestinal microflora and metabolism. J Appl Microbiol 2001; 90:343-352. 

101. Pedrosa MC, Golner BB, Goldin BR, Barakat S, Dallal GE, Russell RM. Survival of yoghurt- 
containing organisms and Lactobacillus gasseri (ADH) and their effect on bacterial enzyme 
activity in the gastrointestinal tract of healthy and hypochlorhydric elderly subjects. Am 
J Clin Nutr 1995; 61:353-359. 

102. Ling WH, Hanninen O, Mykkanen H, Heikura M, Salminen S, von Wright A. Colonization 
and fecal enzyme activities after oral Lactobacillus GG administration in elderly nursing 
home residents. Ann Nutr Metab 1992; 36:162-166. 

103. Hosoda M, He F, Kojima T, Hashimoto H, lino H. Effects of fermented- milk with 
Lactobacillus rhamnosus GG strain administration on defecation, putrefactive metabolites 
and fecal microflora of healthy volunteers. J Nutr Food 1998; 1:1-9. 

103a. Goldin BR, Gorbach SL, Saxelin M, Barakat S, Gualtieri L, Salminen S. Survival of 
Lactobacillus species (strain GG) in human gastrointestinal tract. Dig Dis Sci 1992; 
37:121-128. 

104. Ouwehand AC, Lagstrom H, Suomalainen T, Salminen S. The effect of probiotics on 
constipation, faecal azoreductase activity and faecal mucins. Ann Nutr Metab 2002; 
46:159-162. 



Modifying the Gastrointestinal Microbiota with Probiotics 333 

105. Tomoda T, Nakano Y, Kageyama T. Effect of yogurt and yogurt supplemented with 
Bifidobacterium and/or lactulose in healthy persons: a comparative study. Bifidobact. 
Microflora 1991; 10:123-130. 

106. Goldin BR, Gorbach SL. The relationship between diet and rat faecal bacterial enzymes 
implicated in colon cancer. J Natl Cancer Inst 1976; 57:371-375. 

107. Brigidi P, Vitali B, Swennen E, Bazzocchi G, Matteuzzi D. Effects of probiotic 
administration upon the composition and enzymatic activity of human faecal microbiotia 
in patients with irritable bowel syndrome or functional diarrhea. Res Microbiol 2001; 
152:735-741. 

108. Hughes R, Rowland IR. Metabolic activities of the gut microflora in relation to cancer. 
Microb Ecol Health Dis 2000; 11:179-185. 



18 



Modifying the Intestinal Microbiota 
with Antibiotics 



Asa Sullivan and Carl Erik Nord 

Department of Laboratory Medicine, Karolinska University Hospital, Karolinska 
Instituted Stockholm, Sweden 



INTRODUCTION 

The human host and the microorganisms colonizing skin and mucous surfaces constitute 
dynamic biological communities or ecosystems. The composition of the microbiota is 
relatively stable but fluctuations occur intra-individually over time, and there are also large 
inter-individual differences. The specific microbiota at each ecological habitat is referred to 
as the normal microbiota. The numerically and the most diverse human normal microbiota 
is found in the gastrointestinal tract, and some of the species are potential pathogens that 
may cause disease under certain circumstances (1). One of the functions of the 
gastrointestinal microbiota is to act as a barrier against overgrowth of such organisms 
and also to prevent colonization of pathogenic bacteria from the environment. This 
phenomenon is termed, "colonization resistance" (2). Treatment with antimicrobial agents 
disturbs the ecological balance between the host and the normal microbiota and overgrowth 
of yeasts and Clostridium difficile, or of intrinsically or acquired resistant microorganisms, 
may occur. Horizontal spread of resistance genes by conjugation or transformation to other 
microbial species can take place. The gastrointestinal normal microbiota plays an important 
role in this development (3). 

Orally administered antimicrobial drugs that are incompletely absorbed or excreted 
via bile or transluminally, frequently give rise to a reduced colonization resistance. Other 
factors of importance are the antimicrobial spectrum of the agents, the dose as well as the 
pharmacokinetic properties of the agents. The outcome of antimicrobial treatment with 
respect to disturbances in the intestinal microbiota may further vary between individuals. 
Apart from different anatomical and physiological qualities of the host, the ability of some 
microorganisms to produce substances that inactivate antimicrobial agents and binding of 
agents to intestinal material renders the prediction of the effects difficult (4). However, the 

335 



336 Sullivan and Nord 

ecological impact is of great importance in the clinical situation and guidance may be 
acquired by knowledge of the results from studies on the ecological influence of 
antimicrobial agents on the normal microbiota. 



ANTIMICROBIAL AGENTS THAT INHIBIT THE SYNTHESIS 
OF THE BACTERIAL CELL WALL— p-LACTAM ANTIBIOTICS 



Penicillins 

The effect of penicillins on the gastrointestinal microbiota is summarized in Table 1 



Phenoxymethylpenicillin 

Phenoxymethylpenicillin has been shown to induce minor variations in numbers of aerobic 
and anaerobic gastrointestinal microorganisms in healthy adults (5,6) and in infants treated 
for upper or lower respiratory tract infections or otitis media (7). Penicillin that reaches the 
gastrointestinal tract is destroyed by beta-lactamase produced by the microorganisms. 
Despite the low concentration of the agent in feces, generally under the detection level, 
occasional new colonization with Gram-negative aerobic rods has been observed 
during administration. 



Ampicillin 

Ampicillin has a broader antimicrobial spectrum than phenoxymethylpenicillin and is 
active also against species of Gram-negative microorganisms. The effect on the normal 
gastrointestinal microbiota is moderate with suppressed numbers of enterococci, 
streptococci, corynebacteria and enterobacteria. Minor effects on anaerobic species 
have also been observed in one study. Overgrowth of resistant aerobic Gram-negative rods 
is common and occasionally also of Candida species (8-10). The disturbances are 
increasing with increased doses. 



Ampicillin/Sulbactam 

The impact of ampicillin/sulbactam on the intestinal microbiota has been studied in 
patients undergoing colorectal surgery (11,12). From an ecological point of view it should 
be expected that it would be less favorable to combine ampicillin with a beta-lactamase 
inhibitor like sulbactam since the antimicrobial spectrum increases. The effect in 
particular on the aerobic microbiota has been shown to be mild while the number of 
anaerobic microorganisms was suppressed. With higher doses, overgrowth of yeasts has 
been observed and occasionally also overgrowth of Pseudomonas fluorescens. 



Amoxicillin 

Amoxicillin is an agent closely related to ampicillin and with a similar spectrum. 
Amoxicillin is acid-stable and is therefore better adsorbed. Overgrowth and emergence 
of amoxicillin-resistant enterobacteria have been the main outcome in studies on the 
effects on the normal gastrointestinal microbiota both in patients (13,14,16,19), in healthy 



Modifying the Intestinal Microbiota with Antibiotics 



337 



C3 



O 
O 



c3 

C 
• — < 

4-4 

0) 



pC 
•4— > 

C 

o 

CO 

c 



o 

• i-H 

e 

Oh 

o 

■4— ' 

o 

Oh 





.Q 
(0 



i 

C 



c 
o 
U 



o 

ft 
Ofl 

& 

> 

o 



CJ 

C 

-— * 

. -H 

CO 

o 



cj 
C 

u 

CO) 

ft 

s 

tq 



c 
o 

4-4 
CJ 
c3 
Oh 

a 



O 

C 

<4H 



„ G ,0 bp 

8.S 9«a 

B s r 






SI 

•-■-A 3 

u 






O 
ft 



Q ^ 



■ -- 

4H 



i 

o 

s- 

e y 



i 

O **H 

ft o 





d 
W 


*— < 


1 

o 


i 




!-H 


cd 


c3 


o 

c3 


X) 


• i-H 

■4—4 


d 


o 


< 


X> 




fc 


i 
CJ 


c3 

• i-H 


4— 1 


Cj 


ft 


c 


X> 


QJ 


W 


O 


4—4 



CJ 

cj 
o 
cj 





< 


+ 

a 




<4- 









CO 




53 


4-4 

cj 




B 

d 


d 




Z 




«4-4 

o 


1 

d 


d 
o 


CO 

(T-4 


s 


• i-H 

4-4 

c3 



CO 



8 5 

Q ? 



d 

< 



io vo 



r» OO On 



(imi-ir)\Ohoooo\ 



t-- 



(N <N 



io 



s 

e 

s 

a 



T3 *T3 

V V 



CD CD CD 

dec 



. I 

d ^h 



vo 



(N 



cdcdcdcdcdcdcd 



O c c c c 



a V V 



c c c 



•: v 



CD CD 

c d 



+ + 



+ + I 



+ 



+ 



+ 



+ 



+ + 



+ 



+ 



+ + + + + + + 



+ 



+ 



+ 
+ 



+ 
+ 



+ + + + 



+ 



+ 



+ + + + + + + + + + 



+ + 



+ 



+ 



+ 



o o 



ON o o 



o o 



(N 



Tf 00 so o o o o 



00 Q\ CN ^O VO 



O l> 



(N 
X 

o 

o 
o 



CN 
X 

o 

o 

0O 



^ t> «n 

en 



0D 

B X i 

o o 

o o o 

in n h 



lO CN 



cN"^-»ni>t^t^t>int^ 



o 
o 
o 
m 



m 

X 

o 

o 



o 
o 
o 



IO CN 



en 

X 

o 

o 
o 



en 

X 

o 
o 
m 

o 

o 



CN 

X 

o 

o 
o 



o 

o 
o 



en en en en en en 

X X X X X X 

o o o o o o 

o o o o m >n 



i 


r^ 


en 


r-- 


en 












en 


en 


~o"b 

E 
o 


CN 


X 

o 
>n 

CN 


X 

CN 




o 


c5 

o 



a c 

*-H (- 1 '-H 

Oh 



C cd 

• i-H 4— » 
£-H O 

.a £ 

Oh 3 
C GO 



OJiniOiOiOfNM^OOinm 



CJ 



c -d 
-d d 

?3 * 

•l-H 

X 

c 



^3 "o 

O 03 



< 



338 



Sullivan and Nord 



^3 

C3 



43 

a 



C 

•1-H 

■4— » 



CJ 

-c 

c 

o 

c 



o 

•I-H 

c 
a) 



cj 

c- 
E 



0) 

.q 

(0 

H 



o 

> 
O 



o 

CJ 

c 

in 

a) 



S c <u w) 

§2 ^ 



Cj 



o 
o 

a 

. -H 

CJ 

S-H 



O 

CJ 

C 

o 

00 

S-H 

a 

s 



c 

o 

-1—1 
CJ 

cd 

a 
6 



£ CD 

rt 'o 
PQ ° 



I <3 

CJ 



CJ 

-4—' 

"3 ° 



O "H 

C O 
CD O 

o 

CJ 



c 

tq 



o 

CD X) 



c3 



o 

ctf ctf 



53 



o 






I 
S-H 

CJ 



o 2 

pq o 



CJ 

• i-H 

o 
< 



CO 



CS 



T3 



Q c* 



53 



CJ 

O 

o 

CJ 

+ 

a 






c 

o 



£» S F> 



Kl 



CD ctf 

S 5 



c 
o 
00 

< 



CN CN CO Tt 
(N (N (N (N 



Tf io vo o r- 00 On 
i-h CN (N h (N CN (N 



O O — t CN 

m co en en 



CN 



(N 



CD CD CD CD CD 

d dec d 



V 



CJ 

d 



CJ 

d 



CJ 

d 



„„ 


^. 


CN 






© 
1 


(N 

i 


CN 






T3 


I 

CN 


V 


CD 


aj 


V 






d 


c 



i-H 
I 

V V 



+ 



+ 



+ + + + 



+ + + 



+ 



+ + 



+ + 



+ + 



+ + + + + 



+ 
+ 



+ + + 



+ + + I 



+ + + + + I + 



Tf Tf VD 



OO CN O O VO O O 
_ ^h ^_ CN CN 



o m >n o 



m m t> 



io r- en io °° cn °° 



t> r^ r^ r> 



*co X 
X »n 
io t> c5 



co 
X 

CN 



t> 00 iO O 
CO ^ CN CN 



00 "* 

B X 

00 o 
M O 



<o 



VO 



CO Tt CO 

XXX 

o o o 

O O IT) 



CO CO 

X X 

o o 

o o 

o o 



o 

o 
© 



i-h ^- r- cm m ^ Tt 



c e 



i i % 

> 



C C 



CO 

X 

© 
© 
10 



6 



© 

^t CO CN S 

X X x s 

© © © © 

© © © © 

vo Tf Tt m 



CO 

X 



N 

C3 C3 



CJ 
N ^ 



O 

N 



CD CD aj 

Oh Oh 



CLh H < Oh Oh 






CJ 

CJ 



> 

•1-H 

Cm 



CJ 



CJ 

a 

CJ 

•1-H 

H 



3 

-2 'CJ 
CJ CS 






o 

CJ 

o 
-t— * 
o 

O 



y5 

o 



J2 
O 

c 

cd 

CJ 

cC 



• r. 

CJ 
CJ 

,0 

'-i-H 

00 

a 

CJ 



00 

o 
A 

CJ 

cd 

a 



00 

§ 

+ 
+ 

• c 

CJ 
CJ 

,0 

00 

a 

CJ 



00 

1 
CN 

CJ 

ir, 

6 

CJ 

o 

CD 
CJ 

b 



CJ 

,0 

CJ 
CJ 



03 
CJ fc- 
C3 _CD 







CM 

X5 







1 • 



X 


r- 





Ci 


d 




cd 


<3 




* — 


O 


u. 




'<!( 


-4— > 


:. 


c3 





o 
c 

_ x 



Modifying the Intestinal Microbiota with Antibiotics 339 

volunteers (10,15,17,18) and in infants (7). In patients, in contrast to in healthy persons, 
amoxicillin has also a suppressive effect on the anaerobic microbiota (13,14,19). 



Amoxicillin/ 'Clavulanic Acid 

Administration of amoxicillin/clavulanic acid has been shown to induce increased 
numbers of amoxocillin-resistant enterobacteria both in healthy adults (15,20-23) and in 
child patients (24). In some of the mentioned studies, there were also disturbances in the 
numbers of aerobic cocci, mainly observed as increased numbers of enterococci. 



Bacampicillin, Pivampicillin, and Talampicillin 

Bacampicillin, pivampicillin and talampicillin are esters of ampicillin that are better 
absorbed than ampicillin, and thereby a more favorable ecological effect on the intestinal 
microbiota is expected. 

No major changes in the intestinal microbiota have been observed during long-term 
treatment of patients with bacampicillin (14) or in connection with shorter administration 
to healthy volunteers (25). However, the anaerobic microbiota was affected in some of the 
subjects in the latter study. Subjects receiving bacampicillin in tablets had an undisturbed 
intestinal microbiota in contrast to subjects receiving bacampicillin in syrup. 

Pivampicillin and talampicillin have been shown to give rise to increased numbers 
of enterobacteria in healthy volunteers (10,26) and increased numbers of Candida species 
have been observed in a few subjects during administration with pivampicillin. 



Azlocillin 

The impact of azlocillin on the intestinal microbiota has been studied in connection with 
treatment of patients suffering from skin and soft tissue infections (27). Suppressed 
numbers of both aerobic and anaerobic species were observed and overgrowth of resistant 
enterobacteria occurred in some patients. 



Piperacillin and Piperacillin/Tazobactam 

Piperazillin is excreted in bile leading to high fecal concentrations. Short-term 
administration to patients undergoing colorectal surgery has resulted in marked effects 
on both the aerobic and anaerobic intestinal microbiota (28). Addition of tazobactam to 
piperazillin in treatment of patients reduced the ecological disturbances in the anaerobic 
microbiota while the aerobic microbiota was still suppressed (29). 



Pivmecillinam 

Pivmecillinam has a spectrum including in particular Gram-negative aerobic rods and the 
main impact during administration to healthy volunteers has been seen as reduced 
numbers of gastrointestinal Escherichia coli (30,31). More pronounced changes have been 
observed to occur at higher doses with decreasing numbers of anaerobic species like 
lactobacilli and bacteroides and increasing numbers of enterococci (30). 



340 Sullivan and Nord 

Ticarcillin/Clavulanic Acid 

The effect of ticarcillin/clavulanate on the gastrointestinal microbiota has been evaluated 
in healthy subjects. Only minor disturbances were detected, such as decreased numbers of 
enterobacteria and a concomitant increase of aerobic cocci (32). 



Parenterally Administered Cephalosporins 

The spectra of cephalosporins are broader than that of penicillins. Several cephalosporins 
are excreted biliary and a strong ecological impact can be expected. Enterococci are 
intrinsically resistant to cephalosporins and their numbers usually increase 
during administration. 

The impact of parenterally administered cephalosporins on the gastrointestinal 
normal microbiota is summarized in Table 2. 



Cefazolin 

The impact of intravenously administered cefazolin on the gastrointestinal microbiota has 
been studied in patients at an intensive care unit (33) and in patients undergoing 
gastrectomy (34). Overgrowth of resistant Pseudomonas species was detected in the first 
study while increasing numbers of enterococci, reduced numbers of streptococci and also 
suppressed numbers of some anaerobic species were observed in the second study. 



Cefbuperazone 

Changes in the intestinal microbiota in connection with short-term administration of 
cefbuperazone have been studied in patients undergoing colorectal surgery (35). The agent 
suppressed the aerobic cocci, enterobacteria as well as the anaerobic microbiota. 

Cefepime 

A selective reduction of the numbers of E. coli has been observed during administration of 
cefepime in healthy volunteers (36). 

Cefmenoxime 

Significantly decreased numbers of enterobacteria, bifidobacteria and lactobacilli have 
been observed in connection with parenteral administration of cefmenoxime in healthy 
subjects. Furthermore, there was a concomitant increase in numbers of Clostridia and 
Candida species (37). 



Cefoperazone 

Cefoperazone is mainly excreted in bile giving rise to high fecal concentrations and 
thereby major changes in the intestinal microbiota can be expected. The impact of 
cefoperazone on the fecal microbiota has been evaluated in adult patients (38) and in sick 
children (39,40). The Gram-negative aerobic rods as well as numbers of staphylococci and 
streptococci were markedly suppressed in all studies. Overgrowth of resistant 
enterobacteria, enterococci and Candida species were observed and anaerobic species 
were also suppressed. 



Modifying the Intestinal Microbiota with Antibiotics 



341 



c3 



X> 

o 

u 



c3 

C 

• 1-H 
■*— > 



si 

-t—> 

d 

c 

Cfl 

c 

•1-H 

s 

c 

Oh 

</J 

15 
*d 

Oh 

u 

S-h 

<— i 
CO 



< 

13 

'— 

-(—I 

d 
*— 

Cm 



Dm 



CM 


n 

CO 

H 



c 
•— 

OX) 

s- 

> 

o 



O 

C 

cd 
-*— i 

Gfl 

• 1-H 

1/3 

S- 



o 

d 

OX) 

<d 



w 



d 
c 

■4— ' 

o 

c3 

Oh 



o 

d 

•— 

,o 

'-4— 

<D 

Ch* 



.2 -* 

p "ob 

d c 

O OX) 

S C 

O o3 

u '- 



s 

a 






in M 

<3" s 



i 03 
° 'C 

P-J ^D 



i 

O • — 

O o 





d 
PJ 




6 


i 
o 




S-i 


c-3 


c3 


c3 

C 




•c 

cd 

■4-J 


< 


• i-H 






i 


c3 


C 


X 


CD 


PJ 


o 


■4-J 
•1-H 




o 

•I-H 


o 




X) 


c 




o 


o 




•— 






CD 
< 


+ 




<+H 









c/l 




u 

CD 
X) 


o 

CD 




E 


■§ 

GO 


«H 


i 




o 


•I-H 


d 




d 


c 



^ S 03 

Q ^ § 



CD 03 

S 3 



d 
a) 

OX) 

< 



m 
en 



tj- \r> ^o r- oo on o o ^h m r^ cn m 
mm mm mm^^j-'sj-m m 'xj- ^t 



d 



CD oo 

d d 



CD 0) 

d d 



+ 



CD 

d 



+ 



d 



+ 



+ + 



+ 



+ + 



+ + 



io 



OO O 



oo «n 



m -<t 



r- r- m 

m m ^t 



0) a) ? ^ <ii ai 



d d 



3 A V 



d d 



+ + 



+ 



0) <D I <D 

d d d 



D 0) I (D 

d d d 



+ 



+ + 



d d d 



+ + + 
+ + + 



+ + 



+ + + 



+ 



OO \D V~) \Q ^O 



"> m 



o) o) a) a) d 
d d d d d 



+ 



+ 



+ 



+ 



+ + + + + 



+ + 



+ 



+ + + + + + + 



+ + 



+ 



+ 



+ 
+ 



+ 
+ 



100*0 



OO O OO OO N 





































>n 






-H 


'sl- 


i-H 




00 


m 




Tf- 


f-» « 


S 


c3 


i-H 


m 


(N 


m 




Th 


r-' 


-H 


^h m 


7 














7 


1 






i-H 

1 






(N 

1 










7 


-<t 














r- 








^i- 






OO 










f- 


s 


















OX) 




OX) 


^ 


















m 


(N 




CN 






(N 


^ 




^ 


OX) 

s 



00 




^J- 




O 




(N 




(N 


X 


X 




X 






X 


OX) 

s 




OX) 

s 




X 


1 


O 
O 


m 
X 


X 




X 


00 


OX) 


O 




O 












OX) 





O 


(N 





O 


O O 


1 


M O 


O 




O 


















1 


-^ 





O 










O 


O O 





O 


O 




O 



























O 










O 


m 


^0 




C3 






>< 




(N 

1 


i-H cS 


s 

•^H 




^sO 


^0 


<N 


\D 




d 

c3 


(N 


i 


8 » 

d d 


c 

■H 

'o 




53 

Oh 


QJ 


E 




c 

J 




5-H 


QJ 






E 

03 


C 

• i-H 

*-H 

• i-H 






tH 

Oh 
O 


E 

c 


• i-H 

• i-H 

N 

c3 


O c3 


N 




D 


C 


O- 





Oh 


C 


+-I 






4— ' 


X 






N 


f-H 


2 -2 


f W 




q8 


O 


•^ 


1 


O 


O 


O 






O 


,0 






O 


,Q- 


;-^ .p 


t|H 




N 


M-H 


H-H 


«+H 


N 


«4-H 






«HH 


M-H 






H-H 


«4-H 


«4-H 


U-> m- 


O 




<D 




(D 


<D 




<D 




<D 






<u 


(D 






"D 


<D 


<D 


D <D 


u 




u 




u 


u 




u 




u 






u 


U 






U 


u 


U 


u u 



73 
s 

R 

G 



342 



Sullivan and Nord 



c3 



X) 

o 

o 



o3 

d 

-4—4 



X3 

■4-J 

C 
O 

C 
S-< 

o 

Oh 
C/3 

J3 

-d 

a 

u 

73 

o 
cd 

•4—4 

C/3 



E 

< 

•4—4 

c 

Oh 



cd 

C3 

a 

S 



CM 

.a 
re 



c 

•— 
'— 

cd 
> 



cd 

d 
■*— i 

Gfl 

1/3 

CD 
s- 



CD 
CD 
d 
CD 
OX) 

u 

CD 



PJ 



d 

c 

■4— 4 
CD 

c3 



O 



CD 
CD 

a 

CD 
CD 



|| 

c c 
S c 

O o3 

U '- 



s 



CD 



q -O 

c3 £ 



CQ 



o 

CD 



03 

5- 

CD 



"5 y 



2 o 






03 

d o 



o 

X) 

o 



d 



° d 

(/3 -IH 



c3 



C 
CD 



CD 

f 03 03 



•S3 03 C 

d xi <d 



CD 
CD 

c 

CD 



CD I 






X? 

CO 



d 
c 



&* s $ 



73 

03 C/5 



CD 03 



d 

CD 
< 



^O h 00 ON O ^ ^tOO 





o 


















o 












O 


p 




1 














en 


CD 


73 


CD 


CD 


CD 


CD 


CD 


1 


I 

CN 


d 


V 


c 


d 


d 


d 


d 


d 


d 



+ d + + + 



3 + 



s + 



+ 



o 



CD 
C 



o 

d 



+ 



CD 

d 



d d 



+ + oj + + , 

+ + c + + " l " 



+ + + 



+ + 



CD 
+ d + + + 



+ + + 



O (N 0\ (N 



00 o o 



>n uo o 



c3 



oo 

+ 



O 

o 
o 

(N 



o 

o 
o 



o 

o 
o 



o 
o 
o 



o 
o 
o 



r3 



CN 

X 

o 

o 



X X 

o o 

o o 

o o 



o 

o 
o 



(N CN 



CD CD 

d x> 



o 

X 

03 



u 



03 CD O 

o x «2 

03 03 72 

Ph 2 



o 

c 

d 



c 

o 

• — 

■<— ' 

CJ 

D 

4—> 

CJ 

73 

Cu> 



v. 

CJ 

OX) 

c 

CJ 
4—1 

c 

cd 

CJ 

cd 

. — 

a 
• — 

O 
(= 



CJ 
CJ 

W) 

a 

CJ 

O 

60 

O 

A 

CJ 

r3 



OX) 
C 

O 

— 
■<— • 
y: 



y: 

CJ 
CJ 

0X) 

CJ 

O 

bx) 

o 

•^t 
r 1 , 

CJ 
EQ 

cd 

CJ 
'— 
CJ 
CJ 
TD 

•— 

o 

CJ 

se 

CJ 

•— 

CJ 

c 



CJ 

CJ 
CJ 

■4— ' 

cd 

73 

8 



CD M 

1 s 

a .2 . 

^ w 73 

«S 2 c 

73 p .a 



Modifying the Intestinal Microbiota with Antibiotics 343 

Cefotaxime 

Cefotaxime is excreted in bile to a lesser extent than cefoperazone and the effects on the 
intestinal microbiota are usually more moderate. The numbers of enterobacteria are 
suppressed and overgrowth of Pseudomonas species and occasionally of enterococci have 
been observed (33,40,41). 



Cefotiam 

Cefotiam has been shown to decrease the numbers of intestinal enterobacteria and 
lactobacilli and to increase the numbers of Pseudomonas and Candida species (37). 



Cefoxitin 

Pronounced changes in the gastrointestinal microbiota have been shown to occur after 
cefoxitin prophylaxis of patients undergoing colorectal surgery (42) and in hospitalized 
male patients (43). In both studies the major changes observed were decreased numbers of 
enterobacteria and Gram-negative anaerobic species, while there was a proliferation 
of resistant enterococci and enterobacteria. Growth of C difficile was found in 5 of 6 
hospitalized patients (43). 



Cefozopran 

In patients receiving prophylactic antimicrobial treatment after gastrointestinal surgery, 
cefozopran induced decreased numbers of enterobacteria, streptococci, Veillonella and 
Lactobacillus species and overgrowth of enterococci (34). 

Cefpirome 

Administration of cefpirome to healthy male volunteers suppressed the numbers of E. coli 
below the detection limit (44). No other major changes were observed. 



Ceftazidime and Ceftizoxime 

The impact of a single dose of ceftazidime or ceftizoxime on the intestinal microbiota has 
been investigated in healthy volunteers (37). Ceftazidime significantly reduced the 
numbers of enterobacteria and lactobacilli. The number of enterobacteria was suppressed 
also by administration of ceftizoxime, and resistant enterobacteria like Citrobacter and 
Proteus species proliferated. 



Ceftriaxone and Ceftriaxone/Loracarbef 

Ceftriaxone is, as well as cefoperazone, to a large extent excreted biliary and the agent 
induced marked changes in the intestinal microbiota (40,45-49). Ceftriaxone has been 
shown to give rise to elimination or strong suppression of the numbers of Gram-negative 
aerobic rods, reduced numbers of streptococci and staphylococci and also to reduced 
numbers of anaerobic microorganisms. Overgrowth of species resistant to ceftriaxone like 
enterococci and Candida species is common. 

The ecological effect of ceftriaxone has been compared with a step-down therapy of 
ceftriaxone followed by loracarbef in patients with community-acquired pneumonia (49). 



344 Sullivan and Nord 

Both the aerobic and the anaerobic microbiota were affected in a similar way as with 
ceftriaxone only, although the reduction of enterobacteria occurred to a lesser extent. 



Flomoxef or Moxalactam 

Changes in intestinal microbiota have been investigated after administration of flomoxef 
to patients undergoing gastrectomy (34). The effect on the aerobic microbiota was mainly 
detected as decreased numbers of streptococci and overgrowth of enterococci. Anaerobic 
Gram-positive rods and cocci as well as Gram-negative cocci were suppressed. 

In an earlier study, the effect of a single dose of moxalactam was compared with a 
three-dose prophylaxis (50). In both groups of patients there was a reduction in the 
numbers of enterobacteria and streptococci while enterococci proliferated. Several 
anaerobic species decreased significantly in connection with the administration. 



Perorally Administered Cephalosporins 

Studies on the effects of perorally administered cephalosporins are summarized in Table 3. 



Cefaclor 

Alterations in the intestinal microbiota during administration of cefaclor have been studied 
in patients (19) and in healthy volunteers (51,52). In the microbiota of patients there were 
reduced numbers of both aerobic and anaerobic Gram-positive cocci. Enterococci, 
enterobacteria and Bacteroides species increased and there were also increased numbers of 
Candida albicans. In healthy subjects only minor changes occurred in the anaerobic 
microbiota. 



Cefadroxil 

Reduced numbers of intestinal viridans streptococci have been observed during 
administration of cefadroxil in adult healthy subjects (5). In infants being treated for 
infections, disturbances were restricted to the anaerobic microbiota with reduced numbers 
of bifidobacteria and bacteroides (7). 

Cefetamet/Pivoxil 

Cefetamet has a broad spectrum of activity against both aerobic Gram-positive and Gram- 
negative microorganisms. The modification on the intestinal microbiota during treatment 
of patients has, however, been shown to be slight and nonsignificant (53). 



