Skip to main content

Full text of "كتب في علم الميكروبيولوجي"

See other formats








































I 



• 














PCR METHODS IN FOODS 



FOOD MICROBIOLOGY AND FOOD SAFETY SERIES 

Food Microbiology and Food Safety publishes valuable, practical, and timely 
resources for professionals and researchers working on microbiological topics 
associated with foods, as well as food safety issues and problems. 



Editor-in-Chief 
Michael P. Doyle 

Regents Professor and Director of the Center for Food Safety 
University of Georgia 
Griffin, Georgia 



Editorial Board 
Francis F. Busta 

Director 

National Center for Food Production and Defense 

University of Minnesota 

Minneapolis, MN 



Bruce R. Cords 

Vice President 

Environment, Food Safety & Public Health 

Ecolab Inc. 

St. Paul, MN 



Catherine W. Donnelly 

Professor of Nutrition and Food science 
University of Vermont 
Burlington, VT 



Paul A. Hall 

Senior Director Microbiology & Food Safety 
Kraft Foods North America 
Glenview, IL 



Ailsa D. Hocking 

Chief Research Scientist 
CSIRO 

Food Science Australia 
North Ryde, Australia 



Thomas J. Montville 

Professor of Food Microbiology 
Rutgers University 
New Brunswick, NJ 



R. Bruce Tompkin 

Formerly Vice President-Product Safety 
ConAgra Refrigerated Prepared Foods 
Downers Grove, IL 



PCR METHODS IN FOODS 



Edited by 
John Maurer 

The University of Georgia, Athens 

Athens, GA, USA 




Springer 



Dr. John Maurer 

252 Poultry Diagnostic and Research Center 

College of Veterinary Medicine 

The University of Georgia 

Athens, GA 30602 

USA 



ISBN-10: 0-387-28264-5 
ISBN- 13: 978-0387-28264-0 

Printed on acid-free paper. 



2006 Springer Science+ Business Media, Inc. 
All rights reserved. This work may not be translated or copied in whole or in part without the written 
permission of the publisher (Springer Science+Business Media, Inc., 233 Spring Street, New York, NY 
10013, USA), except for brief excerpts in connection with reviews or scholarly analysis. Use in connection 
with any form of information storage and retrieval, electronic adaptation, computer software, or by similar 
or dissimilar methodology now know or hereafter developed is forbidden. 

The use in this publication of trade names, trademarks, service marks and similar terms, even if they are 
not identified as such, is not to be taken as an expression of opinion as to whether or not they are subject 
to proprietary rights. 

Printed in the United States of America. (SPI/SBA) 

987654321 

springeronline. com 



Preface 



This book will introduce non-molecular biologists to diagnostic PCR-based tech- 
nologies for the detection of pathogens in foods. By the conclusion of this book, 
the reader should be able to: 1) understand the principles behind PCR including 
real-time; 2) know the basics involved in the design, optimization, and imple- 
mentation of PCR in food microbiology lab setting; 3) interpret results; 4) know 
limitations and strengths of PCR; and 5) understand the basic principles behind 
a new fledgling technology, microarrays and its potential applications in food 
microbiology. This book will provide readers with the latest information on PCR 
and microarray based tests and their application towards the detection of 
bacterial, protozoal and viral pathogens in foods. Figures, charts, and tables will 
be used, where appropriate, to help illustrate concepts or provide the reader with 
useful information or resources as an important starting point in bringing 
molecular diagnostics into the food microbiology lab. This book is not designed 
to be a "cookbook" PCR manual with recipes and step-by-step instructions but 
rather serve as a primer or resource book for students, faculty, and other 
professionals interested in molecular biology and its integration into food safety. 



Table of Contents 



Preface 



v 



Chapter 1. PCR Basics 



Amanda Fairchild, M.S., Margie D. Lee DVM, Ph.D., 

and John J. Maurer, Ph.D 1 



Chapter 2. The Mythology of PCR: A Warning to the Wise 



John J. Maurer, Ph.D 



27 



Chapter 3. Sample Preparation for PCR 



Margie D. Lee, DVM, Ph.D. and Amanda Fairchild, M.S 41 



Chapter 4. Making PCR a Normal Routine of the Food Microbiology Lab 



Susan Sanchez, Ph.D 51 



Chapter 5. Molecular Detection of Foodborne Bacterial Pathogens 



Azlin Mustapha, Ph.D. and Yong Li, Ph.D 69 



Chapter 6. Molecular Approaches for the Detection of Foodborne Viral 

Pathogens 



Doris H. D'Souza and Lee-Ann Jaykus 91 



Chapter 7. Molecular Tools for the Identification of Foodborne Parasites 



Ynes Ortega, Ph.D 119 



Index 147 



Vll 



CHAPTER 1 



PCR Basics 

Amanda Fairchild 1 , M.S., Margie D. Lee 12 DVM, Ph.D., 

and John J. Maurer 1 ' 2 *, Ph.D. 

Poultry Diagnostic & Research Center 1 , College of Veterinary Medicine, 

The University of Georgia, Athens, GA 30602 
Center for Food Safety 2 , College of Agriculture and Environmental Sciences, 

The University of Georgia, Griffin, GA 30223 



Introduction 

Using Molecular Methods to Identify Microbial Pathogens 
The Theory Behind PCR 
Thermocycler Technology 
Detection 
Advanced PCR Technologies 

Real-Time PCR 

Multiplex PCR 

Terminal Restriction Fragment Length Polymorphisms 

Microarrays 
Design and Optimization of Diagnostic PCR as Applicable to Food 
Microbiology 

Systematic Approach to Creating Your Own PCR 
Access DNA Databases to Retrieve Sequences or Search for DNA Matches 
References 



INTRODUCTION 



The safety of your food supply is an important goal of the U.S. government and 
diagnostic food microbiologists across the country. Up to 5,000 deaths and 76 
million illnesses in the U.S. each year are associated with the consumption 
of foods laced with pathogenic bacteria (53), costing the U.S. an estimated 
$6.5-$34.9 billion annually (8). Even though bacteria have been shown to be the 
cause of the majority of food-related illnesses, the government does not have a 
mechanism for detecting and accounting for the losses due to other common 
foodborne pathogens, such as viruses and protozoa. Detection, identification, 
and quantification of foodborne pathogens are often made difficult by the low 
numbers of pathogenic organisms and interference from the food matrix that is 
being sampled. Bacterial pathogens of particular importance include Listeria, 
Campylobacter, Escherichia coli, and Salmonella (53), and the norovirus and 



Corresponding author. Phone: (706) 542-5071; FAX: (706) 542-5630; e-mail: jmau- 
rer@vet.uga.edu. 



PCR Basics 



hepatitis A virus are currently regarded as important foodborne viruses (44). 
However, since the advent of the polymerase chain reaction, finding these few 
pathogenic microorganisms in otherwise innocent looking provisions is 
becoming easier, mainstreamed, and second nature to many diagnostic labora- 
tories. The polymerase chain reaction (PCR) is a simple way to quickly amplify 
specific sequences of target DNA from indicator organisms to an amount that 
can be viewed by the human eye with a variety of detection devices. A goal of 
the present-day food microbiology research laboratory is to use the growing 
database of bacterial genomic information, made available by researchers 
mapping unique identifier genes of foodborne pathogens, to design monitor- 
ing systems capable of analyzing various incoming samples for hundreds of 
different organisms accurately and efficiently. 

Using Molecular Methods to Identify Microbial Pathogens. Prior to the 1980s and 
the advent of PCR, identification of microbial pathogens relied on bacteriological 
methods to enrich and isolate the organism from clinical or food sample, and sub- 
sequent biochemical and/or immunological tests to confirm the microbe's identity. 
During the past 30 years, we have gained tremendous insights into how microor- 
ganisms spread and cause disease. In several instances, pathology associated with 
many bacterial illnesses is attributed to a single gene (4, 9, 18, 75, 97). Other 
pathogens like Salmonella are more complex, requiring coordinate regulation of 
several virulence gene sets to cause disease (49). Therefore, the organism's genetics 
or genotype dictates its ability to cause disease, or the severity of the illnesses asso- 
ciated with it. Many of these virulence genes are unique to the pathogen and sub- 
sequently make useful markers for identifying said pathogen (37, 41, 86, 95). With 
several bacterial genomes completed, we now know the genetic basis for pheno- 
types that have been useful markers for distinguishing pathogens from closely 
related commensals that inhabit the same niche. By identifying gene(s) associated 
with phenotype (e.g., 0157 serotype), we have identified a marker with greater 
specificity than afforded by the actual antigen itself, especially when confronting 
cross-reactivity and false positive associated with the immunological test (50). 
Although quite specific, the early molecular-based method, DNA: DNA 
hybridization had limited utility due to its limited sensitivity, time length, or safety 
issues associated with the use of radioactive probes (77, 80). Even with the intro- 
duction of nonradiometric methods for detecting hybridization of probe to its tar- 
get gene, there was still the limitation of sensitivity, [i.e., the ability to detect the 
fewest cells possible (80)]. How could one amplify the target gene enough to detect 
its presence in the sample contaminated with organism X? 



THE THEORY BEHIND PCR 



The concept of the PCR was first described by Panet and Khorana in 1974(64) 
and owes its name to Dr. Kary Mullis and colleagues, who developed the process 
over the course of 4 months in 1983 at the Cetus Corporation. While driving 
down Highway 128 in Mendocino County, California, Dr. Mullis let his mind 



The Theory Behind PCR 



slip back to the lab and a burning question that could not escape his mind. How 
could someone go about reading the sequence known as DNA, the language of 
our genes and blueprint for our existence? Like perfectly crafted ball and socket 
joints, the oligonucleotide base pairs within the DNA molecule bond to one 
another as the entire length of the ladder-shaped molecule twists into a 
corkscrew shape. Dr. Mullis referred to DNA structure as something like a mass 
of "unwound and tangled audio tape on the floor of the car in the dark" (58). 
In 1953, Drs. Watson and Crick had mentioned the biological significance of 
the DNA molecule with its complementary base pairing that suggested "a pos- 
sible copying mechanism" for genetic material (91). One strand of DNA could 
be a template for the formation of a new complementary chain, and in the end 
you could have two DNA ladders, identical in every way. Dr. Mullis' develop- 
ment of PCR has extrapolated the copy machine theory one step further. He 
stated this simply as an analogy to a "'Find' sequence in a computer search" 
(58). This technique would have equivalent power to the latest computer dis- 
playing results of finding a document that consisted of just one word taking up 
20 kilobytes of space on a hard drive the size of 150 Gigabytes littered with files 
of different types and sizes; like finding the code for blue eyes — and that code 
only — within the code that sums up every single trait for a person. 

The second aspect Dr. Mullis had to account for in this process would be the 
ability of the chemical program to display the located sequence in a large 
enough fashion to be detected by the human eye. Dr. Mullis knew that if he 
could produce a short piece of DNA to find a sequence flanking a gene of inter- 
est and then start a process that could make the sequence reproduce itself over 
and over (hence a chain reaction), the concept of PCR could be realized. After 
all, it was already known that DNA innately makes a copy of itself when cells 
divide, so that each daughter cell can have a copy. If he introduced his "find" 
search strings, or primers, into a tube with DNA encouraged to uncoil from its 
natural double helix by heating and a biological glue (polymerase) to attach 
deoxynucleoside triphosphates (dNTPs) to the freshly uncoiled DNA at the 
location, where the primers bonded, a new copy of the DNA would be pro- 
duced as specified by the primers that included the gene(s) of interest as the 
temperature decreased. The DNA polymerase that is used in some PCR reac- 
tions is made from the bacteria Thermus aquaticus, which was found originally 
in Yellowstone thermal water reservoirs. It is stable in temperatures above that 
which denatures DNA, making it a perfect enzyme for the job of attaching free 
dNTPs to make a new strand of DNA. There are other polymerases available 
that can actually proofread the addition of dNTPs, so there will be no errors 
made in the synthesis of longer PCR products. After each cycle in a PCR assay, 
the amount of DNA present doubles, so repeat the cycle and there are 4 copies 
of the gene, repeat again and there are 8 copies. With 30 cycles of this process, 
there would theoretically be just over a billion copies of the sequence in ques- 
tion (2 30 = 1.07 billion). Find a way to tag each copy to make it visible to the 
human eye, with more copies making a stronger visible signal and you have 
proof of the presence of the small sequence embedded within the large DNA 
molecule you started with. A widely used method for viewing PCR products 



PCR Basics 



involves running them on an agarose gel, staining the gel with ethidium bro- 
mide, and observing and photographing the gel on ultraviolet (UV) light source 
(56). The process is relatively fast, dictated by the amount of time it takes to 
heat the DNA strands until they will separate, the time to reduce the tempera- 
ture so the primers bind to the single-stranded DNA, and the time allowed for 
the polymerase to add individual deoxynucleoside triphosphates to extend the 
forming DNA molecule. Dr. Mullis stated that scientists claimed that PCR 
made DNA research boring (57). Even though PCR is often considered "cook- 
book chemistry" because of its simplicity, his suggestion could not be further 
from the truth. For example, PCR has been one of the most important genetic 
tools available to those mapping the human genome and for those attempting 
to detect pathogenic bacteria, viruses, and protozoa. The PCR has made its way 
from the research lab to forensic and diagnostic laboratories worldwide. There 
have been considerable efforts to validate and standardize this tool (17, 38); to 
become a normal routine task/service performed by reference laboratories 
(3, 48, 76), and clinical diagnostic and food microbiology laboratories (1, 33, 38, 
45, 54, 79, 87). 



THERMOCYCLER TECHNOLOGY 



Since the technique of PCR was developed, there have been many evolutions of 
the equipment that makes the process possible, based on the concept that 
strands of DNA denature, or unwind, and anneal, or wind again back into the 
helical corkscrew, in response to fluctuations in temperature. The first success- 
ful PCR reaction took place using water baths at the appropriate temperatures 
for each step in the procedure, with the technician moving vials by hand from 
bath to bath at the appointed time, for 30 or more cycles to get adequate 
amounts of DNA copies that could be detected. Nowadays, thanks to automa- 
tion, PCR reactions can be set up in thermocyclers that over the course of min- 
utes to a few hours reliably yield high numbers of a specific DNA sequence if 
present in a sample. 

The standard thermocycler uses a large heating block, into which microcen- 
trifuge tubes are placed. This type of thermocycler through its computer con- 
trols the heating and coolings of the blocks through the cycles of each reaction. 
Sometimes an oil or wax overlay is put on the samples within the microcen- 
trifuge tubes to keep the sample from escaping the bottom of the tube during 
the heating for the denature step of the PCR reaction. This type of machine is 
not as desirable because of the time it takes to heat and then cool the entire 
block to the appropriate temperature within each cycle. The time required for 
the heat block to uniformly reach each temperature coupled with the slow heat 
transfer rates to the microcentrifuge tubes makes this type of thermocycler vir- 
tually inadequate with today's demand for high-speed accurate amplification of 
PCR products. 

The RapidCycler, manufactured by Idaho Technologies, is an example of 
equipment designed to provide the quick temperature cycling necessary for 



Thermocycler Technology 



PCR reactions. This type of thermocycler uses heat transfer through blasts 
of high-velocity hot air to accomplish the temperature transactions from the 
initial heating of the DNA sample through the annealing of the primers and the 
extension of the new double strand of DNA by the polymerase. There is overall 
temperature uniformity within the cavity of the reaction vessel and rapid heat 
exchange within the sample because individual samples are loaded into micro- 
capillary tubes or thin walled microcentrifuge tubes for the reactions to take 
place. This also allows for a smaller overall volume in each reaction tube, thus 
saving valuable amounts of reaction components such as polymerase and 
primers. After the PCR cycles are complete, the samples are loaded on an 
agarose gel that contains ethidium bromide, and viewed under a UV light source. 

A gradient thermocycler allows the clinician to optimize each of the three 
temperatures needed for the denaturing, annealing and extension of new DNA 
products. Optimization might be required if an existing PCR cycle program 
cannot be located for detection of a particular gene sequence. Optimizing the 
PCR reactions is critical to the success of the production of amplicons and is 
not always the easiest thing to do. The melting temperature can be calculated 
for the primers when they are made, but the denaturing and annealing temper- 
ature of the cycle might have to be determined by educated guess with some 
trial and error, possibly rerunning the same reaction with many different tem- 
peratures before the best fit temperature is found. Luckily, most machines have 
the ability to reach the different temperatures at the same time (thus the gradi- 
ent), so the reactions are run at the same time with results from each tempera- 
ture trial collected at the same time. The block used to hold the samples in this 
type of machine can be programmed to heat over a gradient of about 20° C 
range with the annealing temperature for the PCR reactions increased by an 
increment of 1 or 2°C. 

Another thermocycler type offers PCR product detection at the same time 
as each cycle of the PCR reaction progresses. It allows the technician to track 




Figure 1.1. Example of real-time PCR output graph showing amount of DNA 
sequences produced over 40 PCR cycles. 



PCR Basics 



the increase in products during a PCR reaction as displayed on a graph 
(Fig. 1.1). Higuchi and colleagues introduced this feature, dubbed "Real-Time" 
PCR, and described how the number of cycles necessary to produce a detectable 
fluorescence was "directly related to the starting number of DNA copies" in a 
sample (32). There are a number of companies that offer this technology, which 
combines the rapid cycle polymerase chain reaction with fluorescent detection 
of amplified PCR product in the same closed vessel as the reaction mix. The 
primers are usually labeled with fluorogenic probes, or a DNA-binding dye is 
included in the PCR reaction, which fluoresces under light emitted at a certain 
wavelength. 



DETECTION 



Detection of the PCR product or "amplicon" can be accomplished several 
ways. Following PCR, the sample is loaded into an agarose gel, and the DNA 
fragment(s) or amplicon if present in the sample is separated by electrophoresis 
based on size. Molecular weight, DNA standards are included to estimate size 
of amplicon(s) present in positive samples and positive control. The agarose gel 
and electrophoresis buffer contain a dye, ethidium bromide that binds double- 
stranded DNA and fluoresces upon excitation with UV light. This dye is used 
to visualize the DNA in an agarose gel. As the primers bind to fixed position 
within the target sequence, the expected size for our PCR product/amplicon is 
the distance between the forward and reverse primers. For example, if forward 
and reverse primers bind target gene X at positions 850 and 1000, respectively, 
then one expects to observe an amplicon of 150 bp for positive control or any 
sample that bears organism that contains gene X. The size of the amplicon is 
extrapolated from the DNA standards included in the gel. The sample MUST 
produce an amplicon of expected size predicted for the primers used and cor- 
robated by the positive control before it can be considered positive by PCR. 
There is an inverse linear correlation between the log 10 size of the DNA frag- 
ment (bp) and the distance migrated by the DNA fragment in the agaraose gel. 
The smaller the DNA fragment the farther it migrates through the agarose gel 
during electrophoresis. Therefore one can estimate the amplicon's size from 
DNA standards included with the agarose gel. As most PCRs produce small 
size amplicons (100-1,000 bp), one must use DNA standards that accommo- 
date this size range and agaraose concentration (1.5%) that resolves small DNA 
fragments sufficiently to accurately determine the size for DNA band X. The 
PCR result is recorded photographically with a polaorid or digital camera with 
the appropriate lens filters and exposures for capturing images illuminated by 
the UV light. 

Detection systems are slowly moving towards nongel methods for detecting 
and recording PCR results. Enzyme-linked immunosorbent assay (ELISA) has 
been developed for detecting amplicons (37). In the PCR reaction mix, the stan- 
dard nucleotides have been substituted for chemically "tagged" nucleotides 
(e.g., dioxygenin or DIG). During PCR, the "DIG'-labeled nucleotides are 



Detection 



A 



dsDNA 



94°C 



Heat (Denature DNA) 




regular nucleotide bases 
Dig - labeled nucleotide 

DNA polymerase 
primer 



ssDNA 



30 



C~ ft*) °C^\a P runer Annealing 



o 



m 



w 



65°C~ 72 °C / DNA polymerase(Extension) 




m 



LLi 



A 



A A 



A 



V V 



V V 



B 



A A 



A A 



VV V V 



V 



Denature 

94°C 

Anneal 

50 °C 



► 



A A A A 



A 




Enzymatic 
detection 



AA 

oo 




digoxigenin 
: biotin 
: strepavidin 

IJ : p -nitrophenyl phosphate 

anti-digoxigenin -alkaline phosphatase 

Figure 1.2. PCR-ELISA. Digoxygenin (DIG) labeled, nucleotides are incorporated 
into amplicon during PCR (A). An internal, 3' biotinylated oligoprobe anneals to 
denatured, single-stranded amplicon following PCR. The strepavidin, coating the 
wells, binds to the biotin moiety of the oligoprobe and thus captures the amplicon. 
The amplicon is then detected using anti-DIG antibody enzyme conjugate (B). The 
oligoprobe adds additional specificity to this PCR test. 



incorporated into the amplicon as it is synthesized (Fig. 1.2A). Following the 
last round of PCR, the sample is denatured and allowed to anneal with 5', 
biotin-labeled, internal oligonucleotide. This oligoprobe binds to the comple- 
mentary sequence present within the amplicon. This amplicon-oligoprobe 
hybrid is captured in strepavidin-coated 96 well microplate through the interac- 
tion of the biotin group with the strepavidin (Fig. 1.2B). The bound amplicon 
is visualized colormeterically using anti-DIG antibody enzyme conjugate, usu- 
ally either horseradish peroxidase or alkaline phosphatase. The advantage of 



8 PCR Basics 



this "PCR-ELISA" is that it easy to scale-up for high throughput of samples 
and lends itself quite well to automation. Another nongel method for detecting 
PCR amplicons involves detecting fluorescent dyes bound to or released from 
the amplicon using a fluorometer. This detection method is the basis of real- 
time PCR discussed below. 



ADVANCED PCR TECHNOLOGIES AND MICROARRAYS 



Real-Time PCR. Real-time PCR technology is based on the ability for the detec- 
tion and quantification of PCR products, or amplicons, as the reaction cycles 
progress. Higuchi and colleagues introduced this technology (32) and it is made 
possible by the inclusion of a fluorescent dye that binds the amplicon as it is 
made (Fig. 1.3 A). There are several ways to detect the PCR products under flu- 
orescent detection. In TaqMan PCR, an intact, "internal" fluorogenic oligo- 
probe binds target DNA sequence, internal to the PCR primer binding sites. 
This oligprobe possesses a reporter dye that will fluoresce and a suppressor dye 
known as a quencher that prevents fluorescent activity via fluorescence reso- 
nance energy transfer (FRET). After each PCR cycle, when the double- 
stranded DNA products are made, a measure of fluorescence is taken after the 
fluorogenic probe is hydrolytically cleaved from the DNA structure by the 
exonuclease activity of the Thermus aquaticus DNA polymerase (29, 36). Once 
cleaved, the probe's fluorescent activity is no longer suppressed (Fig. 1.3B). 
FAM (6-carboxyfluorescein) and TAMRA (6-carboxy-tetramethyl-rhodamin) 
are most frequently used as reporter and as quencher, respectively. This PCR is 
often refered to as 5'exonuclease-based, real-time PCR or TaqMan PCR (55). 
When a DNA-binding dye is used, as more DNA copies are made with each 
successive cycle of the PCR, they are all bound, or intercalated, with the dye, 
and the fluorescence increases (Fig. 1.3A). SYBR Green I is the most frequently 
used DNA-binding dye in real-time PCR. 

Two additional advanced methods of amplicon detection are hybridization 
probes and molecular beacons. The hybridization method uses fluorescence reso- 
nance energy transfer from one probe to another after annealing of the primers 
to the template strand of DNA. One probe has a donor dye at the 3' end of the 
oligonucleotide and the other probe has an acceptor dye at the 5' end. When both 
probes anneal to the target sequences, they are situated to have the dyes adjacent 
to one another one base apart. While in that configuration, the energy emitted by 
the donor dye excites the acceptor dye, which emits fluorescent light at a longer 
wavelength. The ratio between the two fluorescent emissions increases as the PCR 
progresses and is proportional to the amount of amplicons produced. Molecular 
beacons are short segments of single-stranded DNA. They use a hairpin shape to 
facilitate quenching of the fluorescent signal until the probe anneals to the com- 
plementary target DNA sequences, produced from PCR. 

Some advantages of real-time technology include high sensitivity with the 
use of an appropriate probe or DNA-binding dye, ability for detection of rela- 
tively small numbers of target DNA copies, and ease of quantification because 



Advanced PCR Technologies and Microarrays 



A 



dsDNA 




940C 



\ 



Heat (Denature DNA) 



— : regular nucleotide bases 



DNA polymerase 



primer 




fluorescent dye 



ssDNA 



300 o 650 C 



\ 



Primer Annealing 



TP 



1*1 



B 



dsDNA 



650C- 720 C 



DNA polymerase (Extension) 

UV 



0= 



G= 







T 



^1 M 



Nffi \y [o^ 





•■k k^l xK 

-^s >is >-■- 

XN M^ X> 

frfr [S?| >p 



^l 



H 



94°C 



Heat (Denature DNA) 



— : regular nucleotide bases 
+ : internal TAQMAN probe 



: DNA polymerase 



: primer 




ssDNA 



▲ : quencher 



30°O 65° C 



Primer Annealing 



m 



fluorescent dye 



/, 



MT1 



65°C~72°C 



♦ 



w 



DNA polymerase (Extension) 



a 



-^ 



n 



■/. 



♦ 




Figure 1.3. "Real-time" PCR detection of amplicons. Using fluorogenic dyes, ampli- 
cons can be detected by a fluorimeter as they are synthesized with each PCR cycle. 
Intially, a fluoresent dye, SYBR Green (A), was used to detect the amplicons. In this 
PCR, SYBR Green binds the double-stranded, DNA amplicon and fluorescences 
upon illumination with ultraviolet light (UV). Subsequently, real-time PCR was devel 
oped using an internal oligoprobe for detecting the amplicons. In TaqMan PCR (B), 
the oligoprobe contains a fluorscent marker and a chemical group that quenches fluo 
rescence of the oligoprobe until the dye is liberated by 3' exonuclease activity of the 
Taq DNA polymerase. This can only occur if the oligo binds the complementary 
sequences present in the target gene and amplicon. 



10 



PCR Basics 



of the lack of post-PCR detection measures. Molecular beacons can even be 
used to detect single nucleotide differences (27). Disadvantages of real-time 
PCR technology lie with the detection of the amplicons. If the DNA-binding 
dye is used, then any double-stranded product is labeled and fluoresces, includ- 
ing primer-dimers, and nonspecific amplicons, whether they are close to the tar- 
get DNA in sequence or have erroneous secondary structure. The effectiveness 
of the fluorogenic probe is also influenced by the creation of primer-dimers, 
and both methods of detection are susceptible to less than optimal design of 
primers for use in the PCR and primer concentration in the master mix. To 
compensate for the unspecific binding of the DNA dye, real-time PCR equip- 
ment has the capability of running a melting curve after the PCR assay, which 
increases the temperature of the vessels in tiny increments until the fluorescence 
is lost due to the DNA denaturing. When the melting temperature of the target 
DNA sequence is reached, a sharp loss of fluorescence will be recorded. If addi- 
tional losses are recorded, there may be PCR contamination, or the parameters 
of the PCR assay were not stringent enough, such as suboptimal primer design 
or temperature choice for the program. This feature of the equipment makes 
DNA-binding dyes a feasible and often cheaper alternative to the other meth- 
ods available. The melting temperature of the amplicons should be known when 
designing primers for the assay and are usually referenced when programming 
the annealing step of the PCR reaction. Accurate primer design and optimiza- 
tion of the PCR reaction conditions for the primers are required in any PCR 
application, but especially with real-time technology. 

Multiplex PCR. Multiplex PCR is a way to amplify two or more amplicons in a 
single PCR reaction. For multiplex PCR, each primer set is designed to its tar- 
get gene to amplify a PCR product of a size unique to the target gene. To per- 
form a multiplex PCR, the concentrations of primers, Mg 2+ , free dNTPs and 
polymerase are altered to allow for the synthesis of the genes of interest, while 
the PCR reaction temperature parameters are optimized to the best average for 
amplicon production for all primer sets. This technique saves time and labor 
since more than one target DNA sequence is detected for each reaction, but 
might not be optimal if the PCR products are close in size and detection 
requires viewing an agarose gel stained with ethidium bromide. In a single PCR 
reaction, one can determine the identity of the organism (28, 39) or genotype 
(21), as the amplicon(s) size is unique to specific organism or gene. Therefore, it 
is possible to detect multiple pathogens in a sample from a single PCR test (65). 

Terminal Restriction Fragment Length Polymorphisms (TRFLP). We can use 

PCR to characterize microbial communities and identify member species using 
a single PCR primer set. This PCR targets the 16S rDNA, a gene that is uni- 
versally conserved among all bacterial species and amplifies a single ~1,500 bp 
amplicon. We can resolve diversity of 16S rDNA amplicons generated from 
this PCR using restriction enzyme(s) that recognize restriction sites within 
genus or species specific sites within this gene and produce DNA fragments, 
whose size corresponds to a specific genus or species (46). This PCR involves 



Advanced PCR Technologies and Microarrays 



11 



using universal 16s rDNA primers in which one of the forward or reverse 
primers is fluorescently tagged (Fig. 1.4). Following PCR, the amplicons are 
digested with a restriction enzyme and subsequently loaded onto capillary bed 
of an automated DNA sequencer. This method has been refined and applied to 
automated DNA sequencers to resolve minor (x bp) differences between DNA 
fragments, monitoring and measuring fluorescence associated with the various 
sized DNA fragments as they elute from the sequencers' capillary bed (Fig. 1.4). 
Fluorescently labeled molecular weight standards are included to calibrate col- 
umn in order to demarcate and identify the molecular weight for each DNA 
fragment separated by on the sequencer's capillary column. Each peak corre- 
sponds to specific genus/species present within the sample. The identity is deter- 
mined from comparisons to an established database of restriction fragments 
predicted from 16S rDNA sequences (47). This database can be generated in 
house, from cloning and sequencing your 16S rDNA library or comparing it 
against an ever-expanding Web-based 16S rDNA database (Michigan State 
University Center for Microbial Ecology; http:ll35.8.164.52lhtmllTAP- 
trflp.html\ 47). The latter has a tool for analyzing your TRFLP profile against 
this database, for various restriction enzymes. Depending on the restriction 
enzyme used, one may not be able to resolve various species or genera with a 



A 



PCR 

(16 S rDNA) 



^B Restriction ^B Separate DNA fragments 
r enzyme digest r column chromatography 





Peak's position: Genus or species 





Fluorescence 








A 



o 



( = 



Elution Time 
DNA fragment size) 



B 



Change in composition of the microbial community 



Sample 1 



Sample 2 



Fluorescence 



Fluorescence 




A 



OU£l^ 





o 



Elution Time 







Elution Time 



Figure 1.4. Characterizing microbial communities and identification of pathogens in 
foods from terminal restriction fragment length polymorphisms (TRFLP) of total 
microbial community 1 6S rDNA. (A) Concept behind TRFLP. (B) Interpretation of 
TRFLP. 



12 PCR Basics 



single restriction enzyme. This is because they produce the same size DNA 
fragment with restriction enzyme X. It may take a number of different TRFLP 
profiles of the same community, generated with different restriction enzymes, 
before genera and/or species differences can be resolved (47). This method is 
currently used in assessing stability and structure of microbial consortiums, 
and it has been recently applied to analyzing changes in the community struc- 
ture of gastrointestinal microflora in response to diet or probiotics (34, 42). 
TRFLP can also identify signature peaks for microbial pathogens (14, 60), 
where differences in 16S rDNA can be discerned between them and closely 
related commensal organisms, exceptions E. coli vs. Salmonella (61). 
Theoretically, TFLP and other molecular ecology tools (e.g., DGGE) will 
prove useful towards analyses of microbial communities present in foods, gas- 
trointestinal tracts of food animals, probiotics and starter cultures and deter- 
mine the impact certain food processes have on their composition, with regards 
to the food's safety for consumers. 

Microarrays. Macroarrays, microarrays, high-density oligonucleotide arrays, and 
microelectronic arrays are all part of a new technology that allows one to screen 
for gene(s), sequence(s) or specific mRNA among myriad of possible sequences 
or genes in a single test (22). DNA hybridization arrays are based on specific posi- 
tioning of a myriad of oligonucleotides or PCR amplicons, representative of a 
complete bacterial genome, on nylon membrane (macroarray), glass slide 
(microarray), or electronic microchip (microelectronic array). Each position on 
this solid support contains an oligonucleotide or PCR product unique to a par- 
ticular gene. Total mRNA or genomic DNA from an organism is fluorescently or 
radioactively labeled and used in hybridization with solid support. The bound 
oligonucleotides or amplicons on the solid support serve to capture labeled probe 
in the RNA: DNA or DNA: DNA hybridization (Fig. 1.5). The labeled nucleic 
acid hybridizes to the position or "spot" on the solid support that contains com- 
plementary sequence for the labeled probe to bind. Identity of gene or sequence 
relates back to the original positioning of the oligonucleotides or amplicons on 
the solid support (Fig. 1.5). This technology has already been applied towards the 
study of bacterial gene expression (30, 71), host-microbe interactions (15, 73, 84), 
bacterial evolution and population genetics (6, 11, 23, 70, 85, 96). Currently, 
microarrays have been applied towards PCR-based detection of pathogens in the 
environment (2, 43, 65, 88). At present, this methodology is experimental, per- 
formed primarily by research laboratories. However, advancement in technologies 
and manufacturing will someday make microarrays affordable and practical for 
use in diagnostic setting, as PCR has now become. 



DESIGN AND OPTIMIZATION OF DIAGNOSTIC PCR 
AS APPLICABLE TO FOOD MICROBIOLOGY 



To perform PCR in any microbiological application, the DNA sequences of an 
infectious agent must be known, and the target sequences must be unique to the 
organism(s) to be detected. For example, if a food sample is suspected to be 



Design and Optimization of Diagnostic PCR as Applicable 



13 



A 




























1 




! * 




J 










i 








D 


1 


2 




J 

















c 



T 



\ 



\ 



PCR 



D 



iAGUui..f: 



B 










A 


1 


£ 


3 




Gene A 


Gene B 


Gen«C 




ATTACT ACT - r l:-, v -^TjrJ- f 

AITACTAf I blAAJpEEu: 
■l i I4L ii*i.7 37AACC3AC 




D 


1 


: 


J 








Gene D 

TCATAAATaCT 
TCaTaaaTaCT 

teATAAATACT 


Gene E 

CCaGCAACCCT 












TTATAjjTiJT 


crjior.i.Anrrr 











* TAW^AC 

TlTCCfAC TaC4CCaC 

T-VGCf-r^r , TAWGCAC 

TACCCCAC TAQUUTAC 

i-. 



TAEGGT *r TACGGrAC 

TAWJCCAC i T*««*C 

TA««AC TAGCGCU 



& 



E 



T 




> SAiLLULAL 
. / ATCCCC 



4- 



F 



EJLbbLi'JH. 
AUTCCCCIC 



EAbbbb-LAi 

at-: err tit 



\ 



J* 



Gene A 

ACTACTf 

iV. 

Ali.l>L I.V. j 

ATTaCTACI 





1 

Gene A 



1 



3 






"^ .t 

2 

Gene B 

flTAAAT^W: 
cn*AAcrcAc: 

"USSSSSL 

GTAaCCGaC 

2 
Gene ft 

TCATAAATaCT 
TCaTaMTACT 

TCATA4ATACT 

H/ATAAATACT 



-- 



3 
Gene C 

* 




ICCCCCTC 



Gene R 

GTACCAArrrr 

CCACGAACCTT 
U'AIWAA4TC'I 



2 
GeneB 



ATTAeTACT 




«tUC»At 


aITaCTaCT 




CTAAJtt&l£ 


a FT ACT ACT 




CTAaCCSaC 


ATTACTACTF 




GTAAC^GAC 


AmrrAtrr 




GTAACPCAr 



2 
GeneB 



Cease D 



Gene C 

r 

!*TCeCCGT5 

T 

ATnrrcGTG 



3 
GeneC 




3 
Gene L 



Figure 1.5. Microarrays. Specific oligonucleotides or PCR amplicons are spotted onto 
defined region on glass side or nylon membrane (A, B). The positioning of this cap- 
ture probe on this solid matrix defines gene or signature sequence for organism X. If 
any of the genes present on slide or membrane are present, then it will be amplified 
during PCR and labeled with fluorescent nucleotide (C) and subsequently bound to 
the complementary sequence present on the solid support (D, E). Position of fluores- 
cent signal (F) identifies gene or organism present in the sample. 



contaminated with bacteria "X", such as E. coli 0157, then a PCR can be used 
to determine the presence of the bacteria if there is a gene that only that bacte- 
ria possesses, such as an identifier gene "XI", or in the case of E. coli, the 0157 
antigen biosynthesis gene (50). If the gene was found in more than one bacteria 
type, say gene "XY4," additional PCRs would have to be performed to separate 
bacteria that harbor that gene (bacteria X and bacteria Y) by looking for a 
unique identifier gene of the target bacteria X, but at additional work, cost and 
time for the clinician. The case of identifying E. coli 0157:H7 might require a 
multiplex PCR approach because of the closely similar genes of the different 
antigen subunit serotypes (21, 26). There are many genes that are shared within 
the same genus and species of bacteria, such as the genes shared among patho- 
genic E. coli strains. Instead of differentiating between bacteria X and Y, the 
researcher is met with finding a uniqueness of bacteria XI versus bacteria X2. 



14 PCR Basics 



Systematic Approach to Creating Your Own PCR. The development and valida- 
tion of PCR is a long and arduous journey from concept to application. It 
involves identification of a candidate marker or allele for pathogen X, whose 
distribution among microbes is strongly associated with the pathogen in ques- 
tion, and the cloning and sequencing of the cognizant gene(s) associated with 
the marker or allele (50). For antigenic variable, surface proteins like flagellin, 
PCR, using primers that recognize conserved sequences flanking sequence vari- 
able regions (19, 81, 83), and subsequent sequencing of the PCR amplicon has 
identified sequences unique to serovar (26, 31) or pathogen (63), which subse- 
quently led to development of serovar or pathogen-specific PCR (26, 31, 63). 
Design and development of PCR is the pursuit of researchers and if a PCR is 
available, commercially or otherwise, it is best to adapt this PCR to your lab 
than having to start from "scratch". Therefore, for most our readers, the inter- 
net, www.ncbi.nlm.nih.gov and the PUBMED search is the best place to look for 
PCRs and protocols for screening foods. 

In the past, PCR design was based on gene(s) or DNA sequences obtained 
from screening plasmid clone (50) or transposon libraries (5, 16) for relevant 
marker, subcloning and sequencing DNA inserts. This approach took consid- 
erable time and resources. Now, in less time, we can sequence the entire genome 
of a single bacterial species, and spend the remainder of our time at the com- 
puter annotating its sequence, searching for signature sequences unique to 
pathogen X. In 1995, the first organism was completely sequenced (20). Since 
that time, 91 bacterial genomes of several, important human pathogens have 
been sequenced, annotated, and published (www.tigr.org; accessed 2/16/05), 
including several foodborne pathogens (10, 12, 35, 52, 59, 66, 69, 74, 82, 92). 
From comparisons of these bacterial genomes, especially between closely 
related commensals and pathogens, several regions within the chromosome 
have been identified that appear to be unique to organism X that is tied to its 
virulence (7, 62, 93), or metabolism (69). With the growing number of bacter- 
ial genomes present in public accessible DNA databases, identification and 
design of PCR for organism can be done in silico, on your desktop computer. 
A priori, of course is that organism X's genome has been sequenced and 
accessible to the user. With advances in PCR and in silico analyses of bacter- 
ial genomes, we can amplify, clone and sequence large regions of the bacterial 
chromosome to quickly identify target DNA sequences for PCR primer design 
(89, 90) 



Access DNA Databases to Retrieve Sequences or Search for DNA Matches. 

For the researcher, the most important resource, second only to the library 
and PUBMED, is the DNA database, GenBank at the National Center 
for Biotechnology Information, National Institutes of Health, Bethesda, 
Maryland. This database can be accessed via the internet at the following 
Website: www.ncbi.nlm.nih.gov, go to the ENTREZ selection at the top of the 
page, and then go to GenBank on the next Web page. One can then search the 
database of sequences by typing in keywords or combination of words for a 
specific organism, serovar, or gene(s). Prior to this search, it is important to do 



Design and Optimization of Diagnostic PCR as Applicable 15 



your initial research in the library, so that your GenBank search is refined and 
specific to pull out select sequences from the millions, probably billions, of data 
base entries present at this Website. The next step is to access a specific 
GenBank accession, for this exercise we will examine the Salmonella enterica 
Typhimurium LT2 genome at NCBI, GenBank Accession # NC 003917 
(Fig. 1.6A, B) and search the annotated genome for the invasion gene invA (24, 
25) by using the search function in Netscape Navigator for the word "invA" to 
find the beginning and end of each gene's open reading frame (ORF) 
(Fig. 1.6C, D). We write down this information and scroll down to the complete 
sequence to find and copy these sequences (Fig 1.7 A). We can paste this 
sequence for the time being into MSWORD, MS WORDPERFECT, or WORD 
Notepad and save this file, giving it the organism/gene name. The first three 
nucleotides should start with ATG, the start codon or rare start codon GTG, 
and end with TAA, TAG, or TGA, the stop codons. It should be noted, espe- 
cially with genome sequences, the gene may be in the opposite orientation on 
the chromosome, requiring inversion of DNA sequence and transcribing the 
opposite DNA strand to identify start and stop of our ORF Many DNA soft- 
ware analysis programs can do this for us. We chose the ORF rather than 
flanking or intergenic regions, because we expect greater selection pressure 
and less chance for sequence divergence among strains of organism X than 
these intergenic regions. This is especially important if we are to identify all 
members of organism X. Now that we have these sequences, we need to deter- 
mine, in silico, whether these sequences are unique to genus Salmonella and 
specifically, the serovar Typhimurium. This can be determined going to 
BLAST on www.ncbi.nlm.nih.gov Website. Click on BLAST and under 
Nucleotide, click on "nucleotide-nucleotide BLAST (blastn) (Fig. 1.7B)." This 
will take you to a new site within NCBI that has a box beside "Search". Paste 
your sequence into this box, and click on the BLAST button (Fig. 1.7B). On the 
next page, select under the "Format" section, the box titled "or select from" and 
chose "Bacteria [ORGN]" and click on the FORMAT button (Fig. 1.7C). 
Allow the BLAST search time to search the database. The time it takes for the 
search is dependent on gene and the amount of "traffic" at this Website; it is a 
very popular site with researchers. The results are returned, outlining how many 
matches there are to your gene sequence. As of 8 February 2005, there were 
222 matches with the closest matches, (>90%) to S. enterica invA representing var- 
ious serovars. Other matches are identified, most notably in homologues, genes 
with similar function, present in Escherichia coli. This is expected, as invA is part 
of the type III secretion system present in many human and plant pathogens (40). 
More importantly, the BLAST results identify for us region of the invA sequence 
to focus on in our primer design. This database search using BLAST is the same 
approach one would use in analyzing DNA sequence generated from the sequenc- 
ing of plasmid clones or PCR amplicons. There are however, no guarantees 
that your gene or sequence will prove useful as a diagnostic marker for organism 
X, based solely on this database search. You find only what is available on the 
database, at the time of your search. It is therefore important experi- 
mentally to determine the distribution of your candidate gene or allele among 



16 



PCR Basics 




* ■ . . , 

s |i || 

.- ,*-£ i 

lift"! 

i I -li I 

tSai -J 1*1- U 

c J S * -c ■ I ■ 

1 1.11,1, liiHHjfl! Jl 



■ 
p 



ill 

alSS 



z 

- 






! 
5 I 

SIS 



2 



• H 

O 



2 o 






^3 



^ 

> 

C 



^*4 






43 






• H 

Q 






r ^3 



• l-H « 

PQ £> 






n rj 

a* 

PQ 

TO 5-H 

<J GO 

PQ 

a 



pa 



p m 



o 
.2 * 



PQ 

5-H 

<D 

-4- > 

u 

o 






gjD 



O 

ca 
<D 
O 
O 

< 
a 

PQ 

o 

a 

o 

bX) 

a 



CO 

Q 
U 

O 

a 
cr 

C/5 



Design and Optimization of Diagnostic PCR as Applicable 



17 



PQ 



On 
CO 




MS 



1 
f 

I 



I 



i 



R 



2 



J II 

i I 1 

■ 1 

Mi 



r. ■ 1* 



71 



■ 1 



b L 




ft 

PQ 

o 

a 

o 

o 

cr 

C/5 

bX) 

a 

• H 

o 






^ 

^ 

C 



*•<> 






bX) 

a 

P. 
O 

u 



o 

GO 



• i-H 

bX) 

a 

O 

C/5 

bX) 

O 

o 

a 

o 

M 

O 

cr 

C/5 

a 

H 

GO 

PQ 

bX) 






PQ 

bX) 

o 
o 

o 

a> 

^— » 

o 

PQ 

-+- » 

u 

o 






3 
gjD 






bX) 

fl 

o 

c/5 

bX) 

O 

o 

a 

o 

M 

O 

cr 

C/5 

a 

H 

GO 

H-l 

PQ 



18 PCR Basics 



a sampling of strains, serovars, and closely and distantly related microbes. 
Despite the presence of invA homolog in other genera and species, sequences 
are divergent enough for this to be a useful genetic marker for detecting 
Salmonella (68, 72). 

Now that you have determined in silico your candidate genetic marker, 
you can proceed to analyze your sequence(s) for the best PCR primer pairs. 
There are several commercially available, as well as Web-based (13; 
http://dbb.nhri.org.tw/primer/) DNA analysis software packages for designing 
PCR primers that vary in price, utility, ease, options, or familiarity to the 
authors. Therefore, we will only provide the reader with general design consider- 
ations. First, let us consider in our design the size we want for our amplicon. This 
consideration is especially important in the development of multiplex PCR 
where the size of the amplicon identifies the gene or organism present in our 
sample. Also PCRs sensitivity is influenced by the size of the amplicon. For sen- 
sitive, real-time PCR, small amplicons, 75-200 bp are preferred. Next thing to 
consider is where to concentrate our search for specific PCR primers. From our 
BLAST search, it appears that the 1st 750 bp of Salmonella invA is ideal for our 
analysis. Also to improve the specificity of our PCR, we need to consider the 
length of each primer (94). Generally, the minimum default value for many of 
the PCR primer design algorithms is 18 bp. This value is generated from the 
probability of finding this exact sequence within the bacterial genome, where for 
this example; we are dealing with an organism with 50% GC content and 
4,000,000 bp genome. The probability of a specific 18 bp sequence is present is 
(1/4) 18 x 4,000,000 = 6 x 10~ 5 . The smaller the sequence, the greater the likelihood 
of finding sequence not just once but multiple times within the genome. That is 
why short 10-mer oligonucleotides have become useful tools for typing bacteria 
by random amplified polymorphic DNA (RAPD) PCR (51), because based on 
size and using our calculations we expect to find these sequences at least 4 times 
within the bacterial genome. Now, having run our analysis, we are presented with 
all possible primer pairs. Our next step is to select primers for the amplicon size 
that we want and screen these primer sets further, to identify those that do not 
form "hairpins" or primer-dimers. We especially want to avoid primers that form 
hairpins at the 3' end as this will interfere with the primers annealing precisely to 
its target sequence and participation of the primer in the DNA extension step in 
PCR. Primer-dimers and hairpins can affect the specificity and sensitivity of 
PCR and should be avoided if possible (78). Once the appropriate primer set(s) 
has been identified, search the GenBank DNA database for match with our 
primers. With the BLAST search, it is recommended with searches of short 
sequences to select Bacteria under "or select from" option. This is to limit con- 
fusion with random and insignificant matches with the larger animal and plant 
genomes (10 9 bp) that sometimes occur. Beyond this point, we generally empiri- 
cally optimize our PCR, using appropriate positive and negative controls, and 
identifying the magnesium concentration and PCR annealing temperature with 
the sensitivity and specificity that is best for detecting organism X. We then ver- 
ify the specificity of our PCR by comparing same strains, serovars, or species 
against different strains, serovars, and closely and distantly related microorgan- 



References 19 



isms to see if same size PCR amplicon is produced only for those groups of bac- 
teria to which the PCR was intended to identify. Ideally, once our PCR has been 
optimized, PCR amplicon, of the expected size is only observed among select 
bacteria that possess the target gene and nothing for all other microorganisms 
that do not possess this gene. It is at this point too that we verify that our ampli- 
con, with size expected based on the primers designed, is the target gene to which 
our primers were intended to amplify. We accomplish this by sequencing the 
PCR amplicon and match resulting sequence against GenBank DNA database 
using the BLAST algorithm. Our amplicon's sequence should match the invA 
sequences present on the DNA database. 



REFERENCES 



1. Abdulmawjood, A., M. Bulte, S. Roth, H. Schonenbrucher, N. Cook, M. 
D'Agostino, M. Burkhard, K. Jordan, S. Pelkonen, and J. Hoorfar. 2004. Toward an 
international standard for PCR-based detection of foodborne Escherichia coliO\57: 
Validation of the PCR-based method in a multicenter interlab oratory trial. J. AOAC 
Int. 87:856-860. 

2. Al-Khaldi, S.F., K.M. Myers, A. Rasooly, and V. Chizhikov. 2004. Genotyping of 
Clostridium perfringens toxins using multiple oligonucleotide microarray hybridiza- 
tion. Mol. Cell Probes 18:359-367. 

3. Anderson, A.D., V.D. Garrett, J. Sobel, S.S. Monroe, R.L. Fankhauser, K.J. Schwab, 
J.S. Bresee, P.S. Mead, C. Higgins, J. Campana, R.I. Glass, and Outbreak 
Investigation Team. 2001. Multistate outbreak of Norwalk-like virus gastroenteritis 
associated with a common caterer. Am. J. Epidemiol. 154:1013-1019. 

4. Benjamin, M.A., J. Lu, G Donnelly, P. Dureja, and D.M. McKay. 1998. Changes in 
murine jejunal morphology evoked by the bacterial superantigen Staphylococcus 
aureus enterotoxin B are mediated by CD4+ T cells. Infect. Immun. 66:2193-2199. 

5. Bilge, S.S., J.C. Vary Jr., S.F. Dowell, and P.I. Tarr. 1996. Role of Escherichia coli 
0157:H7 O side chain in adherence and analysis of an rfb locus. Infect. Immun. 
64:4795-4801. 

6. Borucki, M.K., S.H. Kim, D.R. Call, S.C. Smole, and F. Pagotto. 2005. Selective 
discrimination of Listeria monocytogenes epidemic strains by a mixed-genome DNA 
microarray compared to discrimination by pulsed-field gel electrophoresis, ribotyp- 
ing, and multilocus sequence typing. J. Clin. Microbiol. 42:5270-5276. 

7. Brussow, H., C. Canchaya, and W.D. Hardt. 2004. Phages and the evolution of bac- 
terial pathogens: From genomic rearrangements to lysogenic conversion. Microbiol. 
Mol. Biol. Rev. 68:560-602. 

8. Buzby, J.C. and T. Roberts. 1997. Economic costs and trade impacts of microbial 
foodborne illness. World Health. Stat. Q. 50:57-66. 

9. Cha, J.H., M.Y. Chang, JA. Richardson, and L. Eidels. 2003. Transgenic mice express- 
ing the diphtheria receptor are sensitive to the toxin. Mol. Microbiol. 49:235-240. 

10. Chain, PS., E. Carniel, F.W. Larimer, J. Lamerdin, P.O. Stoutland, W.M. Regala, 
A.M. Georgescu, L.M. Vergez, M.L Land, V.L. Motin, R.R. Brubaker, J. Fowler, 
J. Hinnebusch, M. Marceau, C. Medigue, M. Simonet, V. Chenal-Francisque, 
B. Souza, D. Dacheux, J.M. Elliott, A. Derbise, L.J. Hauser, and E. Garcia. 2004. 
Insights into the evolution of Yersinia pestis through whole-genome comparison 
with Yersinia pseudotuberculosis. Proc. Natl. Acad. Sci. USA 101:13826-13831. 



20 PCR Basics 



11. Chan, K., S. Baker, C.C. Kim, C.S. Detweiler, G. Dougan, and S. Falkow. 2003. 
Genomic comparison of Salmonella enter ica serovars and Salmonella bongori by use 
of an S. enterica serovar Typhimurium DNA microarray. J. Bacteriol. 185:553-563. 