Cefexime 

The ecological effects on the intestinal microbiota of cefixime have been investigated in 
healthy volunteers (51,54) and in patients with exacerbation of chronic bronchitis (53). 
In all three studies, disturbances were observed in the aerobic microbiota as reduced 
numbers of enterobacteria and increased numbers of enterococci. Growth of C difficile 
was common in all studies while the impact on the anaerobic microbiota varied between 
the studies, from reduced numbers of Clostridia to reductions of several species 
including bacteroides. 



Modifying the Intestinal Microbiota with Antibiotics 



345 



cd 



o 

O 



C 

CO 

CD 



a 

c 

1/3 

a 

C 

CO 

o 
13 

a. 

CD 

u 

-a 

•— 

CD 

■«— > 
CO 



< 

13 

s- 

c 

•— 
CD 



cd 

a* 



CO 

a> 
n 

CO 

H 



o 

OD 

CD 
> 

o 



CD 
O 

a 

c3 

*— < 
ex; 

• i-H 
CO 

CD 



CD 
CD 

a 

CD 



W 



c 
o 

o 

C3 

Oh 





CD 




O 




a 




CD 




»H 




.D 




<+H 




<D 




C* 




<D 




W) 


1 

c 


§ &o 


<D 


2 ^ 


O 




a 


fi W) 


o 
U 


.2 E 

-t— < 

o3 




b 




Q 




^ 




•**i4 




t3 




S 




Q 




U 




*} 




*■■«* 




••■■A 




o 




^ 

••-i 5 




~^3 




U 




b? co 




§3 




£' s 




i o3 




ero 
teri 




a ° 




6 •« 




Sh o 




0) o 




c ° 


i 

c 


o 


'— 


03 o3 


CD 

C 


o 3 


< 


4=> 


i 

■4—' 


O 03 

5 ' c 


c 


pO CD 


w 


O ** 




•l-H 




.a 8 




4=) O 




o o 




U 




CD _|_ 




«M 




O M 








1 = 

j CO 




fc 


«+- 


CO 


c 


a S 


CO 


•3 o 


>^ 


§ -d 


cd 

Q 


"2 2 

03 -4— > 




>^ 




03 




CD T3 




CO -««. 




O W) 




Q E 




■fcj 




c 




CD 




bfl 




< 



m 






m t> 



a a V V 



a 



+ 



+ 



+ I + 



I + 



+ 



+ I + 



^O On o o 

C«~) ^H i— < 



>n 



en 



m en en cn =^ 

X X X X 

o o o o 

m io io o 

CN CN (N lO CO 



E 



romH^o\Ti^hrHroooo\OminrH 
ioioio>n^->nioio(Nm>nm^ocN^HVo 



r^ 



00 

00 
CO 

•A 

V 



CN O 

On r- 

<D cd" ^ CD CD 

a c V V d d 



+ 



















« 




(N 






CO 




CN 






CN 
CO 




CO 

1 






i-H 
1 




T— 1 
I 






o 




-i 


QJ 


CD 


■A 


CD 


"A 


CD 


CD 


CN 


CD 


V 


C 


a 


V 


a 


V 


a 


C 


O 


C 



+ 



+ 



+ + + + + + + I + + 



I + + 



+ 



+ 

+ 



+ 



+ 



+ + + + + + + + + I + + 



+++++I++I+++ 



+ 



OOOO l OOOOOO'st-VOOOOOO^OOO l O 

y— ( i—i t— I i-H i-H cN , — I 'sj" CN 



oO'sj-i^r^r^ooocnoor^woi^r-r^^o 

en 



CN Tt 

X X 

o o 

o o 



CN CN CN CN 

X X X X 

o o o o 

o o o o 



o 
o 

Tfr CN CN 



o 
o 



en cN cn cN CN 

X X X X X 

o o o o o 

O m in m in 



in Tt vo CN CN CN CN 



CN 

X 

o 

o 
o 



CN CN ^ 

X X §> 

o o G 

o o o 

cn cn cn 



CD 



o 

1 

c^-H 
CD 

u 



x 
o 

T3 
CD 

u 



5^ ^ 



■4-J 

,o 

CD 

u 



CD 

E 



X 

.$ ( CD 
CD 

u 



x 

o 

C 



CD 

E 

• — 

x 

o 

Qj t ^— l t|— < C<_( 
CD CD CD 

u u u 



CD N 

X O 
O t - 

CD 

u 



c 

CD 

9 



CD 
X 

cd 



2^ «w 

C (D 

-5 03 

CD O 



C 
C 

c 



c 
c 

— ' 

o 

— ' 
0) 

(U 



'V. 

h 

c 

o 

— ' 

c 

o 

cn 

'S 

is: 

C 

c 



4h 

a 

o 

O 

two 

o 

A 

— - 

o 

a. 



C 

c 

— < 



+ 
+ 



u 

o 

4h 
00 

o 
O 

two 

o 

t 

0) 

0) 

•— 

o 

<D 

u_ 
C 

0) 

Cfl 
cd 
0) 
•— 
a 
c 



o 

— ' 

CD 

T3 

. O 

c e 

O O 

•a w 

' ' — — 

c 5 

4H 






o 



•2 <u 






346 Sullivan and Nord 

Cefpodoxime Proxetil 

A marked decrease in numbers of aerobic intestinal microorganisms with disappearance of 
E. coli has been seen during administration of cefpodoxime proxetil in healthy volunteers 
(18,55). The anaerobic microbiota was also affected and after treatment overgrowth of 
enterococci, Candida and C. difficile occurred. 

Cefprozil 

The ecological impact of cefprozil was determined in a double-blind placebo-controlled 
study (56). Analysis of the fecal microbiota revealed mainly a moderate decrease in 
enterobacteria and a few subjects became colonized with C. difficile. 

Ceftibuten 

Ceftibuten administration has been shown to partly affect the aerobic intestinal microbiota 
(57). The numbers of E. coli was significantly reduced while there was an overgrowth of 
enterococci. Four subjects became colonized with yeasts, mainly C. albicans. The 
anaerobic microbiota was disturbed to a lesser degree. However, six volunteers were 
colonized by C. difficile. 



Cefuroxime/Axetil 

The effect of cefuroxime/axetil on the gastrointestinal microbiota has been evaluated in 
patients (53) and in healthy subjects (21,53,58-60). Ecological disturbances have mainly 
been observed as decreased numbers of enterobacteria, overgrowth of enterococci and in 
varied changes in the anaerobic microbiota. In several studies, colonization with Candida 
species and C. difficile has been observed. Fecal concentrations of cefuroxime/axetil, when 
measured, have generally been rather low. In one study though, four subjects had very high 
amounts of the agent in feces and thereby also more pronounced disturbances in the 
microbiota (60). 



Cephradine 

Elimination of staphylococci has been shown to be the major significant change in the 
microbiota occurring during administration of cephradine in healthy volunteers (23). 



Loracarbef 

No major ecological disturbances in the intestinal microbiota have been detected in 
connection with administration of loracarbef as treatment for acute bronchitis (16) or 
in healthy volunteers (61). In patients, new aerobic Gram-negative species were detected 
during the investigation period. However, all strains were susceptible to loracarbef. 
Loracarbef therapy caused increasing levels of enterococci in infants but had no significant 
effect on the anaerobic microbiota (7). 



Monobactams 

The ecological impact of monobactams on the gastrointestinal microbiota is shown in 
Table 4. 



Modifying the Intestinal Microbiota with Antibiotics 



347 



C3 



X 

O 

S-H 

CJ 



c3 

C 
• — < 

03 



X 

<— > 

d 
o 

CJ 

• i-H 

Oh 
03 

Q- 

O 

cj 

5 

d 
a 



c 
cu 

03 
03 

u 

I 

03 

cj 

03 

X) 

o 

c 
o 



<4H 

o 
cj 
a, 

a 



(0 



o 

H 

03 

> 

o 



CJ 

c 

03 

■*— i 

. -H 

03 



O 

c 

03 

01) 

§3 



PJ 



c 
o 

+— I 

cj 
a 

Oh 

a 







CJ 






CJ 






d 






CJ 






S-H 






.13 






«H-H 






CJ 






C* 




03 






OX) 




d 


c3 


01) 


03 


U 


^ 


C 


d 


01) 


o 


_o 


a 


U 


'-4— > 

c3 




'— 






•*— * 


s 

*3 

••"■a 

* ""mi- 

u 




i 

S-H 


C/3 




03 

-<— ' 

o 


03 

73 




c3 

i 


• i-H 

o 

03 




c 
■— 
i) 

■*— > 

d 
ffl 


• i-H 
S-H 

CJ 

■4-J 

CJ 

03 




i 

o 


• ^H 




Jh 


o 




0) 


CJ 




■4— ' 

d 
ffl 


o 

CJ 


6 


i 

o 




S-H 


cd 


03 


03 
c3 
C 


X 
O 


•I-H 

CJ 


< 






S-l 


i 

CJ 


03 

• i-H 


*-l 


C3 


S-H 


c 


X 


CJ 


W 


o 


4-J 
• ^H 




O 


CJ 

CJ 




X) 


O 




O 


CJ 




•— 






03 
< 


+ 

a 




<4- 






c 


C/J 




s- 

03 


CJ 
03 




E 

3 


X? 



C/3 




Z 




<H-H 

o 


i 
"3 


c 

O 


CZ3 

>> 


s 


• i-H 

03 


03 

a 


c3 


S-H 

-<— ' 

C/3 




0) 


03 




Bfl 


T3 




c 






Q 


*5b 

a 

— - 

c 

CJ 

OD 

< 



(N cn cn tj- tj- tj- in 

\o \o ^ ^ ^o \o ^o 



m 

X) 






t^ OO O O C7\ On On 
^O no O t> in VO \Q 



CJ 

d 







o 












o 










^t 


o 


O 




<n 




CN 

1 


A 

i 


o 
1 


o 
I 


^ 


> 


1 


i 
T3 


1 

T3 


i 

73 


1 


d 


V 


V 


V 


V 


d 



I 

d 
V 



CJ 

d 



V 





o 




ra 


o 


O 




o 


O 


r^ 


o 


O 




oo 


O 


On 


oo 


00 






^f 


i-H 


1-H 


T-H 


• 


A 


| 


o 


V 


A 


13 


l 


in 


(N 


1 


1 


d' 


T3 


i— i 


in 


T3 


TD 



+ 



+ + 



+ + + + + 



+ I 



+ + 



+ 



+ + 



+ + + + 

+ + ^ + + ^ ' 



+ + 



+ 



+ + + 



I + + I 



+ + 



+++++++ 



On On O O O O O 



O \D X> O O X> O 

X 

(N 



On On (N in in in (N 

I I 



CN 



m 



CN 



o t> 



CN CN 



\D 



** 













X 




^t 




-<t 


<n cn 


m 










X 

o 




X 

o 


X X 


X 


cn 
X 


cn 
X 


cn 


o 
^ o 






o o 


o 


X 


o o 


■ 


o 


■ — 


o 


o o 


o 


o 


o 


O h 


o 


in 


o 


in 


o o 


o 


o 


o 


o 


o 


o 




o 




CN ^h 


1 ' 


in 


1— < 


CN 


i-H 


in 




m 




6 

c3 










S d 










d 










cj -j-- 










o 










d C<3 










CJ 










CJ 03 










^H 










Oh^: 










-t— » 
N 

< 










| ° 











cn cn ^j- Tf Tt cN cN 

X X X X X X X 

o o m in m o o 

O O CN CN CN O O 

in in ' — i '-h i— < cn "— i 



2a 8 £ 

a Oh o 

* c S 

CJ 03 



CJ 



2 j > 



.s 



Oh 

o 

CJ 

"53 

H 



c 
c 

1) 

d 



c 
•— 

t3 

CJ 

CJ 
73 

CJ 



CJ 



4- 

CJ 



CJ 

c 

• ■-- 

c 
c 



CJ 
CJ 

4h 

00 



CJ 

O 

M 

O 

— 

A 

— i 

6 

0X) 

c 

c 

d 

+ 

+ 

CJ 
0X) 

CJ 

O 

r— 

OX) 

o 

CJ 

-r. 

% 
•8 

5 

CJ 
09 

CJ 



CJ 

CJ 

CJ 

CJ 
— < 

b 

C 

s 



c 
o 

•rt o 

s ^ 

•> 2 

Q S 

"2 + 

03 

•S cj 

c > a 

C3 "^3 CJ 



348 Sullivan and Nord 

Aztreonam 

The effects of aztreonam on the intestinal microbiota have been studied in patient groups 
(62,63) and in healthy volunteers (64). The dominating effects of aztreonam on aerobic 
species have been observed as a marked decrease in numbers of enterobacteria. 
Emergence of aztreonam-resistant enterococci and reduced numbers of anaerobic 
microbiota occurred in connection with higher dosing (64). 



Carbapenems 

The effect of carbapenems on the fecal normal microbiota is shown in Table 4. 



Imipenem 

The effects of parenteral imipenem/cilastin therapy have been evaluated after prophylactic 
treatment of patients undergoing colorectal surgery (65) and in hospitalized patients with 
serious infections (66). In the first study, aerobic Gram-positive cocci, enterobacteria as 
well as several anaerobic species were significantly suppressed. The major effect in the 
latter study was observed as decreased numbers of enterobacteria. 



Meropenem 

The gastrointestinal microbiota has been studied in connection with administration of 
meropenem to healthy male volunteers (67). No measurable concentrations of meropenem 
were found in feces but disturbances were seen both in the aerobic and anaerobic 
microbiota. The numbers of streptococci and enterobacteria decreased while enterococci 
increased. Clostridia, Gram-negative anaerobic cocci and Bacteroides species were 
also suppressed. 



Lenapenem 

In a study where lenapenem was given to healthy male volunteers (68), the antimicrobial 
agent did not influence the total numbers of aerobic or anaerobic bacteria but streptococci 
and Veillonella species were suppressed in numbers. 



OTHER AGENTS WITH INHIBITORY EFFECT ON THE SYNTHESIS 
OF THE CELL WALL— GLYCOPEPTIDES 

Glycopeptides are poorly absorbed and reach very high fecal concentrations and major 
disturbances are expected in the gastrointestinal microbiota. 

A summary of the ecological impact of glycopeptides on the intestinal microbiota is 
shown in Table 4. 



Vancomycin 

Perorally administered vancomycin has been given to healthy subjects and the effects on 
the intestinal microbiota have been analyzed (59,69,70). In the aerobic microbiota the total 
numbers of enterococci and staphylococci have been seen to decrease while resistant 



Modifying the Intestinal Microbiota with Antibiotics 349 

Gram-negative rods and enterococci emerged. Dramatic increase of other naturally 
resistant species like pediococci and lactobacilli has also been observed. Suppressed 
numbers of bacteroides and Bifidobacterium species were seen in two of the studies 
(59,70). 



Teicoplanin 

The ecological impact of teicoplanin has been evaluated in two dosing regimes in healthy 
volunteers (69). Highly glycopeptide-resistant Pediococcus species, enterococci and 
lactobacilli increased during the administration. After the high-dose regimen treatment the 
numbers of staphylococci decreased while enterobacteria increased. 



ANTIMICROBIAL AGENTS INTERFERING WITH THE SYNTHESIS 
OF PROTEINS 

The impact of macrolides, azalide, ketolide, lincosamide and streptogramin on the 
gastrointestinal microbiota is shown in Table 5 and the impact of tetracyclines, 
aminoglycosides, nitrofurantoin and oxazolidone in Table 6. 



Macrolides 



Clarithromycin 

The ecological impact of clarithromycin on the gastrointestinal microbiota has been 
investigated in several studies on healthy volunteers (71-74). In the aerobic microbiota the 
numbers of E. coli have been observed to decrease significantly while there has been a 
concomitant overgrowth of other aerobic Gram-negative species. The degree of the 
disturbances in numbers of enterobacteria has varied depending on the dosing regimen. In 
the study where the lowest dose was applied, there was a suppression also of the number of 
streptococci (74). In the anaerobic microbiota decreased numbers have been detected 
mainly of bifidobacteria, lactobacilli, Clostridia and Bacteroides species. 



Dirithromycin 

The influence of dirithromycin on the normal human intestinal microbiota has been 
evaluated in healthy persons (75). The major route of elimination of the agent is fecal, and 
high fecal concentrations were demonstrated with apparent disturbances in both the 
aerobic and anaerobic microbiota. The numbers of E. coli decreased, streptococci and 
staphylococci increased and there was overgrowth of dirithromycin-resistant enterobac- 
teria. Anaerobic Gram-positive cocci, bifidobacteria, eubacteria and Bacteroides 
decreased while Clostridia and lactobacilli increased during the treatment period. 

Erythromycin 

Marked disturbances have been observed in the intestinal microbiota during oral 
administration of erythromycin in healthy adults (74,76) and in infants (7). The aerobic 
Gram-positive cocci were reduced in numbers and there were marked reductions in the 



350 



Sullivan and Nord 



c3 




■*—> 




O 




•I-H 




X 
o 

S-H 

o 


0) 

o 

q 


• i-H 


cS 


s 


■4— > 

C/3 




C/3 


03 


CD 


e 

• i-H 


tM 


4— > 


o 


C/3 




CD 


0) 


■4-J 


o 


c 


q 


1— 1 


CD 


(D 


ox 


x 

■4— > 

c 
o 


1-4 
CD 

e 



o 

-4— ' 
Oh 
CD 
H 

C/3 

73 

q 

r« 

<D 
73 



c3 

O 

o 

q 

• i-H 
-J 

r- 

<D 
73 

■ H 

o 

<D 
73 

•i-H 

N 
< 

<D 

■H 

'o 

S-H 

o 
c3 



a, 



in 
o 
n 

CO 



2 

OX) 

S3 

> 

O 



o 

•4—4 

o 

c-3 



i 

q 

CD 

o 

q 
o 

U 



i 
o 

< 



CD 

q 

e 

CD 



CD 
OX) 



OX) 

e 



c 
o 

— » 

x 



<D 
O 

CO 



2 

— - 

q 



q 



c 
•— 
o 

— I 

q 
PJ 



o 

o 

•— 
o 

< 



•— 

<D 
X) 



£ 



o -g 

C/3 • — 

^ a 



=3 

c3 



Co 



"a 



CD 

-a 



•i-H 
5-H 

CD 
•*— ' 
CJ 

cd 



2 o 



o 
o 



Q _ 

x -c 



•c 

CD 

•*— ' 
O 

X 



CD 

O 

o 



a 



o 

CO 

x 5 



q 

o 
•*— * 



CD C3 

s s 

Q I 



a 
ox) 



H N m t IT) ^ VO 

t-» r- r- t-» r- r-» r» 



r- 



r- m 



00 ON O 



(N 



hhhhhOOOCOO 



00 



73 

V 



IT) 

00 
(N 






(N 



o 

(N 



q 



73 73 73 73 

V V V V 



+ + 









o 










o 




en 


o 


O 






^t- 




m 


o 






<N 

i 




i-H 
1 


i 




1) 


■A 


ij 


4 


73 


1) 


q 


V 


d 


V 


CN 


V 


q 



£ OO %~- 

JL. — ^ 

\/ On CN 



+ + + 



+ 



+ I 



+ + + + + + + + 



+ I + + 



+ + + + + ^^ + 



+ + + + 
+ + + + 



+ + + + + 



<N 


CM 


(N 


(N 




X 

o 


(N 


OX) 


(N 






en 


^t 


^J- 


^1- 


X 


X 


X 


X 




X 


OX) 

s 


X 






X 


X 


X 


X 


o 


o 


O 


o 


o 


O 


o 


o 


o 


O 


o 


o 


o 


o 


o 


o 


O 


in 


o 


o 


o 


O 


«n 


o 


o 


o 


in 


in 


in 


in 


in 


in 


CN| 


in 


^™ 


in 


^1" 


^ 


in 


00 


^o 


— > 


i— i 


i— i 


q 

■ — 








c 


c 








Pi 


a 


















•1-H 

o 


•^H 

o 






o 


•i-H 

CD 


O 


■ — 








B 

2 








B 

c 


E 
c 






2 




S 
o 


i 
















•— 

X 

■i—> 
•I-H 

s- 

•!-H 


J-H 

X 

— -> 






X 
X 

c 


•i— i 

N 


4— » 
■H 

(1) 


C3 

c 

•-H 








u 








tt 


W 






Csi 


< 


H 


u 









+ + 



+ 



+ 



^^^ + + ^^ + + + + I + + + + 



++++++ 



OCNVOOOOOCNVOVOOinOOeNO 

r^ r-< ^H Ol '— I »-H i— I i— I i— I ^H ^H ^H f>l 



ot^r^c^r^i>r--'sOinrnocnr-r--r-in 

^H | i-H 

cn 



OX) 
OX) 

s 



in 



CN 

X 



^ 



•i-H CO 

3 -a 



o 

d 

d 

. — 

s 

a 
o 
•— 

CJ 

4— I 

CJ 
TD 

CJ 



CJ 

00 



X 

o 



p 

OX) 

• — 

o 



y: 

CJ 
O 

'— 

OX) 



o 

o 

*oo 

o 

A 

c^ 

a. 



0/J 

q 
o 

*3 

+ 
+ 

CJ 

,B 

t|-H 

OX) 

a 

o 

o 

— 
OX) 

o 

CJ 
CO 

cd 

b 
■8 

o 

CJ 
y. 

c^3 
CJ 
— 



O 

CJ 
CJ 

ta 
S3 

. O 

a s 

2 o 

x ♦ j 

CD q 
Q H . 

•2 + 



73 

n 


&5 




— ' 
CO 


• -— . 


73 

CJ 


c 


— 


c 


rt 


^> 


. — 


O 


k 


b 


S 


"C5 



•^ CD 



Modifying the Intestinal Microbiota with Antibiotics 



351 









0) 






















o 






c 


s 


^_^ 


s 


^_^ 


^_^ 


^.^ 


s 


/ 




s 


• l-H 
J3 






2h 


cn 


en 


^~ 


in 


>n 


vo 


o 


VO 


r^ 


l> 


o 






CD 


00 


00 


00 


oo 


00 


OO 


(N 


00 


00 


c 






<H-H 


s — y 


^— ** 


s — y 


s — -' 


*•«■ ^ 


^— ** 


s — y 


^— y 




s — ^ 


o 






0) 






















•l-H 






C. 






















__ 




























03 

c 

•l-H 




























-4-* 

co 
<D 




1 

e 


2 M 






r^ 


o 


o 




o 








■4—" 

c 




o 


"^ » ' ' 






^D 


o 


o 




^D 










c 


C t>0 






in 


i-H 


T-H 




• 












o 


•1 6 

o3 






1 


1 


1 




CN 








CO 




U 


CD 


QJ 


-d 


T3 


TD 


CD 


^ 


6 


CD 


T3 


■4— » 

c 
o 






b 


d 


d 


V 


V 


V 


• 


r^ 


d 


d 


V 




























CO 

<D 


M-H 

o 






+ 


+ 


1 


| 


1 


1 


1 


I 


1 


1 


N 


43 




C "<3 






















03 


-4— » 


























T3 


£ 


























i 


o 

»H 




^ 






















• i-H 


01) 




•~~4 






















o 

•I-H 


CD 
> 






CD 


CD 


! 


6 


d 


QJ 


! 


oJ 


| 


| 


^ 


o 




•i^ 


d 


d 




d 


d 


d 




d 


i 


1 








u 






















03 




























CO 




1 


CO 






















-4— ' 

CO 

•I-H 


CD 

o 


o 

03 


4 -S 


1 


| 


+ 


aj 


o 


oi 


+ 


aj 


1 


1 


O 


e 

03 


CO 


w o 








d 


d 


d 




d 






OD 


-4—' 


























03 


CO 


























-4— ' 

c 

< 

•I-H 

o 


CO 

CD 

S-H 

O 

CD 






+ 


+ 


+ 


■ 


1 


1 


1 


■ 


1 


1 


< 


a 


























o 


CD 


























•I-H 


01) 




i 






















o 


»h 




o # rt 






















Ph 


CD 

a 






1 


1 


1 


1 


1 


1 


1 


1 


1 


1 


,, 




"£ o 


i 


i 


i 


i 


i 


i 


i 


i 


i 


1 


<D 

c 


w 




[§ u 






















o 




























c 




























•I-H 




























X3 




i 


i 






















■H 




o 


o 






















o 

N 






03 03 

X3 -a 


1 


| 


1 


X) 


+ 


QJ 

• 


+ 


QJ 


+ 


I 


03 




WW 

a 


CD 3 












C 


+ 


d 






X 

O 


c 


< 


• i-H 






















c 


o 


o 
























•I-H 

o 

-4-* 


o 

■03 


»h 

CD 

-4—" 




1 


| 


+ 


+ 


+ 


| 


+ 


+ 

i 


1 


1 


c 


Qi 


c 


4-J 4-» 
















+ 






2 


£ 


w 
























3 


HH 


























tfi 




























O 






• i-H 






















•l-H 






4=> o 






















£ 






o o 

S-H 


| 


| 


+ 


+ 


+ 


| 


+ 


| 


1 


1 


CO 






<0 






















3 


























"co 




























O 






M-H 






















O 






O CO 






















J^ 






5-- *-? 






















*W) 






1.8. 

i = 
1 w 


lO 


in 


o 


00 


oo 


t> 


(N 


r- 


oo 


o 


o 






^■^ 


^m^ 


i-H 








i-H 






l-H 


c 

•I-H 

e 
< 


























co" 

c 




<4-H 

o 

CO 


.a e 
c 2 


o 


o 


r-- 


"<t 


"fr 


o 
m 


c-- 


o 
cn 


*? 


T 






03 


6 & 

—3 5-H 


7 

oo 


7 

oo 














cn 


lO 






Q 


T3 j3 

03 






















03 




























S-H 




























-4-J 










ra 


















CD 

H 

'■4- 






>> 

CD 03 
co T3J 


^ 


T-H 

X 




en 


+ 

CN 




<N 


o 




^ cn 


o 

O 

03 






o ■> 


X 

o 


+ 8 


o 


X 

o 


x § 

o cn 


o 


X 

o 


o 

CN 


o 


^X 

B o 






lO 


o 


o 


o 


o 


o 


o 


o 


o 


a- 








(N 


(N 


1— < 


*- ' 


>-H 


i-H 


^o 


^t 


vB 


^t 










CD 






c 

• H 








C3 






CO 

n 

(0 






-4— > 

c 

CD 


•I-H 

CD 

O 

03 

S-H 
-4— < 

1) 






CD 

S 

c3 

S-H 

O 




-H 

o 

• H 


T3 

■H 

o j> 

• l-H 


X 

o 
B 

• H 

s 

6 


CD 

N 


03 

•H 

o M 
& o 


H 






< 


H 


Q 




H 




z 


J 


u 





"9 

a 

s 

•-— < 

♦Hi 

e 

a 



352 



Sullivan and Nord 



c3 



CO 

4—4 

CO 

•a 

o 
top 

o3 



T3 

•-H 

o 

< 

o 

•-H 

o 

5 
c 

o 

a 

X 

O 

c 

• -H 

o 



l-H 

to 



CO 
0) 



"^3 



c 
u 
pa 

> 

o 



o 

c 
c3 

4—4 

CO 

• I-H 

CO 

<D 
S-. 

<4-h 

c 

O 

e 
<d 

E? 



w 



d 

c 

-4— 1 

o 

cd 

Oh 









CD 






O 






a 






CD 






J-l 






,<D 






«4-H 






CD 






0* 




CD 






W) 




c 


c3 


00 




Jh 


^ 


c 


C 


W) 


o 
U 


O 

• I-H 
4— ' 

s- 

4— > 


a 




1 






u 


•»■■* 

u 

CO 


6 


1 


CD 


c3 


f— < 

CD 


T3 


PQ 


4— ' 
1 


O 
o3 




o 
•— 

CD 
•*— < 

C 


•I-H 

5- 
CD 

4W 

o 

X> 




i 

o 


• w* 




s- 


CD 




CD 


O 






o 
o 


6 


i 
o 




u 


c3 


cd 


C3 

C 


X> 

o 


CD 


< 


• i-H 




o 






S-. 


i 


03 


<D 


o 


•c 


<— » 


03 


C 




CD 

4-» 

•l-H 




O 

•I-H 


o 
o 




X> 


o 




o 


o 




•— 






CD 
< 


+ 




tM 






o 


CO 




CD 

3 


-4— » 

o 

CD 
3 

CO 




£ 




o 


i 

CO 

•I-H 


G 


f A 


c 


O 


VJ 


• '— 


•I-H 


03 


£ 


4—4 

03 


Q 


T3 

o3 






CD 


03 




CO 


T3 











Q 


"5h 

a 

4— < 

a 

CD 

< 



00 
00 



ON O 
OO ON 



O 

On 



On 





>^ 




X 


OO 


c 
E 


1 


03 


T3 


T3 T3 


V 


V V 





o 






•— 






P0 






4—4 




1— 1 


•a 




^O 


03 




^ 


o 




I 
OO 


^"^ 


CD 

• 


OO 




c 



+ 



+ 



+ 



+ 



+ + 



+ 



+ 



+ 



+ + 



+ + 



+ 



+ 



+ 



o 



O ^t 



\D 



m 



(N 



r- r-» 



r- 









o 




+ 


X 

o 
o 


X 


o 
o 


X 


o 


"^J- 


o 


o 


o 




m 


o 




OO 




i-H 


^t 




r^D 






1 

c3 




'o 

N 






T3 

• l-H 




03 

• i-H 






O 

-4—4 


CD 


a 






CD 


N 


• i-H 

H 






S 





o 



(N 



(N 



o X o 

o o 



(N 
X 



CJ 

>^ 

X 

IS 

03 



i 
03 



C 

o 
to o 

5 N 



>^ 



o 



73 



CJ 



> 



§ 

■4—4 

cd 

to 

c 

CJ 

c 
o 

u 

cu 
C 

• ^H 

o 



bo 



o 

o 

2h ^o 

Ph 

■j: 

cd 

i 

CO. 



crj 



o 
o 

00 

— 

o 

E 

o 

o 

(N 
CD 

Q 

13 



X) 

— 

CJ 

CJ 

S-H 






cd 
00 






CJ 

d 



a 
c 

• ^H 
•4— I 

u 

CJ 
4-J 

a) 

73 

CJ 

X! 



■v. 