12. Chen, C, K.-M. Wu, Y.-C. Chang, C.-H. Chang, H.-C. Tsai, T.-L. Liao, Y.-M. Liu, 
H.-J. Chen, A.B.-T. Shen, J. - C. Li, T.-L. Su, C.-P. Shao, C.-T. Lee, L.-I. Hor, and 
S.-F. Tsai. 2003. Comparative genome analysis of Vibrio vulnificus, a marine pathogen. 
Gen. Res. 13:2577-2587. 

13. Chen, S.H., C.Y Lin, C.S. Cho, C.Z. Lo, and C.A. Hsiung. 2003. Primer design 
assistant (PDA): A Web-based primer design tool. Nucl. Acids Res. 31:3751-3754. 

14. Christensen, J.E., J. A. Stencil, and K.D. Reed. 2003. Rapid identification of bacte- 
ria from positive blood cultures by terminal restriction fragment length polymor- 
phism profile analysis of the 16S rRNA gene. J. Clin. Microbiol. 41:3790-3800. 

15. Dahan, S., S. Knutton, R.K. Shaw, V.F. Crepin, G Dougan, and G Frankel. 2004. 
Transcriptome of enterohemorrhagic Escherichia coli 0157 adhering to eukaryotic 
plasma membranes. Infect. Immun. 72:5452-5459. 

16. Desmarchelier, P.M., S.S. Bilge, N. Fegan, L. Mills, J.C. Vary, and P.I. Tarr. 1998. 
A PCR specific for Escherichia coli 0157 based on the rfb locus encoding 0157 
lipopolysaccharide. J. Clin. Microbiol. 36:1801-1804. 

17. Dimech, W., D.S. Bowden, B. Brestovac, K. Byron, G. James, D. Jardine, T. Sloots, 
and E.M. Dax. 2004. Validation of assembled nucleic acid-based tests in diagnostic 
microbiology laboratories. Pathol. 36:45-50. 

18. Donohue-Rolfe, A., I. Kondova, S. Oswald, D. Hutto, and S. Tzipori. 2000. 
Escherichia coli 0157:H7 strains that express Shiga toxin (Stx) 2 alone are more neu- 
rotropic for gnotobiotic piglets than are isotypes producing only Stxl or both Stxl 
and Stx 2. J. Infect. Dis. 181:1825-1829. 

19. Fischer, S.H., and I. Nachamkin. 1991. Common and variable domains of the fla- 
gellin gene,/7a^4, in Campylobacter jejuni. Mol. Microbiol. 5:1151-1158. 

20. Fraser, CM., J.D. Gocayne, O. White, M.D. Adams, R.A. Clayton, R.D. Fleischmann, 
C.J. Bult, A.R. Kerlavage, G Sutton, J.M. Kelley, et al. 1995. The minimal gene com- 
plement of Mycoplasma genitalium. Science 270:397-403. 

21. Fratamico, P.M., S.K. Sackitey, M. Wiedmann, and M.Y Deng. 1995. Detection of 
Escherichia coli 0157:H7 by multiplex PCR. J. Clin. Microbiol. 33:2188-2191. 

22. Freeman, W.M., D.J. Robertson, and K.E. Vrana. 2000. Fundamentals of DNA 
hybridization arrays for gene expression analysis. Biotechniques 29:1042-1055. 

23. Fukiya, S., H. Mizoguchi, T. Tobe, and H. Mori. 2004. Extensive genomic diversity 
in pathogenic Escherichia coli and Shigella strains revealed by comparative genomic 
hybridization microarray. J. Bacteriol. 186:3911-3921. 

24. Galan, J.E. and R. Curtiss III. 1989. Cloning and molecular characterization of 
genes whose products allow Salmonella typhimurium to penetrate tissue culture cells. 
Proc. Natl. Acad. Sci. USA 86:6383-6387. 

25. Galan, J.E. and R. Curtiss III. 1991. Distribution of the invA, -B, -C, and -D genes of 
Salmonella typhimurium among other Salmonella serovars: invA mutants of Salmonella 
Typhi are deficient for entry into mammalian cells. Infect. Immun. 59:2901-2908. 

26. Gannon, V.P., S. D'Souza, T. Graham, R.K. King, K. Rahn, and S. Read. 1997. Use 
of the flagellar H7 gene as a target in multiplex PCR assays and improved specificity 
in identification of enterohemorrhagic Escherichia coli strains. J. Clin. Microbiol. 
35:656-662. 

27. Giesendorf, B.A.J., J.A.M. Vet, S. Tyagi, E.J.M.G Mensink, F.J.M. Trijbels, and H.J. 
Blom. 1998. Molecular beacons: A new approach for semiautomated mutation 
analysis. Clin. Chem. 44:482-486. 



References 21 



28. Harmon, K.M., G.M. Ransom, and I.V. Wesley. 1997. Differentiation of 
Campylobacter jejuni and Campylobacter coli by polymerase chain reaction. Mol. 
Cell. Probes 11:195-200. 

29. Heid, C.A., J. Stevens, K.J. Livak, and P.M. Williams. 1996. Real time quantitative 
PCR. Genome Res. 6:986-994. 

30. Herold, S., J. Siebert, A. Huber, and H. Schmidt. 2005. Global expression of prophage 
genes in Escherichia coli 0157:H7 strain EDL933 in response to norfloxacin. 
Antimicrob. Agents Chemother. 49:931-944. 

31. Herrera-Leon, S., J.R. McQuiston, M.A. Usera, P.I. Fields, J. Garaizar, and M.A. 
Echeita. 2004. Multiplex PCR for distinguishing the most common phase- 1 flagel- 
lar antigens of Salmonella spp. J. Clin. Microbiol. 42:2581-2586. 

32. Higuchi, R., C. Fockler, G. Dollinger, and R. Watson. 1993. Kinetic PCR analysis: 
Real-time monitoring of DNA amplification reactions. Biotechnol. 11:1026-1030. 

33. Hirsch, H.H. and W Bossart. 1999. Two-centre study comparing DNA preparation 
and PCR amplification protocols for herpes simplex virus detection in cere- 
brospinal fluids of patients with suspected herpes simplex encephalitis. J. Med. 
Virol. 57:31-35. 

34. Hogberg, A., J.E. Lindberg, T. Leser, and P. Wallgren. 2004. Influence of cereal non- 
starch polysaccharides on ileo-caecal and rectal microbial populations in growing 
pigs. Acta. Vet. Scand. 45:87-98. 

35. Holden, M.T., E.J. Feil, J.A. Lindsay, S.J. Peacock, N.P. Day, M.C. Enright, T.J. Foster, 
C.E. Moore, L. Hurst, R. Atkin, A. Barron, N. Bason, S.D. Bentley, C. Chillingworth, 
T. Chillingworth, C. Churcher, L. Clark, C. Corton, A. Cronin, J. Doggett, L. Dowd, 
T. Feltwell, Z. Hance, B. Harris, H. Hauser, S. Holroyd, K. Jagels, K. D. James, N. 
Lennard, A. Line, R. Mayes, S. Moule, K. Mungall, D. Ormond, M. A. Quail, E. 
Rabbinowitsch, K. Rutherford, M. Sanders, S. Sharp, M. Simmonds, K. Stevens, S. 
Whitehead, B.G Barrell, B.G Spratt, and J. Parkhill. 2004. Complete genomes of two 
clinical Staphylococcus aureus strains: Evidence for the rapid evolution of virulence 
and drug resistance. Proc. Natl. Acad. Sci. USA 101:9786-9791. 

36. Holland, P.M., R.D. Abramson, R. Watson, D.H. Gelfand. 1991. Detection of spe- 
cific polymerase chain reaction product by utilizing the 5'-3' exonuclease activity of 
Thermus aquaticus DNA polymerase. Proc. Natl. Acad. Sci. USA 88:7276-7280. 

37. Hong, Y, M.E. Berrang, T. Liu, C.L. Hofacre, S. Sanchez, L. Wang, and J.J. Maurer. 
2003. Rapid detection of Campylobacter coli, C jejuni, and Salmonella enterica on 
poultry carcasses by using PCR-enzyme-linked immunosorbent assay. Appl. 
Environ. Microbiol. 69:3492-3499. 

38. Hoorfar, J., P. Wolffs, and P. Radstrom. 2004. Diagnostic PCR: Validation and sam- 
ple preparation are two sides of the same coin. APMIS 112:808-814. 

39. Houf, K., A. Tutenel, L. De Zutter, J. Van Hoof, and P. Vandamme. 2000. 
Development of a multiplex PCR assay for the simultaneous detection and identifi- 
cation of Arcobacter butzleri, Arcobacter cryaerophilus, and Arcobacter skirrowii. 
FEMS Microbiol. Lett. 193:89-94. 

40. Hueck, C.J. 1998. Type III protein secretion systems in bacterial pathogens of ani- 
mal and plants. Microbiol. Mol. Biol. Rev. 62:379-433. 

41. Johnson, W.M., S.D. Tyler, E.P. Ewan, F.E. Ashton, D.R. Pollard, and K.R. Rozee. 
1991. Detection of genes for enterotoxins, exfoliative toxins, and toxic shock syn- 
drome toxin 1 in Staphylococcus aureus by the polymerase chain reaction. J. Clin. 
Microbiol. 29:426-430. 

42. Kaplan, C.W, J.C. Astaire, M.E. Sanders, B.S. Reddy, and C.L. Kitts. 2001. 16S ribo- 
somal DNA terminal restriction fragment patter analysis of bacterial communities 



22 PCR Basics 



in feces of rats fed Lactobacillus acidophilus NCFM. Appl. Environ. Microbiol. 
67:1935-1939. 

43. Keramas, G, D.D. Bang, M. Lund, M. Madsen, H. Bunkenborg, P. Telleman, and 
C. B. V. Christensen. 2004. Use of culture, PCR analysis, and DNA microarrays for 
detection of Campylobacter jejuni and Campylobacter coli from chicken feces. 
J. Clin. Microbiol. 42:3985-3991. 

44. Koopmans, M. and E. Duizer. 2004. Foodborne viruses: An emerging problem. Int. 
J. Food Microbiol. 90:23-41. 

45. Mackay, I.M. 2004. Real-time PCR in the microbiology laboratory. Clin. Microbiol. 
Infect. 10:190-212. 

46. Marsh, T.L. 1999. Terminal restriction fragment length polymorphism (T-RFLP): 
An emerging method for characterizing diversity among homologous populations of 
amplification products. Curr. Opin. Microbiol. 2:323-327. 

47. Marsh, T.L., P. Saxman, J. Cole, and J. Tiedje. 2000. Terminal restriction fragment 
length polymorphism analysis program, a Web-based research tool for microbial 
community analysis. Appl. Environ. Microbiol. 66:3616-3620. 

48. Marston, C.K., F. Jamieson, F. Cahoon, G. Lesiak, A. Golaz, M. Reeves, and T. 
Popovic. 2001. Persistence of a distinct Corynebacterium diphtheriae clonal group 
within two communities in the United States and Canada where diphtheria is 
endemic. J. Clin. Microbiol. 39:1586-1590. 

49. Maurer, J.J. and M.D. Lee. 2005. Salmonella. In Mansel Griffiths (ed.), Understanding 
Pathogen Behavior, Woodhead Publishing Limited, Cambridge, United Kingdom 
(In Press). 

50. Maurer, J.J., D. Schmidt, P. Petrosko, S. Sanchez, L. Bolton, and M.D. Lee. 1999. 
Development of primers to O-antigen biosynthesis genes for specific detection of 
Escherichia coli 0157 by PCR. Appl. Env. Microbiol. 65:2954-2960. 

51. Mazurier, S., A. van de Giessen, K. Heuvelman, and K. Wernars. 1992. RAPD 
analysis of Campylobacter isolates: DNA fingerprinting without the need to purify 
DNA. Lett. Appl. Microbiol. 14:260-262. 

52. McClelland, M., K.E. Sanderson, J. Spieth, S.W. Clifton, P. Latreille, L. Courtney, S. 
Porwollik, J. Ali, M. Dante, F. Du, S. Hou, D. Layman, S. Leonard, C. Nguyen, K. 
Scott, A. Holmes, N Grewal, E. Mulvaney, E. Ryan, H. Sun, L. Florea, W. Miller, T 
Stoneking, M. Nhan, R. Waterston, and R. K. Wilson. 2001. Complete genome 
sequence of Salmonella enterica serovar Typhimurium LT2. Nature 413:852-856. 

53. Mead, P.S., L. Slutsker, V. Dietz, L.F. McCaig, J.S. Bresee, C. Shapiro, P.M. Griffin, 
and R.V. Tauxe. 1999. Food-related illness and death in the United States. Emerg. 
Infect. Dis. 5:607-625. 

54. Moscoso, H., S.G Thayer, C.L. Hofacre, and S.H. Kleven. 2004. Inactivation, stor- 
age, and PCR detection of Mycoplasma on FTA filter paper. Avian Dis. 48:841-850. 

55. Mullah, B., K. Livak, A. Andrus, and P. Kenney. 1998. Efficient synthesis of double 
dye-labeled oligodeoxyribonucleotide probes and their application in a real time 
PCR assay. Nucleic Acids Res. 26:1026-1031. 

56. Mullis, K.B., FA. Faloona, S.J. Scharf, R.K. Saiki, GT. Horn, and H.A. Erlich. 
1986. Specific enzymatic amplification of DNA in vitro: The polymerase chain reac- 
tion. Cold Spring Harbor Symp. Quant. Biol. 51:263-273. 

57. Mullis, K.B. 1994. In K.B. Mullis, F Ferre, and R.A. Gibbs (ed.), The Polymerase 
Chain Reaction. Birkhauser Boston, Cambridge, MA. 

58. Mullis, K.B. 1998. Dancing Naked in the Mind Field. Pantheon Books, New York. 

59. Nelson, K.E., D.E Fouts., E.F.Mongodin, J. Ravel, R.T. DeBoy, J.F Kolonay, D.A. 
Rasko, S.V. Angiuoli, S.R. Gill, I.T. Paulsen, J. Peterson, O. White, W.C. Nelson, W. 



References 23 



Nierman, M.J. Beanan, L.M. Brinkac, S.C. Daugherty, R.J. Dodson, A.S. Durkin, 
R. Madupu, D.H. Haft, J. Selengut, S. Van Aken, H. Khouri, N. Fedorova, H. 
Forberger, B. Tran, S. Kathariou, L.D. Wonderling, G.A. Uhlich, D.O. Bayles, J.B. 
Luchansky, and CM. Fraser. 2004. Whole genome comparisons of serotype 4b and 
l/2a strains of the foodborne pathogen Listeria monocytogenes reveal new insights 
into the core genome components of this species. Nucl. Acids Res. 32:2386-2395. 

60. Nilsson, W.B. and M.S. Strom. 2002. Detection and identification of bacterial 
pathogens of fish in kidney tissue using terminal restriction fragment length poly- 
morphism (T-RFLP) analysis of 16S rRNA genes. Dis. Aquat. Organ. 48:175-185. 

61. Nordentoft, S., H. Cristensen, and H.C. Wegener. 1997. Evaluation of a fluores- 
cence-labeled oligonucleotide probe targeting 23S rRNA for in situ detection of 
Salmonella serovars in paraffin-embedded tissue sections and their rapid identifica- 
tion in bacterial smears. J. Clin. Microbiol. 35:2642-2648. 

62. Oelschlaeger, T.A. and J. Hacker. 2004. Impact of pathogenicity islands in bacterial 
diagnostics. APMIS 112:930-936. 

63. Oyofo, B.A., S.A. Thornton, D.H. Burr, T.J. Trust, O.R. Pavlovskiy and P. Guerry 
1992. Specific detection of Campylobacter jejuni and Campylobacter coli by using 
polymerase chain reaction. J. Clin. Microbiol. 30:2613-2619. 

64. Panet, A. and H.G. Khorana. 1974. Studies on polynucleotides. The linkage of 
deoxyribonucleic templates to cellulose and its use in their replication. J. Biol. Chem. 
249:5213-5221. 

65. Panicker, G, D.R. Call, M.J. Krug, and A.K. Bej. 2004. Detection of pathogenic 
Vibrio spp. in shellfish by using multiplex PCR and DNA microarrays. Appl. 
Environ. Microbiol. 70:7436-7444. 

66. Parkhill, J., B.W Wren, K. Mungall, J.M. Ketley, C. Churcher, D. Basham, T 
Chillingworth, R.M. Davies, T. Feltwell, S. Holroyd, K. Jagels, A.V. Karlyshev, S. 
Moule, M.J. Pallen, C.W Penn, M.A Quail, M.A. Rajandream, K.M. Rutherford, 
A.H. van Vliet, S. Whitehead, and B.G Barrell. 2000. The genome sequence of the 
foodborne pathogen Campylobacter jejuni reveals hypervariable sequences. Nature 
403:665-668. 

67. Paton, A.W and J.C. Paton. 1998. Detection and characterization of shiga toxigenic 
Escherichia coli by using multiplex PCR assays for stx v stx T eaeA, entero hemor- 
rhagic E. coli hlyA, rfb QUV and rfb Ql51 . J. Clin. Microbiol. 36:598-602. 

68. Perelle, S., F. Dilasser, B. Malorny, J. Grout, J. Hoorfar, and P. Fach. 2004. 
Comparison of PCR-ELISA and LightCycler real-time PCR assays for detecting 
Salmonella spp. in milk and meat samples. Mol. Cell Probes 18:409-420. 

69. Perna, NT, G Plunkett 3rd, V. Burland, B. Mau, ID. Glasner, D.J. Rose, GF. 
Mayhew, P.S. Evans, J. Gregor, H.A. Kirkpatrick, G Posfai, J. Hackett, S. Klink, A. 
Boutin, Y. Shao, L. Miller, E.J. Grotbeck, N.W. Davis, A. Lim, E.T. Dimalanta, 
K.D. Potamousis, J. Apodaca, T.S. Anantharaman, J. Lin, G. Yen, D.C. Schwartz, 
R.A. Welch, and F.R. Blattner. 2001. Genome sequence of enterohaemorrhagic 
Escherichia coli 0157:H7. Nature 409:529-533. 

70. Porwollik, S., E.F. Boyd, C. Choy, P. Cheng, L. Florea, E. Proctor, and M. 
McClelland. 2004. Characterization of Salmonella enterica subspecies I genovars by 
use of microarrays. J. Bacteriol. 186:5883-5898. 

71. Prouty, A.M., I.E. Brodsky, S. Falkow, and J.S. Gunn. 2004. Bile-salt-mediated 
induction of antimicrobial and bile resistance in Salmonella typhimurium. 
Microbiol. 150:775-783. 

72. Rahn, K., S.A. Grandis, R.C. Clarke, S.A. McEwen, J.E. Galan, C. Ginocchio, R. 
Curtiss III, and C.L. Gyles. 1992. Amplification of invA gene sequence of Salmonella 



24 PCR Basics 



typhimurium by polymerase chain reaction as a specific method of detection of 
Salmonella. Mol. Cell Probes 6:271-279. 

73. Rappuoli, R. 2000. Pushing the limits of cellular microbiology: Microarrays to study 
bacteria-host cell intimate contacts. Proc. Natl. Acad. Sci. USA 97:13467-13469. 

74. Rasko, D.A., J. Ravel, O.A. Okstad, E. Helgason, R.Z. Cer, L. Jiang, K.A. Shores, 
D.E. Fouts, N.J. Tourasse, S.V. Angiuoli, J. Kolonay, W.C. Nelson, A.B. Kolsto, 
CM. Fraser, and T.D. Read. 2004. The genome sequence of Bacillus cereus ATCC 
10987 reveals metabolic adaptations and a large plasmid related to Bacillus anthracis 
pXOl. Nucl. Acids Res. 32:977-988. 

75. Reeves, M.W., R.J. Arko, F.W. Chandler, and NB. Bridges. 1986. Affinity purifica- 
tion of staphylococcal toxic shock syndrome toxin 1 and its pathologic effects in rab- 
bits. Infect. Immun. 51:431-439. 

76. Robertson, B.H., F. Averhoff, T.L. Cromeans, X. Han, B. Khoprasert, O.V. Nainan, 
J. Rosenberg, L. Paikoff, E. DeBess, C.N. Shapiro, and H.S. Margolis. 2000. Genetic 
relatedness of hepatitis A virus isolated during a community- wide outbreak. J. Med. 
Virol. 62:144-150. 

77. Rotbart, H.A., M.J. Levin, L.P. Villarreal, S.M. Tracy, B.L. Semler, and E. Wimmer. 
1985. Factors affecting the detection of enteroviruses in cerebrospinal fluid with 
coxsackievirus B3 and poliovirus 1 cDNA probes. J. Clin. Microbiol. 22:220-224. 

78. Rychlik, W. 1995. Priming efficiency in PCR. Biotechn. 18:84-86, 88-90 

79. Saldanha, X, W. Gerlich, N Lelie, P. Dawson, K. Heermann, A. Heath, and WHO 
Collaborative Study Group. 2001. An international collaborative study to establish 
a World Health Organization international standard for hepatitis B virus DNA 
nucleic acid amplification techniques. Vox Sang. 80:63-71. 

80. Savitt, E.D., M.N. Strzempko, K.K. Vaccaro, W.J. Peros, and C.K. French. 
Comparison of cultural methods and DNA probe analyses for the detection of 
Actinobacillus actinomycetemcomitans, Bacteroides gingivalis, and Bacteroides inter- 
medius in subgingival plaque samples. J. Periodontol. 59:431^138. 

81. Schoenhals, G and C. Whitfield. 1993. Comparative analysis of flagellin sequences 
from Escherichia coli strains possessing serologically distinct flagellar filaments with 
a shared complex surface pattern. J. Bacteriol. 175:5395-5402. 

82. Shimizu, T., K. Ohtani, H. Hirakawa, K. Ohshima, A. Yamashita, T. Shiba, N 
Ogasawara, M. Hattori, S. Kuhara, and H. Hayashi. 2002. Complete genome 
sequence of Clostridium perfringens, an anaerobic flesh-eater. Proc. Natl. Acad. Sci. 
USA 99:996-1001. 

83. Smith, N.H. and R.K. Selander. 1990. Sequence invariance of the antigenic-coding 
central region of the phase 1 flagellar filament gene (fliC) among strains of 
Salmonella typhimurium. J. Bacteriol. 172:603-609. 

84. Stintzi, A., D. Marlow, K. Palyada, H. Naikare, R. Panciera, L. Whitworth, and C. 
Clarke. 2005. Use of genome-wide expression profiling and mutagenesis to study the 
intestinal lifestyle of Campylobacter jejuni. Infect. Immun. 73:1797-1810. 

85. Taboada, E.N., R.R. Acedillo, CD. Carrillo, WA. Findlay, D.T. Medeiros, O.L. 
Mykytczuk, M.J. Roberts, C.A. Valencia, J.M. Farber, and J.H. Nash. 2004. Large- 
scale comparative genomics meta-analysis of Campylobacter jejuni isolates reveals 
low level of genome plasticity. J. Clin. Microbiol. 42:4566^576. 

86. Thomas, E.J., R.K. King, J. Burchak, and V.P. Gannon. 1991. Sensitive and specific 
detection of Listeria monocytogenes in milk and ground beef with the polymerase 
chain reaction. Appl. Environ. Microbiol. 57:2576-2580. 

87. Vinje, J., H. Vennema, L. Maunula, C.H. von Bonsdorff, M. Hoehne, E. Schreier, A. 
Richards, J. Green, D. Brown, S.S. Beard, S.S. Monroe, E. de Bruin, L. Svensson, 



References 25 



and M.R Koopmans. 2003. International collaborative study to compare reverse 
transcriptase PCR assays for detection and genotyping of noroviruses. J. Clin. 
Microbiol. 41:1423-1433. 

88. Vora, G.J., C.E. Meador, D.A. Stenger, and ID. Andreadis. 2004. Nucleic acid 
amplification strategies for DNA microarray-based pathogen detection. 
70:3047-3054. 

89. Wang, L., W. Qu, and P.R. Reeves. 2001. Sequence analysis of four Shigella boydii 
O-antigen loci: Implication for Escherichia coli and Shigella relationships. Infect. 
Immun. 69:6923-6930. 

90. Wang, L., and P.R. Reeves. 1998. Organization of Escherichia coli 0157 O antigen 
gene cluster and identification of its specific genes. Infect. Immun. 66:3545-3551. 

91. Watson, J.D., and EH. Crick. 1953. Molecular structure of nucleic acids; a structure 
for deoxyribose nucleic acid. Nature. 171:737-738. 

92. Wei, J., M.B. Goldberg, V. Burland, M.M. Venkatesan, W Deng, G. Fournier, GF. 
Mayhew, G Plunkett 3rd, D.J. Rose, A. Darling, B. Mau, NT. Perna, S.M. Payne, 
L. J. Runyen-Janecky, S. Zhou, D.C. Schwartz, and F.R. Blattner. 2003. Complete 
genome sequence and comparative genomics of Shigella flexneri serotype 2a strain 
2457T. Infect. Immun. 71:2775-2786. 

93. Whittam, T.S., and A.C. Bumbaugh. 2002. Inferences from whole-genome 
sequences of bacterial pathogens. Curr. Opin. Genet. Dev. 12:719-725. 

94. Wu, D.Y., L. Ugozzoli, B.K. Pal, J. Qian, and R.B. Wallace. 1991. The effect of tem- 
perature and oligonucleotide primer length on the specificity and efficiency of 
amplification by the polymerase chain reaction. DNA Cell. Biol. 10:233-238 

95. Yoo, H.S., S.U. Lee, K.Y. Park, and Y.H. Park. 1997. Molecular typing and epi- 
demiological survey of prevalence of Clostridium perfringens types by multiplex 
PCR. J. Clin. Microbiol. 35:228-232. 

96. Zhang, C, M. Zhang, J. Ju, J. Nietfeldt, J. Wise, P.M. Terry, M. Olson, S.D. 
Kachman, M. Wiedmann, M. Samadpour, and A. K. Benson. 2003. Genome diver- 
sification in phylogenetic lineages I and II of Listeria monocytogenes: Identification 
of segments unique to lineage II populations. J. Bacteriol. 185:5573-5584. 

97. Zhang, X., A.D. McDaniel, L.E. Wolf, GT. Keusch, M.K. Waldor, and D.W 
Acheson. 2000. Quinolone antibiotics induce Shiga toxin-encoding bacteriophages, 
toxin production, and death in mice. J. Infect. Dis. 181:664-670. 



CHAPTER 2 



The Mythology of PCR: 
A Warning to the Wise 

John J. Maurer 1 ' 2 *, Ph.D. 