CJ 

W> 

C 

-a 

u 

4-1 

S3 
O 

a 

b/j 



C 



■j-. 

CJ 
CJ 

r^ 

'•4-H 

u 
O 

— -H 

bi) 

O 

A 

4-4 

CJ 

D- 

6 

• .-H 

b0 

C 

c 



+ 
+ 



CJ 
CJ 

b0 

CJ 

O 

— H 

b0 

O 

CJ 
c^ 

cd 

CJ 
5- 

u 

CJ 
T3 

s- 
C 

CJ 

c^ 
cd 

CJ 
CJ 

C 



CJ 

«4-H 

a) 

CJ 

■4—4 

cd 

l-H 

CJ 

T3 

c 



.2 o 

> ^ 

Q 



+ 



T3 

CJ 
In 
3 - 

C/3 g 

0) 0) 



CJ 

g 
E 

03 
ft) >< 



f3 



C3 -O 






O 

c 



Modifying the Intestinal Microbiota with Antibiotics 353 

numbers of enterobacteria while new species of resistant Gram-negative rods proliferated. 
Several subjects became colonized with yeasts. Anaerobic species like bifidobacteria, 
lactobacilli, Clostridia and Bacteroides were also suppressed to a varying degree. 



Roxithromycin 

The consequences of oral treatment with roxithromycin on the intestinal microbiota are 
more limited than the effects of erythromycin in healthy volunteers (77). The fecal 
concentrations were also lower and the changes were restricted to a decrease in total 
counts of Enterobacteriaceae. 



Azalide 



Azithromycin 

The ecological effect of azithromycin has been compared with the effect of clarithromycin 
in healthy volunteers (73). The main impact of azithromycin was detected as decreased 
numbers of bacterial species in the family Enterobacteriaceae. 



Ketolide 



Telithromycin 

Moderate disturbances in the gastrointestinal microbiota have been recorded during 
administration of telithromycin to healthy subjects (71). The numbers of E. coli were 
significantly reduced and overgrowth of staphylococci and resistant enterobacteria was 
observed. In the anaerobic microbiota there were reduced numbers of lactobacilli and 
bifidobacteria. A selection of highly resistant Bacteroides isolates was also recorded 
during and after treatment. 



Lincosamide 



Clindamycin 

The ecological impact of clindamycin on the fecal microbiota has been studied after 
intravenous clindamycin prophylaxis in patients undergoing colorectal surgery (78) and 
after oral administration in healthy subjects (79-81). Clindamycin is excreted in the bile 
and high fecal concentrations have been detected with marked disturbances, in particular 
in the anaerobic microbiota. Enterococci are not susceptible to clindamycin and their 
numbers have usually increased and so have the numbers of clindamycin-resistant 
enterobacteria. Anaerobic Gram-positive cocci and rods and anaerobic Gram-negative 
rods have been markedly suppressed or eliminated during treatment. Emergence of 
clindamycin-resistant Bacteroides species has been detected in one of the studies and 
colonization with C. difficile was common. 



354 Sullivan and Nord 

Streptogramin 



Quin up ristin/Dalfopristin 

In healthy volunteers treated with quinupristin/dalfopristin (RP59500), the impact on the 
fecal microbiota has been investigated (82). The numbers of enterococci and 
Enterobacteriaceae increased significantly and anaerobic non-sporulating and Gram- 
negative bacteria decreased. The total numbers of quinupristin/dalfopristin-resistant and 
also erythromycin-resistant anaerobes and enterococci increased significantly. The 
observed modifications disappeared within 12 weeks after the administration. 



Tetracyclines 



Tetracycline 

The ecological effect of tetracycline hydrochloride on the gastrointestinal microbiota has 
been examined in healthy volunteers (83). Tetracycline had no major effect on the total 
numbers of intestinal microorganisms although a few subjects acquired new strains of 
C. albicans. However, the major finding was the emergence of resistant E. coli strains in 
10 of 15 subjects. 



Doxycycline 

The effect of doxycycline has been evaluated in two studies in healthy subjects (83,84). 
The results are partly consistent in that new resistant strains were detected during 
treatment. Acquisition of C. albicans occurred in subjects in the first mentioned study and 
new strains of Enterobacteriaceae in the latter. In this study, the aerobic microbiota was 
also suppressed while the anaerobic microbiota was not influenced. However, the number 
of fusobacteria was reduced and a marked emergence of resistance was also observed in 
anaerobic microorganisms (84). 



Aminoglycosides 



Tobramycin 

Two dosing regimens of tobramycin have been compared for the selective decontamina- 
tion effect of the digestive tract in healthy volunteers (85). Both regimens markedly 
suppressed the number of aerobic Gram-negative rods while the higher dose also had an 
effect on the anaerobic microbiota as evidenced by low concentrations of beta- 
aspartylglycine. 

Nitrofurantoin 

Nitrofurantoin has been used for prophylaxis in women with recurrent urinary tract 
infections (86). The effect on the fecal microbiota was examined semi-quantitatively. The 
agent had no effect on the numbers of enterococci or enterobacteria and no resistant strains 
or overgrowth of strains was detected. 



Modifying the Intestinal Microbiota with Antibiotics 355 

Oxazolidinone 



Linezolid 

Linezolid is a relatively new synthetic antimicrobial agent that has been evaluated for the 
effects on the gastrointestinal microbiota in healthy male subjects (20). There was a statis- 
tically significant reduction of enterococci whereas the numbers of resistant Klebsiella 
strains increased. The agent also exerted changes in the anaerobic microbiota with 
decreased numbers of lactobacilli, bifidobacteria, Clostridia and strains of Bacteroides. The 
minimum inhibitory concentrations (MIC) values of Bacteroides fragilis strains increased 
during administration and returned to pre-treatment levels on day 35. 



AGENTS BLOCKING THE METABOLISM OF FOLIC ACID 



The impact of folic acid antagonists is summarized in Table 6. 



Co-trimoxazole 

The ecological effects of co-trimoxazole on the intestinal microbiota have been evaluated 
in a scheme for prophylaxis in women suffering from recurrent urinary tract infections 
and in infants being treated for various infections (7,86). In women there was a marked 
decrease in numbers of Enterobacteriaceae and resistant E. coli strains were detected in 
samples of one woman. In infants, lactobacilli and bifidobacteria were nearly absent but no 
other significant changes were observed. 



ANTIMICROBIAL AGENTS THAT INTERFERE 
WITH THE SYNTHESIS OF DNA 

The ecological impact of nitroimidazoles and combinations of metronidazole and 
penicillin or macrolide is shown in Table 6 and the impact of quinolones is shown in 
Table 7. 



Nitroimidazoles 

Metronidazole 

Only minor alterations of the aerobic and anaerobic gastrointestinal microbiota have been 
shown to occur during metronidazole treatment of patients with different infections (87). 

Tinidazole 

Parenterally administered tinidazole has been used in order to prevent postoperative 
infections after abdominal surgery (88). Analyses of the intestinal microbiota revealed that 
the treatment induced proliferation of the numbers of enterococci and staphylococci. 
Anaerobic Gram-positive cocci, fusobacteria and bacteroides were also significantly 
affected during and immediately after the administration period. In connection with oral 



356 



Sullivan and Nord 



C3 



o 

1-H 
CJ 



c3 
C 

. -H 
4— ' 

CJ 



CJ 

■4-J 

d 
o 

CJ 

C 

o 

'o 
d 

• i-H 
P 



cj 

en 

Oh 






o 
5o 

CD 

> 

O 



o 

d 

■i—> 
CO 

• i-H 
CO 

cj 



o 
o 
d 
cj 

Jh 

CJ 



w 



d 
o 
■*— i 

o 
ed 

a 
S 







<D 






O 






d 






CD 






Jh 






.<L> 






<-4-H 






<d 






Pn 




a> 






bU 




1 

c 


d 

en 


W) 


CJ 

o 


^H 


^ 


c 


d 


W) 


o 
U 


a 

•I-H 

■4-J 

en 


a 




i 


Q 




a 

u 


^3 
•••^ 




u 






CJ 
CJ 


CO 

CD 

T3 




CO 


O 




o 

CJ 


-4— > 




■t— » 

d 


o 

en 
X> 




6 


• I-H 




S-H 


o 




aj 


o 




d 


o 
o 


i 
o 


o 




J- 


en 


a 


CJ 

d 


CJ 


•I-H 
S-H 

CD 


< 


• i-H 




i 

O 
•— 

CJ 


o 

en 


•c 

CD 

-4— > 


a 


X) 


W 




•I-H 




o 

• i-H 


o 
o 




X 


o 




o 


o 




Ih 






CJ 
< 


+ 

a 




«4H 






o 


CO 




cj 

X) 


-4—' 

CJ 
CD 




d 

p 


P 

CO 




Z 




•■+- 


, 




o 


CO 

• i-H 


d 


^ 


d 

• — « 


o 

•I-H 


(Tt 


s 


-4— ' 

en 


Q 


73 

K3 


!-H 
■4-J 




CJ 


en 




o 


T3 




Q 


a 

<D 
< 



on aNaNONONaNONONOOOOOOOOO 



00 

in 

00 
1 


o 
o 


1 
73 


aj ^ 


V 


d v 



+ 

+ 



+ 



(N 



(N 



+ 

X 

O 

in 



o 
^ o 

[--- en 



CD CD CD _. 

d d d m 



m 73 
V 



o 
mom 

«p"?S 

dididiaJdJoJr-O^ 

dd'dddd'^'-Hm 



+ 



CJ 

d 



CJ 

d 



CJ 

d 



+ 



CJ 

d 



+ 



+ I + 



+ 



+ 



+ 



+ + 



+ + 



CJ 

d 



CJ 

d 



+ 
+ 



+ +,+ + +,+ + + + + + + +, 
+ + " h + + + " h + + + + + + + + " h 



+ + + 



+ 



+ 



+ 



dd'tinrjooo^iohhhfiio 



o o 

i— i m 



CN 



CJ 



V-) r- m 



r- ^i- 



oo 
m r^ m m m *sO 



(N 

X 

o 
o 



(N M (N M (N M 

X X X X X X 

o o o o o o 

o o o o o o 



o 
>n 



X) 

CN i— ' CN CN 

X X X X 

o o o o o 

o >n o <n m 



X 

o 



inininmTj-^j-i^mcNmcNcNincN 



X 
O 

5-H •!-( 

Oh O 

• i-H 

u 



Tf- 




^t 




r- 


I-H 

o 
o 




CN 


CN 




X 


| 




o 


o 


o 


o 


o 


CN 


Tf 


d 

•i-H 

o 




d 


en 




• i-H 


X 




CJ 


n 




en 

X 


d 

0) 




u 


*— 




d 


en 



pj a 



Modifying the Intestinal Microbiota with Antibiotics 



357 



00 On O i— ' CS CO cN 't"OT)>«D^^hhOOO\\o^O'- ( CN 



"^5 

c 

• -— I 

e 

a 



d 



CN 



oo 



^1- 


CO 




CO 




o\ 


^o 


a 


o 


« 


1 


1 


r^ 


7 


in 


1 




lO 


1 


CN 


V 


V 




V 





d c d c d c d c co 



o o 
On CO 

CO 



O CO 



o 



CO 

oo 



<D HJ 



c r» c c 



CO 

3 V 



+ I + 



d 



d 



+ 



d 



d 



<3J 

d 



+ 



(D 



+ + + + 

+ ^ ^ + ^ + + 



+ + + + + + + 
+ + + + + + + 



+ + + + + + + 
+ + + + + + + 



+ + I + + 



+ 



+ 



+ 



+ + + + 



00 O CN ^O O O 



CN 



O^^OOOOOOOCNI^O 



>n tj- ^o 

CN 



r- 



t^ c — c^ 



r- 



ooioioininmr-r^r^ 



o 

ro h in 



CO 

X 

o o o o o o 

O CN CN O O O 
Tf CO CO CN lO 'nI" 



X O 

O <G 

53 -3 C 



a a 



i, 

X 
O 

O 
d § ° 



c3 
X 

o 



o 



o 
o 



C3 

X 

o 

qd 



S o o o e 



CN CN CN CN CN CN (N 

X X X X X X X 

o o o o o o o 

o o o o o o o 

^- ^" CN ^1" CN i-h Tt 



o 

X 

o 



o 





CN 








CN 




CN 




X 








X 




X 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


o 


CN 


CN 


^1- 


CN 


c 

•1-H 

o 

X 

o 

o 


CN 


^t 


c 

o 

a 

X 

o 

c: 

Oh 



358 



Sullivan and Nord 



^3 

s 

4— ' 

o 

•I-H 
X) 

o 

U 

o 



c3 

.9 

■i— • 

co 

CJ 



CD 

M 
■*— > 

d 

o 

co 

<D 

c 
c> 

'o 

c 

d 

a 

<4-H 

o 

4— ' 
CJ 
Cd 

o- 
E 



.Q 



o 

S-H 

> 

o 



O 

d 

03 
-I—' 

BO 
& 



u 

C 

8? 



W 



c 
o 

CJ 
c3 
Oh 



u 



c 



I 

e 

4— ' 

C 



O 

d 

J-l 

.1) 

CD 



bo 

§ bp 



i 

d 

CD 

c d 61) 

•9 .2 E 



o3 

*3 






^ 



<u 

4-J 
CJ 
c3 

CQ 



d 



i 
c 



u 



< X5 



o 

c3 
-O 



CJ 

O 

S-H 






d 



CO .2 

o3 _S 

Q 1 



CO 



CO 
CD 

T3 



i o3 

a) £ 

d § 

M-4 Xi 



2 -3 

CD O 

O 

o 



CJ 

03 03 

5- 

CJ 



C3 

• r^ 

s- 

CD 



CJ 
CJ 

C 

CJ 



<D _|_ 



CO 

-t-J 

CJ 

CD 

£? 

CO 



d 
c 



c3 



CD 03 

S 3 



c 

CJ 
< 



^t o> co ^j- >n vo o 

O i-H CN CN CN (N ^h 



r3 



O 









o 




o 


m 




cd 




(N 


O 

co 
1 


d 


O 

co 




CJ 


1 

V 


(N 


d 


^o 


^f 


d 



+ 



+ 



+ 



I + + 



+ 



+ + + 



+ + + + + + 
+ + + + + + 



+ 



+ 



+ 
+ 



+ + 



ID M (N ^3 00 (N (N 



IT) 

co 

I 



O 



CN 
X 

o 
o 



o 

o 



o 

o 



Tt CN 



.9 

o 

X 

c 

3 



I> 00 



o 

O 

O 

CO CN 

X + 

o o 
o o 



o 
o 



o 

o 



CN CN 



d .s 



CJ 

c3 
X 



d , 

O cJJ 



03 X 

x c 

O CC 
03 



> 

+2 5 o 

00 00 H 



CJ 



r3 

. — 

> 

a 

•— 
03 

C 
cd 

4— ' 

c 

cd 
o 






CN 



j-. 

t 



b/j 
S 
O 

m 

CN 

r- 

S-H 

o 






c 
c 

u 

I 

CJ 

■a 



■j~. 

CJ 



X! 



CJ 

b0 



. r 

CJ 

b0 

a 

u 
O 

b0 
O 

A 

4-J 



b/j 

§ 

4—< 
J-, 

. r- 

■j; 

CJ 

o 

CJ 
b0 



o 

O 
i— i 
b0 

O 

—i 

■8 

C 

CJ 
c^ 



u 

CJ 

CJ 
CJ 

4— » 

b 

C 

B 



on 
B 



o 

o 



+ 



CN 

X 



CJ 

o 

bO T3 
S 



cd 



O 
CN 



S 

•2 cj 

2 1 

5 x 

•^ CJ 



Modifying the Intestinal Microbiota with Antibiotics 359 

administration of tinidazole to healthy subjects, no significant changes have been detected 
in the gastrointestinal microbiota (89). 



Metronidazole in Combination with Amoxycillin 

In patients with Helicobacter pylori infection treated with omeprazole, metronidazole and 
amoxycillin, the alterations in the intestinal microbiota have been evaluated (90). Marked 
ecological disturbances were seen. The numbers of enterococci, enterobacteria, other than 
E. coli, and peptostreptococci increased significantly. Several patients became colonized 
with Klebsiella and Citrobacter species as well as with yeasts. 



Metronidazole in Combination with Clarithromycin 

The influence of H. pylori treatment with omeprazole, metronidazole and clarithromycin 
on the intestinal microbiota has been examined in two groups of patients (90,91). In the 
first mentioned study, it was found that the numbers of bifidobacteria, Clostridia and 
species of Bacteroides were significantly decreased during treatment whereas the numbers 
of enterococci increased. Strains of enterococci, Enterobacteriaceae and Bacteroides spp. 
had significantly increased MIC values during the administration. In the second study the 
microbiota was compared to that of healthy subjects. Before treatment, patients were 
characterized by high concentrations of lactobacilli. Immediately after treatment there was 
an increased colonization with yeasts and enterobacteria, other than E. coli, while the 
growth of lactobacilli, Clostridia and bacteroides decreased. Four weeks after the start of 
the study the microbiota of patients was similar to that in healthy subjects. 



Quinolones 

The ecological impact of quinolone administration on the fecal microbiota is described in 
Table 7. 



Ciprofloxacin 

The ecological consequences of ciprofloxacin have been evaluated in patients in connection 
with colorectal surgery (92), in patients with acute leukaemia in remission (96), 
in prevention of bacterial infections in cirrhosis (101,102) and in treatment of travelers' 
diarrhea (103). A number of studies have also been performed on healthy volunteers 
(93-95,97-100,104,105). Ciprofloxacin is excreted in feces in extremely high concen- 
trations and has an activity mainly against Gram-negative aerobic rods. Marked 
suppression or elimination of enterobacteria has also been shown to occur, both in patients 
and in the healthy subjects examined. The extension of disturbances has varied depending 
on the doses. Minor alterations of numbers of Gram-positive aerobic cocci, mainly 
enterococci, have further been observed and in some studies minor alterations were 
detected also in the anaerobic microbiota. Ciprofloxacin-resistant species of Pseudomonas 
and Acinetobacter have been detected during treatment of patients with acute leukaemia 
(96) and in healthy volunteers who were given ciprofloxacin intravenously (98). 
Furthermore, 4 of 7 ciprofloxacin-treated patients with travelers' diarrhea acquired multi- 
resistant E. coli and in 4 subjects increased MIC values of ciprofloxacin for Bacteroides spp. 
were detected (103). 



360 Sullivan and Nord 

Enoxacin 

The effect of enoxacin on the colonic microbiota in human volunteers has been examined 
(106). Enterobacteria were almost completely suppressed during administration of the 
drug whereas other aerobic and anaerobic species were not significantly affected. 



Garenoxacin 

The ecological effect of garenoxacin has been evaluated in healthy individuals receiving 
oral doses ranging between 100 and 1200 mg daily (107). Higher doses resulted in marked 
effects on the intestinal microbiota; the strongest effect was noticed in reduced numbers of 
Bacteroides species. Fecal concentrations of garenoxacin also increased with higher doses 
as well as the selection of resistant strains, mainly enterococci and enterobacteria. In 
comparison, the Bacteroides species strains were less susceptible to the quinolone agent. 



Gatifloxacin 

Gatifloxacin has been given to healthy subjects in order to study the impact on the normal 
intestinal microbiota (108). Gatifloxacin possesses a broad spectrum of antimicrobial 
activity and the administration resulted in not only elimination or strong suppression of 
E. coli strains but also in decreased numbers of enterococci and increased numbers 
of staphylococci. The numbers of Clostridia and fusobacteria decreased significantly in the 
anaerobic microbiota. 



Gemifloxacin 

Gemifloxacin is another agent with a broad spectrum of antimicrobial activity. It is active 
both against Gram-positive and Gram-negative bacteria. The ecological impact of the 
agent has been investigated in a placebo-controlled study in healthy volunteers (109) and 
in a randomized cross-over study where the effect of a single dose was investigated in 
healthy subjects (110). In the first mentioned study, the effect of gemifloxacin was shown 
to be selective with reduced numbers mainly of enterococci, streptococci and 
enterobacteria. The single dose caused a pronounced reduction in the numbers of 
E. coli and to a lesser extent also of enterococci and Bacteroides species. New quinolone- 
resistant isolates of Gram-negative aerobes appeared in some subjects. 



Levofloxacin 

Levofloxacin has been shown to cause a selective reduction in the normal microbiota of 
healthy subjects, mainly directed towards Gram-negative aerobic rods (111,112). The 
numbers of enterococci were reduced to a lesser extent. Increased MIC values against 
strains of Bacteroides was detected in one study (111). 



Lomefloxacin 

Almost a complete eradication of Gram-negative aerobic rods have been shown to occur in 
the intestinal microbiota of volunteers during administration of lomefloxacin (113). 
Aerobic Gram-positive and anaerobic microorganisms were virtually unaffected. 



Modifying the Intestinal Microbiota with Antibiotics 361 

Moxifloxacin 

The ecological impact of moxifloxacin has been evaluated in healthy subjects (72). The 
administration caused significant decreases of enterococci and enterobacteria while no 
other major changes were observed. 

Norfloxacin 

A number of studies have investigated the ecological effects of norfloxacin on the normal 
intestinal microbiota (86,114-119). All studies have been performed in healthy subjects 
and the results have been consistent. Elimination or strong suppression of enterobacteria 
has been observed and slight reductions of enterococci have been detected in connection 
with the highest dosing regimens. Only minor fluctuations of other species have been seen. 



Ofloxacin 

The potential of ofloxacin to disturb the intestinal microbiota has been studied in healthy 
volunteers (112,120) as well as in patients undergoing gastric surgery (121). In 
both volunteers and in patients the numbers of enterobacteria were strongly suppressed 
or eliminated and the numbers of enterococci were significantly reduced. In patients the 
numbers of lactobacilli, bifidobacteria, eubacteria and species of Veillonella and 
Bacteroides were also affected. 



Pefloxacin 

The influence of pefloxacin on the gastrointestinal microbiota with regard to colonization 
resistance has been evaluated in two studies on healthy volunteers (104,122). Gram- 
negative aerobic rods were eliminated during treatment while the numbers of enterococci 
were slightly suppressed. In one of the studies a significant increase of yeasts was detected 
in half of the subjects (122). 



Rufloxacin 

The impact of rufloxacin on intestinal microbiota has been studied in healthy male 
volunteers after a single dose (119) and in connection with prophylactic treatment of 
patients with cancer (123). The single dose significantly reduced the numbers of 
Enterobacteriaceae. This was also observed in patients but the number of Bacteroides 
species was affected as well, however to a lesser extent. The MIC values of rufloxacin for 
enterococci increased significantly during the second week of treatment. 

Sitafloxacin 

Sitafloxacin has been shown to markedly suppress both the aerobic and anaerobic 
intestinal microbiota in healthy persons (124). Most anaerobic microorganisms as well as 
the aerobic Gram-negative rods were eliminated on the third day of administration until 
one day after the discontinuation of the drug. 

Sparfloxacin 

Administration of sparfloxacin to male volunteers has been shown to have a strong impact 
on E. coli and to moderately reduce the numbers of enterococci (125). 



362 Sullivan and Nord 

Trovafloxacin 

The ecological impact of trovafloxacin has been evaluated in connection with multiple 
(126) and single doses (110) administered to healthy males. The numbers of 
Enterobacteriaceae were suppressed in both studies, after long-term use below the 
detection limit. A single dose also resulted in decreased counts of B. fragilis group species 
in some subjects. 



CONCLUSION 

Antibiotics have a profound place in modern medicine and are indispensable in the 
treatment of infectious diseases. However, their antimicrobial properties may also affect 
members of the intestinal microbiota and thereby alter its composition and activity. This 
may lead to unwanted side effects. It is therefore important to select the appropriate 
antibiotic and dose that will cause the eradication of the infectious agent but will 
minimally affect the intestinal microbiota. 



REFERENCES 

1. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet 2003; 361:512-519 
discussion 63-65. 

2. Vollaard EJ, Clasener HA. Colonization resistance. Antimicrob Agents Chemother 
1994; 38:409-414. 

3. Hoiby N. Ecological antibiotic policy. J Antimicrob Chemother 2000; 46:59-62. 

4. van der Waaij D, Nord CE. Development and persistence of multi-resistance to 
antibiotics in bacteria; an analysis and a new approach to this urgent problem. Int 
J Antimicrob Agents 2000; 16:191-197. 

5. Adamsson I, Edlund C, Sjostedt S, Nord CE. Comparative effects of cefadroxil and 
phenoxymethylpenicillin on the normal oropharyngeal and intestinal microflora. 
Infection 1997; 25:154-158. 

6. Heimdahl A, Kager L, Nord CE. Changes in the oropharyngeal and colon microflora in 
relation to antimicrobial concentrations in saliva and faeces. Scand J Infect Dis Suppl 
1985; 44:52-58. 

7. Bennet R, Eriksson M, Nord CE. The fecal microflora of 1-3-month-old infants during 
treatment with eight oral antibiotics. Infection 2002; 30:158-160. 

8. Black F, Einarsson K, Lidbeck A, Orrhage K, Nord CE. Effect of lactic acid producing 
bacteria on the human intestinal microflora during ampicillin treatment. Scand J Infect 
Dis 1991;23:247-254. 

9. Knothe H, Wiedemann B. Die Wirkung von Ampicillin auf die Darmflora des gesunden 
Menschen. Zentralbl Bakteriol Hyg I Abt 1965; 197:234-243. 

10. Leigh DA. Pharmacology and toxological studies with amoxycillin, talampicillin and 
ampicillin and a clinical trial of parenteral amoxycillin in serious hospital infections. 
Drugs Exp Clin 1979; 5:129-139. 

11. Kager L, Malmborg AS, Sjostedt S, Nord CE. Concentrations of ampicillin plus 
sulbactam in serum and intestinal mucosa and effects on the colonic microflora in 
patients undergoing colorectal surgery. Eur J Clin Microbiol 1983; 2:559-563. 

12. Kager L, Liljeqvist L, Malmborg AS, Nord CE, Pieper R. Effects of ampicillin plus 
sulbactam on bowel flora in patients undergoing colorectal surgery. Antimicrob Agents 
Chemother 1982; 22:208-212. 



Modifying the Intestinal Microbiota with Antibiotics 363 

13. Stark CA, Adamsson I, Edlund C, et al. Effects of omeprazole and amoxycillin on the 
human oral and gastrointestinal microflora in patients with Helicobacter pylori infection. 
J Antimicrob Chemother 1996; 38:927-939. 

14. Gipponi M, Sciutto C, Accornero L, et al. Assessing modifications of the intestinal 
bacterial flora in patients on long-term oral treatment with bacampicillin or amoxicillin: 
a random study. Chemioterapia 1985; 4:214-217. 

15. Mittermayer HW. The effect of amoxycillin and amoxycillin plus clavulanic acid on 
human bowel microflora. In: Croydon EAP, Michel MF, eds. Augmentin. Amsterdam: 
Elsevier Science, 1983. 

16. Floor M, van Akkeren F, Rozenberg-Arska M, et al. Effect of loracarbef and amoxicillin 
on the oropharyngeal and intestinal microflora of patients with bronchitis. Scand J Infect 
Dis 1994;26:191-197. 

17. Edlund C, Stark C, Nord CE. The relationship between an increase in beta-lactamase 
activity after oral administration of three new cephalosporins and protection against 
intestinal ecological disturbances. J Antimicrob Chemother 1994; 34:127-138. 

18. Brismar B, Edlund C, Nord CE. Impact of cefpodoxime proxetil and amoxicillin on the 
normal oral and intestinal microflora. Eur J Clin Microbiol Infect Dis 1993; 
12:714-719. 

19. Christensson B, Nilsson-Ehle I, Ljungberg B, et al. Swedish Study Group. A randomized 
multicenter trial to compare the influence of cefaclor and amoxycillin on the colonization 
resistance of the digestive tract in patients with lower respiratory tract infection. 
Infection 1991; 19:208-215. 

20. Lode H, Von der Hoh N, Ziege S, Borner K, Nord CE. Ecological effects of linezolid 
versus amoxicillin/clavulanic acid on the normal intestinal microflora. Scand J Infect Dis 
2001;33:899-903. 

21. Wise R, Bennett SA, Dent J. The pharmacokinetics of orally absorbed cefuroxime 
compared with amoxycillin/clavulanic acid. J Antimicrob Chemother 1984; 
13:603-610. 

22. Motohiro T, Tanaka K, Koga T, et al. Effect of BRL 25000 (clavulanic acid-amoxicillin) 
on bacterial flora in human feces. Jpn J Antibiot 1985; 38:441-480. 

23. Brumfitt W, Franklin I, Grady D, Hamilton-Miller JM. Effect of amoxicillin-clavulanate 
and cephradine on the fecal flora of healthy volunteers not exposed to a hospital 
environment. Antimicrob Agents Chemother 1986; 30:335-337. 

24. Lambert-Zechovsky N, Bingen E, Proux MC, Aujard Y, Mathieu H. Effect of 
amoxycillin combined with clavulanic acid on the fecal flora of children. Pathol Biol 
1984; 32:436-442. 

25. Heimdahl A, Nord CE, Weilander K. Effect of bacampicillin on human mouth, throat 
and colon flora. Infection 1979; 7:S446-S451. 

26. Knothe H, Lembke U. The effect of ampicillin and pivampicillin on the intestinal flora of 
man. Zentralbl Bakteriol [Orig A] 1973; 223:324-332. 

27. Nord CE, Bergan T, Aase S. Impact of azlocillin on the colon microflora. Scand J Infect 
Dis 1986; 18:163-166. 

28. Kager L, Malmborg AS, Nord CE, Sjostedt S. The effect of piperacillin prophylaxis on 
the colonic microflora in patients undergoing colorectal surgery. Infection 1983; 
11:251-254. 

29. Nord CE, Brismar B, Kasholm-Tengve B, Tunevall G. Effect of piperacillin/tazobactam 
treatment on human bowel microflora. J Antimicrob Chemother 1993; 31:61-65. 

30. Knothe H. The influence of pivmecillinam on the human gut flora. Arzneimittel- 
forschung Forschung (Drug Reg) 1976; 26:427-431. 