Poultry Diagnostic & Research Center 1 , College of Veterinary Medicine, 

The University of Georgia, Athens, GA 30602 
Center for Food Safety 2 , College of Agriculture and Environmental Sciences, 

The University of Georgia, Griffin, GA 30223 



Introduction 
Interpretation 

Conventional PCR 

Real Time PCR 
Validation 
Problems and Their Solutions 

False-Positives and Dead vs. Live Bacterial Cell Debate 

PCR Inhibitors, Limits of Detection and False-Negatives 
Conclusions 
References 



INTRODUCTION 



Most diagnostic PCR tests are a qualitative yes or no, presence or absence of 
pathogen X. We know what it means if our sample is positive by PCR, report- 
ing back presumptive positive for organism X and a negative PCR result was 
the end-point for that sample. Were these assumptions correct? The decisions 
we make based on these PCR results require that we know how to interpret 
these results and like any other diagnostic test, know its limitations with regards 
to sensitivity and specificity. Even if your laboratory is only interested in adapt- 
ing existing PCR methods for identification of pathogens in foods, it is impor- 
tant that you know what the results mean, and know how to recognize and 
troubleshoot problems as they occur. You can safe guard or at least be prepared 
to recognize these problems, as they appear, by implementing standard operat- 
ing procedures and including controls recommended by authors in the chapters 
discussed in this book. In this section, I will specifically delve into interpreta- 
tion and understanding of PCR results as well as discuss the limitations, prob- 
lems, and erroneous assumptions associated with PCR and other PCR based 
technologies (e.g., real-time PCR). 



Corresponding author. Phone: (706) 542-5071; FAX: (706) 542-5630; e-mail: jmau- 
rer@vet.uga.edu. 



27 



28 The Mythology of PCR: A Warning to the Wise 



INTERPRETATION 



Conventional PCR. A sample is positive, by PCR, if an amplicon is produced 
with the size expected for the primers used. What if the sample yields an ampli- 
con larger or smaller than the size expected for our PCR primers? Is this sam- 
ple considered positive by PCR? NO!!! This result is referred to in PCR parlance 
as a nonspecific amplicon, it is ignored, AND if we do not observe an amplicon 
with a size expected for primers used, the sample is considered PCR negative. It is 
therefore a requirement to always include DNA molecular weight standards, in 
the appropriate size range for accurately assessing the amplicon's size, and the 
percentage of agaraose and electrophoresis time needed to adequately separate 
the molecular weight standards. One needs to also consider other parameters 
(electrophoresis buffer, buffer strength, voltage, etc.) that affect uniformity of 
DNA separation across the entire width and length of the agarose gel. For wide 
gels with many wells or lanes (>10), one may consider placement of the DNA 
standards in the middle and the outermost wells. With appropriate gel docu- 
mentation software, the user can, using these well placed molecular weight 
standards, correct for electrophoresis migration anomalies that produce 
"smiles" at high voltages. Avoiding electrophoresis at high voltages or circu- 
late/cool the buffer during electrophoresis can prevent this electrophoretic 
anomaly. With every PCR run, ALWAYS include a positive control so that you 
can match your sample with the control, and allow adequate separation of your 
DNA standards, samples, and control so that you do not erroneously report a 
sample with a nonspecific amplicon as positive. If molecular biology is new to 
your laboratory, it is advisable to purchase a general molecular biology manual, 
that details the specifics of gel electrophoresis, includes theory and helps trou- 
ble shoot problems commonly associated with the molecular technique (1, 62). 
For the experienced molecular biologist, this is rather obvious, but for oth- 
ers, especially the novice, it is easy to be lulled into believing the presence of any 
PCR product, regardless of size, on the gel means the sample must be positive 
for organism X. Most genes targeted by PCR have been selected based on their 
conservation and uniformity within a species, subspecies, serovar or pathotype. 
These genes are uniform in size. There are, however, exceptions, genes or DNA 
segments containing repetitive elements or extragenic sequences, the number, 
size, or presence of which varies within the bacterial population (10, 16, 22, 38, 
57, 70). PCRs have been developed to exploit these genetically variable regions 
for the purpose of genus/species identification (10, 16, 24, 35, 57) and strain typ- 
ing (25, 56, 57). Here the different size amplicon identifies the genus or species 
and/or distinguishes strain types. However, a requirement for using any of these 
PCRs is first the isolation of the organism. For PCR screens of foods, it is 
advisable to avoid those PCRs that produce, as designed, these variable size 
amplicons. Unless, an internal probe is included in the PCR screens, for speci- 
ficity, the technician may confuse a true, nonspecific amplicon in a sample as a 
positive and erroneously report the sample as such. 

Real-Time PCR. Results generated by real-time PCR are generally more 
straightforward to interpret for a simple question like: is the organism present 



Interpretation 



29 



in our sample? Rather than visualize the amplicon following PCR, we monitor 
the increase in fluorescence over time as newly synthesized, amplicon binds to 
SYBR Green® or the chemically quenched, fluorescent dye is liberated as the 
amplicon displaces an internally bound, dye-labeled probe. Fig. 2.1 illustrates 
kinetics of real-time PCR. Note the points on the x-axis, "threshold cycle" (C T ) 
where the log-linear phase of fluorescence begins for the different target DNA 
concentrations (43). There is a linear correlation between C T and DNA con- 
centration, making the PCR quantitative. A sample is considered positive pro- 
vided it falls within the range of C T values, demarcated by the PCR's limit of 
detection, and the background fluorescence associated with the negative or no 
DNA controls. While real-time PCR surpasses conventional PCR in speed and 
sensitivity, nonspecific amplicons can result in our erroneously reporting a pos- 
itive result. SYBR Green® binds to double stranded DNA, regardless of 
whether it is the expected amplicon, nonspecific amplicon, or primer-dimers. 
Gradient thermocyclers have become a useful tool in rapidly identifying anneal- 
ing temperature best for PCR amplification of the target gene while avoiding 
primer-dimers. We can distinguish nonspecific amplicon(s) from a true positive 
based on their distinctive DNA melting curves (Fig. 2.2) (59). 



35000 



o 

c 

o 

S-H 

O 



30000 



25000 



20000 



15000 



10000 



5000 








1 



7 10 13 16 19 22 25 28 31 34 37 40 

Cycle # 



Figure 2.1. Detection of foodborne pathogen X in foods by real time PCR. As ampli- 
con is synthesized, the thermocycler continuously measures fluorescences with each 
cycle. The PCR product fluoresces due to binding of fluorescent dye, SYBER Green 
to the double stranded DNA, amplicon as it is formed. When the PCR amplicons are 
first detected during real time, is a function of the target DNA concentration: ■ (100 
pg), ▲ (10 pg), • (1 pg), (0.1 pg), O (0.01 pg), and + NO DNA control. Arrows iden- 
tify the "threshold cycle," C T on the x-axis, # PCR cycles where the log-linear phase of 
fluorescence begins. The cycle numbers the target DNA concentration was plotted rel- 
ative to C T and as shown in the inset, there is a negative, linear correlation between 
DNA concentration and C 



T 



30 



The Mythology of PCR: A Warning to the Wise 



400000 



Tm 54°C 



350000 



300000 



250000 



H 

T3 



200000 



150000 



100000 



50000 








oj> o$> t>> & ^ ^ S % <^ b b ^ <\* ^ <*> % b 

Tm (°C) 



Figure 2.2. Identifying nonspecific PCR amplicons in real time PCR. We can distin- 
guish nonspecific from specific amplicons by measuring the melting temperature (Tm) 
for each amplicon following real-time PCR. The melting temperature is a reflection of 
the amplicons's nucleotide sequence, therefore one looks to see if the DNA melting 
curve for the putative, PCR-positive sample (□) overlaps with that of the positive con- 
trol (♦) or produces a different melting curve (A), that is indicative of a nonspecific 
amplicon. 



When we do not observe, directly or indirectly, any PCR product or ampli- 
con of the expected size, the finding is reported as negative. What does a nega- 
tive result mean? For a pure culture, it means our isolate does not possess the 
gene or gene allele to which our PCR was designed to detect. PCR has become 
an important diagnostic tool not only in identifying medically important gen- 
era (40, 58), but it has been used to identify an organism to species (9, 19, 23, 
40), or serotype level (6, 21, 26, 42, 50, 66) as well as determine its antibiotic 
resistance (20, 27) or virulence potential (2, 55). Depending on the organism 
and gene(s) or gene alleles associated with resistance to drug X, PCR negative 
result may indicate: (1) the organism is susceptible to the antibiotic in question 
(e.g., mycobacterium and isonazid resistance; 27); or (2) PCR negative only 
means the organism does not possess this gene (e.g.,enterococci and strep- 
togramin resistance; 69) and susceptibility cannot be inferred. Gene screens to 
assess, genotypically, drug resistance is challenging due to multiple genes and 
gene alleles associated with resistance to certain antibiotics (8, 15, 64). With 
regards to detection of multi-drug resistant (MDR) pathogens, while it is 



Validation 31 



tempting to select antibiotic resistance genes associated with the MDR as 
probes in PCR screens (31), mobile genetic elements have disseminated these 
drug resistance genes to innocuous commensals also contaminating foods 
(32, 63, 74), providing potential for false positives. Gene screens for these MDR 
loci should therefore be limited to the cultured pathogen. For detection of 
pathogens in foods, it is imperative that we select a target gene or sequence that 
is unique to pathogen X, uniform in its distribution within this bacterial popu- 
lation and genetically stable. 

If this target gene is strongly associated with genus, species or serotype, a 
negative PCR translates to this isolate is NOT the species, strain, or serotype 
identified by this PCR. However, if we apply this very same PCR to screen for 
the presence of the organism that the PCR was designed to identify, does a neg- 
ative result mean it is NOT present? We now are confronted with several ques- 
tions relating to our PCR test's sensitivity and specificity (28, 39), important in 
assessing, validating and finally standardizing our PCR for screening pathogens 
in foods (30). 



VALIDATION 



In optimizing any PCR, we strive to design, identify and develop the primer 
set(s) for discerning the one genus, species, or strain from multitude of micro- 
bial species while being able to detect the fewest cells possible. This is the molec- 
ular biology definition of specificity and sensitivity, respectively. To determine 
specificity, we test our PCR against, many different bacterial strains, closely or 
distantly related species and/or genera. A PCR specific for Salmonella, for 
example, will produce positive results, amplicon of the expected size, for ALL 
Salmonella species, strains, and serovars but will prove negative for all other 
bacterial species, especially closely related species (28, 58). If we continue using 
Salmonella as our example, sensitivity is measured by lowest Salmonella cell 
density detectable by our PCR (28, 39). In its infancy, PCR's specificity and sen- 
sitivity were determined using pure cultures and at best a food product was 
spiked with the offending organism and PCR was performed to detect the 
organism in the processed sample. Only recently have investigators vigorously 
put PCR through its paces in the real world to validate its utility for rapid detec- 
tion of pathogens in foods (30). 

Validation of any diagnostic PCR involves comparison against another 
test, considered the "gold standard" for detection. For food microbiologists, 
the "gold standard" is the bacteriological approach of culture, isolation, and 
the biochemical or serological confirmatory tests. From this comparison, we 
determine statistical specificity (false-/?ositives) and sensitivity (false- 
negatives) of our PCR test (28, 39). A false-positive is when the sample is 
PCR positive but culture negative, while a false-negative is vice-versa: PCR 
negative, culture positive. What is responsible for reporting false-positives 
and false-negatives and what can we do to minimize this in our food microbiol- 
ogy lab? 



32 The Mythology of PCR: A Warning to the Wise 



PROBLEMS AND THEIR SOLUTIONS 



False positives can be attributed to several things, most you cannot control, but 
at least one you can: PCR, template, or sample contamination. As discussed in 
Chapter 4, "Making PCR a Normal Routine of the Food Microbiology Lab," 
preventative measures and standard operating procedures are essential to avoid 
these contamination issues. These measures include physically separating DNA 
and PCR preparation areas from each other as well as from the area where gel- 
electrophoresis is performed; use of barrier tips, disposable gloves; and cleaning 
the PCR preparation area with bleach and/or overnight, ultraviolet illumina- 
tion. As mentioned earlier, PCR amplifies target gene 10 9 -fold, producing more 
than enough molecules per pg-fg of template to serve as template in the next 
PCR reaction. Following a PCR run and upon opening the tube, we create an 
aerosol of amplicons that can quickly contaminate our hands, pipettes, and the 
immediate bench area. Something as simple as disposing of our gloves follow- 
ing the loading of our PCR sample in the agarose gel and before we set up our 
next PCR reaction, can avoid future PCR "carry-over" contamination. PCR 
contamination results in considerable down time for the diagnostic laboratory 
due to the time it takes to identify the source of contamination, and subsequent 
decontamination of the affected area or disposal of the contaminated 
reagent(s). Alternatively, some labs substitute thymidine with uracil in the PCR 
reaction mix and subsequent pretreatment of all PCR reaction mixes with 
uracil DNA glycosylase prior to running these reactions in the thermocycler 
(41). The principle behind this method is that during PCR, amplicons incorpo- 
rate uracil; the amplicon is now susceptible to hydrolysis by uracil DNA glyco- 
sylase, and eliminated prior to each subsequent PCR run. Therefore, erroneous 
reporting of false-positives due to PCR contamination is eliminated. 

As synthesis of the amplicons is identified in "real-time" with newer, PCR flu- 
orescence-based detection technologies, tubes never have to be opened following 
the initial PCR reaction set-up. With conventional PCR, we can identify PCR con- 
tamination when negative or NO DNA controls turn positive. For an experienced 
lab, something is amiss, when the number of PCR positives greatly exceeds fre- 
quency the lab normally encounters for this PCR test or incidence reported in the 
literature AND subsequent culture results do not correlate with the PCR (i.e., 
increase in false positives). This can be observed with real-time PCR as lower C T 
than encountered for past PCR-positive samples, indicative of high-cell density or 
template/target concentration, and fails to yield the organism upon culture. As 
PCR can be extremely sensitive, great care must be taken in sample preparation to 
avoid cross-contamination. Inclusion of a negative control, sample prep with every 
PCR run will be useful in identifying cross-contamination, as a positive PCR for 
this negative control would definitely be indicative of template/sample contamina- 
tion. Anytime when its evident there is PCR contamination, discard the results for 
that PCR run, discontinue any future PCRs, and identify and correct the problem 
immediately before rerunning PCR on any past or future samples. 

Nonspecific PCR amplicons can also result in erroneously reporting a sam- 
ple positive for pathogen X. This can be especially problematic for real-time 



Problems and Their Solutions 33 



PCR using SYBR Green® to detect PCR products and multiplex PCR: the mul- 
tiple gene screens, single PCR test where the size of the amplicon identifies: 
genus, species, serotype, or strain. For real-time PCR, we can identify this prob- 
lem by measuring the amplicon's Tm from the DNA melting peak as stated ear- 
lier or run sample PCR on gel side-by-side with positive control to see any 
differences between the two in their size. Tweaking the PCR conditions to 
improve its stringency can sometimes eliminate these nonspecific amplicons. 
This can be done by empirically identify annealing temperature or MgCl 2 con- 
centration, that eliminates signal for our "false-positive" sample while not 
affecting the positive control. To increase the stringency of the PCR, one 
increases the annealing temperatures and/or lower the MgCl 2 concentrations in 
the reaction mix. 

Another way we can improve the specificity of our PCR and reporting false 
positives, is to use PCR that incorporates internal: nested PCR primers (39); 
DNA: DNA hybridization or capture probes (28, 45); molecular beacon (37); or 
TaqMan probe (29). These PCRs have improved specificity because the internal 
capture or detection probes can distinguish between the real amplicon and non- 
specific amplicons, by binding to the complementary sequence within the target 
amplicon during PCR or at DNA: DNA hybridization step. These internal probes 
also heighten the sensitivity of the PCR at least 100-fold (28, 45). 



False-Positives and Dead vs. Live Bacterial Cell Debate. Even when PCR is run- 
ning optimally, there may not always be 100% agreement between PCR and cul- 
ture. The reasons for false-positives are not completely understood. Several 
explanations have been offered and include: (1) the bacteria are in a viable, non- 
culturable state (61); (2) injury of the bacterial cells during food processing (52); 
or (3) the bacteria are dead (48). One can obtain variable culture results alone 
depending on: (1) whether to include a step(s) that allows for the recovery of 
injured cells (13, 33); (2) the type of enrichment broth (18) and culture conditions 
(12) used, or (3) the use of a delayed, secondary enrichment (72, 73) and may 
explain the disconnect sometimes observed between PCR and culture results. 

Depending on where samples are taken within the continuum of food pro- 
cessing steps, especially at Critical Control Point (CCP) designed to reduce or 
eliminate the pathogen, (e.g., heating), PCR may not be able to distinguish live, 
dead or damaged cells. In fact, we routinely boil bacteria or wash cells in ethanol 
to prepare template for PCR, conditions that readily and rapidly kill bacteria. 
Therefore, one may consider where and when to use PCR in assessing product 
contamination with pathogen X. For a process that readily ruptures or dissolves 
the bacterial cell, pre-DNAse treatment step can remove residual DNA carried 
over from dead, lysed cells (51). However, a significant proportion of heat- 
treated cells remain intact, dead, and suitable as template for PCR (51). We still 
need to know whether CCP was effective at eliminating the pathogen or reduc- 
ing it to an acceptably safe level. PCR still affords us the opportunity to identify 
the few cells still viable following CCP step, (e.g., pasteurization), by using RNA 
as the template. Unlike DNA, RNA has short-half life in the bacteria cell (34), 
as genes are turned on and off as the cell grows and responds to its environment. 



34 The Mythology of PCR: A Warning to the Wise 



Upon cell death, these mRNA transcripts are quickly degraded. There has been 
considerable interest in using RNA as the template for diagnostic PCR to detect 
the few viable cells remaining in the sample (17, 46, 52, 65, 75). This can be 
accomplished by converting RNA to its complementary (c) DNA copy with the 
retroviral reverse transcriptase, at which point the cDNA can serve as template 
in standard PCR. This procedure is referred to as reverse-transcriptase PCR. 
The challenge currently is identifying a constitutively, expressed gene that has 
sequence unique to the organism and has a short, mRNA half-life, especially 
upon death of the bacterial cell (75). RNA turnover in the bacterial cell is 
dependent on its intracellular ribonucleases, and like any enzyme once denatured 
it becomes inactive and the RNA therefore persists, which may explain the long 
half-life of RNAs following thermal inactivation of the bacterial cell (47). 
Therefore, there are times when culture continues to be necessary in assessing 
microbial risk following food processing step at CCP and other instances where 
PCR trumps culture in the detection of foodborne pathogens (see below). 

Finally, we are left to consider viable but ^onculturable (VBNC) bacteria and 
PCR. We know bacteria can enter a physiological state where, with the micro- 
scope, we know they are present and viable, as determined using viability stains, 
but we are unable to plate them from sample X. This VBNC state may result 
from cellular injury (14), adaptation to a harsh, oxygen-poor or nutrient deplete 
environment (5, 7, 71) or subsequent transformation from planktonic to sessile 
state in biofilms (11). In foods, the VBNC state may be the consequence of cel- 
lular injury/damage and may require a recovery period, in a preenrichment 
broth, before the cells can be cultivated. Organisms like Vibrio and 
Campylobacter can readily enter VBNC state, especially in aquatic environments 
(54, 60). Although regarded as a foodborne pathogen, Campylobacter is also 
recognized as the cause of several waterborne outbreaks in the United States (4, 
36). With Campylobacter, the VBNC state may be due to physical or chemical 
agent that damages the cell, or nutrient depletion or limitation triggers a physi- 
ological change to a survival state. When Campylobacter enters the VBNC state, 
its cell morphology changes from helical to coccoidal. Pathogens can revert back 
from this nongrowing, VBNC state into actively growing; cultivatable state, 
under the right conditions in vitro (7, 71) and cause disease in its animal host 
(53). It may be that we are unable to detect it in this state using our current selec- 
tive, enrichment media because of the antibiotics in the media that interfere with 
cellular repair and changes to the cell wall necessary to resume growth (67, 68). 
Where our culture-based approaches currently have failed, PCR offers the 
opportunity for the pathogen's detection, especially in its VBNC state (3, 49, 52). 

PCR Inhibitors, Limits of Detection, and False-Negatives. False-negatives, PCR- 
negative, culture positive samples are attributed to two major factors: PCR 
inhibitors or the PCR's limit of detection. PCR inhibitors may be attributed to 
the food sample itself or the enrichment used to amplify the target organism. 
We can often remove these inhibitors by using simple DNA affinity, spin 
columns to produce clean DNA template for PCR, making samples generally 
recalcitrant to PCR (e.g., soil) pliable for PCR-based screens and analyses. 



Conclusions 35 



Chapter 4, "Sample Preparation for PCR" will go into more detail concerning 
sample preparation and preparation of template that is free of PCR inhibitors. 
More recently, diagnostic PCRs for screening foods have been adapted to include 
an "internal control" in the sample screened in order to eliminate possibility of 
extraneous factors (e.g., PCR inhibitors) from factoring into interpretation of 
PCR negative results. "Internal amplification control" is the cloned, positive- 
control amplicon where an internal region has been removed (44). As template 
in PCR, "internal amplification control" produces a smaller sized-amplicon. The 
plasmid DNA bearing our "internal amplification control" is included with sam- 
ple template in PCR. If the sample is negative for organism X, a single amplicon, 
corresponding in size to that expected for the "internal amplification control." 
However, for a positive sample, two amplicons are produced; one that corre- 
sponds in size to that expected from amplification of the organism X's targeted 
gene and the other corresponds in size to that expected for the internal control. 
For most PCR beginners, false-negatives due to PCR's insensitivity to detect 
a single-cell per sample appear to be a paradoxical, if not a heretical statement. 
You have probably read many research papers and believe their claim that their 
PCR can detect a single cell/ml of a sample. Is this really possible? With PCR, 
we are generally dealing with reaction mix volumes that range between 1 and 
100 |il to which we may add 1 or 10 |il of the sample, once its been processed 
for PCR. What is the probability that you detect lcell/ml by PCR, if you were 
to take 0.001 ml or 1 |il, once from that sample? Knowing Poisson distribution, 
we know that odds are very small that we can detect it. However, if we took 
multiple aliquots from this same sample, a most-probable number approach, we 
would improve our chances of detecting this organism by PCR. The reality is 
that for most PCRs the limit of detection is 1-1000 cells per 1 |il sample tem- 
plate run, which translates to 1,000-1 x 10 6 cells/ml. Therefore, if we relied on 
PCR alone, and discounting PCR inhibitors, does a PCR negative sample mean 
the organism is NOT present? Ideally, one wants to use the PCR that is the most 
sensitive for identifying pathogen X in our food product. How might we 
improve our chances of detecting our pathogen knowing these limitations and 
assuming the organism might be present in our specimens at levels <1000 
cells/ml? One approach is to concentrate cells into a smaller volume, or include 
an enrichment step that amplifies what few cells are present to levels above the 
PCR's threshold for detection (Chapter 3). For the latter, short enrichment 
period may be sufficient to bring cell density of the pathogen above the detec- 
tion limits of the PCR. Enrichments have been especially adapted to PCR pro- 
tocols for foods due to the necessity of processing the large sample volumes 
associated with screening foods from pathogens. 



CONCLUSIONS 



One must be aware of the limitation of any diagnostic test, and PCR is no 
exception. Will PCR soon be the substitute for current culture or immunologi- 
cal tests for foodborne pathogens? Probably not for all pathogens, but it will 



36 The Mythology of PCR: A Warning to the Wise 



become standard tool for detecting foodborne protozoans and viruses, 
pathogens that are currently recalcitrant to culture-based methods of detection. 
PCR will become an important tool in identification of serotypes and patho- 
types. It can be a useful part of any detection scheme, helping with decisions as 
to which samples and enrichments to focus our efforts towards (39). Of course, 
acceptance and implementation of PCR in the diagnostic laboratory requires 
an understanding of its mechanics, meaning of results, the test's limitations, and 
being able to recognize problems and trouble-shoot them as they arise. 



REFERENCES 



1. Ausubel, F.M., R. Brent, R.E. Kingston, D.D. Moore, J.G. Seidman, J. A. Smith, and 
K. Struhl. 1992. Short Protocols in Molecular Biology, 2nd edn. John Wiley & Sons, 
New York. 

2. Bhaduri, S., B. Cottrell, and A.R. Pickard. 1997. Use of a single procedure for selective 
enrichment, isolation, and identification of plasmid-bearing virulence Yersinia enter o- 
colitica of various serotypes from pork. Appl. Environ. Microbiol. 63:1657-1660. 

3. Binsztein, N., M.C. Costagliola, M. Pichel, V. Jurquiza, EC. Ramirez, R. Akselman, 
M. Vacchino, A. Huq, and R. Colwell. 2004. Viable but nonculturable Vibrio 
cholerae Ol in the aquatic environment of Argentina. Appl. Environ. Microbiol. 
70:7481-7486. 

4. Blackburn, B.G., G.F. Craun, J.S. Yoder, V. Hill, R.L. Calderon, N. Chen, S.H. Lee, 
D.A. Levy, and M.J. Beach. 2004. Surveillance for waterborne-disease outbreaks 
associated with drinking water — United States, 2001-2002. MMWR Surveill. 
Summ. 53:23-45. 

5. Boaretti, M., M. del Mar Lieo, B. Bonato, C. Signoretto, and P. Canepari. 2003. 
Involvement of rpoS in the survival of Escherichia coli in the viable but noncultur- 
able state. Environ. Microbiol. 5:986-996. 

6. Borucki, M.K. and D.R. Call. 2003. Listeria monocytogenes serotype identification 
by PCR. J. Clin. Microbiol. 41:5537-5540. 

7. Bovill, R.A. and B.M. Mackey 1997. Resuscitation of 'nonculturable' cells from 
aged cultures of Campylobacter jejuni. Microbiol. 143:1575-1581. 

8. Bradford, PA. 2001. Extended-spectrum beta-lactamases in the 21st century: char- 
acterization, epidemiology, and detection of this important resistance threat. Clin. 
Microbiol. Rev. 14:933-951. 

9. Bubert, A., I. Hein, M. Rauch, A. Lehner, B. Yoon, W. Goebel, and M. Wagner. 
1999. Detection and differentiation of Listeria spp. by a single reaction based on 
multiplex PCR. Appl. Environ. Microbiol. 65:4688^-692. 

10. Bubert, A., S. Kohler, and W. Goebel. 1992. The homologous and heterologous 
regions within the iap gene allow genus- and species- specific identification of 
Listeria spp. by polymerase chain reaction. Appl. Environ. Microbiol. 58:2625-2632. 