31. Sullivan A, Edlund C, Svenungsson B, Emtestam L, Nord CE. Effect of perorally 
administered pivmecillinam on the normal oropharyngeal, intestinal and skin microflora. 
J Chemother 2001; 13:299-308. 



364 Sullivan and Nord 

32. Nord CE, Bergan T, Thorsteinsson SB. Impact of ticarcillin/clavulanate on the intestinal 
microflora. J Antimicrob Chemother 1989; 24:221-226. 

33. Vogel F, Knothe H. Changes in aerobic fecal bacterial flora of severely ill patients during 
antibiotic treatment. Klin Wochenschr 1985; 63:1174-1179. 

34. Takesue Y, Yokoyama T, Akagi S, et al. Changes in the intestinal flora after the 
administration of prophylactic antibiotics to patients undergoing a gastrectomy. Surg 
Today 2002; 32:581-586. 

35. Kager L, Brismar B, Malmborg AS, Nord CE. Impact of cefbuperazone on the colonic 
microflora in patients undergoing colorectal surgery. Drugs Exp Clin Res 1986; 
12:983-986. 

36. Bacher K, Schaeffer M, Lode H, Nord CE, Borner K, Koeppe P. Multiple dose 
pharmacokinetics, safety, and effects on fecal microflora, of cefepime in healthy 
volunteers. J Antimicrob Chemother 1992; 30:365-375. 

37. Knothe H, Dette GA, Shah PM. Impact of injectable cephalosporins on the 
gastrointestinal microflora: observations in healthy volunteers and hospitalized patients. 
Infection 1985; 13:S129-S133. 

38. Alestig K, Carlberg H, Nord CE, Trollfors B. Effect of cefoperazone on fecal flora. 
J Antimicrob Chemother 1983; 12:163-167. 

39. Lambert-Zechovsky N, Bingen E, Proux MC, Aujard Y, Mathieu H. Effects of 
cefoperazone on children's fecal flora. Pathol boil 1984; 32:439-442. 

40. Guggenbichler JP, Kofler J. Influence of third-generation cephalosporins on aerobic 
intestinal flora. J Antimicrob Chemother 1984; 14:67-70. 

41. Lambert-Zechovsky N, Bingen E, Aujard Y, Mathieu H. Impact of cefotaxime on the 
fecal flora in children. Infection 1985; 13:S140-S144. 

42. Kager L, Ljungdahl I, Malmborg AS, Nord CE, Pieper R, Dahlgren P. Antibiotic 
prophylaxis with cefoxitin in colorectal surgery: effect on the colon microflora and septic 
complications-a clinical model for prediction of the benefit and risks in using a new 
antibiotic in prophylaxis. Ann Surg 1981; 193:277-282. 

43. Mulligan ME, Citron D, Gabay E, Kirby BD, George WL, Finegold SM. Alterations in 
human fecal flora, including ingrowth of Clostridium difficile, related to cefoxitin 
therapy. Antimicrob Agents Chemother 1984; 26:343-346. 

44. Knothe H, Schafer V, Sammann A, Badian M, Shah PM. Influence of cefpirome 
on pharyngeal and fecal flora after single and multiple intravenous adminis- 
trations of cefpirome to healthy volunteers. J Antimicrob Chemother 1992; 
29:81-86. 

45. Nilsson-Ehle I, Nord CE, Ursing B. Ceftriaxone: pharmacokinetics and effect on the 
intestinal microflora in patients with acute bacterial infections. Scand J Infect Dis 1985; 
17:77-82. 

46. Cavallaro V, Catania V, Bonaccorso R, et al. Effect of a broad-spectrum cephalosporin 
on the oral and intestinal microflora in patients undergoing colorectal surgery. 
J Chemother 1992; 4:82-87. 

47. de Vries-Hospers HG, Tonk RH, van der Waaij D. Effect of intramuscular ceftriaxone on 
aerobic oral and fecal flora of 11 healthy volunteers. Scand J Infect Dis 1991; 
23:625-633. 

48. Welling GW, Meijer-Severs GJ, Helmus G, et al. The effect of ceftriaxone on the 
anaerobic bacterial flora and the bacterial enzymatic activity in the intestinal tract. 
Infection 1991; 19:313-316. 

49. Vogel F, Ochs HR, Wettich K, et al. Effect of step-down therapy of ceftriaxone plus 
loracarbef versus parenteral therapy of ceftriaxone on the intestinal microflora in 
patients with community-acquired pneumonia. Clin Microbiol Infect 2001; 
7:376-379. 



Modifying the Intestinal Microbiota with Antibiotics 365 

50. Kager L, Malmborg AS, Nord CE, Sjostedt S. Impact of single dose as compared to three 
dose prophylaxis with latamoxef (moxalactam) on the colonic microflora in patients 
undergoing colorectal surgery. J Antimicrob Chemother 1984; 14:171-177. 

51. Finegold SM, Ingram-Drake L, Gee R, et al. Bowel flora changes in humans 
receiving cefixime (CL 284,635) or cefaclor. Antimicrob Agents Chemother 1987; 
31:443-446. 

52. Nord CE, Heimdahl A, Lundberg C, Marklund G. Impact of cefaclor on the normal 
human oropharyngeal and intestinal microflora. Scand J Infect Dis 1987; 19:681-685. 

53. No velli A, Mazzei T, Fallani S, Dei R, Cassetta MI, Conti S. Betalactam therapy and 
intestinal flora. J Chemother 1995; 7:25-31. 

54. Nord CE, Movin G, Stalberg D. Impact of cefixime on the normal intestinal microflora. 
Scand J Infect Dis 1988; 20:547-552. 

55. Orrhage K, Sjostedt S, Nord CE. Effect of supplements with lactic acid bacteria and 
oligofructose on the intestinal microflora during administration of cefpodoxime proxetil. 
J Antimicrob Chemother 2000; 46:603-612. 

56. Lode H, Muller C, Borner K, Nord CE, Koeppe P. Multiple-dose pharmacokinetics of 
cefprozil and its impact on intestinal flora of volunteers. Antimicrob Agents Chemother 
1992; 36:144-149. 

57. Brismar B, Edlund C, Nord CE. Effect of ceftibuten on the normal intestinal microflora. 
Infection 1993; 21:373-375. 

58. Edlund C, Brismar B, Sakamoto H, Nord CE. Impact of cefuroxime-axetil on the normal 
intestinal microflora. Microb Ecol Health Dis 1993; 6:185-189. 

59. Edlund C, Barkholt L, Olsson-Liljequist B, Nord CE. Effect of vancomycin on intestinal 
flora of patients who previously received antimicrobial therapy. Clin Infect Dis 1997; 
25:729-732. 

60. Leigh DA, Walsh B, Leung A, Tait S, Peatey K, Hancock P. The effect of cefuroxime 
axetil on the fecal flora of healthy volunteers. J Antimicrob Chemother 1990; 
26:261-268. 

61. Nord CE, Grahnen A, Eckernas SA. Effect of loracarbef on the normal 
oropharyngeal and intestinal microflora. Scand J Infect Dis 1991; 23:255-260. 

62. Jones PG, Bodey GP, Swabb EA, Rosenbaum B. Effect of aztreonam on throat and 
stool flora of cancer patients. Antimicrob Agents Chemother 1984; 26:941-943. 

63. Kager L, Brismar B, Malmborg AS, Nord CE. Effect of aztreonam on the colon 
microflora in patients undergoing colorectal surgery. Infection 1985; 13:111-114. 

64. de Vries-Hospers HG, Welling GW, Swabb EA, van der Waaij D. Selective 
decontamination of the digestive tract with aztreonam: a study of 10 healthy volunteers. 
J Infect Dis 1984; 150:636-642. 

65. Kager L, Brismar B, Malmborg AS, Nord CE. Imipenem concentrations in colorectal 
surgery and impact on the colonic microflora. Antimicrob Agents Chemother 1989; 
33:204-208. 

66. Nord CE, Kager L, Philipson A, Stiernstedt G. Impact of imipenem/cilastatin therapy on 
fecal flora. Eur J Clin Microbiol 1984; 3:475-477. 

67. Bergan T, Nord CE, Thorsteinsson SB. Effect of meropenem on the intestinal microflora. 
Eur J Clin Microb Infect Dis 1991; 10:524-527. 

68. Nakashima M, Uematsu T, Kosuge K, Nakagawa S, Hata S, Sanada M. Pharmacoki- 
netics and safety of BO-2727, a new injectable 1 -beta-methyl carbapenem antibiotic, and 
its effect on the fecal microflora in healthy male volunteers. J Antimicrob Chemother 
1994; 33:987-998. 

69. Van der Auwera P, Pensart N, Korten V, Murray BE, Leclercq R. Influence of oral 
glycopeptides on the fecal flora of human volunteers: selection of highly glycopeptide- 
resistant enterococci. J Infect Dis 1996; 173:1129-1136. 



366 Sullivan and Nord 

70. Lund B, Edlund C, Barkholt L, Nord CE, Tvede M, Poulsen RL. Impact on human 
intestinal microflora of an Enterococcus faecium probiotic and vancomycin. Scand 
J Infect Dis 2000; 32:627-632. 

71. Edlund C, Alvan G, Barkholt L, Vacheron F, Nord CE. Pharmacokinetics and 
comparative effects of telithromycin (HMR 3647) and clarithromycin on the 
oropharyngeal and intestinal microflora. J Antimicrob Chemother 2000; 46:741-749. 

72. Edlund C, Beyer G, Hiemer-Bau M, Ziege S, Lode H, Nord CE. Comparative effects of 
moxifloxacin and clarithromycin on the normal intestinal microflora. Scand J Infect Dis 
2000; 32:81-85. 

73. Matute AJ, Schurink CA, Krijnen RM, Florijn A, Rozenberg-Arska M, Hoepelman IM. 
Double-blind, placebo-controlled study comparing the effect of azithromycin with 
clarithromycin on oropharyngeal and bowel microflora in volunteers. Eur J Clin 
Microbiol Infect Dis 2002; 21:427-431. 

74. Brismar B, Edlund C, Nord CE. Comparative effects of clarithromycin and erythromycin 
on the normal intestinal microflora. Scand J Infect Dis 1991; 23:635-642. 

75. Eckernas SA, Grahnen A, Nord CE. Impact of dirithromycin on the normal oral and 
intestinal microflora. Eur J Clin Microbiol Infect Dis 1991; 10:688-692. 

76. Heimdahl A, Nord CE. Influence of erythromycin on the normal human flora and 
colonization of the oral cavity, throat and colon. Scand J Infect Dis 1982; 
14:49_56. 

77. Pecquet S, Chachaty E, Tancrede C, Andremont A. Effects of roxithromycin on fecal 
bacteria in human volunteers and resistance to colonization in gnotobiotic mice. 
Antimicrob Agents Chemother 1991; 35:548-552. 

78. Kager L, Liljeqvist L, Malmborg AS, Nord CE. Effect of clindamycin prophylaxis on the 
colonic microflora in patients undergoing colorectal surgery. Antimicrob Agents 
Chemother 1981; 20:736-740. 

79. Heimdahl A, Nord CE. Effect of erythromycin and clindamycin on the indigenous 
human anaerobic flora and new colonization of the gastrointestinal tract. Eur J Clin 
Microbiol 1982; 1:38-48. 

80. Orrhage K, Brismar B, Nord CE. Effect of supplements with Bifidobacterium longum 
and Lactobacillus acidophilus on the intestinal microbiota during administration of 
clindamycin. Microb Ecol Health Dis 1994; 7:17-25. 

81. Sullivan A, Barkholt L, Nord CE. Lactobacillus acidophilus. Bifidobacterium lactis and 
Lactobacillus F19 prevent antibiotic-associated ecological disturbances of Bacteroides 
fragilis in the intestine. J Antimicrob Chemother 2003; 52:308-311. 

82. Scanvic-Hameg A, Chachaty E, Rey J, et al. Impact of quinupristin/dalfopristin 
(RP59500) on the fecal microflora in healthy volunteers. J Antimicrob Chemother 2002; 
49:135-139. 

83. Bartlett JG, Bustetter LA, Gorbach SL, Onderdonk AB. Comparative effect of 
tetracycline and doxycycline on the occurrence of resistant Escherichia coli in the 
fecal flora. Antimicrob Agents Chemother 1975; 7:55-57. 

84. Heimdahl A, Nord CE. Influence of doxycycline on the normal human flora and 
colonization of the oral cavity and colon. Scand J Infect Dis 1983; 15:293-302. 

85. Mulder JG, Wiersma WE, Welling GW, van der Waaij D. Low dose oral tobramycin 
treatment for selective decontamination of the digestive tract: a study in human 
volunteers. J Antimicrob Chemother 1984; 13:495-504. 

86. Mavromanolakis E, Maraki S, Samonis G, Tselentis Y, Cranidis A. Effect of norfloxacin, 
trimethoprim-sulfamethoxazole and nitrofurantoin on fecal flora of women with 
recurrent urinary tract infections. J Chemother 1997; 9:203-207. 

87. Nord CE. Ecological impact of narrow spectrum antimicrobial agents compared to broad 
spectrum agents on the human intestinal microflora. In: Nord CE, Heidt PJ, Rusch VC, 



Modifying the Intestinal Microbiota with Antibiotics 367 

van der Waaij D, eds. Old Herborn University Monograph: Consequences of 
Antimicrobial Therapy for the Composition of the Microflora of the Digestive Tract. 
Herborn: Institute for Microbiology and Biochemistry, 1993:8-19. 

88. Kager L, Ljungdahl I, Malmborg AS, Nord CE. Effect of tinidazole prophylaxis on the 
normal microflora in patients undergoing colorectal surgery. Scand J Infect Dis Suppl 
1981;26:84-91. 

89. Heimdahl A, Nord CE, Okuda K. Effect of tinidazole on the oral, throat, and colon 
microflora of man. Med Microbiol Immunol (Berl) 1980; 168:1-10. 

90. Adamsson I, Nord CE, Lundquist P, Sjostedt S, Edlund C. Comparative effects of 
omeprazole, amoxycillin plus metronidazole versus omeprazole, clarithromycin 
plus metronidazole on the oral, gastric and intestinal microflora in Helicobacter 
pylori-infected patients. J Antimicrob Chemother 1999; 44:629-640. 

91. Buhling A, Radun D, Muller WA, Malfertheiner P. Influence of anti-Helicobacter triple- 
therapy with metronidazole, omeprazole and clarithromycin on intestinal microflora. 
Aliment Pharmacol Ther 2001; 15:1445-1452. 

92. Brismar B, Edlund C, Malmborg AS, Nord CE. Ciprofloxacin concentrations and impact 
of the colon microflora in patients undergoing colorectal surgery. Antimicrob Agents 
Chemother 1990; 34:481-483. 

93. Bergan T, Delin C, Johansen S, Kolstad IM, Nord CE, Thorsteinsson SB. 
Pharmacokinetics of ciprofloxacin and effect of repeated dosage on salivary and fecal 
microflora. Antimicrob Agents Chemother 1986; 29:298-302. 

94. Brumfitt W, Franklin I, Grady D, Hamilton-Miller JM, Iliffe A. Changes in the 
pharmacokinetics of ciprofloxacin and fecal flora during administration of a 7-day 
course to human volunteers. Antimicrob Agents Chemother 1984; 26:757-761. 

95. Ljungberg B, Nilsson-Ehle I, Edlund C, Nord CE. Influence of ciprofloxacin on the 
colonic microflora in young and elderly volunteers: no impact of the altered drug 
absorption. Scand J Infect Dis 1990; 22:205-208. 

96. Rozenberg-Arska M, Dekker AW, Verhoef J. Ciprofloxacin for selective decontamina- 
tion of the alimentary tract in patients with acute leukemia during remission induction 
treatment: the effect on fecal flora. J Infect Dis 1985; 152:104-107. 

97. Enzensberger R, Shah PM, Knothe H. Impact of oral ciprofloxacin on the fecal flora of 
healthy volunteers. Infection 1985; 13:273-275. 

98. Krueger WA, Ruckdeschel G, Unertl K. Influence of intravenously administered 
ciprofloxacin on aerobic intestinal microflora and fecal drug levels when administered 
simultaneously with sucralfate. Antimicrob Agents Chemother 1997; 41:1725-1730. 

99. Pecquet S, Ravoire S, Andremont A. Fecal excretion of ciprofloxacin after a single oral 
dose and its effect on fecal bacteria in healthy volunteers. J Antimicrob Chemother 1990; 
26:125-129. 

100. Holt HA, Lewis DA, White LO, Bastable SY, Reeves DS. Effect of oral ciprofloxacin on 
the fecal flora of healthy volunteers. Eur J Clin Microbiol 1986; 5:201-205. 

101. Borzio M, Salerno F, Saudelli M, Galvagno D, Piantoni L, Fragiacomo L. Efficacy of 
oral ciprofloxacin as selective intestinal decontaminant in cirrhosis. Ital J Gastroenterol 
Hepatol 1997; 29:262-266. 

102. Esposito S, Barba D, Galante D, Gaeta GB, Laghezza O. Intestinal microflora changes 
induced by ciprofloxacin and treatment of portal-systemic encephalopathy (PSE). Drugs 
Exp Clin Res 1987; 13:641-646. 

103. Wistrom J, Gentry LO, Palmgren AC, et al. Ecological effects of short-term 
ciprofloxacin treatment of travellers' diarrhoea. J Antimicrob Chemother 1992; 
30:693-706. 

104. Van Saene JJ, Van Saene HK, Geitz JN, Tarko-Smit NJ, Lerk CF. Quinolones and 
colonization resistance in human volunteers. Pharm Weekbl Sci 1986; 8:67-71. 



368 Sullivan and Nord 

105. van de Leur JJ, Vollaard EJ, Janssen AJ, Dofferhoff AS. Influence of low dose 
ciprofloxacin on microbial colonization of the digestive tract in healthy volunteers 
during normal and during impaired colonization resistance. Scand J Infect Dis 1997; 
29:297-300. 

106. Edlund C, Lidbeck A, Kager L, Nord CE. Effect of enoxacin on colonic microflora of 
healthy volunteers. Eur J Clin Microbiol 1987; 6:298-300. 

107. Nord CE, Gajjar DA, Grasela DM. Ecological impact of the des-F(6)-quinolone, BMS- 
284756, on the normal intestinal microflora. Clin Microbiol Infect 2002; 8:229-239. 

108. Edlund C, Nord CE. Ecological effect of gatifloxacin on the normal human intestinal 
microflora. J Chemother 1999; 11:50-53. 

109. Barker PJ, Sheehan R, Teillol-Foo M, Palmgren AC, Nord CE. Impact of gemifloxacin 
on the normal human intestinal microflora. J Chemother 2001; 13:47-51. 

110. Garcia-Calvo G, Molleja A, Gimenez MJ, et al. Effects of single oral doses of 
gemifloxacin (320 milligrams) versus trovafloxacin (200 milligrams) on fecal flora in 
healthy volunteers. Antimicrob Agents Chemother 2001; 45:608-611. 

111. Inagaki Y, Nakaya R, Chida T, Hashimoto S. The effect of levofloxacin, an optically- 
active isomer of ofloxacin, on fecal microflora in human volunteers. Jpn J Antibiot 1992; 
45:241-252. 

112. Edlund C, Sjostedt S, Nord CE. Comparative effects of levofloxacin and ofloxacin on 
the normal oral and intestinal microflora. Scand J Infect Dis 1997; 29:383-386. 

113. Edlund C, Brismar B, Nord CE. Effect of lomefloxacin on the normal oral and intestinal 
microflora. Eur J Clin Microbiol Infect Dis 1990; 9:35-39. 

1 14. Leigh DA, Emmanuel FXS, Tighe C, Hancock P, Boddy S, Pharmacokinetic studies on 
norfloxacin in healthy volunteers and effect on the fecal flora. 14th International 
Congress of Chemotherapy, Kyoto, Japan, 1985. 

115. Pecquet S, Andremont A, Tancrede C. Selective antimicrobial modulation of the 
intestinal tract by norfloxacin in human volunteers and in gnotobiotic mice associated 
with a human fecal flora. Antimicrob Agents Chemother 1986; 29:1047-1052. 

116. De Vries-Hospers HG, Welling GW, Van der Waaij D. Norfloxacin for selective 
decontamination: a study in human volunteers. Prog Clin Biol Res 1985; 181:259-262. 

117. Meckenstock R, Haralambie E, Linzenmeier G, Wendt F. Die Beeinflussung der Darmflora 
diirch norfloxacin bei gesunden Menschen. Z Antimikr Antineoplast Chemother 1985; 
1:27-34. 

118. Edlund C, Bergan T, Josefsson K, Solberg R, Nord CE. Effect of norfloxacin on human 
oropharyngeal and colonic microflora and multiple-dose pharmacokinetics. Scand 
J Infect Dis 1987; 19:113-121. 

119. Marco F, Gimenez MJ, Jimenez de Anta MT, Marcos MA, Salva P, Aguilar L. 
Comparison of rufloxacin and norfloxacin effects on fecal flora. J Antimicrob Chemother 
1995; 35:895-901. 

120. Pecquet S, Andremont A, Tancrede C. Effect of oral ofloxacin on fecal bacteria in human 
volunteers. Antimicrob Agents Chemother 1987; 31:124-125. 

121. Edlund C, Kager L, Malmborg AS, Sjostedt S, Nord CE. Effect of ofloxacin on oral and 
gastrointestinal microflora in patients undergoing gastric surgery. Eur J Clin Microbiol 
Infect Dis 1988;7:135-143. 

122. Vollaard EJ, Clasener HA, Janssen AJ. Influence of pefloxacin on microbial colonization 
resistance in healthy volunteers. Eur J Clin Microbiol Infect Dis 1992; 11:257-260. 

123. D' Antonio D, Pizzigallo E, Lacone A, et al. The impact of rufloxacin given as 
prophylaxis to patients with cancer on their oral and fecal microflora. J Antimicrob 
Chemother 1996; 38:839-847. 

124. Inagaki Y, Yamamoto N, Chida T, Okamura N, Tanaka M. The effect of DU-6859a, a 
new potent fluoroquinolone, on fecal microflora in human volunteers. Jpn J Antibiot 
1995; 48:368-379. 



Modifying the Intestinal Microbiota with Antibiotics 369 

125. Ritz M, Lode H, Fassbender M, Borner K, Koeppe P, Nord CE. Multiple-dose 
pharmacokinetics of sparfloxacin and its influence on fecal flora. Antimicrob Agents 
Chemother 1994; 38:455-459. 

126. van Nispen CH, Hoepelman AI, Rozenberg-Arska M, Verhoef J, Purkins L, 
Willavize SA. A double-blind, placebo-controlled, parallel group study of oral 
trovafloxacin on bowel microflora in healthy male volunteers. Am J Surg 1998; 
176:27S-31S. 



19 



The Intestinal Microbiota of Pets 
Dogs and Cats 



Minna Rinkinen 

Department of Clinical Veterinary Sciences, Faculty of Veterinary Medicine, 
University of Helsinki, Helsinki, Finland 



INTRODUCTION 

The knowledge of canine and feline intestinal microbiota is relatively scarce and based 
mainly on data from laboratory animals, on responses to dietary interventions, or on 
animals suffering from chronic intestinal disorders believed to be of bacterial nature. Most 
of the studies are performed on quite low numbers of animals that were often sacrificed 
and samples of intestinal material collected post-mortem (1,2). 

As obtaining fecal samples is much more feasible than sampling the contents of upper 
intestinal tract, most of the papers have focused on fecal microbiota, which may not be 
considered to represent the whole intestinal microecology. In addition, observations based 
on the cultivation of luminal contents may not reflect the microbiota adhered to mucosa. 

Most of the bacterial studies have been performed with traditional cultivation and 
characterization methods, which may have biased the identification and taxonomy of 
microbiota. In humans, it is estimated that only 40% of intestinal bacteria are culturable (3); 
a similar outcome can be expected also in dogs and cats. In addition, the bacterial 
taxonomy and nomenclature have changed during time, so bacteria identified in earlier 
studies may currently be re-classified under a different name. For a more in-depth 
description on the analysis of the intestinal microbiota, see the chapter by Ben- Amor and 
Vaughan in this book. 

Proximal small intestine harbors total bacteria of 10 ~ CFU/ml of luminal 
content. The number of intestinal bacteria increases distally, reaching up to 10 14 CFU/g 
in feces. In the small intestine aerobic and facultative aerobic bacteria outnumber anaerobic 
bacteria (4). When moving aborally in the gut, anaerobic bacteria start to dominate and 
finally gain numbers as high as 10 10 of CFU anaerobic bacteria/g fecal material (5). 



DEVELOPMENT OF INTESTINAL MICROBIOTA IN DOGS AND CATS 

Although there is paucity of research data concerning the development of intestinal 
microbiota of dogs and cats, it can be considered to follow a similar pattern as known for 

371 



372 Rinkinen 

other mammals. Intestinal colonization is a gradual process starting immediately after 
birth. In newborn puppies and kittens the alimentary canal is sterile but is quickly 
inhabited by bacteria from birth canal and environment. The dam usually licks the 
newborn thoroughly thus transferring its own indigenous bacteria to her offspring. Within 
24 hours the numbers of bacteria in various parts of the gastrointestinal tract of a newborn 
puppy are similar to those of an adult dog (2). 

The indigenous intestinal microbiota is considered an integral part of the host 
defense mechanisms. It forms a barrier against pathogen colonization and also influences 
the host's immunological, biochemical, and physiological features (6). 

Once the microbiota has become established, it is relatively stable. Oral antibiotics 
may have a marked effect on the homeostasis of intestinal microbiota. However, these 
changes will be re-established relatively soon (7-9). Disturbances in the gut microbiota 
may result in diarrhea, malabsorption, and chronic intestinal inflammation (10). Acute 
diarrhea may be fatal as pathogens may invade the host's tissues resulting in bacteremia 
and sepsis. 

Ageing has documented effects on the constitution of intestinal microbiota in dogs. 
Numbers of bifidobacteria and peptostreptococci diminish with ageing whereas Clostri- 
dium perfringens and streptococci are more prevalent in the large bowel of elderly dogs (1). 



CANINE AND FELINE GASTROINTESTINAL MICROBIOTA 

Gram-Positive Intestinal Bacteria 

Amongst Gram-positive bacteria residing in the gut, lactic acid bacteria (LAB) make up 
the largest and most important part of the intestinal microbiota. Although they have a 
significant protective function in the gut, the present knowledge of canine and feline 
Gram-positive intestinal microbiota is scant. 

Most of the canine LAB belong to the genera Streptococcus and Lactobacillus. In a 
recent study, Streptococcus alactolyticus was found to be a predominant culturable LAB 
in jejunal and fecal samples of four beagle dogs. In addition, Lactobacillus animalis, 
L. reuteri, L. murinus, L. ruminus and S. bovis are reported to harbor in the gut (11,12). 

The presence of bifidobacteria in canine GI tract is controversial. Many papers 
report absence of bifidobacteria in the canine fecal samples (11,13), whereas others 
described bifidobacteria as a substantial part of canine fecal microbiota (14-17). Willard 
and co-workers isolated fecal bifidobacteria from dogs inconstantly and independent on 
the diet. It was concluded that bifidobacteria may be only sporadically present in the feces 
of healthy dogs (18). 

In healthy cats, the total number of duodenal microbiota is reported to range from 
10 5 to 10 9 cfu/ml, most of the bacteria being anaerobic (10,19). The most common 
anaerobic isolates belonged to groups Bacteroides, Clostridium, Eubacteria and 
Fusobacteria, whereas Pasteurella spp were the most prevailing aerobic bacteria in 
feline proximal small intestine. In addition, Acinetobacter spp, Pseudomonas spp and 
Lactobacillus spp were detected in the duodenal samples of healthy cats (10,19). 
Lactobacilli were also isolated from feline fecal samples (20). 



Intestinal Pathogenic Bacteria 

Bacteria are seldom the sole pathogenic factor in canine and feline gastrointestinal 
disturbances. Some of the pathogens have been linked to clinical disease, but these 
pathogenic organisms are frequently isolated also in healthy individuals (21-26). 



The Intestinal Microbiota of Pets 373 

Escherichia Coli 

Escherichia coli is a normal intestinal inhabitant in warm-blooded animals, including cats 
and dogs, although its clinical significance as canine and feline enteropathogen is not very 
well documented. Colonization is believed to take place within the first days of a newborn 
animal. Certain strains of E. coli may act as intestinal pathogens causing gastrointestinal 
infections. Enteropathogenic E. coli and enterotoxigenic E. coli are known to associate 
with canine diarrhea, especially in young dogs (27-30). However, these strains have been 
isolated from non-diarrheic animals, too (28,30,31). 

Enterohemorrhagic E. coli (EHEC) has been isolated occasionally from dogs. Most 
of these reports are from dogs living in contact with cattle. EHEC has never been 
documented in cats (24). 



Clostridia 

Clostridium perfringens 

Clostridium perfringens is an anaerobic, spore-forming bacillus associated with acute and 
chronic diarrhea in dogs and cats. However, the role of C perfringens as an intestinal 
pathogen is questionable, as it commonly harbors in the intestinal tract of healthy dogs, 
too (23,32). C. perfringens produces toxins, which are classified in five toxigenic types 
(A-E). C. perfringens enterotoxin (CPE) is the best characterized virulence factor and 
coregulated with sporulation. All C. perfringens types can produce CPE, but type A strains 
are most frequently involved. CPE has been reported to cause nosocomial diarrhea, severe 
hemorrhagic enteritis, and acute and chronic large bowel diarrhea in dogs (33). On the 
other hand, CPE is also found in feces of non-diarrheic animals (23,32), although a 
significant association was present with diarrhea and detection of CPE (23). 

One study reports C perfringens carrying 62 toxin gene (cpb2) isolated from 
diarrheic dogs, suggesting 62 toxin alone or together with CPE may play a role in canine 
clostridial diarrhea (34). 



Clostridium difficile 

C difficile is associated with diarrhea in dogs, although it has been frequently isolated from 
dogs with no signs of diarrhea (23,35). C difficile-related diarrhea in humans is principally 
associated with hospitalization and use of antimicrobials. In dogs, no significant 
association was found in the prevalence of C. difficile along with hospitalization and 
antibiotic administration, but increased carriage rate was observed in non-hospitalized 
dogs receiving antibiotics (23). 