11. Buswell, CM., YM. Herlihy, L.M. Lawrence, XT. McGuiggan, PD. Marsh, C.W. 
Keevil, and S.A. Leach. 1998. Extended survival and persistence of Campylobacter 
spp. in water and aquatic biofilms and their detection by immunofluorescent-anti- 
body and rRNA staining. Appl. Environ. Microbiol. 64:733-741. 

12. Carlson, V.L., GH. Snoeyenbos, B.A. McKie, and C.F. Smyser. 1967. A comparison 
of incubation time and temperature for the isolation of Salmonella. Avian Dis. 
11:217-225. 



References 37 



13. Chang, V.R, E.W. Mills, and C.N. Cutter. 2003. Comparison of recovery methods 
for freeze-injured Listeria monocytogenes, Salmonella Typhimurium, and 
Campylobacter coli in cell suspensions and associated with pork surfaces. J. Food 
Prot. 66:798-803. 

14. Chaveerach, P., A.A. ter Huurne, L.J. Lipman, and F. van Knapen. 2003. Survival 
and resuscitation of ten strains of Campylobacter jejuni and Campylobacter coli 
under acid conditions. Appl. Environ. Microbiol. 69:711-714. 

15. Chopra, I. and M. Roberts. 2001. Tetracycline antibiotics: Mode of action, applica- 
tions, molecular biology, and epidemiology of bacterial resistance. Microbiol. Mol. 
Biol. Rev. 65:232-260. 

16. Christensen, H., K. Jorgensen, and J.E. Olsen. 1999. Differentiation of 
Campylobacter coli and C jejuni by length and DNA sequence of the 16S-23S rRNA 
internal spacer region. Microbiol. 145:99-105. 

17. Coutard, F, M. Pommepuy, S. Loaec, and D. Hervio-Heath. 2005. mRNA detection 
by reverse transcription-PCR for monitoring viability and potential virulence in a 
pathogenic strain of Vibrio parahaemolyticus in viable but nonculturable state. 
J. Appl. Microbiol. 98:951-961. 

18. D'Aoust, J.Y., A.M. Sewell, and D.W. Warburton. 1992. A comparison of standard 
cultural methods for the detection of foodborne Salmonella. Int. J. Food Microbiol. 
16:41-50. 

19. Di Pinto, A., G. Ciccarese, G. Tantillo, D. Catalano, and V.T. Forte. 2005. A colla- 
genase- targeted multiplex PCR assay for identification of Vibrio alginolyticus, 
Vibrio cholerae, and Vibrio parahaemolyticus. J. Food Prot. 68:150-153. 

20. E-Hajj, H.H., S.A. Marras, S. Tyagi, F.R. Kramer, and D. Alland. 2001. Detection 
of rifampin resistance in Mycobacterium tuberculosis in a single tube with molecular 
beacons. J. Clin. Microbiol. 39:4131-4137. 

21. Fratamico, P.M., S.K. Sackitey, M. Wiedmann, and M.Y. Deng. 1995. Detection of 
Escherichia coli 0157:H7 by multiplex PCR. J. Clin. Microbiol. 33:2188-2191. 

22. Frenay, H.M.E., J.P.G Theelen, L.M. Schouls, CM. J.E. Vandenbroucke-Grauls, 
J. Verhoef, W.J. van Leeuwen, and F.R. Mooi. 1994. Discrimination of epidemic and 
nonepidemic methicillin-resistant Staphylococcus aureus strains on the basis of pro- 
tein A gene polymorphisms. J. Clin. Microbiol. 32:846-847. 

23. Gonzalez, L, K.A. Grant, P.T. Richardson, S.F Park, and M.D. Collins. 1997. 
Specific identification of the enteropathogens Campylobacter jejuni and 
Campylobacter coli by using a PCR test based on the ceuE gene encoding a putative 
virulence determinant. J. Clin. Microbiol. 35:759-763. 

24. Graham, T., E.J. Golsteyn-Thomas, V.R Gannon, and J.E. Thomas. 1996. Genus- 
and species-specific detection of Listeria monocytogenes using polymerase chain 
reaction assays targeting the 16S/23S intergenic spacer region of the rRNA operon. 
Can. J. Microbiol. 42:1155-1162. 

25. Gurtler, V. 1993. Typing of Clostridium difficile strains by PCR-amplification of 
variable length 16S-23S rDNA spacer regions. J. Gen. Microbiol. 139:3089-3087. 

26. Herrera-Leon, S., J.R. McQuiston, M.A. Usera, P.I. Fields, J. Garaizar, and M.A. 
Echeita. 2004. Multiplex PCR for distinguishing the most common phase- 1 flagel- 
lar antigens of Salmonella spp. J. Clin. Microbiol. 42:2581-2586. 

27. Herrera-Leon, L., T. Molina, P. Saiz, J.A. Saez-Nieto, and M.S. Jimenez. 2005. New 
multiplex PCR for rapid detection of isoniazid-resistant Mycobacterium tuberculosis 
clinical isolates. Antimicrob. Agents Chemother. 49:144-147. 

28. Hong, Y, M. Berrang, T. Liu, C. Hofacre, S. Sanchez, L. Wang, and J.J. Maurer. 
2003. Rapid detection of Campylobacter coli, C jejuni and Salmonella enterica on 



38 The Mythology of PCR: A Warning to the Wise 



poultry carcasses using PCR-enzyme-linked immunosorbent assay. Appl. Environ. 
Microbiol. 69:3492-3499. 

29. Hoorfar, J., P. Ahrens, and P. Radstrom. 2000. Automated 5' nuclease PCR assay for 
identification of Salmonella enterica. J. Clin. Microbiol. 38:3429-3435. 

30. Hoorfar, X, P. Wolffs, and P. Radstrom. 2004. Diagnostic PCR: Validation and sam- 
ple preparation are two sides of the same coin. APMIS 112:808-814. 

31. Khan, A. A., M.S. Nawaz, S.A. Khan, and C.E. Cerniglia. 2000. Detection of mul- 
tidrug-resistant Salmonella typhimurium DTI 04 by multiplex polymerase chain reac- 
tion. FEMS Microbiol. Lett. 182:355-360. 

32. Keyes, K., C. Hudson, J.J. Maurer, S.G Thayer, D.G. White, and M.D. Lee. 2000. 
Detection of florfenicol resistance genes in Escherichia coli isolated from sick chick- 
ens. Antimicrob. Agents Chemother. 44:421-424. 

33. Knabel, S.J., H.W Walker, P.A. Hartman, and A.F. Mendonca. 1990. Effects of 
growth temperature and strictly anaerobic recovery on the survival of Listeria mono- 
cytogenes during pasteurization. Appl. Environ. Microbiol. 56:370-376. 

34. Kushner, S.R. 1996. mRNA decay, pp. 849-860. In EC. Neidhardt (ed.), Escherichia 
coli and Salmonella: Cellular and Molecular Biology. ASM Press, Washington, DC. 

35. Kuwahara, T, I. Norimatsu, H. Nakayama, S. Akimoto, K. Kataoka, H. Arimochi, 
and Y. Ohnishi. 2001. Genetic variation in 16S-23S rDNA internal transcribed 
spacer regions and the possible use of this genetic variation for molecular diagnosis 
of Bacteroides species. Microb. Immunol. 45:191-199. 

36. Lee, S.H., D.A. Levy, G.F. Craun, M.J. Beach, and R.L. Calderon. 2002. 
Surveillance for waterborne-disease outbreaks — United States, 1999-2000. Morb. 
Mort. Week. Rep. 51(SS-8):l-66. 

37. Liming, S.H. and A.A. Bhagwat. 2004. Application of a molecular beacon-real-time 
PCR technology to detect Salmonella species contaminating fruits and vegetables. 
Int. J. Food Microbiol. 95:177-187. 

38. Liu, T, M. Garcia, S. Levisohn, D. Yogev, and S.H. Kleven. 2001. Molecular vari- 
ability of the adhesion-encoding gene pvpA among Mycoplasma gallisepticum 
strains and its application in diagnosis. J. Clin. Microbiol. 39:1882-18888. 

39. Liu, T, K. Liljebjelke, E. Bartlett, C.L. Hofacre, S. Sanchez, and J.J. Maurer. 2002. 
Application of nested PCR to detection of Salmonella in poultry environments. 
J. Food Prot. 65:1227-1232. 

40. Logan, J.M., K.J. Edwards, N.A. Saunders, and J. Stanley. 2001. Rapid identification 
of Campylobacter spp. by melting peak analysis of bioprobes in real-time PCR. 
J. Clin. Microbiol. 39:2227-2232. 

41. Longo, M.C., M.S. Berninger, and J.L. Hartley. 1990. Use of uracil DNA glycosy- 
lase to control carry-over contamination in polymerase chain reactions. Gene 
93:125-128. 

42. Luk, J.M., U. Kongmuang, PR. Reeves, and A.A. Lindberg. 1993. Selective ampli- 
fication of abequose and paratose synthase genes (rfb) by polymerase chain reaction 
for identification of Salmonella major serogroups (A, B, C2, and D). J. Clin. 
Microbiol. 31:2118-2123. 

43. Mackay, I.M., K.E. Arden, and A. Nitsche. 2002. Real-time PCR in virology. 
Nucleic Acids Res. 30:1292-1305. 

44. Malorny, B., J. Hoorfar, C. Bunge, and R. Helmuth. 2003. Multicenter validation of 
the analytical accuracy of Salmonella PCR: Towards an international standard. 
Appl. Environ. Microbiol. 69:290-296. 

45. Maurer, J. J., D. Schmidt, P. Petrosko, S. Sanchez, L. Bolton, and M.D. Lee. 1999. 
Development of primers to O-antigen biosynthesis genes for specific detection of 
Escherichia coli 0157 by PCR. Appl. Env. Microbiol. 65:2954-2960. 



References 39 



46. Mclngvale, S.C., D. Elhanafi, and M.A. Drake. 2002. Optimization of reverse tran- 
scriptase PCR to detect viable shiga-toxin producing Escherichia coli. Appl. 
Environ. Microbiol. 68:799-806. 

47. McKillip, J.L., L.A. Jaykus, and M. Drake. 1998. rRNA stability in heat-killed and 
UV-irradiated enterotoxigenic Staphylococcus aureus and Escherichia coli 0157:H7. 
Appl. Environ. Microbiol. 64:4264-4268. 

48. McKillip, J.L., L.A. Jaykus, and M. Drake. 1999. Nucleic acid persistence in heat-killed 
Escherichia coli 0157:H7 from contaminated skim milk. J. Food Prot. 62:839-844. 

49. Moore, J., P. Caldwell, and B. Millar. 2001. Molecular detection of Campylobacter 
spp. in drinking, recreational and environmental water supplies. Int. J. Hyg. Environ. 
Health 204:185-189. 

50. Nachamkin, I., H. Ung, and CM. Patton. 1996. Analysis of HL and O serotypes of 
Campylobacter strains by flagellin gene typing system. J. Clin. Microbiol. 34:277-281. 

51. Nogva, H.K., A. Bergh, A. Hoick, and K. Rudi. 2000. Application of the 5'-nucle- 
ase PCR assay in evaluation and development of methods for quantitative detection 
of Campylobacter jejuni. Appl. Environ. Microbiol. 66:4029-4936. 

52. Novak, IS., and V.K. Juneja. 2001. Detection of heat injury in Listeria monocyto- 
genes Scott A. J. Food Prot. 64:1739-1743. 

53. Oliver, J.D., and R. Bockian. 1995. In vivo resuscitation, and virulence towards mice, 
of viable but nonculturable cells of Vibrio vulnificus. Appl. Environ. Microbiol. 
61:2620-2623. 

54. Oliver, J.D., F. Hite, D. McDougald, NX. Andon, and L.M. Simpson. 1995. Entry 
into, and resuscitation from, the viable but nonculturable state by Vibrio vulnificus 
in an estuarine environment. Appl. Environ. Microbiol. 61:2624-2630. 

55. Pass, M A., R. Odedra, and R.M. Batt. 2000. Multiplex PCRs for identification of 
Escherichia coli virulence genes. J. Clin. Microbiol. 38:2001-2004. 

56. Payne, R.E., M.D. Lee, D.W. Dreesen, and H.M. Barnhart. 1999. Molecular epi- 
demiology of Campylobacter jejuni in broiler flocks using randomly amplified poly- 
morphic DNA-PCR and 23S rRNA-PCR and role of litter in its transmission. Appl. 
Environ. Microbiol. 65:260-263. 

57. Pourcel, C, Y. Vidgop, F. Ramisse, G. Vergnaud, and C. Tram. 2003. 
Characterization of a tandem repeat polymorphism in Legionella pneumophila and 
its use for genotyping. J. Clin. Microbiol. 41:1819-1826. 

58. Rahn, K., S.A. Grandis, R.C. Clarke, S.A. McEwen, J.E. Galan, C. Ginocchio, R. 
Curtiss III, and C.L. Gyles. 1992. Amplification of invA gene sequence of 
Salmonella typhimurium by polymerase chain reaction as a specific method of detec- 
tion of Salmonella. Mol. Cell Probes 6:271-279. 

59. Ririe, K.M., R.P. Rasmussen, and C.T. Wittwer. 1997. Product differentiation by 
analysis of DNA melting curves during the polymerase chain reaction. Anal. 
Biochem. 245:154-160. 

60. Rollins, D.M. and R.R. Colwell. 1986. Viable but nonculturable stage of 
Campylobacter jejuni and its role in survival in the natural aquatic environment. 
Appl. Environ. Microbiol. 52:531-538. 

61. Sails, A.D., F.J. Bolton, A.J. Fox, D.R. Wareing, and D.L. Greenway 2002. Detection 
of Campylobacter jejuni and Campylobacter coli in environmental waters by PCR 
enzyme-linked immunosorbent assay. Appl. Environ. Microbiol. 68:1319-1324. 

62. Sambrook, J, E.F. Fritsch, and T. Maniatis. 1989. Molecular Cloning: A Laboratory 
Manual, 2nd edn. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. 

63. Sanchez, S., M.A. McCrackin Stevenson, C.R. Hudson, M. Maier, T. Buffington, Q. 
Dam, and J.J. Maurer. 2002. Characterization of multi-drug resistant Escherichia 
coli associated with nosocomial infections in dogs. J. Clin. Microbiol. 40:3586-3595. 



40 The Mythology of PCR: A Warning to the Wise 



64. Shaw, K.J., P.N. Rather, R.S. Hare, and G.H. Miller. 1993. Molecular genetics of 
aminoglycoside resistance genes and familial relationships of the aminoglycoside- 
modifying enzymes. Microbiol. Rev. 57:138-163. 

65. Sheridan, G.E.C., E.A. Szabo, and B.M. Mackey 1999. Effect of post-treatment 
holding conditions on detection of tufA mRNA in ethanol-treated Escherichia colt 
Implications for RT-PCR-based indirect viability tests. Lett. Appl. Microbiol 
29:375-379. 

66. Shi, F., Y.Y. Chen, T.M. Wassenaar, W.H. Woods, P.J. Coloe, and B.N. Frye. 2002. 
Development and application of a new scheme for typing Campylobacter jejuni and 
Campylobacter coli by PCR-based restriction fragment length polymorphism analy- 
sis. J. Clin. Microbiol. 40:1791-1797. 

67. Signoretto, C, M.M. Lleo, and P. Canepari. 2002. Modification of the peptidogly- 
can of Escherichia coli in the viable but nonculturable state. Curr. Microbiol. 
44:125-131. 

68. Signoretto, C, M.M. Lleo, M.C. Tafi, and P. Canepari. 2000. Cell wall chemical 
composition of Enterococcus faecalis in the viable but nonculturable state. Appl. 
Environ. Microbiol. 66:1953-1959. 

69. Simjee, S., D.G. White, J. Meng, D.D. Wagner, S. Qaiyumi, S. Zhao, J.R. Hayes, and 
PR McDermott. 2002. Prevalence of streptogramin resistance genes among 
Enterococcus isolates recovered from retail meats in the Greater Washington DC 
area. J. Antimicrob. Chemother. 50:877-882. 

70. Spinaci, C, G. Magi, C. Zampaloni, L.A. Vitali, C. Paoletti, M.R. Catania, M. 
Prenna, L. Ferrante, S. Ripa, PE. Varaldo, and B. Facinelli. Genetic diversity of cell- 
invasive erythromycin-resistant and -susceptible group A streptococci determined 
by analysis of RD2 region of the prtFl gene. J. Clin. Microbiol. 42:639-644. 

71. Wai, S.N., Y. Mizunoe, A. Takade, and S. Yoshida. 2000. A comparison of solid and 
liquid media for resuscitation of starvation- and low-temperature-induced noncul- 
turable cells of Aeromonas hydrophila. Arch. Microbiol. 173:307-310. 

72. Waltman, WD., A. Home, C. Pirkle, and T. Dickson. 1991. Use of delayed second- 
ary enrichment for the isolation of Salmonella in poultry and poultry environments. 
Avian Dis. 35:88-92. 

73. Waltman, W.D., A.M. Home, C. Pirkle. 1993. Influence of enrichment incubation 
time on the isolation of Salmonella. Avian Dis. 37:884-887. 

74. White, D.G, C.R. Hudson, 11 Maurer, S. Ayers, S. Zhao, M.D. Lee, L.F. Bolton, T. 
Foley, and J. Sherwood. 2000. Characterization of chloramphenicol and florfenicol 
resistance in bovine pathogenic Escherichia coli. J. Clin. Microbiol. 38:4593^4598. 

75. Yaron, S. and K.R. Matthews. 2002. A reverse transcriptase-polymerase chain reac- 
tion assay for detection of viable Escherichia coli 0157:H7: Investigation of specific 
target genes. J. Appl. Microbiol. 92:633-640. 



CHAPTER 3 



Sample Preparation for PCR 

Margie D. Lee 12 *, DVM, Ph.D. and Amanda Fairchild 1 , M.S. 

Poultry Diagnostic & Research Center 1 , College of Veterinary Medicine, 

The University of Georgia, Athens, GA 30602 

and 
Center for Food Safety 2 , College of Agriculture and Environmental Sciences, 

The University of Georgia, Griffin, GA 30223 



Introduction 

How Do You Get Started? 

What Conditions Affect the Success of the PCR? 

What Are PCR Inhibitors? 

Potential Solutions to the Challenges of Using PCR to Detect Pathogens in 

Foods 

References 



INTRODUCTION 



Adding PCR-detection to a laboratory's repertoire of tools can improve sample 
turn-around time and accuracy. Yet, PCR is not a universal solution for pathogen 
detection problems. For pathogens that are rapidly growing and contaminate 
foods in high numbers, culture onto selective and differential media may actually 
be more rapid and cost-effective than PCR. However, PCR can greatly improve 
turn-around time in instances of slow-growing pathogens and can improve detec- 
tion of pathogens present at low concentrations. Nevertheless, like any other pro- 
tocol, correct preparation of the samples is key to PCR's success. Figure 3.1 
shows a sample processing strategy for PCR. Specific steps in the processing strat- 
egy will vary depending on pathogen and foodstuff. Prior research may have 
shown whether pathogen amplification steps, such as enrichment culture, are 
needed prior to PCR, so check the published literature for relevant protocols. 



HOW DO YOU GET STARTED? 



Sample preparation serves several functions for PCR detection (14, 19). It ini- 
tially decreases sample volume and concentrates the PCR template into a work- 
able volume. The first challenge in choosing a good sample preparation protocol 
is to know whether the pathogen contaminates food at high levels or whether it 



Corresponding author. Phone: (706) 583-0797; FAX: (706) 542-5630; e-mail: 
leem@vet.uga.edu. 



41 



42 



Sample Preparation for PCR 



Collection of 
food sample 



I 



Process food 
sample 

Homogenization 
Washing 



I 



Concentrate Pathogen 

Enrichment 

Immunocapture 

Buoyant density centrifugation 



+ 



Template Extraction 

Heat 

Detergent 

Chemical 

Solvent 



I 



will be necessary to amplify the bacteria 
with an enrichment culture. For exam- 
ple, very few Listeria cells may be pres- 
ent on a slice of deli meat but these few 
bacteria may be enough to cause serious 
illness in a pregnant woman. It may be 
impossible to directly collect a few bac- 
terial cells and detect them using PCR. 
An enrichment culture can amplify the 
bacterial cells and the PCR can detect 
the bacteria in the enrichment broth 
more rapidly than they can be identified 
using standard bacteriological methods. 
In this instance PCR can aid in the rapid 
detection of Listeria and the sample 
preparation protocol will include per- 
forming the enrichment culture, collec- 
tion of bacteria from the enrichment 
broth, extraction of DNA from the bac- 
terial cells and then performing the PCR 
test. 

Once the pathogen is collected, 
PCR template must be prepared from 
its DNA (or RNA). The first step in 
preparing template from a pathogen 
requires lysis (rupture) of the cells (or 
viruses) to release the nucleic acids 
(DNA and RNA). Specific organisms 
may require specific protocols for effi- 
cient template extraction. For example, 
there are a few basic approaches to 
extracting nucleic acids from bacteria 
but their effectiveness depends on sev- 
eral features of the bacterial cell wall. Gram-negative bacteria lack a thick cell 
wall, thus heat or detergent can lyse the cells. Many of the published protocols for 
E. coli, Salmonella, and Campylobacter use this approach for lysis of the cells (see 
Table 3.1 for applications). Gram-positive bacteria have a thick cell wall that must 
be removed or disrupted in order to lyse the cells. Lysozyme (plus lysostaphin for 
Staphylococcus) digestion is commonly used, prior to detergent treatment, for 
nucleic acid extraction from gram-positive and gram-negative bacteria. A third 
method of bacterial cell lysis involves a high salt/chemical lysis with guanidium 
salts. This method is most commonly used for gram-positive bacteria but will 
work for gram-negative cells as well. Solvent extraction of nucleic acids, with 
organic solvents such as ether, can be used for bacteria, viruses, and protozoa. 
Commercial PCR detection kits will incorporate one or some derivation of these 
methods, but the methods have to be optimized for the specific organism. 



Concentrate Template 

Alcohol precipitation 
Binding matrices 



I 



PCR 



Figure 3.1. Sample preparation for PCR 
detection of foodborne pathogens. 



HOW DO YOU GET STARTED ? 



43 



Table 3.1. Sample preparation of foods for PCR 







Method to 


DNA (RNA) 




Food Category 




Concentrate 


Extraction 


Pathogens 


( Challenges) 


Sample 


Pathogen 


method 


(Reference) 


Dairy 










PCR inhibitors 


Skim milk, 


Differential 


Solvent-based 


E. coli 0157 


(fat, protein, 


pasteurized 


centrifugation 


nucleic acid 


(16) 


calcium, chelators), 


milk, dry 


or none 


extraction or 


Listeria (12) 


dead cells, low 


milk, hard and 




guanidinium 


Staphylococcus, 


numbers of 


soft cheese, 




isothiocyanate 


Yersinia (20) 


pathogen cells, 


reconstituted 




extraction 


Campylobacter 


other bacteria 


whey powder 






(24) 




Raw milk 


Centrifugation 


Boiled cells 
with Chelex- 
100 removal 
of inhibitors; 
Tth polyme- 
rase improved 
sensitivity 


Staphylococcus 
(10) 




Raw milk 


Enrichment 

and 

centrifugation 


Commercial 
kits 


Salmonella (5) 




Soft cheese 


None 


Detergent lysis 
with Nal 
extraction 
of DNA 


Listeria (15) 


Meat and poultry 










rinses 










PCR inhibitors (fat, 


Chicken 


Enrichment 


Commercial 


Listeria 


protein, collagen, 


carcass rinses, 


and 


kits 


Salmonella 


blood), small num- 


red meat 


centrifugation 




E. coli (4, 5) 


bers of bacteria, 












Homogenates 


Buoyant 


Guanidinium 


Campylobacter 




of chicken skin, 


density 


isothiocyanate 


(24) 




whole chicken 


centrifugation 


and detergent 






leg, chicken 




extraction 






sausages, turkey 










leg meat, 










ground beef, 










mince meat, 










beef, pork 










Raw whole 


Buoyant 


Boiled cells 


Campylobacter 




chicken rinses 


density centri- 
fugation and 
enrichment 
culture 




(27) 




Chicken and 


Enrichment 


Multiple meth- 


Salmonella (6) 




turkey muscle, 




ods firmed 






skin, internal 




including boiled 






organs; raw 




cells, alkaline 






carcasses 




lysis, and 
commercial kits 





Continued 



44 



Sample Preparation for PCR 



Table 3.1. Sample preparation of foods for PCR — cont'd 



Food Category 
( Challenges) 



Sample 



Method to 

Concentrate 

Pathogen 



DNA (RNA) 

Extraction 

method 



Pathogens 
(Reference 



Meat and poultry 










rinses 


Ground beef 


Buoyant 

density 

centrifugation, 

immunoma- 

gnetic 

separation, 

enrichment 


Boiled cells 
or Chelex- 
extraction 


E. coli (25) 




Ham 


Immuno- 
magnetic 
separation 


Lysozyme 
and detergent 
extraction 


Listeria (8) 




Minced Pork 


Enrichment 


Commercial 


Yersinia (13) 




meat, raw 


and buoyant 


extraction 






whole pork leg 


density 
centrifugation, 


buffer and 
heat 






Ground pork 


Enrichment 


Chelex resin- 
based 

commercial 
kit 


Yersinia (26) 




Sausage and 


Homogeni- 


Commercially 


Clostridium (11) 




meat rolls 


zation of 


available kits 






(Korean ethnic 


food then 


but increased 






foods) 


filtration and 
centrifugation 


Mg++ levels in 
samples 






Deli meats: 


None 


Commercial 


Norwalk-like 




ham, turkey, 




extraction 


virus 




roast beef 




solution 


Hepatitis A 
virus (22) 


Seafood 










PCR inhibitors 


Smoked 


Homogeni- 


Detergent 


Listeria (23) 


(phenolics, 


salmon 


zation of 


extraction 




cresols, aldehydes, 




food 


and Tween 




proteins, fats), 






20 facilitator; 




low numbers of 






PCR 




bacteria 






inhibitors 
removed by 
solvent 
extraction 
or column 
purification 






Fish cakes, 


Enrichment 


Detergent and 


Listeria (1) 




fish pudding, 




boiling for 






peeled frozen 




extraction 






shrimp, salted 










herring, 










marinated and 










sliced coalfish 










in oil 









What Conditions Affect the Success of the PCR 



45 



Produce 
PCR inhibitors 
(chelators), few 
bacteria 



Shellfish: 


Homogeni- 


Guanidinium 


Norwalk-like 


muscles and 


zation of 


thiocyanate 


virus, 


oysters 


food then 


and silica 


Adenovirus 




high-speed 


purification 


Enterovirus, 




centrifugation 




Hepatitis 
A virus (7) 


Raw Oysters 


Homogeni- 


Commercial 


Norovirus (17) 




zation then 


kit 






buoyant 








density 








centrifugation 






Whole 


Column 


Commercial 


Protozoa (18) 


raspberries 


filtration and 


kit (FTA 






centrifugation 


filter) 




Lettuce 


Homogeni- 


Commercial 


Hepatitis A 




zation, centri- 


kits 


virus 




fugation, and 




Norwalk virus 




precipitation 




(21) 




with 








polyethylene 








glycol 







The next step in sample processing is to concentrate the template and reduce 
the concentration of PCR inhibitors. The specific approach will vary depending 
on whether DNA or RNA is desired as template and the chemical composition 
of the PCR inhibitors present in the sample. Phenol/chloroform extraction 
steps can reduce protein and lipid inhibitors. Other chemical inhibitors can be 
diluted by washing bound (silica beads or column matrices) or precipitated 
(ethanol or propanol) nucleic acids. An effective protocol for removing 
inhibitors must be developed for each specific food. Then the specific PCR can 
be performed for the pathogens of interest. 



WHAT CONDITIONS AFFECT THE SUCCESS OF THE PCR? 



The purpose of PCR is to detect the organism's specific nucleic acids in the sam- 
ple so that time-consuming biochemical and immunological assays are not 
needed. PCR causes the synthesis of DNA using an enzymatic reaction that 
cycles over and over due to the temperature cycling of the thermal cycler. 
Enzymes, including the polymerases that are used in PCR, must have specific 
chemical conditions in order to do work effectively. One of the major concepts 
of PCR is that the polymerase can exponentially increase the amount of DNA 
in the sample because of the temperature cycling (9). However, if the conditions 
are not optimal, the polymerase may not be able to synthesize enough DNA for 
the reaction to be detected as positive; these are called "false-negative" reac- 
tions. There are many situations where the PCR reaction can be suboptimal and 



46 Sample Preparation for PCR 



produce false negative results. Incorrect primers, buffer composition, cation 
(Mg++) concentration, nucleotide concentration (dNTPs), the wrong annealing 
temperature, extension cycles that are too brief, and incorrect template can 
cause the reaction to be falsely positive or negative. Always include two nega- 
tive controls: a different organism's DNA and a control with no DNA template. 
These will help you determine the specificity of your PCR and whether you 
have sample contamination. In addition, always include a positive control with 
DNA template that you know will amplify in the PCR. These controls can help 
identify the problem when the PCR is inhibited. For example, different poly- 
merases need different cations in the buffer in order to synthesize DNA. Taq 
polymerase uses magnesium (Mg++) therefore too little Mg++ in the master mix 
will result in a negative PCR reaction. The nucleotide concentration is impor- 
tant as well; they will chelate the Mg++ if you use too much of the dNTP mix. 
However, too much Mg++ will also result in a false negative reaction because 
there is a narrow window of effectiveness for the PCR to work. Every PCR 
reaction must be tested to determine the optimal concentration of Mg++ for the 
specific primers, buffer, and cycling temperatures. A similar situation also exists 
for the template concentration, too much or too little will result in a false neg- 
ative reaction. You should optimize the PCR, by running different concentra- 
tions of cation and template to find the concentrations that will produce the 
amplicon of the correct size (or melting temperature if you are using real-time 
PCR). If you are setting up a new PCR that you found in published literature, 
do not just assume that the published conditions will work for you. If you get 
into the habit of optimizing your PCR reaction conditions, you will seldom 
have a problem with your routine PCRs that you cannot quickly solve. 



WHAT ARE PCR INHIBITORS? 



The PCR reaction can be inhibited when substances bind (chelate) or degrade 
a component in the reaction and prevent it from participating in the synthesis 
of DNA (9, 28). These substances are called "PCR inhibitors" and include 
chelators of cations and substances that bind or degrade the polymerase or the 
DNA template. When pathogens are grown to high levels in culture, PCR tem- 
plate can often be made directly by chemical or enzymatic lysis of the organ- 
ism's cells. For example, DNA can easily be extracted from gram-negative 
bacteria by boiling the cells in water and using the boiled lysate as template. One 
important caveat, most enrichment broths and selective agars contain sub- 
stances that inhibit PCR so it will be important to wash the cells collected from 
an enrichment or agar plate. You can do this with bacteria by pelleting the cells 
using centrifugation, removing the liquid and resuspending the cells in saline or 
water for the DNA extraction. 

The real challenge is to isolate the pathogen and/or its DNA directly from a 
food matrix. The great variety of foodstuffs complicates any quest to produce a 
single sample preparation protocol that will work for every application. Unique 
PCR inhibitors are found in just about any food type including meat, milk, 



Potential Solutions to the Challenges of Using PCR to Detect Pathogens 47 



cheese, produce, and spices (28). Many of these have not been identified but 
some are known substances. For example, milk contains high levels of cations 
(Ca++), proteases, nucleases, fatty acids, and DNA. In addition, heme, bile salts, 
fatty acids, antibody, and collagen are PCR inhibitors that may be present in 
meat or liver. The inhibitors have variable effects on the PCR reaction but in 
general, they will make it more difficult to detect low numbers of bacterial cells 
or viruses. A good sample preparation protocol will focus on collecting the 
pathogen, removing the inhibitors present in the foodstuff (or culture medium) 
and concentrating the template for PCR. In addition, use of a polymerase that 
is less susceptible to the effects of inhibitory substances is a possible solution to 
some PCR problems. For example a number of the newer polymerases, such as 
Tfl and rTth, are more reliable than Taq polymerase when using PCR template 
made from meat or cheeses (2). Moreover, the activity of the polymerases, in the 
presence of inhibitors, can be improved with the use of some facilitators such 
as bovine serum albumin (BSA), dimethyl sulfoxide (DMSO), Tween 20 and 
betaine (2, 3, 19, 28). If you are trying to adapt a published PCR to a different 
food type, you may have to consider adding a facilitator or using a different 
polymerase to enhance sensitivity of the reaction. 



POTENTIAL SOLUTIONS TO THE CHALLENGES 
OF USING PCR TO DETECT PATHOGENS IN FOODS 



Foods differ greatly in their composition. The presence of fats, proteins, 
enzymes, chemical additives, fiber, and bacteria as well as ranges of pH influ- 
ence your ability to isolate the organism, its nucleic acids, and amplify its nucleic 
acids using PCR. In addition, nonpathogenic organisms present in fermented 
foods, the contaminating soil and manure organisms present on produce, and 
fecal contamination of meat will produce competing DNA that may reduce the 
sensitivity of the PCR reaction. Unless you are experienced in developing PCR 
reactions, you may not want to solve all of these problems yourself. Use proto- 
cols developed and validated by reputable labs. Note specific steps in the proto- 
col. Are enrichment steps such as immunomagnetic capture or enrichment 
culture needed to collect the organism from the sample? How are the organisms 
collected from the sample or the enrichment? How is the DNA extracted from 
the organism? What are the specific conditions of the PCR reaction? How will 
you detect the PCR amplicon? Do you have a positive control organism (or tem- 
plate) for the reaction? Once you have dissected out these important compo- 
nents from the publication or protocol, you can determine, which components 
can be modified for your specific needs. 

Table 3.1 illustrates some of the challenges and solutions for PCR detection 
of pathogens that contaminate foods. If the challenges are acknowledged, then 
possible solutions become feasible. If the pathogen contaminates the food in low 
numbers, then the pathogen must be amplified in some way. The important thing 
to know is whether the PCR can detect very low numbers of bacteria. 
Theoretically PCR can detect 1 pathogen in the reaction. Yet realistically, 



48 



Sample Preparation for PCR 



because of PCR inhibitors and other factors, you will usually need a substantial 
amount of template, from a few hundred to thousands of pathogen cells or 
viruses, in the PCR reaction in order to reliably detect the presence of the 
pathogen. In addition, if you consider that some food samples may only contain 
a few hundred pathogen cells per gram of food, the need for an enrichment step 
becomes apparent. Figure 3.2 shows how common pathogen amplification 
methods work to concentrate the pathogen in a volume that can be used for the 
next steps in the sample preparation . Enrichment culture is commonly used to 
amplify the bacterial numbers although immunological capture can theoretically 
be used for bacteria, viruses, and protozoa. Nevertheless, immunocapture 
requires the availability of an antibody that is specific for the pathogen. An 
immunocapture system works by binding one end of the antibody to a handling 
apparatus (such as magnetic beads in immunomagnetic separation) then expos- 
ing the other end of the antibody to the contaminated food. If the pathogen is 
present in the food, many of the cells or virus particles will be bound to the anti- 
body. This means that the immunocapture system can concentrate the pathogen 
onto the magnetic beads, which can then be used for enrichment or directly 
processed in a DNA extraction for PCR. Similarly, pathogens can be isolated 
from liquid samples by using centrifugation protocols that either float the 
bacteria or virus particles out of the sample (buoyant density centrifugation) or 
pellet the cells in the tube. Filtration of liquid samples may be effective for 



Centrifugation 



8000x£ 



► 



Suspension of Pellet of 
pathogen cells pathogen cells 



Enrichment culture 




Food sample 
(at least lgr) 



Incubate 



► 



Enrichment 
broth 



<J 



Suspension of 
pathogen cells 



Buoyant density centrifugation 



Immunocapture 




Homogenized 
food sample 



■* f- r 
; = = = 



1-5000xp 




► 



Layer of 

pathogen cells 

and food sample 



-i - r 



<— Layer of 
pathogen cells 



Homogenized 
food sample 



<j 



Add antibody-y 
coated beads^P^ 



► 



Suspension of 

food sample and 

pathogen cells 




Pellet of 
pathogen cells 



Figure 3.2. Approaches to concentrate or amplify pathogens. 



References 



49 



concentrating pathogens that pass through a 0.45 micron filter (such as 
Campylobacter and virus particles). For some pathogens where the infectious 
dose is very low, more than one concentration step may be needed in order to 
amplify the pathogen to a detectable level. Many studies have addressed these 
detection issues in developing PCRs for specific applications (Table 3.1). 



REFERENCES 



1. Agersborg, A., R. Dahl, and I. Martinez. 1997. Sample preparation and DNA 
extraction procedures for polymerase chain reaction identification of Listeria mono- 
cytogenes in seafoods. Int. J. Food Microbiol. 35:275-280. 

2. Al-Soud, W.A. and P. Radstrom. 1998. Capacity of nine thermostable DNA poly- 
merases to mediate DNA amplification in the presence of PCR-inhibiting samples. 
Appl. Environ. Microbiol. 64:3748-3753. 

3. Al-Soud, W.A. and P. Radstrom. 2000. Effects of amplification facilitators on diag- 
nostic PCR in the presence of blood, feces, and meat. J. Clin. Microbiol. 
38:4463-4470. 

4. Bhaduri, S. and B. Cottrell. 2001. Sample preparation methods for PCR detection of 
Escherichia coli 0157:H7, Salmonella typhimurium, and Listeria monocytogenes on 
beef chuck shoulder using a single enrichment medium. Mol. Cell. Probes 15:267-274 

5. Chen, S., A. Yee, M. Griffiths, C. Larkin, C.T. Yamashiro, R. Behari, C. Paszko- 
Kolva, K. Rahn, and S.A. De Grandis. 1997. The evaluation of a fluorogenic poly- 
merase chain reaction assay for the detection of Salmonella species in food 
commodities. Int. J. Food Microbiol. 35:239-250. 

6. De Medici, D., L. Croci, E. Delibato, S. Di Pasquale, E. Filetici, and L. Toti. 2003. 
Evaluation of DNA extraction methods for use in combination with SYBR Green I 
Real-Time PCR to detect Salmonella enterica Serotype Enteritidis in poultry. Appl. 
Environ. Microbiol. 69:3456-3461. 

7. Formiga-Cruz, M., G. Tofino-Quesada, S. Bofill-Mas, D.N. Lees, K. Henshilwood, 
A.K. Allard, A.-C. Conden-Hansson, B.E. Hernroth, A. Vantarakis, A. Tsibouxi, 
M. Papapetropoulou, M.D. Furones, and R. Girones. 2002. Distribution of human 
virus contamination in shellfish from different growing areas in Greece, Spain, 
Sweden, and the United Kingdom Appl. Environ. Microbiol. 68:5990-5998. 

8. Hudson, J.A., R.J. Lake, M.G Savill, P. Scholes and R.E. McCormick. 2001. Rapid 
detection of Listeria monocytogenes in ham samples using immunomagnetic separa- 
tion followed by polymerase chain reaction. J. Appl. Microbiol. 90:614-621. 

9. Kainz, P. 2000. The PCR plateau phase — Towards an understanding of its limita- 
tions. Biochimica et Biophysica Acta 1494:23-27. 

10. Kim, C.H., M. Khan, D.E. Morin, W.L. Hurley, D.N. Tripathy, M. Kehrli Jr., A.O. 
Oluoch, and I. Kakoma. 2001. Optimization of the PCR for detection of 
Staphylococcus aureus nuc gene in bovine milk. J. Dairy Sci. 84:74-83. 

11. Kim, S., R.G Labbe, and S. Ryu. 2000. Inhibitory effects of collagen on the PCR 
for detection of Clostridium per fringens. Appl. Environ. Microbiol. 66:1213-1215. 

12. Koo, K. and L.A. Jaykus. 2003. Detection of Listeria monocytogenes from a model 
food by fluorescence resonance energy transfer-based PCR with an asymmetric flu- 
orogenic probe set. Appl. Environ. Microbiol. 69:1082-1088. 

13. Lantz, P.G., R. Knutsson, Y. Blixt, W.A. Al-Soud, E. Borch, and P. Radstrom. 1998. 
Detection of pathogenic Yersinia enter ocolitica in enrichment media and pork by a 



50 Sample Preparation for PCR 



multiplex PCR: A study of sample preparation and PCR-inhibitory components. 
Int. J. Food Microbiol. 45:93-105. 

14. Lantz, P.G., W.A. Al-Soud, Ri. Knutsson, B. Hahn-Hagerdal, and P. Radstrom. 
2000. Biotechnical use of polymerase chain reaction for microbiological analysis of 
biological samples. Biotechnol. Ann. Rev. 5:130. 

15. Makino, S.I., Y. Okada, and T. Maruyama. 1995. A new method for direct detection of 
Listeria monocytogenes from foods by PCR. Appl. Environ. Microbiol. 61:3745-3747. 

16. McKillip, J.L., L.A. Jaykus, and M.A. Drake. 2000. A comparison of methods for 
the detection of Escherichia coli 0157:H7 from artificially contaminated dairy prod- 
ucts using PCR. J. Appl. Microbiol. 89:49-55 

17. Nishida,T., H. Kimura, M. Saitoh, M. Shinohara, M. Kato, S. Fukuda, T. 
Munemura, T. Mikami, A. Kawamoto, M. Akiyama, Y. Kato, K. Nishi, K. Kozawa, 
and O. Nishio. 2003. Detection, quantitation, and phylogenetic analysis of 
Noroviruses in Japanese oysters. Appl. Environ. Microbiol. 69:5782-5786. 

18. Orlandi, P.A., and K.A. Lample. 2000. Extraction-free, filter-based template prepa- 
ration for rapid and sensitive PCR detection of pathogenic parasitic protozoa. 
J. Clin. Microbiol. 38:2271-2277. 

19. Radstrom, P., R. Knutsson, P. Wolffs, M. Lovenklev, and C. Lofstrom. 2004. Pre- 
PCR processing strategies to generate PCR-compatible samples. Mol. Biotechnol. 
26:133-146. 

20. Ramesh, A., B.P. Padmapriya, A. Chandrashekar, and M.C. Varadaraj. 2002. 
Application of a convenient DNA extraction method and multiplex PCR for the 
direct detection of Staphylococcus aureus and Yersinia enter ocolitica in milk samples 
Mol. Cell. Probes 16:307-314. 

21. Sair, A.I., D.H. D'Souza, C.L. Moe, and L.A. Jaykus. 2002. Improved detection of 
human enteric viruses in foods by RT-PCR. J. Vir. Meth. 100:57-69. 

22. Schwab, K.J., F.H. Neill, R.L. Fankhauser, N.A. Daniels, S.S. Monroe, D.A. 
Bergmire-Sweat, M.K. Estes, and R.L. Atmari. 2000. Development of methods to 
detect "Norwalk-Like Viruses" (NLVs) and Hepatitis A Virus in delicatessen foods: 
Application to a foodborne NLV outbreak. Appl. Environ. Microbiol. 66:213-218. 

23. Simon, K.C., D.I. Gray, and N. Coo. 1996. DNA extraction and PCR Methods for 
the detection of Listeria monocytogenes in cold-smoked salmon. Appl. Environ. 
Microbiol. 62:822-824. 

24. Uyttendaele, M., J. Debevere, and R. Lindqvist. 1999. Evaluation of buoyant den- 
sity centrifugation as a sample preparation method for NASBA-ELGA detection of 
Campylobacter jejuni in foods. Food Microbiol. 16:575-582. 

25. Uyttendaele, M., S. van Boxstael, and J. Debevere. 1999. PCR assay for detection of the 
E. coli 0157:H7 eae-gene and effect of the sample preparation method on PCR detec- 
tion of heat-killed E. coli 157:117 in ground beef. Int. J. Food Microbiol. 52:85-95. 

26. Vishnubhatla, A., D.Y.C. Fung, R.D. Oberst, M.P. Hays, T.G. Nagaraja, and S.J.A. 
Flood. 2000. Rapid 5' nuclease (TaqMan) assay for detection of virulent strains of 
Yersinia enter ocolitica. Appl. Environ. Microbiol. 66:4131-4135. 

27. Wang, FL, J.M. Farber, N. Malik, and G. Sanders. 1999. Improved PCR detection of 
Campylobacter jejuni from chicken rinses by a simple sample preparation procedure 
Int. J. Food Microbiol. 52:39-45. 

28. Wilson, I.G 1997. Minireview: Inhibition and facilitation of nucleic acid amplifica- 
tion. Appl. Environ. Microbiol. 63:3741-3751. 



CHAPTER 4 



Making PCR a Normal Routine 
of the Food Microbiology Lab 

Susan Sanchez, Ph.D. 

Athens Diagnostic Laboratory, and the Department of Infectious Diseases, 
College of Veterinary Medicine, The University of Georgia, Athens, GA 30602 



Introduction 

Setting up Your Laboratory for PCR 
Physical Set-up of PCR 
Personnel 

Real-Time vs. Standard Format PCR 
Equipment 

Reagents and Disposables 
Quality Control and Quality Assurance 
Where to Locate Vendors 
Noncommercial Tests for Foodborne Pathogens 
Validation 
Standardization 
Available Commercial PCR Tests for Foodborne Pathogens 
Real Time PCR 

BAX Dupont-Qualicon 
Food-proof Roche 
IQ-Check BioRad 
PCR-ELISA 
Probelia BioRad 

AnDiaTec Salmonella sp. PCR-ELISA 
References 



INTRODUCTION 



In food microbiology, polymerase chain reaction (PCR) should not be consid- 
ered a substitute for conventional microbiology techniques. The rationale of 
employing PCR technology, as well as any other molecular diagnostic tech- 
nique, should be founded on the following key consideration: (1) simplicity, (2) 
throughput, (3) cost, (4) speed, and (5) appropriateness (37). Conventional 
bench microbiology is often considered to be less technically demanding than 
polymerase chain reaction, however, in reality, the techniques of PCR are eas- 
ier to master and usually requires less time to achieve competence than conven- 
tional microbiology. Experience in our laboratory shows that personnel of 
varied technical and educational backgrounds, and absolutely no training in 
microbiology, can master polymerase chain reaction in no more than a week. By 



51 



52 Making PCR a Normal Routine of the Food Microbiology Lab 



contrast, competence in conventional microbiology techniques and their inter- 
pretation requires significant training and experience. Polymerase chain reac- 
tion can be mastered in less than a week with very little or no previous training 
in microbiology, whereas conventional microbiological techniques will require 
several weeks of training and a background in microbiology. A technique, as 
simple as PCR, can be applied in more laboratories and it is more amenable to 
field applications, which could allow for data collection right at the food-pro- 
cessing plant. The food microbiologists' constant aim is to make our food sup- 
ply safer; this is only achieved with large-scale screening of foodstuffs. 
Polymerase chain reaction is a test process that allows for high throughput and 
is amenable to automation. Over the past few years, molecular reagents have 
become more affordable, and easier to obtain, with longer shelve life (37). 
Naturally, the cost and accessibility varies greatly with location. Except for 
primers and probes, most PCR reagents can be used and shared among differ- 
ent PCR tests, as well as for other molecular-based techniques used in food 
microbiology (e.g., strain typing) (37). The decision to use PCR needs to be 
made by the laboratory based on clients' requirements for quick turnaround, 
reliability, and confidence in the laboratory for correctly reporting results. Real- 
time PCR allows not only the detection of suspect pathogen in food but its 
enumeration as well (36, 37, 38). These molecular tools are essential to a con- 
temporary food laboratory. Although the cost to equip a laboratory is high, 
PCR complements and enhances the traditional microbiological methods; by 
increasing speed, sensitivity, and, specificity for detecting pathogens in foods. 
Polymerase chain reaction can be performed rapidly in the field, and limits the 
number of cultures and isolations to the few samples identified as positive by 
PCR (28). This reduces labor, conserves resources, and holds down costs. 



SETTING UP YOUR LABORATORY FOR PCR 



The PCR is a very sensitive, exceptionally powerful, and relatively simple 
method that can unlock the door to many a genetic mystery after a few rounds 
of cycling temperatures. However, PCR can return false positives or negatives if 
care is not taken in the proper setup, standardization, and implementation of 
quality controls. Careful planning needs to be given to standardizing a routine 
protocol(s) for processing and testing of samples by PCR. This includes: the 
physical setup of the PCR laboratory; quality control; storage and selection of 
reagents; deciding which PCR detection platform to use; and finally, a critical 
component — well-trained personnel. With good laboratory practices, the diag- 
nostic laboratory avoids the possible pitfalls with PCR that lead to erroneous 
reporting of laboratory results. 

Physical Setup of PCR. Avoiding sources of PCR contamination is paramount 
when dedicating laboratory space to PCR. Ideally there should be 4 to 5 distinctly 
separate rooms or areas in the laboratory for: (1) sample preparation, which 
includes sample enrichment, clean reagent preparation, and DNA extraction; (2) 
PCR setup; (3) thermocycler; and (4) detection of PCR products or amplicons by 



Setting Up Your Laboratory for PCR 



53 



agarose gel electrophoresis, or enzyme-linked immunosorbent assay (ELISA). 
Realistically, having this much room may not be possible in existing food micro- 
biology laboratory, although it should be kept in mind if future renovations are 
planned, or a new building is being designed. In most instances, this physical sep- 
aration can be accomplished by the development of one-way traffic flow (Fig. 1) 
within a laboratory, assigning particular areas to each step in the process. This will 
hopefully prevent cross-contamination from sample to sample and PCR to PCR. 
The assignment of areas and/or rooms needs to be intuitive and easy to follow so 
that it does not influence the speed and normal work flow of the laboratory. 
Ideally, personnel will start the day in the clean area and move to those areas 
where there is higher risk of aerosols that could be transported to clean areas and 
become sources of PCR contamination. In those laboratories that process a very 
large number of samples, dedicated personnel to each area is a good idea with 
rotation of these people to a different area every week to avoid boredom and 
maintain full competence in all aspects of the PCR protocol. Laboratory coats or 
scrubs should be worn always, but these garments need to remain in each area 
except when cleaned. DNA present on a bench top can easily contaminate a 
sleeve. If a technologist later uses the same laboratory coat, then the jacket could 
serve as the source of PCR carryover contamination when the individual sets up 
the next PCR reaction. Gloves should be used at all times and should never be 
worn in the different PCR areas. The sample preparation area is one of the most 
important areas in the laboratory and it should be divided in three individual areas 
for: ( 1 ) food sample preparation, (2) clean reagent preparation for DNA extraction, 
where no sample or template should ever be present; and (3) processing and extrac- 
tion of DNA from foods or enrichments. This final step should be conducted in a 
biological safety cabinet to avoid sample cross-contamination. The technician can 
inadvertently contaminate sample(s), from which PCR template is prepared by 



Sample 
handling and 
enrichment 
steps 



Clean reagent for PCR 
preparation area 



Clean reagent 
for DNA 
extraction 
preparation 
area 





PCR product 
detection 



Figure 4.1. Chart delineating the workflow in the PCR laboratory. Some of the PCR 
work areas need to be physically separated in order to minimize the potential for PCR 
carryover contamination, or sample cross-contamination. DNA template or PCR 
product should never enter the clean reagent preparation area. 



54 Making PCR a Normal Routine of the Food Microbiology Lab 



touching a small droplet from an enrichment broth, positive for the microbe in 
question, and subsequently can transfer this to another tube containing a differ- 
ent sample, by touching the second sample with contaminated gloved hand. This 
cross-contamination is insidious as it will not be detected by our PCR controls 
and, even if contamination is suspected, retesting of the contaminated sample 
template will only yield the same spurious false-positive results. The sample will 
have to be reextracted, adding time and cost to the process. The PCR can be 
exquisitively sensitive, even detecting a few cells that may cross-contaminate a 
"negative" sample. To avoid this scenario, care should be taken in cleaning the 
outside of the sample tubes with a disinfectant to kill bacteria and DNAse, or 
10% bleach solution to destroy any contaminating DNA. It is also recommended 
that the cabinet and the pipettes are wiped with bleach solution to eliminate sam- 
ple cross-contamination, and subsequently wiped clean with ethanol or water to 
avoid corrosion by the bleach, before the next DNA extractions (13). 
Contamination by aerosols can also be minimized if supernatants are aspirated 
and not decanted. When large volumes of sample need to be handled, disposable 
individually wrapped sterile pipettes should be used. DNA extraction from 
already enriched samples is one of the PCR steps that is amiable to automation, 
which reduces the risk for intersample cross-contamination and sample exchange 
(30, 49). The PCR setup and thermoycler area can be physically separated but they 
can also be placed together as long as there is a biosafety cabinet in this area, or 
a PCR box for the PCR setup. "PCR clean hoods" are relatively inexpensive, sit 
on a table or bench and they are easy to clean. Their relatively small size allows 
for their placement in most laboratories. PCR clean hoods also have germicidal 
ultraviolet (UV) lights, which aid in maintaining sample purity by denaturing 
DNA contaminants (13). The use of UV light to decontaminate the PCR setup 
area, where reagents are openly handled can minimize the risk of carryover con- 
tamination (40). 