Salmonella 

Both healthy and diarrheic dogs and cats may carry Salmonella. Prevalence in healthy 
dogs is reported to be between 1% and 38% (24,36). Furthermore, Salmonella isolation 
rates in dogs with clinical enteritis is reported low (21,25,37). 

The prevalence of Salmonella in canine fecal isolates examined has reduced during 
the past decades. This most likely reflects the change in feeding of dogs, as commercial pet 
foods have replaced raw meat and offal (36). Feeding bones and raw food diet yielded a 
30% Salmonella isolation rate in stool samples of dogs consuming this type of diet. 
Feeding raw chicken and meat to dogs may therefore be a risk for potential transfer of 
Salmonella to humans, too (38,39). 



374 Rinkinen 

Salmonella is regarded relatively rare in cats, isolation prevalence varying between 
0.8% and 18%; in most reports it is approximately 1%. Also cats may be asymptomatic 
carriers (22,24,40). An outbreak of Salmonella enterica serovar Typhimurium in cats was 
reported in Sweden, where salmonellosis was probably transmitted from wild infected 
birds hunted by the cats (41). 



Campylobacters 

Campylobacters are regarded as important zoonotic pathogens. Most of the human 
infections are food- or water-borne, but infections from pets may also be of concern, 
especially with immunocompromised people (42-44). Campylobacters have been 
associated with acute and chronic diarrhea in dogs and cats (43). However, as they are 
frequently isolated from both healthy and diarrheic animals, it is suggested they are not 
primary pathogens but more likely opportunistic microbes producing clinical signs in 
predisposing conditions, such as poor nutrition or housing, or high animal density (45,46). 
Young dogs seem to be more prone to carry Campylobacters, carriage rate being up to 75% 
of dogs less than 12 months old, whereas the isolation rate in adult dogs was only 32.7% 
(47,48). 

Campylobacter shedding correlates clearly with diarrhea in young dogs, but for dogs 
older than 12 months there was no evident correlation with shedding and clinical disease. 
In cats, no significant association was found between campylobacteriosis and diarrhea in 
any age group (49,50). 

In cats and dogs, C. helveticus, C jejuni, and C. upsaliensis are most prevalent 
Campylobacter strains. C helveticus has been isolated in healthy cats and dogs (47,51,52). 
One study reported C helveticus to inhabit 21.7% of the cats examined, being the most 
prevalent Campylobacter species isolated (47). In addition, C coli, and C lari have been 
isolated to lesser extent (43,45,48,50,53-55). However, the traditional phenotypic 
identification methods have been criticized for being unreliable when identifying thermo- 
philic Campylobacters (56). The clinical relevance of these Campylobacters is unclear. 

Campylobacter upsaliensis 

C upsaliensis is a catalase-negative thermotolerant Campylobacter recognized as an 
emerging human pathogen. In humans it is associated with gastroenteritis and bactere- 
mia (57). It was first isolated from canine feces (54) and some years later also from feline 
feces (58). It has been reported to be the most prevalent Campylobacter in dogs (47,50,56) 
and cats (50,56). Thus, it is of interest whether household pets may comprise a reservoir 
for this zoonotic pathogen although human and canine strains are reported to be 
genotypically distinct (51). 

C. upsaliensis has been isolated from feces of both diarrheic and healthy dogs and 
cats. It is documented to infect puppies at approximately six weeks of age without causing 
a clinical disease when puppies were raised separately in a breeding kennel, presumably in 
acceptable conditions. Poor sanitation and high animal density are marked risk factors, 
increasing the carriage rate of C. upsaliensis up to 2.6-fold. These findings support the 
opportunistic nature of this organism as a canine and feline pathogen (51,59). 

Helicobacters 

Helicobacter spp. are Gram-negative, microaerophilic curved or spiral-shaped motile 
bacteria. Many gastric Helicobacter-like organisms (GHLO) are frequently found in cats 



The Intestinal Microbiota of Pets 375 

and dogs. Virtually all dogs can be expected to harbor gastric GHLO (60,61), although 
most of the dogs are asymptomatic. Additionally, the clinical signs in dogs suffering from 
gastritis may persist despite the eradication of helicobacters. Therefore the role of GHLO 
as an etiological factor in canine gastritis is currently unclear (62,63). 

In dogs, H. felis, H. bizzozeronii, H. salomonis, "Flexispira rappini, " H. bilis, and 
"H. heilmannii" have been reported to inhabit the gastric mucosa. The human pathogen 
H. pylori has not yet been isolated in canine gastric biopsies. However, a recent paper 
reports presumably non-cultivable H. pylori, or a closely related Helicobacter in two 
dogs, results based on its 16S rRNA sequence (64). Unlike dogs, cats have been 
documented to acquire H. pylori, although very infrequently. Feline H. pylori infection has 
been suggested to be an anthroponosis, i.e., cats are infected by humans carrying H. pylori 
(63,65-67). 

In addition to GHLOs, dogs and cats are reported to have also enteric helicobacters. 
H. canis has been isolated from diarrheic cats and dogs (68,69), and H. marmotae from cat 
feces (70). 



MODIFYING THE INTESTINAL MICROBIOTA: PRE- AND PROBIOTICS 

First documented studies of dietary manipulation of canine and feline intestinal microbiota 
date back to the beginning of the twentieth century (71). 

Today, there is growing interest in modifying their gut microbiota towards what is 
considered a healthy composition, i.e., increase in LAB and bifidobacteria, and decrease in 
potential pathogenic bacteria (72). Many commercial pet foods now contain prebiotics 
(e.g., fructo-oligosaccharides, FOS). In addition, probiotics are also marketed for dogs 
and cats. 

Prebiotics 

Prebiotics are reported to have a variable impact on canine fecal and intestinal microbiota. 
Supplementing dogs' food with FOS and mannanoligosaccharides increased ileal 
lactobacilli and fecal lactobacilli and bifidobacteria concentrations (73). Feeding short 
chain FOS to dogs increased the total number of fecal anaerobes and lowered the number 
of Clostridium perfringens (17,74). Similar outcome was achieved with arabinogalactan 
supplementation (15). On the other hand, no significant differences were noticed in the 
denaturing gradient gel electrophoresis analysis of fecal bacterial profiles when dogs were 
fed a diet containing 10% fiber (16), and another study revealed no significant effect of 
FOS supplementation on canine fecal Clostridium spp (18). 

FOS supplementation increased fecal lactobacilli and decreased numbers of E. coli 
in healthy cats, but did not alter the duodenal microbiota (75,76). This supports the notion 
that, as FOS are nondigestible fibers fermented in the proximal gut in humans (mainly in 
the large intestine) (77), also in cats FOS have only a minimal effect on the microbes 
residing in the proximal part of GI tract. In a study of eight cats, feeding lactosucrose 
increased fecal lactobacilli and bifidobacteria counts significantly, while numbers of 
Clostridia and Enterobacteriaceace decreased significantly (78). 

Probiotics 

Currently, there are no commerically available probiotics fulfilling the species specificity 
criterion applied to probiotics as stated by Saarela and co-workers (79). Despite that, 



376 Rinkinen 

probiotics are utilized in pet animals in the hope to create beneficial alterations in the 
intestinal microbiota. 

Enterococcus faecium SF68 has been documented to enhance specific immuno- 
logical responses in young dogs (80) and E. faecalis FK-23 stimulated non-specific 
immune functions in healthy adult dogs (81). E. faecium is also reported to have an effect 
on canine enteropathogens. It significantly decreased the canine in vitro mucus adhesion 
of C. perfringens (82). This finding was supported also in vivo (83). On the other hand, 
E. faecium increased both the in vitro adhesion and fecal shedding of Campylobacters 
(82,83). Pasupathy and co-workers (84) evaluated the effect of Lactobacillus acidophilus 
on the digestibility of food and growth of puppies. They concluded that Lactobacillus 
supplementation has a favorable effect during the active growth period, although 
differences between the study group and control group were not significant. 



CONCLUSION 

In the recent years the interest in canine and feline gastrointestinal microbiota has 
increased, resulting in a fair amount of documented information. However, the current 
knowledge of canine and feline gastrointestinal microbiota is still rather scarce. The 
growing interest in pre- and probiotics together with the novel microbiological methods 
has already made a scientific contribution to the field of small animal intestinal 
microbiology. With this trend likely to continue in the future, our knowledge of the canine 
and feline gastrointestinal microbiota and the factors related to its regulation will expand. 



REFERENCES 

1. Benno Y, Nakao H, Uchida K, Mitsuoka T. Impact of the advances in age on the 
gastrointestinal microflora of beagle dogs. J Vet Med Sci 1992; 54:703-706. 

2. Buddington RK. Postnatal changes in bacterial populations in the gastrointestinal tract of dogs. 
Am J Vet Res 2003; 64:646-651. 

3. Tannock GW, Munro K, Harmsen HJM, Welling GW, Smart J, Gopal PK. Analysis of the fecal 
microflora of human subjects consuming a probiotic product containing Lactobacillus 
rhamnosus DR20. Appl Environ Microbiol 2000; 66:2578-2588. 

4. Delles EK, Willard MD, Simpson RB, et al. Comparison of species and numbers of bacteria in 
concurrently cultured samples of proximal small intestinal fluid and endoscopically obtained 
duodenal mucosa in dogs with intestinal bacterial overgrowth. Am J Vet Res 1994; 
55:957-964. 

5. Davis CP, Cleven D, Balish E, Yale CE. Bacterial association in the gastrointestinal tract of 
beagle dogs. Appl Environ Microbiol 1977; 34:194-206. 

6. Tannock GW. The normal microflora: an introduction. In: Tannock GW, ed. Medical 
Importance of Normal Microflora. London: Kluwer Academic Publishers, 1999:1-23. 

7. Lode H, Von der Hoh N, Ziege S, Borner K, Nord CE. Ecological effects of linezolid versus 
amoxicillin/clavulanic acid on the normal intestinal microflora. Scand J Infect Dis 2001; 
33:899-903. 

o 

8. Sullivan A, Edlund C, Nord CE. Effect of antimicrobial agents on the ecological balance of 
human microflora. Lancet Infect Dis 2001; 1:101-114. 

9. Nord CE, Gajjar DA, Grasela DM. Ecological impact of the des-F(6)-quinolone, BMS-284756, 
on the normal intestinal microflora. Clin Microbiol Infect 2002; 8:229-239. 

10. Johnston KL. Small intestinal bacterial overgrowth. In: Simpson KW, ed. The Veterinary 
Clinics of North America, Small Animal Practice. Saunders, Philadelphia: Progress in 
Gastroenterology, 1999:523-550. 



The Intestinal Microbiota of Pets 377 

11. Greetham HL, Giffard C, Hutson RA, Collins MD, Gibson GR. Bacteriology of the Labrador 
dog gut: a cultural and genotypic approach. J Appl Microbiol 2002; 93:640-646. 

12. Rinkinen ML, Koort JMK, Ouwehand AC, Westermarck E, Bjorkroth JK. Streptococcus 
alactolyticus is the dominating culturable lactic acid bacterium species in canine jejunum and 
feces of four fistulated dogs. FEMS Microbiol Lett 2004; 230:35-39. 

13. Martineau B. Comparison of four media for the selection of bifidobacteria in dog fecal samples. 
Anaerobe 1999;5:123-127. 

14. Beynen AC, Baas JC, Hoekemeijer PE, et al. Faecal bacterial profile, nitrogen excretion, and 
mineral absorption in healthy dogs fed supplemental oligofructose. J Anim Physiol Anim Nutr 
2002; 86:298-305. 

15. Grieshop CM, Flickinger EA, Fahey GC, Jr. Oral administration of arabinogalactan affects 
immune status and fecal microbial populations in dogs. J Nutr 2002; 132:478-482. 

16. Simpson JM, Martineau B, Jones WE, Ballam JM, Mackie RI. Characterization of fecal 
bacterial populations in canines: effects of age, breed, and dietary fiber. Microb Ecol 2002; 
44:186-197. 

17. Swanson KS, Grieshop CM, Flickinger EA, et al. Fructooligosaccharides and Lactobacillus 
acidophilus modify gut microbial populations, total tract nutrient digestibilities, and fecal 
protein catabolite concentrations in healthy adult dogs. J Nutr 2002; 132:3721-3731. 

18. Willard MD, Simpson RB, Cohen ND, Clancy JS. Effects of dietary fructooligosaccharide on 
selected bacterial populations in feces of dogs. Am J Vet Res 2000; 61:820-825. 

19. Johnston KL, Swift NC, Forster-van Hijfte M. Comparison of the bacterial flora of the 
duodenum in healthy cats and cats with signs of gastrointestinal tract disease. J Am Vet Med 
Assoc 2001; 218:48-51. 

20. Hartemink R, Rombouts FM. Comparison of media for the detection of bifidobacteria, 
lactobacilli, and total anaerobes from faecal samples. J Microbiol Methods 1999; 36:181-192. 

21 . Buogo C, Burnens AP, Perrin J, Nicolet J. Presence of Campylobacter spp Clostridium difficile, 
C. perfringens, and salmonellae in litters of puppies and in adult dogs in a shelter. Schweiz 
Arch Tierheilkd 1995; 137:165-171. 

22. Spain CV, Scarlett JM, Wade SE, McDonough P. Prevalence of enteric zoonotic agents in cats 
less than 1 year old in central New York State. J Vet Intern Med 2001; 15:33-38. 

23. Marks SL, Kather EJ, Kass PH, Melli AC. Genotypic and phenotypic characterization of 
Clostridium perfringens and Clostridium difficile in diarrheic and healthy dogs. J Vet Intern 
Med 2002; 16:533-540. 

24. Sanchez S, Lee MD, Harmon BG, Maurer JJ, Doyle MP. Animal issues associated with 
Escherichia coli 0157:H7. J Am Vet Med Assoc 2002; 221:1122-1126. 

25. Hackett T, Lappin MR. Prevalence of enteric pathogens in dogs of north-central Colorado. 
J Am Anim Hosp Assoc 2003; 39:52-56. 

26. Staats JJ, Chengappa MM, DeBey MC, Fickbohm B, Oberst RD. Detection of Escherichia coli 
Shiga toxin (stx) and enterotoxin (estA and elt) genes in fecal samples from non-diarrheic and 
diarrheic greyhounds. Vet Microbiol 2003; 94:303-312. 

27. Drolet R, Fairbrother JM, Harel J, Helie P. Attaching and effacing and enterotoxigenic 
Escherichia coli associated with enteric colibacillosis in the dog. Can J Vet Res 1994; 
58:87-92. 

28. Hammermueller J, Kruth S, Prescott J, Gyles C. Detection of toxin genes in Escherichia coli 
isolated from normal dogs and dogs with diarrhea. Can J Vet Res 1995; 59:265-270. 

29. Beutin L. Escherichia coli as a pathogen in dogs and cats. Vet Res 1999; 30:285-298. 

30. Starcic M, Johnson JR, Stell AL, et al. Haemolytic Escherichia coli isolated from dogs with 
diarrhea have characteristics of both uropathogenic and necrotoxigenic strains. Vet Microbiol 
2002; 85:361-377. 

31. Holland RE, Walker RD, Sriranganathan N, Wilson RA, Ruhl DC. Characterization of 
Escherichia coli isolated from healthy dogs. Vet Microbiol 1999; 70:261-268. 

32. Weese JS, Staempfli HR, Prescott JF, Kruth SA, Greenwood SJ, Weese HE. The roles of 
Clostridium difficile and enterotoxigenic Clostridium perfringens in diarrhea in dogs. J Vet 
Intern Med 2001; 15:374-378. 



378 Rinkinen 

33. Sasaki J, Goryo M, Asahina M, Makara M, Shishido S, Okada K. Hemorrhagic enteritis 
associated with Clostridium perfringens type A in a dog. J Vet Med Sci 1999; 61:175-177. 

34. Thiede S, Goethe R, Amtsberg G. Prevalence of (2 toxin gene of Clostridium perfringens type 
A from diarrhoiec dogs. Vet Rec 2001; 149:273-274. 

35. Struble AL, Tang YJ, Kass PH, Gumerlock PH, Madewell BR, Silva J, Jr. Fecal shedding of 
Clostridium difficile in dogs: a period prevalence survey in a veterinary medical teaching 
hospital. J Vet Diagn Invest 1994; 6:342-347. 

36. Cave NJ, Marks SL, Kass PH, Melli AC, Brophy MA. Evaluation of a routine diagnostic fecal 
panel for dogs with diarrhea. J Am Vet Med Assoc 2002; 221:52-59. 

37. van Duijkeren E, Houwers D. Salmonella enteritis in dogs, not relevant? Tijdschr Diergeneeskd 
2002; 127:716-717. 

38. LeJeune JT, Hancock DD. Public health concerns associated with feeding raw meat diets to 
dogs. J Am Vet Med Assoc 2001; 219:1222-1225. 

39. Joffe DJ, Schlesinger DP. Preliminary assessment of the risk of Salmonella infection in dogs 
fed raw chicken diets. Can Vet J 2002; 43:441-442. 

40. Hill SL, Cheney JM, Taton-Allen GF, Reif JS, Bruns C, Lappin MR. Prevalence of enteric 
zoonotic organisms in cats. J Am Vet Med Assoc 2000; 216:687-692. 

41. Tauni MA, Osterlund A. Outbreak of Salmonella typhimurium in cats and humans associated 
with infection in wild birds. J Small Anim Pract 2000; 41:339-341. 

42. Ketley JM. Pathogenesis of enteric infection by Campylobacter. Microbiology 1997; 143:5-21. 

43. Steinhauserova I, Fojtikova K, Klimes J. The incidence and PCR detection of Campylobacter 
upsaliensis in dogs and cats. Lett Appl Microbiol 2000; 31:209-212. 

44. Alios BM. Campylobacter jejuni infections: update on emerging issues and trends. Clin Infect 
Dis2001; 32:1201-1206. 

45. Torre E, Tello M. Factors influencing fecal shedding of Campylobacter jejuni in dogs without 
diarrhea. Am J Vet Res 1993; 54:260-262. 

46. Fernandez H, Martin R. Campylobacter intestinal carriage among stray and pet dogs. Rev 
Saude Publica 1991; 25:473-475. 

47. Moser I, Rieksneuwohner B, Lentzsch P, Schwerk P, Wieler LH. Genomic heterogeneity and 
O-antigenic diversity of Campylobacter upsaliensis and Campylobacter helveticus strains 
isolated from dogs and cats in Germany. J Clin Microbiol 2001; 39:2548-2557. 

48. Engvall EO, Brandstrom B, Andersson L, Baverud V, Trowald-Wigh G, Englund L. Isolation 
and identification of thermophilic Campylobacter species in faecal samples from Swedish 
dogs. Scand J Infect Dis 2003; 35:713-718. 

49. Burnens AP, Angeloz-Wick B, Nicolet J. Comparison of Campylobacter carriage rates in 
diarrheic and healthy pet animals. J Vet Med 1992; 39:175-780. 

50. Sandberg M, Bergsjo B, Hofshagen M, Skjerve E, Kruse H. Risk factors for Campylobacter 
infection in Norwegian cats and dogs. Prev Vet Med 2002; 55:241-253. 

51. Stanley J, Jones C, Burnens A, Owen RJ. Distinct genotypes of human and canine isolates of 
Campylobacter upsaliensis determined by 16S rRNA gene typing and plasmid profiling. J Clin 
Microbiol 1994; 32:1788-1794. 

52. Shen Z, Feng Y, Dewhirst FE, Fox JG. Coinfection of enteric Helicobacter spp. and 
Campylobacter spp. in cats. J Clin Microbiol 2001; 39:2166-2172. 

53. Prescott JF, Munroe DL. Campylobacter jejuni enteritis in man and domestic animals. J Am 
Vet Med Assoc 1982; 81:1524-1530. 

54. Sandstedt K, Ursing J, Walder M. Thermotolerant Campylobacter with no or weak catalase 
activity isolated from dogs. Curr Microbiol 1983; 8:209-213. 

55. Duim B, Vandamme PA, Rigter A, Laevens S, Dijkstra JR, Wagenaar JA. Differentiation of 
Campylobacter species by AFLP fingerprinting. Microbiology 2001; 147:2729-2737. 

56. Engvall EO, Brandstrom B, Gunnarsson A, Morner T, Wahlstrom H, Fermer C. Validation of a 
polymerase chain reaction/restriction enzyme analysis method for species identification of 
thermophilic Campylobacters isolated from domestic and wild animals. J Appl Microbiol 2002; 
92:47-54. 



The Intestinal Microbiota of Pets 379 

57. Patton CM, Shaffer N, Edmonds P, et al. Human disease associated with "Campylobacter 
upsaliensis" (catalase -negative or weakly positive Campylobacter species) in the United 
States. J Clin Microbiol 1989; 27:66-73. 

58. Fox JG, Maxwell KO, Taylor NS, Runsick CD, Edmonds P, Brenner DJ. "Campylobacter 
upsaliensis" isolated from cats as identified by DNA relatedness and biochemical features. 
J Clin Microbiol 1989; 27:2376-2378. 

59. Baker J, Barton MD, Lanser J. Campylobacter species in cats and dogs in South Australia. Aust 
Vet J 1999; 77:662-666. 

60. Happonen I, Linden J, Saari S, et al. Detection and effects of helicobacters in healthy dogs and 
dogs with signs of gastritis. J Am Vet Med Assoc 1998; 213:1767-1774. 

61. Strauss-Ayali D, Simpson KW. Gastric Helicobacter infection in dogs. Vet Clin North Am 
Small Anim Pract 1999; 29:397-414. 

62. Happonen I, Linden J, Westermarck E. Effect of triple therapy on eradication of canine gastric 
helicobacters and gastric disease. J Small Anim Pract 2000; 41:1-6. 

63. Neiger R, Simpson KW. Helicobacter infection in dogs and cats: facts and fiction. J Vet Intern 
Med 2000; 14:125-133. 

64. Buczolits S, Hirt R, Rosengarten R, Busse HJ. PCR-based genetic evidence for occurrence of 
Helicobacter pylori and novel Helicobacter species in the canine gastric mucosa. Vet 
Microbiol 2003; 24:259-270. 

65. Handt LK, Fox JG, Stalis IH, et al. Characterization of feline Helicobacter pylori strains and 
associated gastritis in a colony of domestic cats. J Clin Microbiol 1995; 33:2280-2289. 

66. El-Zaatari FA, Woo JS, Badr A, et al. Failure to isolate Helicobacter pylori from stray cats 
indicates that H. pylori in cats may be an anthroponosis — an animal infection with a human 
pathogen. J Med Microbiol 1997; 46:372-376. 

67. Simpson KW, Strauss-Ayali D, Straubinger RK, et al. Helicobacter pylori infection in the cat: 
evaluation of gastric colonization, inflammation and function. Helicobacter 2001; 6:1-14. 

68. Stanley J, Linton D, Burens AP, et al. Helicobacter canis sp. nov. a new species from dogs: an 
integrated study of phenotype and genotype. J Gen Microbiol 1993; 139:2495-2504. 

69. Foley JE, Marks S, Munson L, et al. Isolation of Helicobacter canis from a colony of Bengal 
cats with endemic diarrhea. J Clin Microbiol 1999; 37:3271-3275. 

70. Fox JG, Shen Z, Xu S, et al. Helicobacter marmotae sp. nov. isolated from livers of 
woodchucks and intestines of cats. J Clin Microbiol 2002; 40:2513-2519. 

71. Torrey JC. The regulation of the intestinal flora of dogs through diet. J Med Res 1918; 
39:415-477. 

72. Hussein HS, Flickinger EA, Fahey GC, Jr. Petfood applications of inulin and oligofructose. 
J Nutr 1999; 129:1454S-1456S. 

73. Swanson KS, Grieshop CM, Flickinger EA, et al. Effects of supplemental fructooligosacchar- 
ides plus mannanoligosaccharides on immune function and ileal and fecal microbial 
populations in adult dogs. Arch Anim Nutr 2002; 56:309-318. 

74. Flickinger EA, Schreijen EM, Patil AR, et al. Nutrient digestibilities, microbial populations, 
and protein catabolites as affected by fructan supplementation of dog diets. J Anim Sci 2003; 
81:2008-2018. 

75. Sparkes AH, Papasouliotis K, Sunvold G, et al. Bacterial flora in the duodenum of healthy cats, 
and effect of dietary supplementation with fructo-oligosaccharides. Am J Vet Res 1998; 
59:431-435. 

76. Sparkes AH, Papasouliotis K, Sunvold G, et al. Effect of dietary supplementation with fructo- 
oligosaccharides on fecal flora of healthy cats. Am J Vet Res 1998; 59:436-440. 

77. Molis C, Flourie B, Ouarne F, et al. Digestion, excretion, and energy value of 
fructooligosaccharides in healthy humans. Am J Clin Nutr 1996; 64:324-328. 

78. Terada A, Hara H, Kato S, et al. Effect of lactosucrose (4G-beta-D-galactosylsucrose) on fecal 
flora and fecal putrefactive products of cats. J Vet Med Sci 1993; 55:291-295. 

79. Saarela M, Mogensen G, Fonden R, Matto J, Mattila-Sandholm T. Probiotic bacteria: safety, 
functional and technological properties. J Biotechnol 2000; 84:197-215. 



380 Rinkinen 

80. Benyacoub J, Czarnecki-Maulden GL, Cavadini C, et al. Supplementation of food with 
Enterococcus faecium (SF68) stimulates immune functions in young dogs. J Nutr 2003; 
133:1158-1162. 

81. Kanasugi H, Hasegawa T, Goto Y, Ohtsuka H, Makimura S, Yamamoto T. Single 
administration of enterococcal preparation (FK-23) augments non-specific immune responses 
in healthy dogs. Int J Immunopharmacol 1997; 19:655-659. 

82. Rinkinen M, Jalava K, Westermarck E, Salminen S, Ouwehand AC. Interaction between 
probiotic lactic acid bacteria and canine enteric pathogens: a risk factor for intestinal 
Enterococcus faecium colonization? Vet Microbiol 2003; 92: 111—119. 

83. Vahjen W, Manner K. The effect of a probiotic Enterococcus faecium product in diets of 
healthy dogs on bacteriological counts of Salmonella spp. Campylobacter spp. and Clostridium 
spp. in faeces. Arch Anim Nutr 2003; 57:229-233. 

84. Pasupathy K, Sahoo A, Pathak NN. Effect of Lactobacillus supplementation on growth and 
nutrient utilization in mongrel pups. Arch Tierernahr 2001; 55:243-253. 



20 



The Gastrointestinal Microbiota 
of Farm Animals 



Alojz Bomba, Zuzana Jonecova, Sofia Gancarcikova, and Radomfra Nemcova 

Institute of Gnotobiology and Prevention of Diseases in Young, 
University of Veterinary Medicine, Kosice, Slovak Republic 



INTRODUCTION 

The colonization of the digestive tract in animals begins soon after birth or hatching and 
the normal microbiota changes dramatically during the life of the host. The composition 
of gastrointestinal microbiota differs between animal species, between individuals within 
the same species and between the body sites of the host. The gut microbiota is a complex 
interactive community of organisms and its functions are the result of activities of all 
microbial components. Together with the host, the microorganisms constitute an 
ecological system, beneficial for the host, as well as for the microbial species. In 
principle, the role of gut microbiota in animals is the same as in humans — salvaging 
energy from the undigested feed components through fermentation, providing the basis 
for a barrier that prevents pathogenic bacteria from invading the gastrointestinal tract, 
protective functions together with the gut immune system, a role in metabolism 
of xenobiotics and contribution to the vitamin and amino acids requirements of the 
animals (1). Some of these functions are emphasized in farm animals with regard to their 
environment, character of their feed and the economy of farm animals' rearing. The 
composition and metabolism of the gastrointestinal microbiota affects the performance of 
farm animals in many ways, especially in the young, which are subjected to many 
stressful conditions. 

Farm animals can be divided into three main groups according to the degree of 
development of their gastrointestinal tract and efficacy of feed digestion: (1) omnivorous 
animals — the feed of plant origin with small content of cellulose and lignin, as well as the 
feed of animal origin is easily and quickly digested with a help of enzymes produced in 
the gastrointestinal tract of the animal (pigs), (2) carnivorous animals — under natural 
conditions they consume mostly feed of animal origin, (3) herbivorous animals — consume 
feed of plant origin with high content of cellulose and lignin, which the animal is able to 
digest exclusively through microbial fermentation by its gastrointestinal microbiota 
(ruminants, horses). Herbivorous animals have some part of their gastrointestinal tract 
adapted to microbial fermentation. The ruminants are polygastric animals with foregut 

381 



382 Bomba et al. 

capacity 150-180L in adult cows. In horses, which are monogastric, the caecum with 
capacity 100-140L is developed for microbial fermentation of lignin and cellulose. 

The greatest differences in the composition of the microbiota of the gastrointestinal 
ecosystem have been shown to occur between ruminants and monogastric animals. 
Gradual changes in the composition of the gastrointestinal microbiota that take place 
within an animal species are related to age (2). At an early age the microbiota of the 
digestive tract of young animals is very similar. With the exception of poultry, this 
similarity is related to the intake of maternal milk. During the suckling period, bacteria, 
which can utilize the components of milk, predominate in the upper tract, and the milk 
constituents evidently largely determine which microbe can be implanted in the 
intestines. The forestomachs of ruminants have not yet started functioning and 
the physiology of the digestive tract compares to that of monogastric animals. After 
the animals start to consume creep feed and they are finally weaned, an adult type of 
microbiota begins to develop in the upper and lower intestinal tract. At the same time the 
main site of bacterial fermentation changes from the stomach to the large intestine or, in 
ruminants, to the rumen. 

Due to progressing of age, changes in the composition of the ingested feed and a 
different morphological and functional development of the gastrointestinal tract, certain 
differences gradually occur in the composition of the microbiota in calves, lambs, suckling 
piglets and chicks that are typical for the given farm animal species. The gut ecosystem of 
adult animals is stable and changes only due to the effects of external factors of an 
adequate intensity (long-lasting change of feeds, stress, administration of antibiotics). 