The detection area itself, where tubes or capillaries are opened and loaded 
into agaraose gels or ELISA microplates, can serve as a source of PCR con- 
tamination. PCR can amplify DNA from very small amounts of template to 
large quantities of amplicon, which once aerosolized, aspirated, or spilled, can 
easily contaminate surfaces. The glass capillaries used with the hot-air thermo- 
cyclers can break within their carrier during transport to and from thermocy- 
cler and detection area, or required glasscutter to free it from its block. 
Breakage can result in contamination of this block with amplicon. Therefore, 
pipettes as well as racks and carriers, used to transport samples from PCR setup 
to thermocycler to detection area and back, can provide an opportunity for 
contamination of the next scheduled PCR run. This issue is largely avoided with 
thin-walled microcentrifuge tubes and other specialized microtubes for real- 
time PCR. These tubes are not likely to break and contaminate the holders, the 
only piece of equipment that will go back in the PCR setup area for future use. 
Barrier tips can prevent inadvertent suction of fluids into the barrel of pippet- 
tors. Also a separate set of pipettors also eliminates pipettors as vehicles for 
introducing PCR carryover contamination during setup. Real-time PCR avoids 
carryover contamination due to the "real-time" detection of the amplicons as 



Real-Time vs. Standard Format PCR 55 



they are produced, and thus eliminates the necessity from ever having to open 
the tube once the PCR reaction has been set up to run. 

With each additional PCR run, the risk of false-positives increases for that 
test. Therefore, minimizing the risk using the best work flow and work practices 
possible is paramount. The size and the number of working areas that can be 
used simultaneously at one given time will have to be anticipated based on the 
number of samples that the laboratory plans to process daily, and how many 
personnel will be working in each area at the same time. 

Personnel. The PCR is simpler than traditional microbiology, not only regarding 
the procedure itself, but also because mastering this new technology takes less 
time than required to learn conventional microbiology. It can be mastered in less 
than a week with very little or no previous training in microbiology, whereas con- 
ventional techniques will require several weeks of training and a background in 
microbiology. Competent conventional microbiologists can be easily trained to 
perform PCR and will quickly discover the advantages of this methodology. It 
can provide rapid same or next day results and, as an initial screen, it can stream- 
line culture and isolations to the few presumptively positive samples (Fig. 2). If 
we only check PCR-positive samples, we decrease our workload. For the labora- 
tory, this reduces labor, resources, and finally cost. Labor costs rise exponentially 
as the number of microorganisms to be ruled out increases when comparing 
PCR to bacterial culture methods (47). Multiplex PCR and microarrays dis- 
cussed in Chapter 1, reduces these pathogen screens to a single test (12, 26, 41, 
46). Unfortunately, several of the commercial tests discussed later in this chapter 
are detection tests for single foodborne pathogen, and single multipathogen- 
detection systems are in their infancy. While detection and final confirmation of 
foodborne pathogen may take up to 6 days with standard microbiology proto- 
cols, PCR can provide clientele with preliminary results while the microbiology 
lab continues to work up the submission. Laboratory supervisory personnel need 
to be knowledgeable in interpreting PCR results, recognizing and correcting 
problems as they develop, and in explaining the significance of PCR results to 
the clientele. Chapter 2 discusses interpretation of PCR results. Personnel with 
enough molecular training needs monitor result output and keep track of the 
number of positive samples, to request the retesting of positive samples sus- 
pected of being cross-contaminated. The personnel should also keep abreast of 
the literature regarding PCR to discover improvements and problems with cur- 
rent tests and identify new primers, PCR assays, or methodology available for 
foodborne pathogens confronting the food industry. 



REAL-TIME VS. STANDARD FORMAT PCR 



The PCR amplicons are detected either in "real time" as they are synthesized, 
or following PCR run on an agarose gel, or in an ELISA microplate. The suit- 
ability of either real-time or standard format PCR will differ according to the 
laboratory's needs, resources, and expertise. 



56 



Making PCR a Normal Routine of the Food Microbiology Lab 



A 




Tetrathionate broth 
41 C; 24 h 



* *... 




► 




X 



24 h 




BGN 



XLT4 & BGN 

37 C; 24 h 




Salmonella Poly-O 
Antisera 



+ 



u 



Culture-positive" 



a 



AND 



5? 



48 h 



a 



longer 



?? 



Biochemical Verification 
Serotyping 



v 




Salmonella 
PCR 



+ --- + - - - + 



^ 2h 







24 h 



48 h 



V 



B 



Salmonella Poly-O 
Antisera 



Figure 4.2. PCR as tool to identify samples or enrichments, which need to be 
processed further. (A) Steps and time required to identify sample(s) contaminated with 
PCR. (B) Incorporation of PCR into screen of samples, or culture enrichments to 
identify samples requiring additional culture/platings to identify the foodborne 
pathogen, in this example Salmonella enter ica. 



Amplicon detection can be done without agarose gels. In PCR-ELISA, the 
amplicon hybridizes with a specific, internal oligo probe tagged with biotin, 
which is subsequently bound and captured by streptavidin-coated ELISA 
microplate. As the amplicon was labeled with digoxygenin-tagged nucleotides 
during PCR, the bound amplicon-oligoprobe complex is detected coloromet- 
ricly with antidigoxygenin antibody conjugated either to the enzymes alkaline 
phosphatase or horseradish peroxidases. Color change is subsequently recorded 
using an ELISA plate reader. With real-time PCR, a fluorimeter is built into the 
thermocycler to monitor fluorescence as the machine cycles through its dena- 
turing, annealing, and extension step. As the double stranded amplicon is 



Real-Time vs. Standard Format PCR 57 



produced, a fluorescent dye, called SYBR Green, or specific probes labeled with 
a fluorescent dye bind to the PCR product and the detector measures the result- 
ing fluorescence. The advantageous of either PCR-ELISA or real-time PCR 
over agarose gels in the detection of amplicons is their amendability to 96-well- 
microplate formats and automation (31, 43, 46). However, the need for agarose 
gels for detecting amplicons is not eliminated with either PCR detection format, 
as it still has a use in validation; the occasional trouble shooting, as problems 
arise (see Chapter 2); and implementation of real-time PCR or PCR-ELISA 
into the laboratory's routine. 

Equipment. When talking about the laboratory adopting PCR, the first and in 
most cases the only piece of equipment that comes to mind to most readers is 
the thermocycler. Although, by far it is the most expensive individual piece of 
equipment, we should not lose sight of other expenses accrued in setting up the 
laboratory to perform PCR. Several sets of pipettes are needed for DNA extrac- 
tion and PCR setup, a minimum of four sets. If the laboratory does not own 
enough biosafety cabinets, PCR clean hoods can be used. Electrophoresis 
equipment is required for amplicon detection in gels, the microwave oven 
needed for melting agarose, and enough -20°C freezer space to keep all the nec- 
essary PCR reagents separated from the sample template to be tested. For PCR- 
ELISA, it will be necessary to also possess an ELISA reader, although this is an 
essential component already in place for any laboratory that does serology (46). 
Finally, the laboratory will need to record PCR results. For standard PCR for- 
mat, where agarose gels are used to detect amplicons, the laboratory will need 
UV transilluminator and a camera to capture the gel image. 

There are a number of factors that will influence which type of thermo- 
cycler is purchased. The overall cost of the machine itself might be a major 
deciding factor in a laboratory's ability to take advantage of PCR technology at 
all. A simple, hot-air, or conventional heating block thermocycler cost a few 
thousand dollars. However, the price can vary depending on user's needs and 
requirements regarding sample throughput (48 vs. 96), sample format (tube vs. 
96-well microplate), and PCR reaction volume. The laboratory also needs to 
consider the cost of the PCR assay with regards to the individual reagents, dis- 
posables and ancillary components (e.g., wax beads or mineral oil) in selecting 
a thermocycler. For example, thermocyclers with heated lids allow for smaller 
PCR reaction volumes and eliminates the need for mineral oil in the PCR reac- 
tion. The speed of a 30-cycler program, 10 vs. 90 min, may be another factor in 
the purchase of a thermocycler. The laboratory may want to consider the ben- 
efits of a gradient thermocycler, which allows users to perform, simultaneously, 
several PCR tests that require different PCR cycle parameters. These gradient 
thermocylers also have the advantage of identifying optimal-annealing temper- 
ature for a single or multiple PCR primer sets in a single run, due to the 
machine's ability to assign separate programs to each well. The price for real- 
time PCR technology jumps to between $30,000 and $140,000, depending on 
the components of the unit such as 96-well format and automation module. In 
selecting a thermocycler, the laboratory also needs to determine if the PCR will 



58 Making PCR a Normal Routine of the Food Microbiology Lab 



be an in-house validated assay or a commercial assay, as the latter will require 
specific PCR equipment. The following questions need to be addressed before 
purchasing a PCR thermocycler: Will there be many samples submitted on a 
regular basis or only periodically that require this type of technology? Will the 
samples only be a part of the usual submitted workload? Will the samples 
require detection of only one gene of interest, or are there various genes to iden- 
tify, which would require different programs for the thermocycler? The PCR 
format chosen by the laboratory is a big deciding factor to determine the needs 
for equipment as we have seen before, because initial cost of equipment and 
applications of the equipment vary. 

All instruments, from pipettes to freezers, must be calibrated and certified 
according to the respective institutional standard operating procedures, which 
in most cases are dictated by the institutions accreditation agency. For most 
institutions it is at least once a year. Most large equipment manufacturers offer 
yearly contracts for calibration of their equipment. 

Reagents and Disposables. Reagents for the PCR consist of those used for the 
reaction that takes place in the thermocycler, and those used for the detection 
of amplicons. Purchasing of reagents should be done from a reputable company 
and molecular grade should be requested for any reagent that is going to be 
used for PCR. Primers and probes as well as Taq DNA polymerase can be con- 
taminated with extraneous DNA. Top quality components should only be used 
and when handling these reagents, gloves should be worn as to prevent the 
introduction of metal ions, nucleases, or other contaminants to the PCR reac- 
tion (48). Water is an important component. Always use it as aliquoted, single 
use volumes that have been filtered (0.22 |im), and autoclaved. If money is not 
a problem, then ideally DNA-free and DNAse-free water should be purchased 
for PCR. We must not forget that the PCR reaction requires a DNA template, 
therefore; the required reagents for extraction or cell lysis need to be taken into 
account. Consumption and cost of reagents is related to the machine and 
the DNA extraction method that is used and the number of samples to be ana- 
lyzed. Depending on the machine, volumes for each reaction can vary from 10 
to 100 |il. The average volume used in PCR reactions is 25-50 |il. The smaller 
the PCR reaction volume, the cheaper the cost per PCR test is. Costs for Taq 
DNA polymerase, dNTPs, and other components of the PCR can turn PCR 
into an expensive venture for a laboratory very quickly. Lastly, one must con- 
sider the cost of labor for PCR setup as well as for sample preparation. It has 
been reported that the overall costs of the reagents and materials involved in 
identifying specific bacterial agents by PCR were 2 to 5 times higher than the 
costs involved with bacterial culture identification (17). As molecular reagents 
are continuously getting better in quality and longer in shelf life making them 
overall less expensive, this may not hold true. A good example of this is the 
reduction in cost of oligonucleotide synthesis. 

In cost analysis, one must consider whether certain alternative traditional 
microbiological methods are feasible or practical with the laboratory's current 
resources and manpower. For example, Salmonella serotyping requires the lab- 



Real-Time vs. Standard Format PCR 59 



oratory to keep an extensive battery of antisera to identify the thousands of 
different Salmonella serovars. Considering the limited shelf-life of the neces- 
sary serotyping reagents, the laboratory's sample volume, and time it takes to 
identify the Salmonella serovar, a PCR-based approach might be a more time- 
and cost-effective approach (19, 21). Likewise, several of foodborne pathogens 
described in Chapters 6 and 7 are either recalcitrant to current culture, or iso- 
lation methods and PCR is more cost effective compared to the alternative iso- 
lation procedures. Where PCR trumps culture-based detection methods is that 
it can provide rapid preliminary results for making important decisions (15, 39, 
41). For the laboratory, the decision as to which samples to work up further, and 
for the clientele, which lots to hold and which ones to ship. 

The thermocycler and its throughput capacity will dictate the type of con- 
tainer used to hold PCR reaction: glass capillary tubes vs. thin-walled plastic; 
PCR microfuge tubes vs. 96-well microplate. Pipette tips must have filter barri- 
ers to avoid contamination of the inside of the pipette. 

All newly prepared and purchased PCR reagents require quality control 
before use. This is necessary for any food microbiology laboratory to become 
successfully proficient at PCR. 

Quality Control and Quality Assurance. Food microbiology laboratories are 
used to working under standard operating procedures (SOPs), as their labora- 
tories are accredited to perform food microbiology. Each SOP not only contains 
the information on precisely how to conduct the procedure, but will also have 
information regarding when and how much quality control (QC) needs to be 
performed. A QC program usually requires all products and reagents (from 
DNA extraction to PCR) of each lot received to be tested to make sure they 
meet the same standard as previous lots so they can be utilized with confidence 
in the procedures. A good practice is to aliquot all reagents in single-use format 
after QC testing. Sometimes the testing of several aliquots is a good idea, espe- 
cially if the laboratory does large volume of PCR routinely. Preparation of an 
aliquoted ready-to-use PCR master mix, that has been QC tested, is a good 
practice because this minimizes technician error resulting from miscalculation, 
or from forgetting to add a key component in the master mix. Commercially 
available kits should come with all reagents previously tested and this informa- 
tion if not in the packet insert should be available upon request. SOPs must 
include information regarding the required positive and negative amplification 
controls, as these will determine the stringency and accuracy of our PCR tests. 
The basic PCR controls are: DNA extraction controls, sample purposely spiked 
with the organism of interest, and another spiked with a different, unrelated 
organism; and PCR controls, pure, known amount of DNA from the organism 
of interest, and negative, no DNA, control. A QC for PCR can be made even 
more robust if an internal amplification control is included to check for the 
presence of PCR inhibitors in our samples (22, 23, 34). For more information 
about PCR inhibitors (see Chapters 3 and 4). Good SOPs and a good QC pro- 
gram will help minimize mistakes due to bad reagents and human error. Every 
laboratory should run a quality control program that is applicable and relevant 



60 Making PCR a Normal Routine of the Food Microbiology Lab 



for this methodology, and which is capable of detecting deficiencies at any level. 
This process can be made simpler most times by consulting with other labs per- 
forming similar PCR assays and purchasing reagents from suggested reputable 
vendors, although this will not eliminate the need for QC testing. SOPs are good 
training guides for new staff. Good record keeping is essential to any laboratory, 
where a bound laboratory notebook is kept with detailed and dated descriptions 
of protocols, reagents (including lot numbers, purchase, and expiration dates), 
controls, and results (32). Furthermore, a good quality assurance (QA) program 
at the institution ensures that there is compliance with SOPs for the PCR assays 
and consistency is achieved. Additional measures to consider are addressed in 
Chapter 2, and for a more thorough review of PCR laboratory setup, see 
Methods in Molecular Medicine, Vol. 16: Clinical Applications of PCR (29). 

Where to Locate Vendors. For setting up your laboratory to do diagnostic PCR, 
you will need equipment and reagents to routinely perform PCR. Reagent-wise, you 
will need the enzyme, buffers, nucleotides, and barrier tips for dispensing 
reagents into thin- walled PCR tubes that contain the PCR reaction. In addition 
to this, you will need a source for custom synthesis of your oligonucleotides and 
probes needed for PCR. You will need to purchase agaraose gel electrophoresis 
and photo documentation equipment (film or digital-based), for analysis and 
documentation of conventional PCR results, as well as source for agarose, elec- 
trophoresis buffers, loading dye, and molecular weight (MW) standards. We 
have listed in Table 1, several sources for reagents and equipment listed above. 
This list of companies is not an endorsement of the companies or their products; 
rather, the table provides the reader an idea of what will be needed to implement 
PCR into food microbiology laboratory 



NONCOMMERCIAL TESTS FOR FOODBORNE PATHOGENS 



Your laboratory after much consultation has decided that you need PCR as an 
additional method to evaluate your food samples. Now you need to make a 
decision which PCR format you want, provided you have a good physical infra- 
structure that will allow you to have the required physically separated areas in 
your laboratory, and you also have capable and trained personnel and a good 
QC program. Your laboratory has also decided that the use of a commercial test 
is not applicable, but instead the laboratory is going to use currently published 
primers, or even design for a much better PCR primer set. Published data by 
one laboratory can sometimes be difficult to reproduce due to the nature of the 
reagents, the variation in equipment, and the personnel training. Validation 
based on consensus criteria, detection limit, diagnostic accuracy (the degree 
of correspondence between the response obtained by the PCR method and 
the response obtained by the reference method on identical culture samples 
[AC = (PA + NA)/total number of samples; where PA = positive agreement; 
NA = negative agreement], diagnostic sensitivity, diagnostic specificity, and 
robusteness, is a must for a successful microbiology laboratory (24, 25, 33). Your 



Noncommercial Tests for Foodborne Pathogens 



61 



Table 4.1. Molecular biology vendors of PCR 



Vendor 



Web Address 



Pro duct (s 



BIO-RAD 



www.bio-rad.com 



Dupont Qualicon www.qualicon.com 



EPICENTRE 

Fisher Scientific 
Co. 



Idaho Technology 
Inc. 

Invitrogen 

MO BIO 

Laboratories Inc. 

Molecular Probes, 
Inc. 

Promega 



Roche Applied 
Science 

SeqWright 

Sigma-Genosys 



Thermocyclers, Electrophoresis 
apparatus, buffers, and MW standards, 
Photo documentation system 

PCR diagnostic tests (e.g., Bax 
Salmonella) 

www.epicentre.com PCR reagents, DNA cloning reagents 



www. fisher sci . com 



www.idahotech.com 



PCR tubes, barrier tips, etc. 
Electrophoresis apparatus, Photo 
documentation, Agarose, Electro-phoresis 
buffer, Micropipettors, 
-20 °C Freezer 

Thermocyclers, design and synthesis 
of primers and probes 



www.invitrogen.com PCR reagents, PCR cloning vectors 



www.mobio.com 



Nucleic acid extraction kits 



www.probes.com 



Fluorescent dyes 



www.promega.com 



www.roche-applied- 
science.com 



PCR reagents, PCR cloning vectors, 
MW standards, gel loading dye 

PCR reagents, Thermocycler 



www.seqwright.com DNA sequencing 



www.sigma- 
genosys.com 



Custom oligonucleotide synthesis 



USA/Scientific Inc. www.usascientific 

com 



PCR clean hood 



laboratory will need to implement and then validate the PCR tests, bench- 
marking performance by demonstrating that the new method can generate 
results that are equal, or better, than those obtained by the current gold stan- 
dard for detection. For a commercial PCR test that has already been validated, 
implementation is the only step required by your laboratory (34). One last 
point, when deciding which PCR format your laboratory is going to select, 
keep in mind the compatibility of the PCR tests chosen with the laboratory's 
current instrumentation and training. Molecular diagnostic tests that require 
new equipment, more laboratory space, and more training may not be the best 
choice. Therefore, a concerted effort at the initial planning stages should be 
made to foresee future demands. 



Validation. There is no perfect PCR test and interlaboratory variation in per- 
formance of a PCR does occur. However, before diagnostic labs accept a PCR, 



62 Making PCR a Normal Routine of the Food Microbiology Lab 



there is a requirement for multilaboratory confirmation of the tests, specificity, 
sensitivity, and reproducibility. (See Chapter 2, discussion of validation.) Spiked 
samples as well regular samples should be used in the validation process as to 
mimic as much as possible everyday samples and situations. The extent your lab 
plays in the validation process of a PCR will be directly related to the target 
organism, food matrix, and previous work describing validation of PCR in 
peer-reviewed publications. 

Currently, there is not a single harmonized validation protocol available. In 
1999, the European Union (EU), through an initiative titled "the FOOD-PCR 
Project" (http://www.PCR.dk), set out to validate and standardize PCR for the 
detection of pathogenic bacteria in food using nonproprietary primers. The 
intended outcomes of this project were production of guidelines and kits for 
proficiency testing of different brands and types of thermocyclers, method for 
DNA extraction and purification, production of reference DNA material, and 
an online database containing validated PCR protocols. These protocols and 
results are available from their website. A further attempt has been made by the 
EU through the ISO/TC34 committee in collaboration with CEN/TC275, 
through the proposal EN ISO/FIDS 16140 "Microbiology of food and animal 
feeding stuffs — Part 42: Protocol for the validation of alternative methods" (1). 
Validation through this method seems more suited for commercially developed 
tests as the process is costly for nonproprietary, "home brew" PCR (34). Several 
commercial diagnostic tests have been validated in a very extensive manner 
and have been accredited by organizations on standards both at the interna- 
tional and national level (14, 20, 44). Current commercially available tests for 
the detection of foodborne pathogens will be reviewed later in this chapter. 

Standardization. Standardization of PCR tests and extraction protocols at the 
national and international level will allow for accurate interlaboratory com- 
parison. This can be achieved either with commercial tests, or with validated 
published primers. Standardization allows for fast implementation of tests, war- 
ranted accuracy, and detection limits, as well as known strength to allow for 
some variation in the tests procedure without giving misleading results. 
Standardization will also guarantee continued research and improvement of the 
PCR assay and protocols. Thus, the PCR test will fulfill its promise of being 
simple, high-yield, fast, appropriate, and even cheaper than the traditional 
culture (24, 34). 



AVAILABLE COMMERCIAL PCR TESTS FOR FOODBORNE 
PATHOGENS 



There is a wealth of ready-to-use PCR-based tests for the most common food- 
borne pathogens: Salmonella spp., Listeria sp. and E. coli 0157:H7. The avail- 
ability of commercial PCR tests for other foodborne pathogens is sparse and 
laboratory "home-brewed" PCR tests may be a better, if not the only, option. 
Chapters 5-7 describe several published PCR tests for bacterial, viral, and 



Available Commercial PCR Tests for Foodborne Pathogens 



63 



protozoal-foodborne pathogens. Several real-time and standard format PCR 
are commercially available. These two formats need to be studied carefully by 
each individual laboratory introducing PCR into their routine, chiefly to deter- 
mine whether the equipment cost in comparison with potentially improved 
diagnostic ability is a worthwhile endeavor to pursue. One or more national and 
international agencies have validated commercial PCR tests described in this 
section (14, 20, 44). Their sensitivities and specificities are well known (14, 18, 
20, 35, 41, 44, 45), and information is readily available form the products 
websites (www.andiatec.com; www.bio-rad.com; www.qualicon.com/bax.html; 
and www.roche-applied-science.com). Once a decision has been made on a PCR- 
detection format, then the choice of brands should be determined by the individ- 
ual laboratories based on following consideration: (1) simplicity, (2) throughput, 
(3) cost, (4) speed, and (5) appropriateness (37). Table 2 lists the currently avail- 
able PCR-based diagnostic tests and identifies which type of format they are 
based. The commercial PCR tests described in the next section is not an endorse- 
ment of any one product, but rather presents the reader with the commercial kits 
available for PCR detection of pathogens in foods. 

Real-Time PCR. BAX Dupont-Qualicon This PCR test is based on the use of 
pathogen-specific primers combined with a dye that allows for detection 
of amplicon formation during each cycle. A selective enrichment step appropri- 
ate for each food is required before the DNA extraction step. Testing is carried 



Table 4.2. Commercial validated and approved test for the detection of bacterial 
pathogens in food 



Test Format 


Brand Name 


Certification Agency 


Organisms Detected 


Real-Time 


BAX Dupont- 


AOAC-RI 


Salmonella, E. coli 


PCR 


Qualicon 




0157:H7, 






USDA-FSIS 


Listeria monocytogenes 






AFNOR 








NorVal 






Food-proof 


AOAC-RI 


Salmonella, E. coli 




Roche 




0157:H7, Listeria 




IQ-Check BioRad 


AFNOR 


Salmonella 


PCR-ELISA 


Probelia BioRad 


AFNOR 


Salmonella, E. coli 
0157:H7, Listeria monocy- 
togenes, Campylobacter 
jejuni and Campylobacter 
coli. 




AnDiaTec 




Salmonella 

Listeria monocytogenes 



Notes: AOAC-RI: Association of Official Analytical Chemists-Registration International; AFNOR: 
Association Francaise de Noramlisation; NorVal: Nordic Validation Organ; USDA-FSIS: United 
States Department of Agriculture- Animal and Food Safety and Inspection Service. 



64 Making PCR a Normal Routine of the Food Microbiology Lab 



out in a 96-well type matrix and up to 94 samples can be tested at once with one 
positive and one negative control. The product amplification is detected real 
time by including the fluorescent SYBR Green I. This fluorogenic reporter dye 
is not specific for the desired target molecule, therefore, post-PCR melting curve 
analysis is required in the protocol, and spurious, nonspecific amplicons are 
easy to identify. The BAX system has been developed for the detection of 
Salmonella spp., Listeria monocytogenes, E. coli 0157:H7, and Campylobacter 
spp(12, 16, 20) (Table 2). 

The BAX Salmonella spp has been accepted as an official method by several 
accreditation bodies; the Association of Official Analytical Chemists (AOAC) 
has accepted it as an official method (#2003.09) for use in raw beef, raw chicken, 
raw frozen fish, cheese, frankfurters, and orange juice (5). The AOAC-RI 
Performance Tested Method license #100201 applies to food: tested on milk, 
black pepper, chilled ready meal, chipped ham, chocolate, cooked chicken, 
cooked fish, custard, dry pet food, elbow macaroni, frozen peas, hot dogs, non- 
fat dry milk, orange juice, peanut butter, pizza dough, seafood-prawns, alfalfa 
sprouts, ground beef, and liquid egg (3). The USDA-FSIS (MLG 4C.00) has 
adopted the BAX Salmonella spp. for use in ready-to-eat meat, poultry, and 
pasteurized eggs (6). The AFNOR (certificate QUA- 18/3- 11/02) applies to all 
human and animal food (2). The NordVal (certificate 2003-2-5408-00023) 
applies to all foods and animal feed (7). 