MICROBIOTA OF THE GASTROINTESTINAL TRACT IN FARM ANIMALS 

The gastrointestinal ecosystem of animals is a complex, open, interactive system involving 
the animal's environment and diet, the animal itself, and many microbial species. This 
system regulates the course of the successional events and the population levels and 
geographic distribution of the climax communities once they are formed. In adult animals 
the microbial communities occupy many niches in habitats distributed from the center 
of the lumen to the depths of the crypts, and from the oral cavity to the anus. Depending 
upon the animal species any or all habitats may be occupied. The microbial communities 
occupying the habitats are usually composed of autochthonous (indigenous) microbes. 
A sample from any given habitat may at any given time yield allochthonous (non- 
indigenous) microbes as well as indigenous ones. The allochthonous microbes derive from 
what the animal ingested (feed, water, faeces) or from habitats above the one in question. 

The gastrointestinal microbiota interact profoundly with their animal host, 
influencing its early development, quality of life, ageing and resistance to infectious 
diseases. One of the functions of the microbiota is to degrade dietary components such as 
fiber in order to provide short-chain fatty acids and other essential nutrients that are 
absorbed by the host. Animal hosts have incubation chambers such as the rumen (cattle, 
sheep, goat) or the caecum (horse, chicken) in which bacterial fermentation proceeds 
under optimal conditions. Those animals that have only small caeca, (pigs), have a 
microbiota which has adapted to use "fast food" such as simple carbohydrates and proteins 
that are consumed with the diet and available in the host's secretions such as saliva or 
mucus (3). 

In horses and poultry, so-called hind gut fermenters, the caecum fulfill a function 
that is similar to that of the rumen in ruminants. The caecum is found in the anterior part of 



The Gastrointestinal Microbiota of Farm Animals 383 

the large intestine and its microbial activity can provide for about 30% of the nutritional 
requirements of these animals. 

In monogastric animals, the enzymes of the host ensure digestion of the feed despite 
the fact that their digestive tract is rather short. Of the farm animal species pigs are typical 
representatives of this group of animals. Humans are equipped with a similar type of 
digestive tract. The large intestinal microbiota of pigs is the most numerous and most 
varied one. Recent knowledge indicates a pronounced similarity of the ruminal, caecal and 
large intestinal microbiota in animals. 

Regulation of the composition and localization of microbial communities in the 
gastrointestinal tract is a multi-factorial process in which any or all of these numerous 
forces may come into play (4). Stability of the microecosystem of the digestive tract is 
maintained by the interrelations of the microecosystem and the macroorganism as well as 
by the interactions of the microorganisms in the ecosystem. On the part of the host, 
both endogenous (age, host immunity, digestive tract motility and length, acidity) and 
exogenous factors (diet) play an important role (5). On the other hand, the microbiota of the 
digestive tract greatly affects the development of the host animal, mainly at an early age, 
and plays a very important role in the animal's resistance to infectious diseases. The 
interactions between microorganisms are mediated by competition for gut receptors and 
nutrients as well as by the production of antimicrobial substances (6,7). The mechanisms of 
bacterial interactions also mediate the barrier effect (8) or competitive exclusion (9), which 
is the ability of the indigenous microbiota to prevent the implantation of allochthonous 
microbes in the gastrointestinal tract. Knowledge of the mechanism of bacterial 
interactions is an inevitable presupposition if optimization of the composition of the 
gastrointestinal microbiota and stimulation of the beneficial effects of the latter on the host 
animal are desired (10). 

Pigs 

The gastrointestinal tract of the piglets at parturition is sterile, but the gut microbiota 
develops very rapidly. The first bacteria, which become established in the digestive tract of 
the piglet, originate from the dam or the environment, but they are not the most abundant 
ones of the ecosystems encountered by the young (11). The newborn possesses very 
efficient selection systems enabling it to favor certain bacterial species among the bacteria 
of the different ecosystems. Many factors might be involved in this selection — diet, 
environmental conditions such as hygienic stage, temperature, the microbial interactions 
in the digestive tract and the barrier effect of the dominant microbiota against the 
environmental bacteria. 

The indigenous microbiota exerts a profound influence on both the morphological 
structure and on the digestive and absorptive capabilities of the gastrointestinal tract (12). 
From the stomach of suckling piglets significant populations of microorganisms have been 
isolated upto 10 viable counts per 1 cm of the tissue (13). The microbial population 
adhering to the pars esophagea varies little from birth until after weaning and the anaerobic 
microbiota, particularly lactobacilli, might be important in maintaining the pars esophagea 
free from colonization by other microorganisms. The stratified squamosus epithelium, of 
which the pars esophagea is composed, is continuously desquamating releasing cells with 
attached bacteria into the lumen and may serve as a continuous inoculum of specific lactic 
acid bacteria into the gastric contents (14). 

In the small intestine, a fast transit time and digestive secretions such as bile acids 
limit bacterial numbers and diversity. The gastrointestinal microbiota of the young piglets 
is composed of facultatively anaerobic microorganisms in the proximal intestine 



384 Bomba et al. 

(duodenum, jejunum) whose number ranges from 10 to 10 per g content (11). This 
number increases progressively in the ileum, and in the last parts of the digestive tract 
strictly anaerobic bacteria are found among the dominant microbiota. In very young 
piglets, Escherichia coli is the dominant microbe of all gut segments, together with species 
of the genera Lactobacillus and Streptococcus. The microbiota of the piglet progressively 
changes with age, the number of Escherichia coli decreases in all segments and the 
lactobacilli and streptococci constitute the dominant microbiota of the proximal intestine. 
The presence of lactobacilli as a constituent of the normal microbiota of the 
gastrointestinal tract is considered to be beneficial to the porcine host (15). The strictly 
anaerobic microbiota becomes more diversified in the distal segments, where Bacteroides, 
Eubacterium, Peptostreptococcus and many Clostridium species are found (11). 

The change of the gut environment occurs in connection to weaning of the piglets. 
Weaning and weaning age have significant effects on microbial population and volatile 
fatty acids concentration (16). During the first week after weaning, pH and the content of 
dry matter decrease, as well as the count of lactobacilli, while the number of coliform 
bacteria increases (17). These changes contribute to low weight gains and predisposition to 
diarrhea. Associated with weaning there are marked changes to the histology and 
biochemistry of the small intestine, such as villous atrophy and crypt hyperplasia, which 
caused decreased digestive and absorptive capacity (18) and contribute to post- weaning 
diarrhea. The major factors implicated in the etiology of these changes are: change in 
nutrition, stress due to separation from mother and littermates, new environment, the 
withdrawal of milk-borne growth promoting factors, as well as enteropathogens and their 
interactions with the gut microbiota. Enterotoxigenic Escherichia coli strains are generally 
considered to be the main cause of diarrhea at weaning and the period immediately 
thereafter. The colonizing of the small intestine by enterotoxigenic E. coli strains may be 
possible for several reasons (19): (1) the brush border of the intestinal epithelium of newly 
weaned pigs may be damaged by components in the feed or by viruses allowing E. coli to 
adhere and colonize the damaged epithelium, (2) after weaning the pigs are no longer 
protected by the milk of the sow, an important factor that prevents E. coli colonization 
during the suckling period, (3) newly weaned pigs have a shortage of digestive enzymes 
and feed is poorly digested and absorbed. 

Concentrations of bacteria in contents of the gastrointestinal tract of pigs are much 
higher in the caecum and in colon than in more proximal portions of the tract. The 
microbiota is dominated by strict anaerobes and the most numerous species are members 
of the genera Bacteroides, Selenomonas, Butyrivibrio, Lactobacillus, Peptostreptococus 
and Eubacterium (20). The development of a complex microbiota in the large intestine 
takes 2-3 weeks after weaning. Starch and some oligosaccharides are mainly digested in 
the small intestine of monogastric animals by enzymes of the salivary glands, pancreas and 
intestinal brush border. Cellulose, hemicelluloses, pectins and some oligosaccharides are 
partly digested by the microbiota of the large intestine. Fiber total digestibility varies 
considerably and depends on the nature of the fiber and the animal species. It is less than 
10% in chickens, whereas pigs seem to digest fibers as well as sheep (21). Dietary fiber 
may contribute up to 30% of the maintenance energy needs of growing pigs. Higher 
energy contributions may be obtained from dietary fiber fed to sows, along with some 
improvements in reproduction, health, and well-being. Swine microbiota constitutes 
highly active ruminal cellulolytic and hemicellulolytic bacterial species, which include 
Fibrobacter succinogenes (intestinalis), Ruminococcus albus, Ruminococcus flavefaciens, 
Butyrivibrio species, and Prevotella (Bacteroides) ruminicola (22). Additionally, a new 
highly active cellulolytic bacterium, Clostridium herbivorans, has been isolated from pig 
large intestine (23). The populations of these microorganisms are known to increase in 



The Gastrointestinal Microbiota of Farm Animals 385 

response to the ingestion of diets high in plant cell wall material. The numbers of 
cellulolytic bacteria from adult animals are approximately 6 to 7 times greater than those 
found in growing pigs. None of these highly active cellulolytic bacterial species are found 
in the human large intestine. Thus, the pig large intestinal fermentation of fiber seems to 
more closely resemble that of ruminants than that of humans (22). 

Poultry 

Bacterial colonization of the intestinal tract of poultry occurs after hatching when 
the young bird starts to receive the feed. The esophagus of gallinaceous poultry creates the 
crop, which serve as a store of the feed. The ingested feed in the crop is softened by water 
and by secretion of salivary glands and the glands of esophagus. In water poultry, the 
esophagus is able to widen throughout its length. The gastric juice produced in the gizzard 
helps in chemical digestion of the feed. The gut of poultry is short and the caecum is 
doubled. Soft feed passes through the digestive tract very fast (2 to 4 hours), crude feed 
takes much longer (up to 20 hours). The poultry should be fed with feed of high nutritive 
value due to the shortness and fast transit time of the intestinal content. 

Lactobacillus microbiota lining the crop of the chicken gastrointestinal tract 
becomes established within a few days after hatching and the specific adherence of avian 
associated lactobacilli onto the crop epithelium plays a role in the colonization (24). From 
the third day of life, large numbers of lactobacilli are present throughout the alimentary 
tract (25). Recent research showed that freshly isolated lactobacilli from chickens are able 
to adhere to the epithelium of crop, as well as to the follicle-associated epithelium and the 
apical surface of mature enterocytes of intestinal villi (26). 

Enterobacteriaceae and enterococci are present in large numbers in 3 -day-old 
broilers but they start to decrease with the age. Lactobacilli, however, remain stable during 
the growth of broilers. The presence of volatile fatty acids is responsible for the reduction 
of Enterobacteriaceae in the broiler chicken. The amounts of acetate, butyrate and 
propionate increase from undetectable amounts in 1 -day-old broilers to high 
concentrations in 15-day-old broilers (27). Facultative anaerobic microbiota (streptococci, 
lactobacilli and E. coli) comprise the predominant microbiota of the small intestine and 
Salanitro and coworkers (28) found that the above-mentioned bacteria represent 60-90% 
of the isolated bacteria. While the number of aerobic and anaerobic bacteria in duodenum 
and ileum were in their study very similar, they found 10 1 ] anaerobic bacteria per g of dry 
tissue in the caecum and the latter exceeded aerobe plate count by at least a factor 100. The 
use of anaerobic methods developed for rumen bacteria have shown that the dominant 
microbiota of the caecum is composed of strict anaerobes and the most frequently isolated 
genera were Eubacterium, Clostridium, Eusobacterium, Bacteroides, Bifidobacterium, 
Peptostreptococcus, and Lactobacillus (28,29). Scanning electron microscopy of the 
intestinal epithelia of 14-day-old chickens revealed populations of microbes on the 
duodenal, ileal and caecal mucosa surfaces (28). 

The study of intestinal microbiota composition has relied almost exclusively on the 
quantitative cultivation of microbes from samples. Culture results obtained in these studies 
compose between 50 and 80% of total microscopic counts (30). Culture-based techniques 
can be very selective, but never capture the total microbial community of complex 
anaerobic habitats such as the avian gastrointestinal tract. Apajalahti and coworkers (31) 
analyzed broiler chickens from eight commercial farms in Southern Finland for the 
structure of their gastrointestinal microbial community by a non-selective DNA-based 
method, percent G + C-based profiling and, in addition, a phylogenetic 16S rRNA gene- 
based study was carried out to aid interpretation of the percent G + C profiles. Most of the 



386 Bomba et al. 

16S rRNA sequences found could not be assigned to any previously known bacterial genus 
or they represented an unknown species of one of the taxonomically heterogeneous genera 
such as Clostridium, Bacteroides and Eubacterium. Bacteria related to ruminococci and 
streptococci were the most abundant members observed. The source of the feed and feed 
amendment changed the bacterial profile significantly. 

Horses 

The intestinal tract of horses and other monogastric herbivores is characterized by a 
combination of a large caecum and an even larger colon where fermentation and 
absorption occurs. Bacteriological studies have shown that the equine intestinal 
ecosystems contain several hundreds of microbial species, of which most are strict 
anaerobes (32) and metabolic products from this microbiota provide the horse with a 
significant part of its energy requirements. There is little information about the microbiota 
of the small intestine in horses. However, like in the other species of animals, the total 
microbial counts as well as E. coli and streptococci rise continuously from duodenum to 
ileum; lactobacilli predominate in the duodenum (33). The acetate concentration increases 
along the length of the small intestine and molar proportion of acetate, propionate and 
butyrate 85:10:3 were found in hindgut (34). Acetate is a common fermentation end 
product from intestinal anaerobes of the genera Bacteroides, Bifidobacterium, 
Eubacterium, Propionibacterium and Selenomonas (35), and it is indicative for a diet 
that is low in rapidly fermentable sugars or concentrates. From the data given by Colinder 
and coworkers (36), horses have a lower total concentration of faecal short-chain fatty 
acids than pigs, rats and man and even lower than the values in cows. The significantly 
higher proportion of acetate can depend on its correlation to high-fiber diets and reflects a 
difference in diets between horses and other monogastric species. Reduced faecal 
excretion of absorbable compounds, as short-chain fatty acids, is probably due to 
prolonged stay of digesta in the hindgut; four days or more (37). Daly and Shirgazi- 
Beechey (38) obtained quantitative data on the predominant bacterial populations 
inhabiting the equine large intestine by using group-specific oligonucletide probes. Results 
showed the Spirochetaceae, the Cytophaga-Flexibacter '-Bacteroides assemblage, the 
Eubacterium re dale- Clostridium coccoides group and unknown cluster C of Clostridia- 
ceae to be the largest populations in the equine gut, each comprising 10-30% of the total 
microbiota in each horse sampled. Other detected notable populations were the Bacillus- 
Lactobacillus-Streptococcus group, Fibrobacter and unknown cluster B, each comprising 
1-10% of the total microbial community. 

Ruminants 

The forestomach of cattle, sheep and goats consists of the reticulum, rumen and omasum 
that are followed by the abomasum; the latter is an analogy of the stomach of monogastric 
animals. 

In young ruminants after birth, only the fourth stomach (abomasum) is functional and 
its capacity is about twice that of the other compartments. In the adult ruminants, abomasum 
represents only 8% of the total capacity. The volume of the rumen represents 80% of the 
total (39). The difference between ruminants and non-ruminant animals results from the 
morphological adaptation of their gastrointestinal tract to the consumption and utilization of 
cellulose as well as their adaptation to utilization of the end products from the rumen 
fermentation. The rumen provides an ideal environment for fermentation with relative 
stable temperature and a continuous supply of the nutrients (40). The ruminal pH value in a 



The Gastrointestinal Microbiota of Farm Animals 387 

healthy animal is 6.2-6.8 and it is influenced by food, buffer capacity of the saliva, by 
products of fermentation and by the animals' ability to absorb the latter through the rumen 
wall. The microbial ecosystem of the rumen is one of the most complex, with wide variety of 
interactions between microorganisms, between microorganisms and the host and between 
microorganisms and the feed (41). The rumen microbial population consists of bacteria, 
protozoa and fungi. The amount of rumen protozoa depends on the diet, but usually ranges 
from 10 4 to 10 7 per ml of rumen digesta. Because of their sensitivity to low pH and sufficient 
amount of nutrients, they can completely disappear from the rumen content. The rumen 
anaerobic fungi take part in rumen fiber digestion (42). 

The population of rumen bacteria is characteristic and indispensable for the 
ruminal ecosystem. Bacteria in the rumen adhere to the epithelium of the rumen wall, to 
feed particles, or they move freely in the contents (43). Bacteria adhering to the 
epithelium of the rumen wall are considered to be the regulating factor of the rumen 
microbiota (44). At the age of 9 to 13 weeks the ruminal microbiota of the calf is similar 
to that of an adult animal. The number of rumen bacteria ranges from 10 9 to 10 11 per ml 
of rumen digesta and depends on the diet and the time of sampling after feeding (45). 
The permanent microbiota consists of more than 60 species of bacteria and the 
concentration of dominant species ranges from 10 to 10 per ml of rumen digesta. The 
most important species are divided in to metabolic groups according to their main 
substrates which they are able to ferment (46) — cellulolytic (Bacteroides succinogenes, 
Ruminococcus albus, Ruminococcus flavefaciens), amylo- and dextrinolytic (Bacteroides 
amylophylus, Streptococcus bovis, Succinomonas amylolytica, Succinivibrio dextrino- 
solvens), saccharolytic (Bacteroides ruminicola, Butyrivibrio fibrisolvens, Megasphaera 
elsdenii, Selenomonas ruminantium) and hydrogen-utilizing bacteria (Methanobacter 
ruminantium, Vibrio succinogenes). The most important attributes of the ruminal 
microbiota are the ability to hydrolyse cellulose, synthesize amino acids, produce 
volatile fatty acids and vitamins. In the young of ruminants, lactate-utilizing bacteria, 
among them Megasphaera elsdenii, Veillonella alcalescens and Selenomonas 
ruminantium (47), are of great importance. Comparative Polymerase Chain Reaction 
(PCR) assays were developed for enumeration of the rumen cellulolytic bacterial species: 
Fibrobacter succinogenes, Ruminococcus albus and Ruminococcus flevefaciens (48). 
Enumeration of the cellulolytic species in the rumen and alimentary tract of sheep found 
Fibrobacter succinogenes dominant; 10 per ml of rumen digesta compared to 
Ruminococcus species (10 4-6 per ml). All three species were detected in the rumen, 
omasum, caecum, colon and rectum, the numbers at these sites varied within and 
between animals. 



INFLUENCING THE ECOSYSTEM OF THE DIGESTIVE TRACT 
IN FARM ANIMALS 

In farm animals the microbiota of the digestive tract plays an important role both in 
the process of optimal development and growth of the organism as well as in securing the 
resistance of animals to diseases. However, due to various adverse impacts, disturbances 
of optimum growth, production and health state of the animals are rather frequent in 
animal production. 

Abrupt change of feed, weaning, stress, administration of antibiotics at therapeutical 
dosage and pathogenic microorganisms can all be classified among these adverse factors. 
All of them disturb the stability and composition of the natural microbiota of the digestive 
tract, thus disturbing physiological processes and resistance of the organism to diseases; 



388 Bomba et al. 

they slow down growth, decrease the performance or lead to diseases of farm animals. 
From these facts it is obvious that in order to minimize the negative effects of adverse 
factors it is essential to give targeted and efficient support to the beneficial microbiota of 
the digestive tract that plays an important part in the physiological processes and in the 
resistance of the organism to diseases. In order to ensure optimum growth, production and 
health of the farm animals the beneficial microbiota of the ecosystem of the digestive tract 
can be supported by manipulation of the diet and application of probiotic microoraganisms. 
Growth-promoting antibiotics will be banned in the European Union by 2006 and similar 
measures may be expected in other countries in the future. From this point of view, it is 
necessary to search for naturally occurring alternatives to antibiotics. The manipulation of 
the gastrointestinal microbiota by diet and application of probiotics could represent such 
safe alternative to antimicrobials. 

Manipulation of the Gastrointestinal Microbiota by Diet 

Dietetic methods can be used to positively influence the development of the rumen 
microbiota of young ruminants during the period of milk nutrition and transition from milk 
to plant feeding; in monogastric animals, mainly pigs, these methods can be used for the 
same purpose mainly at the time of weaning. 

The influence of the feed amount and quality upon the ecosystem of the digestive 
tract is of extraordinary importance (49). If the diet is changed from roughage to grain, the 
rumen microbiota and microfauna and the final products of these elements undergo 
changes as well (50). Dietetic stimulation of the rumen microbiota of ruminants comprises 
several ways of manipulating the feeds offered to the animals, among them changing the 
composition of the feeds, the form of the feeds as well as the time of starting feeding dry 
feeds to milk-fed animals. Adverse factors such as regulation of milk feeding or feeding 
frequency may also be used to influence the development of the rumen microbiota or 
rumen digestion. A gradual decrease of the amount of milk forces the animals to 
supplement the missing nutrients by taking in dry grain and later forage feeds, which 
accelerates the functional and morphological development of the rumen (51-53). 
Cruy wagen and Horn (54) point at the possibility of influencing dry fodder intake by the 
composition of the liquid diet. According to these authors a factor is present in the bovine 
colostrum that stimulates the intake of dry concentrate feed. Bush and Nicholson (55) also 
stated that it would be possible to increase the intake of dry feeds during the period of milk 
nutrition and thus to affect changes in the microbiota of the digestive tract of calves by the 
addition of formic acid. In these animals feeding a pre-starter mixture and weaning at an 
early age have a very positive effect on the functional development of the rumen (56). 

Feed composition is of decisive importance for the stimulation of rumen digestion in 
ruminant young in the period of predominant milk nutrition. The amount of dry feeds is 
only of secondary importance. Easily fermentable grains are vital for the development of 
the amylolytic microbiota while roughage, silage, hayage and hay are decisive for the 
cellulolytic one. With respect to the development of the functions of the forestomachs 
intake of high-quality hay and grain is of vital importance (57,58). Since calves do not 
consume great amounts of hay in the first eight weeks of life, the level of rumen 
metabolism during the period of milk nutrition can be positively affected mainly by a 
suitable composition of the starter mixture (59). With progressing age and maturation of 
the rumen, cellulolytic microbiota gradually develops and increased amounts of hay, 
hayage and silage can be offered to the calves. The cellulolytic activity of rumen bacteria 
is stimulated by isoacids that develop during the catabolism of certain amino acids. Isoacid 
levels in the rumen can be increased by a diet that is rich in concentrate and proteins (60). 



The Gastrointestinal Microbiota of Farm Animals 389 

Dietetic methods can also be used to influence the microeco system of the intestinal 
tract in piglets during weaning. At this period important morphological and functional 
changes occur in the digestive tract of piglets that are also accompanied by changes in the 
composition of the gut microbiota (17,61). In the first days after weaning Lactobacillus 
populations decrease considerably whereas the numbers of coliforms increase. In piglets 
the brush border of the intestinal epithelium can be damaged by feed components (62) or 
viruses (63); such damage enables enterotoxigenic E. coli to colonize the injured 
epithelium. Important factors that the piglets had been receiving by maternal milk and that 
prevented E. coli from colonizing the gut (64) are no more at the animals' disposal. All 
these changes support the tendency to low weight gain and predispose to the occurrence of 
the diarrheic syndrome. Several researchers tried to influence the morphological and 
functional development of pigs during the weaning period in order to optimize digestion 
and to minimize the danger of the post- weaning diarrheic syndrome. Adjustment of the 
form of feeds seems to positively influence morphological development of the intestinal 
epithelium in weaned piglets. On days 8 and 1 1 after weaning Deprez and coworkers (65) 
observed the intestinal villi to be higher in the piglets fed pulpy feeds than in those 
receiving the same composition in pellets. The higher villi observed in the piglets 
receiving pulpy feeds may reflect an increased level of energy intake. This assumption has 
been confirmed by the findings of Partridge and coworkers (66) who stated weanlings 
receiving dry feed in the form of a pulp consume more feed and grow more rapidly than 
piglets receiving the same feed as pellets. Beers-Schreurs and coworkers (67) concluded a 
decreased energy intake during the post-weaning period to be the main cause of villar 
atrophy. If it is our aim to influence the development of the digestive tract during the 
weaning period, then the finding of Kelly and coworkers (12) according to whom 
continuous presence of feeds in the lumen plays an important part in the integrity of 
intestinal morphology and function is of extreme importance. McCracken (68) stated that a 
low intake of feed after weaning might cause morphological and functional changes in the 
intestinal tract. Pluske (69) pointed out that if nutritional stress caused by discontinuation 
of feed intake at weaning could be overcome, transition from maternal milk to solid feeds 
would be less traumatic to the piglets. Milk intake after weaning seems to have 
pronounced stimulating effects upon growth and functioning of the mucosa; it promotes 
the integrity of the small intestine and supports the growth of piglets by increasing or 
maintaining the digestive and absorption capacity. Pluske and Williams (70) demonstrated 
that the height of villi and depth of crypts in weanlings can be maintained by feeding fresh 
cow's milk at two-hour intervals immediately after weaning. 

It is important to stress that current modern methods of rearing frequently employ 
early and abrupt weaning, which increases the predisposition to diseases of the digestive 
tract. The most pronounced changes in the morphology of the intestine, its enzyme 
capacity, in the physiology of digestion and the microbiota of the digestive tract occur in 
the period after weaning. For this reason the composition of feeds during the period of 
transition from milk to plant-based nutrition should take into account the morphological 
changes of the digestive tract and the level of its functional development. 

Manipulation of the Gastrointestinal Microbiota by Application 
of Probiotic Microorganisms 

Administration of preparations based on autochthonous microorganisms is a very effective 
method of affecting the microbiota of the gastrointestinal tract in farm animals. In this way 
development of the microbiota of the young at an early age and around weaning can 
be influenced. 



390 Bomba et al. 

Development of the rumen microbiota in calves and lambs can be supported by 
microbial preparations mainly at the start of dry feeding. Effective use of microbial 
preparations in the young depends also on the level of knowledge of the so-called 
environmental factors in the rumen which determine the age at which a given 
microorganism may colonize the rumen and enable the development of cellulolytic 
microbiota (71). The specificity of using probiotics in calves, lambs and goatlings consists 
in the possibility of influencing the formation of the ruminal ecosystem; application of 
selected strains of rumen microorganisms lays the foundation of a future population 
showing a high fermentation activity. Colonization with selected cultures of living 
microorganisms should enable an earlier and more stable onset of the ruminal type of 
digestion. Controlled action on the rumen microbiota in the young during milk nutrition is 
mainly related to the effect upon development of the microbiota adhering to the epithelium 
of the rumen wall. The effects of stimulation can be expected to be most pronounced at the 
period of the most rapid development of the adherent microbiota, at 2 to 3 weeks of age. 
Autochthonous species colonizing the rumen immediately after birth are of decisive 
importance. This microbiota, though simple at the beginning, enables the development of 
a cellulolytic population and that of ruminal digestion. Strains of Streptococcus bovis may 
be used to stabilize rumen fermentation. During a 4-week administration of a colonizing 
preparation containing S. bovis AO 24/85 to lambs the numbers of S. bovis germs adhering 
to the rumen epithelium were significantly increased (p< 0.001) and so was their alpha- 
amylase activity (72). In order to promote the development of the ruminal microbiota 
Kopecny and Simunek (73) used a mixture of rumen bacteria that contained amylolytic, 
cellulolytic, hemicellulolytic, saccharolytic, proteolytic and lactate-utilizing strains. 

It is of great importance to influence the intestinal microbiota of calves, piglets and 
poultry at an early age since this is the period when the danger of diarrhea-accompanied 
diseases of the digestive tract reaches its maximum. Due to their high morbidity and 
mortality rates such diseases present an extraordinarily serious health and economic issue. 
Preventive application of probiotics at an early age helps to optimize the composition of the 
gut microbiota and has an inhibitory effect upon the pathogens of the digestive tract in the 
young of farm animals. Preventive application of Lactobacillus casei at a dose of 1.10 
germs decreased the counts of enterotoxigenic E. coli O101:K99 adhering to the small 
intestinal mucosa of gnotobiotic lambs by 99. 1 % and 76.0% on day 2 and 4 after inoculation, 
respectively (74). Perdigon and coworkers (75) found the preventive effect of L. casei and 
yoghurt against Salmonella typhimurium infections in mice to depend on the duration of 
administration. The short-term preventive application of Lactobacillus paracasei (76) 
induced slight decrease in number of E. coli adhered to jejunal mucosa of gnotobiotic 
piglets, while continuous application led to significant (p < 0.05) decrease (Fig. 1). Thomke 
and El winger (77) and Mead (78) suggested that it seems possible to lower enteropathogens 
(E. coli and Salmonella) but not to control them by administering Lactobacillus acidophilus. 
Increased lactic acid production in the small intestine of pigs fed lactobacilli and 
yeast caused a decrease in intestinal pH and the presence of E. coli within in intestinal 
content (79). 

Potentiation of the probiotic effect of microorganisms seems to be possible by 
combining them with synergically acting components of natural origin. As such, prebiotics 
(mainly oligosaccharides), substrates and metabolites of microorganisms and phyto- 
components are taken into consideration. Bomba and coworkers (80) showed that the 
administration of L. paracasei alone had almost no inhibitory effect on the adhesion of 
E. coli to the jejunal mucosa of gnotobiotic and conventional piglets while L. paracasei 
administered together with maltodextrin decreased the number of E. coli colonizing the 



The Gastrointestinal Microbiota of Farm Animals 



391 



log 10. cm- 2 




short-term 



continual 



Figure 1 Colonization of the jejunal mucosa of gnotobiotic piglets by Escherichia coli 08: K88 at 
short-term and continual preventive application of Lactobacillus paracasei. ( □ ) Control group E; 
( ■ ) experimental group L-E. Source: From Ref. 76. 



jejunal mucosa of conventional piglets by 2.7 logarithm (4.75 log 10/cm ) in comparison 
to the control group (7.42 log 10/cm , p<0.05). 

Findings reported by Nemcova and coworkers (81) pointed at the fact that the 
probiotic effect of microorganisms could be potentiated by combining them with 
prebiotics. The application of L. paracasei combined with fructooligosaccharides to 
piglets for the first 10 days of life and 10 days after weaning revealed an effect upon 
bacterial counts in the faeces that was significantly more positive than that of lactobacilli 
only. With this combination significantly increased counts of Lactobacillus species, 
Bifidobacterium species, total anaerobes and aerobes as well as significantly decreased 
counts of enterococci were stated in the faeces when compared to the control as well as the 
Lactobacillus only group. Comparison with the controls revealed the combination of 
lactobacilli and fructooligosaccharides to result in a significant decrease of Clostridium 
and Enterobacteriaceae and an insignificant decrease of coliform counts in the faeces of 
piglets. These results prove a synergically positive effect of L. paracasei and 
fructooligosaccharides in the faecal microbiota of piglets (Table 1). 

Our results showed that the application of L. paracasei combined with 
fructooligosaccharides and maltodextrin decreased the preweaning mortality of piglets 
(Fig. 2). The field trial lasted eight months and comprised 4000 heads of 1-35 days old 
piglets and the results were compared with the same period of the previous year in which 
antibiotic feed additivies were used. 

Competition for receptors on the intestinal wall is one of the mechanisms that 
mediates the inhibitory effect of probiotic microorganisms on the adhesion of pathogens to 
the intestinal mucosa. Based on this fact it can be hypothesized that an increase in the 
number of probiotic microorganisms colonizing the intestinal epithelium may potentiate 
their probiotic effect. From this point of view the findings of Ring0 and coworkers (82) 
about the effects of lipids containing feeds on the gastrointestinal microbiota and 
especially on the population of lactobacilli are of great interest. According to Kankaanpaa 
and coworkers (83) higher concentrations of polyunsaturated fatty acids inhibited the 
growth and mucus adhesion of selected lactobacilli whilst growth and mucus adhesion of 
Lactobacillus casei Shirota was promoted by low concentrations of y-linolenic acid and 
arachidonic acid. In gnotobiotic piglets oral administration of oil that contained 
polyunsaturated fatty acids significantly increased the numbers of Lactobacillus paracasei 



392 



Bomba et al. 



Table 1 Composition of Fecal Microbiota in Weanling Pigs Receiving Lactobacillus paracasei 
and Mixture of Lactobacillus paracasei and Fructooligosaccharides 



Organisms 


Group 1 


Group 2 


Group 3 


Total anaerobes 


9.8±0.2 


9.8 + 0.3 


10.2 + 0.2 a*, b* 


Total aerobes 


8.0 + 0.5 


8.2 + 0.2 


9.3 + 0.7 a*, b* 


Bifidobacterium 


7.5 + 0.3 


7.1+0.7 


8.3 + 0.3 a*, b* 


Lactobacillus 


9.9 + 0.1 


9.9 + 0.3 


10.3 + 0.1 a**, b* 


Enterococcus 


9.3 + 0.1 


9.3 + 0.3 


8.2 + 0.2 a ***,b*** 


Clostridium 


8.1+0.1 


7.4 + 0.4 a* 


7.7 + 0.3 a* 


Enterobacteriaceae 


7.9 + 0.4 


6.5 ±0.9 a* 


5.9 + 0.9 a" 


Coliforms 


6.8±0.7 


6.3 + 0.7 


5.8 + 0.7 



Values are mean + SEM of log bacteria counts per gram of wet feces (n = 7). Group 1- 
Group 2 — Lactobacillus paracasei. Group 3 — Lactobacillus paracasei and FOS. 

(a) Significantly different from control group. 

(b) Significantly different from Lactobacillus paracasei group. 
*p<0.05; **p<0.01; ***p<0.001. 

Source: From Ref. 81. 



-control. 



adhering to the jejunal mucosa as compared to the control group (84). It is suggested that 
polyunsaturated fatty acids could modify the adhesion sites for gastrointestinal 
microorganisms by changing the fatty acid composition of the membranes of the 
intestinal epithelial cells (82). The ability of probiotics to adhere to mucosal surfaces is a 
presupposition of their health-promoting effects. The stimulatory effect of polyunsaturated 
fatty acids upon the adhesion of lactobacilli could be used to enhance the effectiveness of 
probiotics in inhibiting the pathogens of the digestive tract. 

Early colonization of the gut by an autochthonous microbiota protects chickens from 
Salmonella infection. The direct competition for the site of attachment is suggested to be 
the prime mechanism for the competitive exclusion (85) and development of a bionTm 
of protective microbiota was observed using scanning electron microscopy. The method of 
competitive exclusion constitutes an additional prophylactic method that may be applied 
directly in the animal to enhance its resistance towards Salmonella infection (86). It is also 
considered a possible application in preventing colonization of poultry with E. coll 0157 



% 




August September October November December January February 



Figure 2 Total preweaning mortality of the piglets during control period July 2000-February 
2001 and during experimental period July 2001-February 2002. (□) 2000, 2001 (■) 2001, 2002. 



The Gastrointestinal Microbiota of Farm Animals 393 

and Campylobacter jejuni (78). Optimal protection against S. typhimurium was observed 
when broiler chicks were treated with a culture of caecal microbiota in combination with 
dietary lactose (87). The same results were described in turkey poultry (88) and layer 
chicks (89). In poultry, lactose can also be considered a prebiotic because of absence of the 
endogenous lactase. The lactose is converted into lactic acid by fermentation of hindgut 
microbiota. The decrease of intestinal pH results in reduction of the S. typhimurium 
concentration. 



THE USE OF GNOTOBIOTIC ANIMALS IN STUDIES OF THE 
GASTROINTESTINAL MICROBIOTA IN FARM ANIMALS 

Gnotobiotic animals proved to be a very useful model for studying the physiology of 
the digestive tract. They mainly enable observation of the role of microorganisms in the 
process of the functional and morphological development of the digestive tract and 
the investigation of bacterial interactions and their influence on the macroorganism. A key 
experimental strategy for defining the conversations that occur between microorganisms 
and their hosts is to first define cellular function in the absence of bacteria (under germ-free 
conditions) and then to evaluate the effects of adding a single or defined population of 
microbes. The power of germ-technology lies in the ability to control the composition of the 
environment in which a multicellular organism develops and functions. The combined use 
of genetically manipulatable model organisms and gnotobiotic has the potential to provide 
new and important information about how bacteria affect normal development, establish- 
ment and maintenance of the mucosa-associated immune system, and epithelial cell 
functions. Gnotobiology can help to provide new insights into the aetiology of infectious 
diseases. The combination of gnotobiotics and molecular genetics should provide a deeper 
understanding of how pathogens arise, how they gain control of their habitat, and what 
contributions are made by the "normal" gut inhabitants to the pathogenesis of diseases. 
Such understanding, in turn, could lead to the development of novel chemicals and 
microbes for use in prebiotic and probiotic strategies in order to prevent or cure infectious 
diseases and perhaps also immune disorders. For a more extensive review on research with 
germ-free and gnotobiotic animals, see the chapter by Norin and Midtvedt. 

Gnotobiotic Ruminants in Studies into the Microbiota 
of the Gastrointestinal Tract 

Gnotobiotic ruminants can be used to observe the development of the rumen ecosystem as 
well as to study the relations between rumen and its microbiota. The rumen microbiota 
directly affects the development of the rumen epithelium and the level of intermediary 
metabolism by the action of rumen fermentation and its final metabolites. Fonty and 
coworkers (90), using meroxenic lambs demonstrated that the functions of the rumen 
and the stability of the ecosystem depended on the complexity and diversity of the 
microbiota. In the light of the present knowledge it is not possible to precisely determine 
the composition of the minimum microbiota enabling rumen development and function. 
Fonty and coworkers (91) also studied the role of rumen microbiota in the development of 
the rumen ecosystem and functional development of the rumen at an early age. Their 
results suggest that the rumen microbiota of the very young lamb plays an essential role in 
the establishment of the rumen ecosystem and in the onset of the digestive functions. 
Those bacterial species that colonize the rumen immediately after birth when this organ is 
not yet active, contribute to a biotope favoring the establishment of cellulolytic strains and 



394 Bomba et al. 

the set-up of digestive processes that affect both degradation of the lignocellulose-rich 
feeds and fermentation of the resulting soluble compounds. Ecological factors controlling 
the establishment of cellulotytic bacteria and ciliate protozoa in the lamb rumen were 
studied in meroxenic lambs (92). The results obtained in this study suggested that the 
establishment of cellulolytic bacteria and protozoa required an abundant and complex 
microbiota and was favored by an early inoculation of the animals. All above-mentioned 
results point at the extremely important role of the microbiota in the development of the 
rumen. There is a good relationship between the development of rumen function and 
the complexity of its microbiota. The presence of a simple microbiota cannot assure the 
digestive function as properly as a complex microbiota can. Bomba and coworkers (93) 
used the gnotobiotic approach to observe the development of rumen fermentation in lambs 
from birth up to 7 weeks of age in association to the complexity of the digestive tract 
ecosystem. The results obtained indicated that complexity of rumen microbiota 
significantly affected the development of rumen fermentation both from the quantitative 
and the qualitative viewpoint. 

The fact that early inoculation of the animals is a factor favoring fermentation and 
digestive activities in the rumen is probably related to the action of bacteria on the 
development of papillae, the rumen mucosa and the digestive tract (94). A complex 
microbiota presents an inevitable presupposition of optimal development of the alimentary 
tract in ruminants. 

Colonization of the individual gut segments by lactobacilli and the inhibitory effect 
of Lactobacillus casei upon the adhesion of enterotoxigenic E. coli K 99 to the intestinal 
wall were also studied in gnotobiotic lambs (74). Soares and coworkers (95) and Lysons 
and coworkers (96) compared several parameters of the morphological and functional 
development in germ-free, gnotobiotic and conventional lambs. 

Monogastric Gnotobiotic Animals in Studies 
of the Gastrointestinal Microbiota 

Monogastric gnotobiotic animals were also used to study the functional and morphological 
development of the digestive tract. Nemcova and coworkers (97) studied the colonization 
ability of selected strains of lactobacilli in the small intestine of gnotobiotic piglets. 
Studies were also aimed at the effects of lactobacilli on the intestinal metabolism during 
the first 3 weeks of life (98). The numbers of lactobacilli adhering to the jejunal and ileal 
mucosa and found in the jejunal and ileal contents were comparable to the data reported by 
other authors (99,100) in conventional and gnotobiotic piglets. Bomba and coworkers 
(101) investigated the effect of the inoculation of three Lactobacillus strains upon organic 
acid levels in the mucosal film and intestinal contents of gnotobiotic pigs. In the jejunum 
of inoculated animals, the mucosal film revealed significantly increased levels of lactic, 
propionic and acetic acids when compared to the contents. In the ileum of gnotobiotic pigs 
propionic acid levels in the mucosal film were significantly higher than those in the 
contents. The above results suggest that significantly increased levels of the lactobacilli- 
produced organic acids in the intestinal mucosal film may present an efficient barrier to 
inhibit the adherence of digestive tract pathogens to the intestinal mucosa. 

Gnotobiotic animals present a very good model to determine bacterial interactions in 
the digestive tract. The interactions of lactobacilli and enterotoxigenic E. coli in the 
intestinal tract of gnotobiotic piglets were observed by Bomba and coworkers (80). 
In experiments carried out in gnotobiotic animals the interest focused on the effects of 
the microbiota upon morphology, motility, secretion and absorption in the digestive 
tract (102,103). The use of germ-free, gnotobiotic and conventional animals facilitated 



The Gastrointestinal Microbiota of Farm Animals 395 

considerable progress in the knowledge of the complex ecological system of the 
gastrointestinal tract in birds (104). 



CONCLUSION 

The gastrointestinal microbiota plays a very important role in the physiology of farm 
animals. Despite substantial knowledge of this ecosystem, it is necessary to obtain 
additional information on the mechanisms mediating their interactions. Such knowledge 
will facilitate the optimization of the development and function of gastrointestinal 
microbiota of, especially young, farm animals. It can be expected, that new 
biotechnological and natural methods for manipulation of gastrointestinal microbiota 
will be developed. These methods will enable to replace prophylactic antibiotic use in 
farm animals' diet and will contribute to the production of healthy and safe foods while at 
the same time benefiting the environment. Several useful in vitro methods are used to 
study gastrointestinal microbiota. It seems that germ-free and gnotobiotic animals could 
represent, in conjunction with in vitro methods, a helpful base for the complex study of 
gastrointestinal ecosystem in farm animals. 



REFERENCES 

1. Salminen S, Boulez C, Boutron-Ruault M-C, et al. Functional food science and 
gastrointestinal physiology and function. Brit J Nutr 1998; 1:147-171. 

2. Smith HW. The development of the flora of the alimentary tract in young animals. J Path Bact 
1965;90:495-513. 

3. Carman RJ, Roger L, van TMS, Tracy DW. The normal intestinal microflora: ecology 
variability and stability. Vet Human Toxicol 1993; 35:11-14. 

4. Savage DC. Microbial ecology of the gastrointestinal tract. Ann Rev Microbiol 1977; 
31:107-133. 

5. Tannock GW. Effects of dietary of environmental stress on the gastrointestinal microbiota. In: 
Hentges DJ, ed. Human Intestinal Microbiota in Health and Disease. London: Academic 
Press, 1983:517-539. 

6. Piard JC, Desmazeaud M. Inhibiting factors produced by lactic acid bacteria. 1. Oxygen 
metabolites and catabolism and products. Lait 1991; 71:525-541. 

7. Freter R. Factors affecting the microecology of the gut. In: Fuller R, ed. Probiotics: The 
Scientific Basis. London: Chapman and Hall, 1992:111-114. 

8. Ducluzeau R, Bellier M, Raibaud P. Transit digestif de divers inoculums bacteriens into duits 
"per os" chez des souris axeniques ou "holoxeniques" (conventionelles) effect antagoniste de 
la microteore dutractus gastrointestinal. Zbl Bacteriol I Abt Orig 1970; 213:533-548. 

9. Lloyd AB, Cumming RB, Kent RD. Prevention of Salmonella typhimurium infection in 
poultry by pretreatment of chickens and poults with intestinal extracts. Austral Vet J 1977; 
53:82-87. 

10. Stavric S, Kornegay ET. Microbial probiotic for pigs and poultry. In: Wallace RJ, Chesson A, 
eds. Biotechnology in Animal Feeds and Animal Feeding. Weinheim, Germany: VCH 
Verlagsgesellschaft mbH, 1995:205-231. 

1 1 . Ducluzeau R. Implantation and development of the gut flora in the newborn piglet. Pigs News 
Inf 1985; 4:415-418. 

12. Kelly D, Begbie R, King TP. Postnatal intestinal development. In: Valey MA, Williams PEV, 
Lawrence TLJ, eds. Neonatal Survival and Growth. Edinburgh, UK: Occasional Publication, 
British Society of Animal Production, 1992; 15:63-79. 



396 Bomba et al. 

13. McGillvery DJ. Anaerobic microflora associated with the pars oesophagea of the pig. Res Vet 
Sci 1992;53:110-115. 

14. Fuller R, Barrow PA, Brooker BE. Bacteria associated with the gastric epithelium of neonatal 
pigs. Appl Environ Microb 1978; 35:582-591. 

15. Tannock GV. The microecology of lactobacilli inhabiting the gastroinetstinal tract. Adv 
Microb Ecol 1990; 11:147-171. 

16. Franklin MA, Mathew AG, Vickers JR, Clift RA. Characterization of microbial populations 
and volatile fatty acid concentrations in the jejunum, ileum, and cecum of pigs weaned at 17 
versus 24 days of age. J Anim Sci 2002; 80:2904-2910. 

17. Jensen BB. The impact of feed additions on the microbial ecology of the gut in young pigs. 
J Anim Feed Sci 1998; 7:45-64. 

18. Pluske JR, Hampson DJ, Williams IH. Factors influencing the structure and function of the 
small intestine in the weaned pig: a review. Liv Prod Sci 1997; 51:215-236. 

19. Nabuurus MJA. Microbiological, structural and functional changes of the small intestine of 
pigs at weaning. Pig News Inf 1995; 16:93-97. 

20. Robinson IM, Allison MJ, Bucklin JA. Characterization of the cecal bacteria of normal pigs. 
Appl Environ Microbiol 1981; 41:950-955. 

21. Champ M. Carbohydrate digestion in monogastric animals. Reprod Nutr Dev 1985; 25:819-842. 

22. Varel VH, Yen JT. Microbial perspective on fiber utilization by swine. J Anim Sci 1997; 
75:15-22. 

23. Varel VH, Tanner RS, Woese CR. Clostridium herbivorans sp.nov. a cellulolytic anaerobe 
from the pig intestine. Int J Syst Bacteriol 1995; 45:490-494. 

24. Fuller R. Ecological studies of the Lactobacillus flora associated with the crop epithelium of 
the fowl. J Appl Bacteriol 1973; 36:131-139. 

25. Barnes EM, Impey CS, Cooper DM. Manipulation of the crop and intestinal flora of the newly 
hatchet chicks. Am J Clin Nutr 1980; 33:2426-2433. 

26. Edelman S, Westerlund-Wikstrom B, Leskela S, et al. In vitro adhesion specificity of 
indigenous lactobacilli, within the avian intestinal tract. Appl Environ Microb 2002; 
68:5155-5159. 

27. van der Wielen PW, Biersterveld S, Notermans S, Hofstra H, Urlings BA, van Knapen F. Role 
of volatile fatty acids in development of the cecal microflora in broiler chickens during 
growth. Appl Environ Microb 2000; 66:2536-2540. 

28. Salanitro JP, Blake IG, Muirhead PA, Maglio M, Goodman JR. Bacteria isolated from 
duodenum, ileum and cecum of young chicks. Appl Environ Microb 1978; 35:782-790. 

29. Adami A, Cavazzoni V. An experimental standard pattern of assessing the cecal microflora of 
chicken. Ann Microb Enzimol 1993; 43:329-336. 

30. Tannock GW. Molecular assesment of intestinal microflora. Am J Clin Nutr 2001; 
73:410-414. 

31. Apajalahti JH, Kettunen A, Bedford MR, Holben WE. Percent G + C profiling accurately 
reveals diet-related differences in the gastrointestinal microbial community of broiler 
chickens. Appl Environ Microb 2001; 67:5656-5667. 

32. Julliand V. Microbiology of the equine hindgut. 1 . European conference on the nutrition of the 
horse. Pferdeheilkunde: Sonderausgabe, 1992:42-47. 

33. Frey K, Sasse HHL. Zur darmflora des pferdes-eine literaturstudie. Pferdeheilkunde 1996; 
6:855-863. 

34. Mackie RI, Wilkins CA. Enumeration of anaerobic bacterial microflora of the equine 
gastrointestinal tract. Appl Environ Microb 1988; 54:2155-2160. 

35. Holdeman LV, Cato EP, Moore WEC. 4th ed. Anaerobe laboratory manual. Blacksburg: 
Virginia Polytechnic Institute and State University, 1977. 

36. Collinder E, Lindholm A, Midtvedt T, Norin E. Six intestinal microflora-associated 
characteristics in sport horses. Equine Vet J 2000; 32:222-227. 

37. Cuddeford D, Pearson RA, Archibald RF, Miurhead RH. Digestibility and gastro-intestinal 
transit time of diets containing different proportions of alfalfa and oat straw given to 
Thoroughbreds, Shetland ponies, highland ponies and donkeys. Anim Sci 1995; 61:407-417. 



The Gastrointestinal Microbiota of Farm Animals 397 

38. Daly K, Shirazi-Beechey SP. Design and evaluation of group-specific oligonucleotide probes 
for quantitative analysis of intestinal ecosystems: their application to assessment of equine 
colonic microflora. FEMS Microb Ecol 2003; 44:243-252. 

39. Roy JHB. The calf. London: Butterwors, 1980. 

40. Russell JB, Hespell RB. Microbial rumen fermentation. J Dairy Sci 1981; 64:1153-1169. 

41. Gall LS. Significance of microbial interaction in control of microbial ecosystem. Biotech 
Bioeng 1970; 12:333-340. 

42. Bauchop T. The anaerobic fungi in rumen fiber digestion. Agri Environ 1981; 6:339-348. 

43. Cheng KJ, McCowan RP, Costerton JW. Adherent epithelial bacteria in ruminants and their 
roles in digestive tract function. Amer J Clin Nutr 1979; 32:139-148. 

44. Cheng KJ, Stewart CS, Dinsdale D, Costerton JW. Electron microscopy of bacteria involved 
in the digestion of plant cell walls. Anim Feed Sci Tech 1984; 10:93-120. 

45. Bartos S. Microbiology and biochemistry of digestion in rumen of ruminants. Praha: 
Academia, 1987. 

46. Baldwin RL, Allison MJ. Rumen metabolism. J Anim Sci 1983; 2:461-477. 

47. Kmet V, Baran M, Kalacnjuk GI. Manipulation of rumen ecosystem in calves and lambs by 
microbiological preparations. Bratislava: Veda, 1990. 

48. Koike S, Kobayashi Y. Development and use of competitive PCR assays for the rumen 
cellulolytic bacteria: Fibrobacter succinogenes, Ruminococcus albus and Ruminococcus 
flavefaciens. FEMS Microb Lett 2001; 204:361-366. 

49. Smith HW. The development of the bacterial flora of the faeces of animals and man: the 
changes that occur during ageing. J Appl Bact 1961; 24:235-241. 

50. Berger LL. Effects of diet composition on rumen fermentation. ISI Atl Sci Anim Plant Sci 
1988; 1:178-182. 

51. Lynch GP, Bond J. Relationship of lactose to nitrogen matabolism of artificially reased beef 
calves. J Dairy Sci 1983; 66:2544-2550. 

52. Stewart GD, Schingoethe DJ. Evaluation of high starch and high fat rations for dairy calves. 
J Dairy Sci 1984; 3:598-605. 

53. Kertz AF, Reutzel LF, Mahoney JH. Ad libitum water intake by neonatal calves and its 
relationship to calf starter intake, weight gain, feces score and season. J Dairy Sci 1984; 
67:2964-2969. 

54. Cruywagen CW, Horn JG. Pre-weaning growth and feed intake of dairy calves receiving 
different combinations of soya bean flavour, whey powder and colostrum. S Afr J Anim Sci 
1985; 15:11-14. 

55. Bush RS, Nicholson JWG. Effect of two acids and formalin in calves milk on faed 
consumption and performance. Can J Anim Sci 1987; 67: 1 129—1 131. 

56. Klein RD, Kincaid RL, Hodgson AS, Harrison JH, Hillers JK, Cronrath JD. Dietary fiber and 
early weaning on growth and rumen development of calves. J Dairy Sci 1987; 70:2095-2104. 

57. Laksesvela B, Slagsvold P, Krogh A, Ommundsen A, LandsverkT. Indigestion in young calves. 
II. The influence of ground barley, coarse and fine hay. Acta Veter Scand 1977; 18:416-425. 

58. Jagos P, Dvorak R, Skrivanek M. The influence of early intake of concentrates and roughage 
feeds on development of digestive processes of calves in high concentration calves house. Vet 
Med (Praha) 1986; 31:257-264. 

59. Bomba A, Laukova A, Reiffova K. The effect of starter diet formula on rumen metabolism in 
calves. Ziv Vyr 1993; 38:1003-1114. 

60. Flachowsky G, Matthey M, Ochrimenko WI, Schneider M. Profile of isoacids in rumen fluid 
and influence of added isoacids on in sacco dry matter disappearance of untreated and 
ammonia treated wheat straw. Arch Anim Nutr Berlin 1988; 38:431-439. 

61. Risley CR, Kornegay ET, Lindermann HD, Wood CM, Eigel WN. Effects of feeding organic 
acids on selected intestinal content measurements at varying times postweaning in pigs. 
J Anim Sci 1992; 70:196-206. 

62. Kik MJ, Huisman J, van der Poel AF, Mouwen MJ. Pathologic changes of the small intestine 
mucosa of piglets after feeding of Phase olus vulgaris beans. Vet Pat 1990; 27:329-334. 



398 Bomba et al. 

63. Lecce JG, Balsbaugh RK, Clare DA, King MW. Rotavirus and hemolytic enterophatogenic 
Escherichia coli in weanling diarrhoea of pigs. J Clin Microbiol 1982; 16:715-723. 

64. Deprez P, Van den Hende C, Muylle E, Oyaert W. The influence of the administration of sows 
milk on the post weaning excretion of the hemolytic, E. coli in the pig. Vet Res Com 1986; 
10:469-478. 

65. Deprez P, Deroose P, van den Hende C, Muylle E, Oyaert W. Liquid verces dry feeding in 
weaned piglets: The influence on small intestine morphology. J Vet Med 1987; B34:254-259. 

66. Partridge GG, Fischer J, Gregory H, Prior SG. Automated wet feeding of weaner pigs versus 
conventional dry feeding effects on growth rate and food consumption. Anim Prod 1992; 
54:484 (abstract). 

67. Beers-Schreurs van, HMG, Nabuurs MJA, Vellenga L, Breukink HJ. The effect of weaning 
and diets on vilous height and crypt depth in the small intestines of piglets. In: Proceedings of 
the IX-th International conference on Production Diseases in farm animals. Berlin: Germany, 
1995; 103. 

68. McCracken KJ. Effect of diet compositions on digestive development of early weaned pigs. 
Proc Nutr Soc 1984; 43:109. 

69. Pluske JR. Psychological and nutritional stress in pigs at weaning: Production parameters, 
the stress response and histology and biochemistry of the small intestine. Ph.D. Thesis, The 
University of Western Australia, 1993. 

70. Pluske JR, Williams IH. Reducing stress in piglets as a means of increasing production after 
weaning: administration ton of amperozide or co-mingling of piglets during lactation? Anim 
Sci 1996;62:121-130. 

71. Fonty G, Gouet P, Jouany J, Senaud J. Ecological factors determing establishment of 
cellulolytic bacteria and protozoa in the rumens of meroxenic lambs. J Gen Microb 1983; 
129:213-223. 

72. Kmet V, Bomba A. Control of rumen microflora development. Ann Rep Inst Anim Phys Slov 
Acad Sci 1987; 7:44-52. 

73. Kopecny J, Simunek J. Probiotics in calves based on rumen microorganisms. In: Proceeding 
on new knowledge in using of additives in calves nutrition. Uhrineves: CSVTS, 1989:82-88. 

74. Bomba A, Kravjansky I, Kastel R, et al. Inhibitory effects of Lactobacillus casei upon the 
adhesion of enterotoxigenic Escherichia coli K99 to the intestinal mucosa in gnotobiotic 
lambs. Small Ruminant Res 1996; 23:199-206. 

75. Perdigon G, Alvarez S, Rachid M, Agnero G. Immune system stimulation by probiotics. 
J Dairy Sci 1995; 78:1597-1606. 

76. Bomba A, Gancarcikova S, Kastel R, Nemcova R, Herich R. Concentration of organic acids in 
the digestive tract of gnotobiotic piglets after preventive application of lactobacilli. Slov Vet 
Cas 1998; 23:321-326. 

77. Thomke S, Elwinger K. Growth promotants in feeding pigs and poultry. III. Alternatives to 
antibiotic growth promotants. Ann Zootech 1998; 47:245-271. 

78. Mead GC. Prospects for "competitive exclusion" treatment to control salmonellas and other 
feedborne pathogens in poultry. Vet J 2000; 159:111-123. 

79. Kovacs- Zomborszky M, Kreinzinger F, Gombo S, Zomborszky Z. Data on the effects of the 
probiotic "Lacto-Sacc". Acta Vet Hung 1994; 42:3. 

80. Bomba A, Nemcova R, Gancarcikova S, Herich R, Kastel R. Potentiation of the effectiveness 
of Lactobacillus casei in prevention of E. coli induced diarrhea in conventional and 
gnotobiotic pigs. In: Paul PS, Francis DH, eds. Mechanisms in the Pathogenesis of Enteric 
Diseases 2. New York: Kluwer Academic/Plenum Publishers, 1999:185-190. 

81. Nemcova R, Bomba A, Gancarcikova S, Herich R, Guba P. Study of the effect of 
Lactobacillus paracasei and FOS on the faecal microflora in weanling piglets. Berl Munch 
Tieraztl Wschr 1999; 112:225-228. 

82. Ring0 E, Bendiksen HR, Gausen SJ, Sundsfjord A, Olsen RE. The effect of dietary fatty acids 
on lactic acid bacteria associated with the epithelial mucosa and from faecalia of Artie charr, 
Salvelinus alpinus (L.). J Appl Microb 1998; 85:855-864. 



The Gastrointestinal Microbiota of Farm Animals 399 

83. Kankaanpaa PE, Salminen SJ, Isolauri E, Lee YK. The influence of polyunsaturated fatty 
acids on probiotic growth and adhesion. FEMS Microb Lett 2001; 194:149-153. 

84. Bomba A, Nemcova R, Gancarcikova S, et al. The influence of to- 3 polyunsaturated fatty 
acids (to-3 pufa) on lactobacilli adhesion to the intestinal mucosa and on immunity in 
gnotobiotic piglets. Berl Munch Tieraztl Wschr 2003; 116:312-316. 

85. Soerjadi AS, Rufner R, Snoeyenbos GH, Weinack OM. Adherence of Salmonellae and native 
gut microflora to the gastroinestinal mucosa of chicks. Avian Dis 1982; 26:576-584. 

86. Mether U. Administration of autochthonus intestinal microflora — a method to prevent 
Salmonella infections in poultry. Dtsch Tierarztl Wochenschr 2000; 107:402-408. 

87. Nisbet DJ, Corrier DE, DeLoach JR. Effect of mixed cecal microflora maintained in 
continuous culture and of dietary lactose on Salmonella typhimurium colonization in broiler 
chicks. Avian Dis 1993; 37:528-535. 

88. Corrier DE, Hinton A, Jr., Kubena LF, Ziprin RL, DeLoach JR. Decreased Salmonella 
colonization in turkey poults inoculated with anaerobic cecal microflora and provided dietary 
lactose. Poult Sci 1991; 70:1345-1350. 

89. Corrier DE, Hargis B, Hinton A, Jr., et al. Effect of anaerobic cecal microflora and dietary 
lactose on colonization resistance of layer chicks to invasive Salmonella enteritidis. Avian Dis 
1991; 35:337-343. 

90. Fonty G, Gouet P, Ratefiarivelo H, Jouany JP. Establishment of Bacteroides succinogenes and 
measurement of the main digestive parameters in the rumen of gnotoxenic lambs. Can 
J Microbiol 1988; 34:39-46. 

91 . Fonty G, Jouany JP, Chavarot M, Bonnemoy F, Gouet P. Development of the rumen digestive 
functions in lambs placed in a sterile isolator a few days after birth. Reprod Nutr Develop 
1991; 31:521-528. 

92. Fonty G, Gouet P, Jouany JP, Senaud J. Ecological factors determining the estabilishment of 
cellulolytic bacteria and protozoa in the rumen of meroxenic lambs. J Gen Microbiol 1983; 
129:213. 

93. Bomba A, Zitrlan R, Koniarova I, et al. Rumen fermentation and metabolic profile in 
conventional and gnotobiotic lambs. Arch Anim Nutr 1995; 48:231-243. 

94. Lysons RJ, Alexander TJL, Wellstead D, Hobson PN, Mann SO, Stewart CS. Defined 
bacterial populations in the rumens of gnotobiotic lambs. J Gen Microbiol 1976; 94:257-269. 

95. Soares JH, Leffel EC, Larsen RK. Neonatal lambs in a gnotobiotic environment. J Anim Sci 
1970; 31:733-740. 

96. Lysons RJ, Alexander TJL, Wellstead D, Jennings W. Observations on the alimentary tract of 
gnotobiotic lambs. Res Vet Sci 1976; 20:70-76. 

97. Nemcova R, Bomba A, Herich R, Gancarcikova S. Colonization capability of orally 
administered Lactobacillus strains in the gut of gnotobiotic piglets. Dtsch Tierarztl Wschr 
1997; 105:199-200. 

98. Bomba A, Gancarcikova S, Nemcova R, et al. The effect of lactic acid bacteria on intestine 
metabolism and metabolic profile of gnotobiotic pigs. Dtsch Tieraztl Wschr 1998; 
105:384-389. 

99. Sarra PG, Cantalupo R, Massa S, Trovatelli LD. Colonization of the gastrointestinal tracts of 
conventional piglets by Lactobacillus strains. J Gen Appl Microb 1991; 37:219-223. 

100. Tortuero F, Rioperex J, Fernandez E, Rodriguez ML. Response of piglets to oral 
administration of lactic acid bacteria. J Food Protect 1995; 58:1369-1374. 

101. Bomba A, Kastel R, Gancarcikova S, Nemcova R, Herich R, Cizek M. The effect of 
lactobacilli inoculation on organic acid levels in the mucosal film and the small intestine 
contents in gnotobiotic pigs. Berl Munch Tieraztl Wschr 1996; 109:428-430. 

102. Gordon HA, Pesti L. The gnotobiotic animal as a tool in the study of host parasite 
relationships. Bacteriol Rev 1971; 35:390-429. 

103. Yokota H, Coates ME. The uptake of nutrients from the small intestine of gnotobiotic and 
conventional chicks. Br J Nutr 1982; 47:349-356. 

104. Vanbelle M, Teller E, Focant M. Probiotics in animal nutrition: a review. Arch Anim Nutr 
1990; 40:543-567. 



Index 



Abomasum, 386 

Acetate, 53, 324, 386 

Achlorhydria, 30, 226 

ACNQ. See 2-amino-3-carboxy-l, 

4-naphthoquinone 
Acquired immunity, 96 
Actinobacteria, gene analysis, 2 
Actinomyces israellii, 30 
Acute leukemia, 360 
Adhesins, 130 

Adhesion, 129-130, 276, 292, 297, 390 
Aging, 77 
Aglycones, 156 
Allergies 
diet, 198 
infants, 189-199 
pathogens, 197 
Allochthonous microbes, 127 
2- Amino-3-carboxy- 1 ,4-naphthoquinone 

(ACNQ), 287 
Amino acid homeostasis, 53 
Amino acid metabolism, 139-140 
Aminoglycosides, 355-356 
Amoxicillin, 336 
Ampicillin, 336 
Androgens, 143 
Angiogenins, 104 
Animal models, 253-265 
Anthocyanins, 162 
Antibiotic-associated diarrhea, 296, 

318,319 
Antibiotics, 27, 94, 116, 189, 296, 

335-363 
Antibodies, 27 

Antigen-presenting cells (APC), 96, 99, 112 
Anti-rotavirus response, 107-108 
Apoptosis, 226, 230, 264 
Arachidonic acid, 391 
Aromatization, 144 



Asthma, 113, 190 
Atopic diseases, 319 
Atopy, 190, 194 
Autochthonous microbes, 127 
Autoimmune diseases, 115-116 
Azalide, 354 
Azithromycin, 354 
Azlocillin, 340 
Azoreductase, 146, 326 
Aztreonam, 348 
Azulfidine, 148 

Bacampicillin, 340 

Bacillus, 123 

Bacterial adhesion, 129-130 

Bacterial cholesterol metabolism, 142 

Bacterial enzymes, 230-231 

Bacterial nitrozation, 147 

Bacteriocins, 323 

Bacteroides 

carbohydrate fermentation, 138-139 
diet, 55 
feces, 44 

large intestine, 42 
volatile fatty acids, 36 
B. distasonis, 55 
B. fragilis 
diet, 55 

gene analysis, 12 
B. thetaiotaomicron 

cytokine expression, 212 
fucose, 123 

genome sequencing, 178 
host-microbe interactions, 53, 181 
PCR analysis, 64 
B. vulgatus, 3, 55, 64 
Bacteroides, 52, 123, 208 
Bacteroidetes , gene analysis, 2, 8 
Batch-type simulators, 238 



401 



402 



Index 



Beta-aspartylglycine, 279 
Bifidobacteria, 52, 76, 286, 315 

adults, 292-294 

aging, 77, 78, 79-88, 95 

colonization, 54, 76 

diet, 55 

immune response, 107-108 

infants, 107, 130, 291 

large intestine, 42 

mucus adhesion, 80-81 

PCR techniques, 65 

pets, 372 

prebiotics, 295 

probiotics, 317 

taxonomy, 79 
Bifidobacterium 

B. adolescentis, 79, 87 
IL-12 production, 82 
PCR analysis, 64 

B. angulatum, 79 

B. bifidum, 79 

B. breve, 79 

B. catenulateum, 79 

B. dentium, 79 

B. infantis, 212 

B. lactis, 159, 319 

B. longum 

genome sequencing, 178 
PCR analysis, 64 

B. parvulorum, 79 

B. pseudocatenulateum, 79 
Bifidogenic factors, 286 
Bile acid metabolism, 27, 277, 279 
Bile acids, 143,231-232 
Bile acid test, 39-40 
Bile pigments, 143, 276 
Biomarkers, 230 
Biopsy 

esophagus, 29 

large intestine, 43-44 

small intestine, 34-35 

stomach, 30-31 
Biotin, 141 
Birth control, 150 

Bottle feeding. See formula- fed babies 
Breast feeding, 54, 76, 94, 107 
Breath tests, 39-42 
Butyrate, 53, 229, 232, 297, 324 
Bystander suppression, 101 



Caecum, 254, 263, 275, 382 
Caesarian section delivery, 54, 94, 198 
Calcium, 232, 298 



Calves, 388, 390 
Campylobacter, 43, 374 

C. jejuni, 128, 393 

C. upsaliensis, 374 
Cancer, 225-233, 263-264 
Candida 

allergies, 191 

C. albicans, 28, 299 
Candidiasis, 296 
Cannulation, 263 
Capsule biopsies, 36 
Carbapenems, 348 
Carbohydrates, 52, 301 

fermentation, 138-139 
Carbon-carbon bond cleavage, 144 
Carcinogens, 145 
Carnivorous animals, 381 
Catechin, 162 
Cats, 371-376 
CD14, 102 

CD. See Crohn's disease 
CD AD. See Clostridium dijficile-associated 

diarrhea 
14 C-D-xylose breath test, 40 
Cefaclor, 344-345 
Cefadroxil, 347 
Cefazolin, 343 
Cefbuperazone, 343 
Cefepime, 343 
Cefetamet, 347 
Cefexime, 347 
Cefmenoxime, 343 
Cefoperazone, 343 
Cefotaxime, 343 
Cefotiam, 343 
Cefoxitin, 343 
Cefozopran, 344 
Cefpirome, 344 
Cefpodoxime proxetil, 347 
Cefprozil, 347 
Ceftazidime, 344 
Ceftibuten, 347 
Ceftizoxime, 344 
Ceftriaxone, 344 
Cefuroxime/axetil, 347 
Cell-mediated immunity (CMI), 8 1 
Cell signaling, 209 
Cellular fatty acid (CFA) analysis, 78 
Cellulose, 387 
Cephalosporins, 340-348 
Cephradine, 348 
CFA. See cellular fatty acid 
14 C-glycocholate breath test, 39-40 
Chemokines, 95 



Index 



403 



Chlorogenic acid, 157 
Cholera, 44 

Cholesterol, 142, 277-278, 298 
Cigarette smoking, 227 
Ciprofloxacin, 360-361 
Cirrhosis, 343, 360 
Clarithromycin, 350, 354 
Clindamycin, 27, 355 
Clonal libraries, 59 
Clostridia, 52, 208 

gene analysis, 2 

large intestine, 42 

volatile fatty acids, 36 
Clostridium 

C. coccoides 

fecal samples, 44 
gene analysis, 8, 10 

C. difficile, 128, 296 

associated diarrhea (CDAD), 78, 294 
large intestine, 43 
pets, 373 

C. herbivorans, 384 

C. histolyticum, allergies, 192 

C. innocum, 191 

C. leptum, gene analysis, 8, 10 

C. orbiscindens, 161 

C. ramosum, 276 
Clostrium 

C. clostridioform, PCR analysis, 64 

C. perfringens, 11 

enterotoxin (CPE), 373 
CMI. See cell-mediated immunity 
Colitis, 210 

Collinsella serogaciens, 55 
Colon cancer, 55, 297, 319 
Colonization 

elderly, 75-79 

farm animals, 381, 390 

infants, 94, 128-129, 208 

pets, 372 
Colonization resistance, 53, 299, 335 
Colorectal cancer (CRC), 225, 228-229 
Colorectal surgery, 343, 360 
Community profiling assays, 59 
Constipation, 302, 315, 318 
Coprophagy, 255 
Coprostanol, 277 
Corticosteroids, 143 
Co-trimoxazole, 356 
Cow milk allergy, 192 
COX-2. See cyclooxygenase, 2 
CPE. See Clostrium perfringens 

enterotoxin 
CRC. See colorectal cancer 



Crohn's disease (CD), 208 
molecular ecology, 3 
probiotics, 217-218 
susceptibility gene, 209 

Crosby capsule, 36 

Cross-talk, 53, 130-131 

Cryptosporidium, 28 

Cycasin, 145 

Cyclamate, 149 

Cyclooxygenase-2 (COX-2), 227 

Cytokines, 95, 190, 196, 212, 321 

Cytomegalovirus, 28 



DC. See dendritic cells 

Decarboxylation, 140 

Dendritic cells (DC), 95, 99, 100, 106-107, 

196, 212 
Denaturing gradient gel electrophoresis 

(DGGE), 59 
Desulfovibrio, 144 
DGGE. See denaturing gradient gel 

electrophoresis 
Diarrhea, 43, 300, 315, 374, 390 
Diet 

allergies, 198 

cancer, 225 

farm animals, 388-389 

fecal microbiota, 55 
Dietary fibers, 290, 301, 384 
Diethylstilbesterol, 149 
Digoxin, 149 
3,4-Dihydroxyphenylalanine (DOPA), 

147-148 
Dipstick analysis, 44 
Dirithromycin, 354 
DNA microarrays, 12 
DNA synthesis, 356-363 
Dogs, 371-376 

DOPA. See 3,4-dihydroxyphenylalanine 
Dot-blot hybridization, 8, 66 
Doxycycline, 355 
Drug metabolism, 147-150 
Duodenal string test, 37-38 



Ecology, 1 

Eczema, 114, 190, 194,292 

EHEC. See enterohemorragic 

Escherichia coli 
Elderly, 75-88, 294-295 
Endoscopy, 34 
Enoxacin, 361 
Enteric infections, infants, 107 



404 



Index 



Enterobacteriaceae 

allergies, 191 

elderly, 77 

large intestine, 42 

poultry, 385 
Enterococcus, 29, 52, 55, 208 

E.fecalis, 82, 170 
Enterocytes, 124 
Enterohemorragic Escherichia coli (EHEC), 

43, 128, 373 
Enteropathogenic Escherichia coli (EPEC), 

43, 128, 132 
Enterotest, 37-38 

Enterotoxic Escherichia coli (ETEC), 33 
Enterotoxin, 319 
Enzymes, 26 

EPEC. See enteropathogenic Escherichia coli 
Epifluorescence microscopy, 9 
Epithelium-bacterial cross talk, 111 
Erythromycin, 354 
Escherichia coli, 123, 208 

colonization, 76 

diet, 55 

gene analysis, 12 

host-microbe interactions, 170 

hydroxycinnamates, 159 

infants, 94 

PCR analysis, 64 

pets, 373 

pigs, 384 
Escherichia coli, 157, 392-393 
Esophagus, 28-29 
Estrogens, 143, 150 

ETEC. See enterotoxic Escherichia coli 
Eubacterium, 42, 52, 123 

E. biforme, PCR analysis, 64 

E. hallii, gene analysis, 10 

E. lentum, 149 

E. limosum, PCR analysis, 64 

E. ramulus, 161 

E. rectale 

fecal samples, 44 
gene analysis, 10 

FAE. See follicle-associated epithelium 

Faecalibacterium prausnitzii, 2 

Farm animals, 381-395 

FCM. See flow-cytometry 

Fecal biomarkers, 230 

Fecal bulking, 287, 299 

Fecal enzymes, 324, 326 

Fecal samples, 44, 55 

Fecal water, 232-233 

Fermentative activity, 297 



Fermentor-type simulators, 238-241 

Ferrulic acid, 157 

Fetus, 76 

FISH. See fluorescent in situ hybridization 

Flavan-3-ols, 161-162 

Flavonoids, 159-162 

Flavonols, 159-161 

Flomoxef, 344 

Flow-cytometry (FCM), 14-15 

Fluorescent in situ hybridization (FISH), 8-11, 

66, 106 
Folic acid, 141 
Folic acid antagonists, 356 
Follicle-associated epithelium (FAE), 98 
Food allergies, 94, 110, 191 
Food dyes, 146 

Formula-fed babies, 54, 76, 94, 107, 292 
FOS. See fructo-oligosaccharides 
Fructans, 290 

Fructo-oligosaccharides (FOS), 300, 375, 391 
Fucose, 123 

Fucosylated glycoconjugates, 103 
Full thickness biopsy, 35 
Fusobacterium, 52, 123 
F. prausnitzii, 64 

GAC. See germ-free associated characteristics 

Galacto-oligosaccharides, 290, 298 

GALT. See gut-associated lymphoid tissue 

Garenoxacin, 361 

Gas chromatography, 36 

Gastric Helicobacter-Yike organisms (GHLO), 

374-375 
Gastroenteritis, 318 
Gatifloxacin, 361 
Gemifloxacin, 361 
Gene analysis, 2-4, 173-177 
Gene deficient animals, 263 
Gene expression, 14, 230 
Genistein, 142 

Genome sequencing, 177-180 
Germ-free animals, 260-261, 274 
Germ-free associated characteristics (GAC), 

261, 274 
Germ-free life, 273-281 
GFP. See green fluorescent protein 
GHLO. See gastric Helicobacter-like 

organisms 
Giardia lamblia, Enterotest, 38 
Glucose hydrogen breath test, 41-42 
Beta-glucuronidase, 145-146, 230-231, 326 
Glycopeptides, 350 
Glycoproteins, 126, 287 
Glycosidase, 145 



Index 



405 



Gnotobiotic mice, 104, 181 
Gnotobiotic ruminants, 393-394 
Goblet cells, 124 
Gram-negative bacteria 

immunosenescence, 82 

phase variation, 130 

sulphate metabolism, 143 
Gram-positive bacteria 

immunosenescence, 82 

pets, 372 

small intestine, 5 1 
Green fluorescent protein (GFP), 14 
Group-specific PCR-DGGE, 5-6 
Growth rates, 13 
Gut-associated lymphoid tissue (GALT), 53, 

98, 256, 280 
Gut epithelium, 212 

Haemophilus influenzae, genome sequencing, 

177 
Hay fever, 113 
Helicobacter, 374-375 
H. pylori, 30, 5 1 

antibiotics, 360 

cancer, 226-227 

cross talk, 131 

sampling, 28, 30-31 

urea breath test, 31-32 
Hepatic encephalopathy, 299, 302 
Herbivorous animals, 381 
Herpes simplex virus, 28 
HFA. See human flora-associated animals 
Hippuric acid, 158 
Histoplasma capsulatum, 28 
HMOs. See human milk oligosaccharides 
Hormone replacement therapy, 150 
Horses, 382, 386 

Host-microbe interactions, 53, 169-183 
Human flora-associated animals (HFA), 

261-262 
Human milk oligosaccharides (HMOs), 

287, 291 
Hybridization techniques, 7-8 
Hydrogen breath test, 39 
Hydroxybenzoates, 159 
Hydroxycinnamates, 157-159 
Hygiene hypothesis, 113, 189 
Hypersensitivity, 100 
Hypochlorhydria, 226 
Hypocholesterolemea, 318 



IgA-secreting cells, 105 

IIS. See intestinal immune system 

IL. See interleukin 

Imipenem, 348 

Immune modulation, 93-117, 287, 301 

Immune response 

maturation, 292 

probiotics, 319 

regulation, 112-116 
Immunosenescence, 81, 87 
Infants 

allergies, 189-199 

bifidobacteria, 107, 130, 291 

colonization, 54-55, 76, 94, 112, 208 

eczema, 114 

enteric infections, 107 

fecal ecosystem, 5 

feeding, 63, 76 

Thl/Th2 balance, 113 
Infectious gastritis, 30 
Inflammation 

chronic, 225 

esophagus, 28 
Inflammatory bowel disease (IBD), 93, 
209-211,285 

molecular ecology, 3 

prebiotics, 293-294 

probiotics, 207-219, 319 

tolerance, 102 
Innate immunity, 95-96, 112 
Interferon- y, 190 
Interleukin (IL-10), 196, 211, 213 
Interleukin (IL-12), 82, 196 
Intestinal immune system (IIS), 93, 97-103 
Intestinal intubation, 37 
Intestinal microbiota, 76 
Intestinal motility, 52 
Intestinal permeability, 103, 213 
Intra-epithelial lymphocytes (IEL), 98 
Inulin, 213, 289, 298, 300 
In vitro models, 237-250 
In vivo expression technology (IVET), 173, 

174-177 
Isoflavones, 141-142 
IVET. See in vivo expression technology 

Kaempferol, 159 

Kantvik colon simulator (KCS), 240-241 

Ketolide, 354 

Klebsiella pneumoniae, 174 



IBD. See inflammatory bowel disease 
IEL. See intra-epithelial lymphocytes 



LAB. See lactic acid bacteria 
Lactate, 324 



406 



Index 



Lactic acid bacteria (LAB), 230, 372 
Lactobacillus, 76, 123, 286 

cytokine production, 197 

diet, 55 

elderly, 78 

gene analysis, 8 

gnotobiotic piglets, 394 

L. acidophilus 
elderly, 78 
gene analysis, 12 
PCR analysis, 64 

L. bulgaricus, 212 

L. casei 

community profiling, 63 
probiotics, 212 

L. gasseri, 159 

L. plantarum, 319 

gene analysis, 13, 177 
host-microbe interactions, 171 
IL-12 production, 82 

L. re uteri, 319 

L. rhamnosus, 63, 319 

L. salivarius UCC, 118, 212 

poultry, 385 

probiotics, 315, 317 
Lactococcus lactis, 214 
Lactoferrin, 287 
Lactose intolerance, 315 
Lactulose, 40, 290, 298, 299, 302 
Lactulose hydrogen breath test, 40-41 
Lambs, 390 
Lamina propria, 125 
Large intestine, A2-AA 

cancer, 228-230 

function, 125 
Lectins, 130 
Lenapenem, 350 
Levofloxacin, 361 
Lignans, 142 
Lincosamide, 355 
Linezolid, 356 
y-Linolenic acid, 391 
Lipid metabolism, 140, 298 
Lipopoly saccharides, 173 
Listeria monocytogenes, 170, 174, 299 
Lomefloxacin, 361 
Loracarbef, 344, 348 
Luminal washes, esophagus, 29 

MAC. See microflora associated 

characteristics 
Macrolides, 350, 354 
Macrophages, 95 
Magnesium, 298 



MALT. See mucosa-associated lymphoid 

tissue 
Maltodextrin, 391 
Mannanoligosaccharides, 375 
Maternal antibiotics, 94 
M cells, 125 

ME. See Milieu exterieur 
Membrane- array technique, 68 
Meropenem, 348-349 
Mesenteric lymph nodes (MLN), 256 
Metabolic activity, 13-15 
Metronidazole, 148, 360 
MI. See Milieu interieur 
Microflora associated characteristics (MAC), 

261, 274 
Migrating motor complex (MMC), 27 
Milieu exterieur (ME), 274 
Milieu interieur (MI), 274 
Milk, 287, 315 
Mineral absorption, 287, 298 
MLN. See mesenteric lymph nodes 
MMC. See migrating motor complex 
Molecular ecology, 1-16 
Molecular fingerprinting, 4-6, 68, 296 
Monobactams, 348-350 
Moxalactam, 344 
Moxifloxacin, 362 
Mucin, 278, 321 

Mucosa-associated lymphoid tissue (MALT), 99 
Mucosal brushings, 35 
Mucosal interactions, 123-132 
Mucus, 125-127 
Mucus adhesion, 80-81 
Mutagens, 144-147 
Mycobacterium 

M. avium, 28, 30 

M. paratuberculosis, 210 

M. tuberculosis, 30 

Natural immunoglobulins, 114-115 

Natural killer cells (NK), 82, 95 

NDOs. See non-digestible oligosaccharides 

Neutrophils, 95 

Nitrate, 231 

Nitrate reductase, 326 

Nitric oxide (NO), 95, 321 

Nitrites, 147 

Nitrofurantoin, 356 

Nitrogen metabolism, 139-140 

Nitroimidazoles, 356, 360 

Nitroreductase, 146-147, 326 

NK. See natural killer cells 

NF-kB. See nuclear factor kB 

NO. See nitric oxide 



Index 



407 



Non-digestible oligosaccharides (NDOs), 287, 

294, 301 
Non-fermentor models, 241-242 
Nonsteroidal anti-inflammatory drug (NSAID), 

227 
Norfloxacin, 362 
Normal microbiota, 51-69 
NSAID. See nonsteroidal anti-inflammatory 

drug 
Nuclear factor kB (NF-kB) pathway, 111, 211, 

212-213 
Nutritional stress, 389 

Ofloxacin, 362 

Oligosaccharides, 286 

Omnivorous animals, 381 

Oral tolerance, 100-101, 109-110, 209 

Orocecal transit time, 40 

Oxazolidinone, 356 

p53 role, 227 

PAMPs. See pathogen-associated molecular 

patterns 
Paneth cells, 125 
Pantethine, 287 
Parkinson's disease, 147 
Pathogen-associated molecular patterns 

(PAMPs), 131 
Pathogens, 26, 256 

adhesion, 297 

allergies, 197 

enteric, 128 

esophagus, 28-29 

large intestine, 43, 128 

pets, 372-374 

probiotics, 323 

small intestine, 33-34 

translocation, 300 
PCR-cloning, 64-66 
PCR-denaturing gradient gel electrophoresis 

4-5, 64, 296 
Pediatrics, Enterotest, 38 
Pefloxacin, 362 
Penicillins, 336-340 
Peptic ulcers, 51, 227 
Peptide nucleic acid (PNA), 9 
Peptidoglycan, 196 
Peptococcus, 52 
Peptostreptococcus, 52, 123 

P. productus, 55 
Peristalsis, 25 

Preoperative needle aspiration, 35-36 
Peroral intubation, 38 



Peroxisome proliferator-activated receptor 

(PPARy), 111 
Pets, 371-376 

Peyer's patches (PPs), 98, 101 
Pharmabiotics, 211 
Phase variation, 130 
Phenolic acids, 157-159 
Phenoxymethylpenicillin, 336 
Phylochips, 12 
Physiology, 26-28 
Pigs, 383-385 
Piperacillin, 340 
Pivampicillin, 340 
Pivmecillinam, 340 
PNA. See peptide nucleic acid 
Polyphenols, 155-164 
Polyunsaturated fatty acids, 392 
Postnatal development, 27 
Pouchitis, 210, 216-217 
Poultry, 382, 385-386 
PPARy. See peroxisome proliferator-activated 

receptor 
PPs. See Peyer's patches 
Prebiotics, 285-302, 375, 391 
Proanthocyanidins, 163 
ProbeBase, 7 
Probiotics, 1, 296-297, 315-327 

diarrhea, 319-320 

farm animals, 390 

immune response, 114 

immunosenescence, 87 

inflammatory bowel disease, 207-219 

pets, 375-376 

pouchitis, 216-217 
Procyanidins, 163 
Progesterone, 143 
Propionate, 324 

Propionibacteriumfreudenreichii, 170, 287 
Prostaglandin, 321 
Protein, 52 
Proteus, 17, 123 
Pseudomembranous colitis, 296 
Pseudomonas, 29 
Pyxigraphy, 44 

Quantitative reverse transcriptase PCR, 180 
Quercetin, 159 
Quinic acid, 144 
Quinolones, 360-363 
Quinupristin/dalfopristin, 355 

Raffinose, 292 
Rapid urease test, 3 1 
Reading simulator, 239 



408 



Index 



Real time PCR, 12 
Rectum, 125 
Regulatory T cells, 190 
Resistance, 335 
Rhinoconjunctivitis, 190 
Rodents, 254, 255 
Rotavirus, 108, 319 
Rothia, 29 
Roxithromycin, 354 
Rufloxacin, 362 
Rumen, 382, 386-387 
Rumen simulators, 244-249 
Ruminants, 381-382, 386-387 
Ruminococcus, 52 

gene analysis, 12 

R. obeum, 67 

R. productus, diet, 55 

Saccharomyces boulardii, 217 ', 319 
Salicylazosulfapyridine, 148 
Salmonella, 43, 128 

Enterotest, 38 

host-microbe interactions, 170 

pets, 373 

poultry, 392-393 

S. enterica, 174 

S. enteritidis, 299 
Sampling, 28-46 

SBBO. See small bowel bacterial overgrowth 
SCFAs. See short-chain fatty acids 
SCOTS. See selective capture of transcribed 

sequences 
SCW. See streptococcal cell wall 
Secretory IgA antibodies, 102 
Selective capture of transcribed sequences 

(SCOTS), 173 
Self- antigens, 211 
Shigella, 43, 128 
SHIME model, 239-240 
Short-chain fatty acids (SCFAs), 52, 53, 140, 

229, 232, 279, 287, 324 
Signature tagged mutagenesis (STM), 172 
SIP. See stable isotope probing 
Sitafloxacin, 362 
Skin diseases, 319 
Small bowel aspiration, 36-38 
Small bowel bacterial overgrowth (SBBO), 

27, 34, 36 
Small intestine, 32-42, 124-125 
Soybean oligosaccharides, 289 
Sparfloxacin, 362 

16S rRNA gene analysis, 2-4, 66, 169, 385 
Stable isotope probing (SIP), 13 
Staphylococcus, 29, 55, 123 



STM. See signature tagged mutagenesis 
Stomach, 29-32, 76 

cancer, 226-227 
Streptococcal cell wall (SCW)-induced 

arthritis, 115 
Streptococci, diet, 55 
Streptococcus, 29, 77, 94 

S. alactolyticus, 372 

S. bovis, 229, 390 

S. mitis, 82 

S. parasanguinis, 59 

S. salivarius, 59 
Streptogramin, 355 
Strickland reaction, 139 
Subspecies differentiation, 68 
Succession, elderly, 75-79 
Sulphate metabolism, 143-144 
Surgically modified animals, 262-263 
Synbiotics, 289, 294-295 

Talampicillin, 340 
Taxonomy, Bifidobacteria, 79 
Tazobactam, 340 
Teicoplanin, 350 
Telithromycin, 354 
Terminal-restriction fragment length 

polymorphism (T-RFLP), 6 
Tetracycline, 355 

Thl/Th2 balance, 96-97, 112-113, 256 
Thiamine, 141 
Ticcarcillin, 340 
Tinidazole, 360 
TLRs. See Toll-like receptors 
Tobramycin, 355-356 
Tolerance, 27 

Toll-like receptors (TLRs), 95, 131 
Transit time, 52 
Traveler's diarrhea, 318, 360 
Treponema pallidum, 30 
T-RFLP. See terminal-restriction fragment 

length polymorphism 
Tropheryma whipplei, 33 
Trovafloxacin, 363 
Trypsin, 276 
Tryptophan, 147 
Type B gastritis, 51 
Tyrosine, 147 

UC. See ulcerative colitis 
Ulcerative colitis (UC), 148, 208 

probiotics, 215-216 
Ulcer bleeding, 227 
Urea breath test, 31-32 



Index 



409 



Urinary excretion tests, 38 

Urobilins, 276 

Urogenital tract infection, 319 



Water retention, 299 
Weaning, 63, 76, 292, 384, 389 
Whipple's disease, 33 



Vancomycin, 350 
Veillonella, 123 
Vibrio cholerae, 33, 44 
Vitamin B 12 , 141 
Vitamin C, 227 
Vitamin K, 141, 274-275 
Vitamins, 26 
Volatile fatty acids, 36 



Yeasts 

antibiotics, 336 

diet, 55 
Yersinia 

Y. enter ocolitica, 43, 128 

Y '. pseudotuberculosis, 131 
Yogurt, 315