The BAX Listeria monocytogenes has been approved by the AOAC. It has 
been accepted as an official method (#2003.12 AOAC-RI Performance Tested 
Method license #070202) for use in a wide variety of foods including raw meats, 
fresh produce/vegetables, processed meats, seafood, dairy cultured/noncultured, 
egg and egg products, and fruit juices (4). The USDA-FSIS (MLG 8A.00) has 
adopted the BAX Listeria monocytogenes for use in red meat, poultry, egg, and 
environmental samples (11). The BAX E. coli 0157:H7 has been approved by 
the AOAC, and has accepted as an official method (#2004.8 AOAC-RI 
Performance Tested Method license #010402) for use in apple cider, orange 
juice, and ground beef (9). The USDA-FSIS is currently in the process of vali- 
dating this technology. 

Food-proof Roche This PCR test is based on real-time detection of either 
Salmonella spp. or Listeria monocytogenes DNA in raw materials and food 
samples through the use of a combination of primers and sequence-specific taq- 
man probes with hot start methodology. An internal control is added to each 
sample prior to extraction, in order to assess the presence of PCR inhibitors. 
Additionally, this commercial test contains uracil-DNA glycosidase to avoid 
PCR carryover contamination. The Salmonella spp. test method is certified by 
the AOAC-RI with license #12030 (8), as a performance-tested method for 
detecting Salmonella in food products. Some raw materials are highly inhibitory 
for the PCR reaction and the use of a proprietary sample preparation kit (High 
Pure Food-proof kit; Roche; Indianapolis, IN) seems to ensure DNA of high 
quality for PCR. The Listeria monocytogenes test method has also been certi- 
fied by the AOAC-RI with license #12030 (10) as a performance-tested 
method for the detection Listeria monocytogenes in food products when used in 



References 65 



combination with ShortPrep foodproof II Kit. These foodstuffs include peanut 
butter, dried whole eggs, dry whole milk, dry pet food, milk chocolate, melon 
cubes, white cabbage, pizza, vanilla ice cream, paprika emulsion dye, spaghetti, 
sausage, gravlax, "harzer" cheese, raw ground chicken, raw ground pork, bean 
sprouts, parsley flakes, ham, and Pollack fillet. 

IQ- Check BioRad This commercial test (BioRad; Hercules, CA) uses 
primers and a molecular beacon probe tagged with a fluorescent label specific 
for the target organism. Amplified products are detected real time by detection 
of the fluorescence. This system also contains an internal control, present in the 
amplification mix that assesses the presence of PCR inhibitors. The internal 
control is detected real time using another florescent beacon labeled with a dif- 
ferent fluoroprobe (27, 45). The BioRad's IQ-Check Salmonella detection kit 
has been approved by AFNOR as a valid method for the detection of 
Salmonella in all human and animal food products, and environmental samples. 

PCR-ELISA. Probelia BioRad This PCR test is based on the enzymatic detec- 
tion of a PCR product that combines DNA: DNA hybridization with its cap- 
ture and in a microtitier plate with an internal oligoprobe. For the detection of 
Salmonella, this PCR-ELISA can detect 3 CFU/25g sample with 99.6% speci- 
ficity, following an 18 h of preenrichment step (14, 18). It includes an internal 
control to evaluate PCR inhibitors in samples, which are monitored in a paral- 
lel well. Results depend on the optical density obtained on the detection 
microplate relative to the internal control well. Salmonella and Listeria applica- 
tions have been approved AFNOR for all foodstuffs. 

AnDiaTec Salmonella sp. PCR-ELISA This commercial kit (AnDiaTec 
GmbH & Co.; Kornwestheim, Germany) comes in two modules. Module one 
includes all reagents needed for DNA extraction, amplification mixture in a 
ready-to-use format, and negative and positive controls. The second module con- 
sists of a microtiter plate, probes, the peroxidase conjugate, and all the buffers 
required for DNA: DNA hybridization and enzymatic detection of the amplified 
PCR products. For Salmonella spp. detection, this test has a demonstrated 98% 
agreement with bacterial culture when it is conducted according to the ISO 6579 
standards. Only samples that had high levels of inhibitors, such as bitter choco- 
late and herbs required a different extraction method than the one included in 
the tests kit (35). There is also a kit for the detection of Listeria monocytogenes. 



REFERENCES 



1. Anonymous. 2002. Microbiology of food and animal feeding stuffs — Protocol for 
the validation of alternative methods (EN ISO/FIDS 16140). European Committee 
for Standardization, AFNOR, Paris, France. 

2. Anonymous. 2002b. AFNOR Certificate QUA- 18/3- 11/02: Dupont Qualicon 
BAX® system PCR assay for screening Salmonella, www.afnor.fr/portail.asp. 

3. Anonymous. 2002c. BAX® system with automated detection PCR assay for screen- 
ing for Salmonella, www.aoac.org/testkits/testedmethods.html. 



66 Making PCR a Normal Routine of the Food Microbiology Lab 



4. Anonymous. 2002d . BAX® system with automated detection PCR assay for screen- 
ing for L. monocytogenes, www.aoac.org/testkits/testedmethods.html. 

5. Anonymous. 2003a. Evaluation of the BAX® system for the detection of Salmonella 
in selected foods (DuPont Qualicon, Inc.; Willmington, DE). www.aoac.org/vmeth/ 
newsmtd.htm#faoma. 

6. Anonymous. 2003b. FSIS procedure for the use of the BAX® system PCR assay for 
screening Salmonella in raw meat, carcass sponge samples, whole bird rinses, ready- 
to-eat meat and poultry products and pasteurized egg products, www.fsis.usda.gov/ 
ophs/microlab/mlg4c0 1 .pdf. 

7. Anonymous. 2003c. NordVal Certificate 2003-20-5408-00023: Dupont Qualicon 
BAX® system PCR assay for screening Salmonella. www.nmkl.org/NordVal/ 
NordVal.htm. 

8. Anonymous. 2003d. Roche Diagnostics LightCycler® foodproof Salmonella detec- 
tion kit for Salmonella spp. in combination with ShortPrep foodproof I Kit. 
www.aoac.org/testkits/testedmethods.html 

9. Anonymous. 2004a. BAX® system PCR assay for screening E. coli 0157:H7 kit. 
www.aoac.org/testkits/testedmethods.html. 

0. Anonymous. 2004b. Roche/BIOTECON diagnostics LightCycler® Listeria monocy- 
togenes detection systems in combination with the Listeria ShortPrep foodproof® II 
kit. www.aoac.org/testkits/testedmethods.html. 

1. Anonymous. 2005. FSIS procedure for the use of Listeria monocytogenes BAX® 
screening test. www.fsis.usda.gov/Ophs/Microlab/Mlg_8A_01.pdf. 

2. Bhagwat, A.A. 2003. Simultaneous detection of Escherichia coli 0157:H7, Listeria 
monocytogenes and Salmonella strains by real-time PCR. Int. J. Food Microbiol. 
84:217-224. 

3. Borst, A., A.T. Box, and A.C. Fluit. 2004. False-positive results and contamination 
in nucleic-acid amplification assays: Suggestions for a "prevent and destroy" strat- 
egy. Eur. J. Clin. Microbiol. Infect. Dis. 23:289-299. 

4. Coquard, D., A. Exinger, and J.M. Jeltsch. 1999. Routine detection of Salmonella 
species in water: Comparative evaluation of the ISO and PROBELIA polymerase 
chain reaction methods. J. AOAC Int. 82:871-876. 

5. Cui, S., CM. Schroeder, D.Y. Zhang, and J. Meng. 2003. Rapid sample preparation 
method for PCR-based detection of Escherichia coli 0157:H7 in ground beef. 
J. Appl. Microbiol. 95:129-134. 

6. Englen, M.D. and P.J. Fedorka-Cray. 2002. Evaluation of commercial diagnostic 
PCR for the identification of Campylobacter jejuni and Campylobacter coli. Lett. 
Appl. Microbiol. 55:353-356. 

7. Erlandsson, A., A. Backman, E. Tornqvist, and P. Olsen. 1997. PCR assay or cul- 
ture for diagnosis of Bordetella pertussis in the routine diagnostic laboratory? 
J. Infect. 35:221-224. 

8. Fach, P., F. Dilasser, J. Grout, and J. Tache. 1999. Evaluation of a polymerase chain 
reaction-based test for detecting Salmonella spp. in food samples: Probleia 
Salmonella spp. J. Food Prot. 62:1387-1393. 

9. Herrera-Leon, S., J.R. McQuiston, M.A. Usera, P.I. Fields, J. Garaizar, and M.A. 
Echeita. 2004. Multiplex PCR for distinguishing the most common phase- 1 flagel- 
lar antigens of Salmonella spp. J. Clin. Microbiol. 42:2581-2586. 

20. Hochberg, A.M., A. Roering, V. Gangar, M. Curiale, and W.M. Barbour. 2001. 
Sensitivity and specificity of the BAX for screening! Listeria monocytogenes 
assay: Internal validation and independent laboratory study. J. AOAC Int. 
84:1087-1097. 



References 67 



21. Hong, Y, T. Liu, C. Hofacre, M. Maier, S. Ayers, D.G. White, L. Wang, and 
J. J. Maurer. 2003. A restriction fragment length polymorphism based polymerase 
chain reaction as an alternative to serotyping for identifying Salmonella serotypes. 
Avian Dis. 47:387-395. 

22. Hoorfar, J., N. Cook, B. Malorny, M. Wagner, M.D. De, A. Abdulmawjood, and 
P. Fach. 2003. Making internal amplification control mandatory for diagnostic 
PCR. J. Clin. Microbiol. 41:5835. 

23. Hoorfar, J., N. Cook, B. Malorny, M. Wagner, M.D. De, A. Abdulmawjood, and P. 
Fach. 2004. Diagnostic PCR: Making internal amplification control mandatory 
Lett. Appl. Microbiol. 38:79-80. 

24. Hoorfar, J. and N. Cook. 2003. Critical aspects of standardization of PCR. In 
K. Sachese and J. Frey (eds.), PCR Detection of Microbial Pathogens: Methods in 
Molecular Microbiology, Vol. 216, pp. 51-64. 

25. Josefsen, M.H., S.T. Lambertz, S. Jensen, and J. Hoorfar. 2003. Food-PCR. 
Validation and standardization of diagnostic PCR for detection of Yersinia entero- 
colitica and other foodborne pathogens. Adv. Exp. Med. Biol. 529:443-449. 

26. Keramas, G., D.D. Bang, M. Lund, M. Madsen, H. Bunkenborg, P. Telleman, and 
C.B. Christensen. 2004. Use of culture, PCR analysis, and DNA microarrays for 
detection of Campylobacter jejuni and Campylobacter coli from chicken feces. 
J. Clin. Microbiol. 42:3985-3991. 

27. Liming, S.H. and A.A. Bhagwat. 2004. Application of a molecular beacon-real-time 
PCR technology to detect Salmonella species contaminating fruits and vegetables. 
Int. J. Food Microbiol. 95:177-187. 

28. Liu, T., K. Liljebjelke, E. Bartlett, C.L. Hofacre, S. Sanchez, and J.J. Maurer. 2002. 
Application of nested PCR to detection of Salmonella in poultry environments. 
J. Food Prot. 65:1227-1232. 

29. Lo, YM. 1998. Methods in Molecular Medicine: Clinical Applications of PCR, Vol. 
16. Humana Press Inc., Totowa, NX 

30. Loeffler, J., K. Schmidt, H. Hebart, U. Schumacher, and H. Einsele. 2002. Automated 
extraction of genomic DNA from medically important yeast species and filamentous 
fungi by using the MagNA pure LC system. J. Clin. Microbiol. 40:2240-2243. 

31. Luk, J.M., U. Kongmuang, R.S. Tsang, and A.A. Lindberg. 1997. An enzyme-linked 
immunosorbent assay to detect PCR products of the rfbS gene from serogroup 
D salmonella: A rapid screening prototype. J. Clin. Microbiol. 35:714-718. 

32. Macrina, F.L. 1995. Scientific Integrity: An Introductory Text with Cases. ASM 
Press, Washington, DC. 

33. Malorny, B., E. Paccassoni, P. Fach, C. Bunge, A. Martin, and R. Helmuth. 2004. 
Diagnostic real-time PCR for detection of Salmonella in food. Appl. Environ. 
Microbiol. 70:7046-7052. 

34. Malorny, B., P.T. Tassios, P. Radstrom, N Cook, M. Wagner, and J. Hoorfar. 2003. 
Standardization of diagnostic PCR for the detection of foodborne pathogens. Int. 
J. Food. 

35. Metzger-Boddien, C, A. Bostel, and J. Kehle. 2004. AnDiaTec Salmonella sp. PCR- 
ELISA for analysis of food samples. J. Food Prot. 67:1585-1590. 

36. Nogva, H.K., K. Rudi, K. Naterstad, A. Hoick, and D. Lillehaug. 2000. Application 
of 5'-nuclease PCR for quantitative detection of Listeria monocytogenes in pure cul- 
tures, water, skim milk, and unpasteurized whole milk. Appl. Enivorn. Microbiol. 
66:4266-4271. 

37. Riley, L. W 2004. Molecular epidemiology of infectious diseases: principles and 
practices. ASM Press, Washington, DC. 



68 Making PCR a Normal Routine of the Food Microbiology Lab 



38. Rudi, K., H.K. Hoidal, T. Katla, B.K. Johansen, J. Nordal, and K.S. Jakobsen. 
2004. Direct real-time PCR quantification of Campylobacter jejuni in chicken fecal 
and cecal samples by integrated cell concentration and DNA purification. Appl. 
Environ. Microbiol. 70:790-797. 

39. Sails, A.D., A.J. Fox, F.J. Bolton, D.R. Wareing, and D.L. Greenway. 2003. A real- 
time PCR assay for the detection of Campylobacter jejuni in foods after enrichment 
culture. Appl. Environ. Microbiol. 69:1383-1390. 

40. Sarkar, G. and S.S. Sommer. 1993. Removal of DNA contamination in polymerase 
chain reaction reagents by ultraviolet irradiation. Methods Enzymol. 218:381-388. 

41. Sergeev, N., M. Distler, S. Courtney, S.F. Al-Khaldi, D. Volokhov, V. Chizhikov, and 
A. Rasooly. 2004. Multipathogen oligonucleotide microarray for environmental and 
biodefense applications. Biosens. Bioelectron. 20:684-698. 

42. Shearer, A.E., CM. Strapp, and R.D. Joerger. 2001. Evaluation of a polymerase 
chain reaction-based system for detection of Salmonella enteritidis, Escherichia coli 
0157:H7, Listeria spp, and Listeria monocytogenes on fresh fruits and vegetables. 
J. Food Prot. 64:788-795. 

43. Shi, P.Y., E.B. Kauffman, P. Ren, A. Felton, J.H. Tai, A.P. Dupuis, II, S.A. Jones, 
K.A. Ngo, D.C. Nicholas, J. Maffei, G.D. Ebel, K.A. Bernard, and L.D. Kramer. 

2001. High-throughput detection of West Nile virus RNA. J. Clin. Microbiol. 
39:1264-1271. 

44. Silbernagel, K., R. Jechorek, C. Carver, W.M. Barbour, and P. Mrozinski. 2003. 
Evaluation of the BAX system for detection of Salmonella in selected foods: 
Collaborative study. J. AOAC Int. 86:1149-1159. 

45. Uyttendaele, M., K. Vanwildemeersch, and J. Debevere. 2003. Evaluation of real- 
time PCR vs. automated ELISA and a conventional culture method using a semi- 
solid medium for detection of Salmonella. Lett. Appl. Microbiol. 37:386-391. 

46. Wellinghausen, N, B. Wirths, A. Essig, and L. Wassill. 2004. Evaluation of the 
Hyplex Bloodscreen multiplex PCR-enzyme-linked immunosorbent assay system 
for direct identification of gram-positive cocci and gram-negative bacilli from posi- 
tive blood cultures. J. Clin. Microbiol. 42:3147-3152. 

47. Wiedbrauk, D.L. and R.L. Hodinka. 1998. Applications of the polymerase chain 
reaction. In S. Specter, M. Bendinelli, and H. Friedman (eds.), Rapid Detection of 
Infectious Agents. Plenum Press, New York. 

48. Wilson, I.G 1997. Inhibition and facilitation of nucleic acid amplification. Appl. 
Environ. Microbiol. 63:3741-3751. 

49. Wolk, D.M., S.K. Schneider, NX. Wengenack, L.M. Sloan, and J.E. Rosenblatt. 

2002. Real-time PCR method for detection of Encephalitozoon intestinalis from 
stool specimens. J. Clin. Microbiol. 40:3922-39288. 



CHAPTER 5 



Molecular Detection of Foodborne 

Bacterial Pathogens 

Azlin Mustapha*, Ph.D. and Yong Li, Ph.D. 

Food Science Program, University of Missouri-Columbia, 1 Columbia, MO 65211 
l Food Science and Human Nutrition Program, University of Manoa, Honolulu, 

HI 96822 



Introduction 

Conventional PCR Detection of Foodborne Pathogens 

Multiplex PCR Detection of Foodborne Pathogens 

Reverse Transcriptase PCR Detection of Foodborne Pathogens 

Real-Time PCR Detection of Foodborne Pathogens 

Conclusions 

References 



INTRODUCTION 



Since its discovery in the mid 1980s, the Nobel prize-winning polymerase chain 
reaction (PCR) has gained acceptance as a powerful microbial detection tool. 
The application of automated PCR technology in the medical and pharmaceu- 
tical industries has a longer history than its use in the food industry. In recent 
years, however, PCR technology has become more recognized for its potential at 
becoming a powerful alternative to cultural methods of pathogen detection in 
foods. The BAX PCR system manufactured by Dupont Qualicon, Inc. and the 
TaqMan Pathogen Detection kits by Applied Biosystems Co. are two examples 
of automated PCR systems that have found application in the food industry. The 
BAX PCR is currently an AOAC-approved PCR-based method that can be used 
to detect Listeria monocytogenes, Listeria species, Salmonella and Escherichia 
coli 0157:H7 in food products, and is used by the USDA Food Safety and 
Inspection Service (FSIS) for detecting L. monocytogenes and Salmonella in 
meat, poultry, egg, and ready-to-eat meat products. 

The advantage of using PCR techniques for food products is the specificity 
and rapidity of the tests as compared to traditional cultural techniques. 
However, the sensitivity of a PCR-based test for detection of pathogens in foods 
will depend on the type of food matrix involved. Because of the complexity of 
food matrices, many compounds in foods can prove to be inhibitory to PCR 
reactions (74). Thus, in most PCR assays, including the automated BAX and 



Corresponding author. Food Science Program, 256 William Stringer Wing, University of 
Missouri-Columbia, Columbia, MO 65211, Phone: (573) 882-2649; FAX: (573) 882-0596; 
e-mail: MustaphaA@missouri.edu. 



69 



70 Molecular Detection of Foodborne Bacterial Pathogens 



TaqMan systems, food samples are enriched prior to the amplification steps in 
order to overcome this hurdle. In addition to increasing the number of target 
microorganisms, and thus the detection sensitivity, enrichment is also helpful 
at reducing the risk of amplifying nucleic acids from dead or nonculturable 
cells (11). Following PCR, target species can be detected by agarose gel elec- 
trophoresis or hybridizations with labeled DNA probes (60). 

Despite its limited use in the food industry, numerous studies have been 
reported in the literature on the development of PCR-based detection methods 
for foodborne pathogens. The following text will discuss the various PCR detec- 
tion methods that have been successful at detecting various pathogens in different 
types of foods. Table 1 illustrates the target genes, primer sequences, tested foods, 
detection limits, specificity, and references of certain published PCR protocols. 



CONVENTIONAL PCR DETECTION OF FOODBORNE 
PATHOGENS 



Conventional PCR relies on the amplification of nucleic acids via a single pair 
of primers to detect one pathogen at a time, and the PCR reaction is optimized 
for the specific food product tested. With enrichment of tested samples, con- 
ventional PCR assays can detect Clostridium perfringens at 10 cfu/g in meat, 
milk, and salad (17), enterohemorrhagic E. coli 0157:H7 at ca 10" 1 cfu/g in beef 
(55), enterotoxigenic E. coli at 1 cfu/ml in milk (70), L. monocytogenes at ca 10" 2 
to 10° cfu/g in various foods (19, 30, 48, 63), Salmonella at ca 10 _1 to 10 1 cfu/g 
in milk and meat products (11, 22, 43, 44), Shigella at 2 cfu/g in mayonnaise 
(71), Staphylococcus aureus at 5-15 cfu/g in skim milk and cream (65), and 
10 cfu/ml in raw milk and curd (16), and Vibrio parahaemolyticus at 10 cfu in 
fish (31). However, other studies have found much higher detection limits of 
PCR assays for foods, without enrichment, including 4 x 10 2 to 4 x 10 3 cfu/g for 
C. perfringens in Korean ethnic foods (32), 10 cfu/ml for L. monocytogenes in 
milk (3), 5 x 10 1 to 5 x 10 2 cfu/ml for Shigella in various produce washes (36), 
10 2 cfu/g for S. aureus in skim milk and cheddar cheese (67), 3 x 10 2 cfu/g for 
V. parahaemolyticus in shellfish (74), and 4 x 10 4 cfu/g for Yersinia enterocolitica 
in pork (37). A PCR based on degenerate primers targeting known nonribo- 
somal peptide synthetases (NRPS) has also been successfully developed for 
detecting emetic strains of Bacillus cereus (15). 



MULTIPLEX PCR DETECTION OF FOODBORNE 
PATHOGENS 



In multiplex PCR, two or more gene loci are simultaneously amplified in one 
reaction. This technique has been used widely to characterize pathogenic bac- 
teria on the basis of their virulence factors and antigenic traits. Fratamico 
et al. (21) employed primers for a plasmid-encoded hemolysin gene (hlyA 933 ), 



Multiplex PCR Detection of Foodborne Pathogens 



71 



C/3 

bO 

o 

-4— > 
•i-H 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-H 

a 

O 

-*— > 



u 

Ph 
> 

en 
<D 

5-H 















3 



^3 






3 

<^> 

3 



ft. 



Is 



3 Q 

oq ft, 



M2 



oq 



s 



M 



Qq 



>3 

CO 



3 

oq 



O 






On 



oq 



oq 



< 

z 



< 

z 



\1 

< 

z 



z 



u 



u 

En 
En 
O 
Eh 
Eh 
Eh 



m 



a 


P 


a 


a 


3 


2 


3 


=5 


o 


o 


o 


o 


5-1 


5-1 


5-1 


5-1 


bJO 


bJO 


bO 


bJO 


=>5 


>3 


>3 


»2 


s 


3 


3 


s 


<^> 


<^> 


<^> 


<^> 


Sk. 


^ 


^ 


^ 


<^> 


<^> 


<^> 


<^> 


O 


<L> 


<L> 


o 



oq 



<N 



a 



o 



-*— > 

5-i 
-*— > 

D 
O 

O 

o 



CO 



bo 



oq 



<o 



o 



bJO 



o o § 

go 

w pQq 






< 



hpHh^hrtfepHpHpH 



z 



oo 






^ 



en 






»0 









3 



I 






• • 



^1- 






^ 



< 

Z 



< 

z 



U Eh 

U EH 



Eh 
U 



in 






u u u u 



u 

Eh 

I 

EH 
EH 

S3 



^ Pm ffJ 



CN CN 



CO CO 

Pm pd 









^r 



3 



*> 



<; 









•1*^ 

o 
^J 



u u 



Eh 

EH 

Eh 

S3 



P4 « 



3 






72 



Molecular Detection of Foodborne Bacterial Pathogens 



R 



CO 

00 

O 

-4— > 

P 

•i-H 

5-1 

<D 

-*— > 

O 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

CO 

O 

-*— > 



P^ 

u 

Ph 
> 

en 
<D 

5-H 









R 






R 



^3 



V 
V 



R 

<^> 

CO 



iV. 

a. 






t? *R 

^o h~>> 






Eh 
U 
O 
Eh 

S3 

O 
Eh 
Eh 
Eh 
En 

Eh 
Eh 

8 



^ 

^ 

5T 



EH 



Eh Eh 



Eh 
Eh 
Eh 
O 
Eh 
Eh 

o 

Eh 
En 

$ 

Eh 

EH 

U 



U O 






en co 
Ph Ph* 






r 



^ 



U 



< 



CN 






P4 



s a 



^ 



O O co 5-h C 



S3 
S 

1 

o 

Eh 
Eh 

• • 

Ph 



Ph 



■^ 


^ 


s 


© 


<5J 


^> 


^J 


<o 



En 

i 

EH 



Eh 
U 
U 

| 

PQ CJ 

O O 

« O 

Ph Eh 



• • 



CO 



CN 



uo 



I 

a 



< 



< 






Cn| 



•^r 









^ 



»2 

R^ 



uu 



X 

o o 



CNl 



m 



^ a 



O rtj 



h Ph P4 









Q 

° o 

PQ £ 

u .a 

>-. H 






<; 



• • 



P< Ph 



3 






^H 

u 



R ^ 

^ bo 



R 

5^ 






u 



•• 



o 



to 
R 

bo 
R 

^H 

u 



bX) 



5-H ^H-T rt 

XS Ph S JJ « 

P O 



° O S 



I 1 



• • 



Ph" fe 



K 


^2 


^K^ 




^ 


^s 



Multiplex PCR Detection of Foodborne Pathogens 



73 



Eh 

u 
o 

Eh 
U 
U 

% 

I 

u 

Eh 

i 



i-H CN CN 

tf Pm tf 






t> 



R 
<^> 

bo 
R 









S3 

Eh 
Eh 
O 
U 
U 
O 
Eh 

ii 

Eh 
O 
En 



h 






GO 

•i-H 

X 

'o rt 

o fe 



tf 



CN 
Ph 






CN CO CO 

tf Ph tf 






Eh 
Eh 



ST 



Eh 



Eh Eh 
U U 
En 
U 
Eh 
Eh 
Eh 
Eh 
Eh 
U 
Eh 

O 
En 
Eh 
Eh 
En 
En 



Ph tf 



en 



o 

•<-H 

S R 

O '^ 

w o 



CM 



m 



<N 



,W) X 

X ,2 in 

M— I 



^ ° GO 



Eh 






• • 



^t 






u 









-5P 

o S 



O « 

00 rD 



o 
o 

Eh 
U 
O 

o 

En 
U 
O 

u 

Eh 

• • 



« Ph 



9 



"i-* 



R-R 

to J*. 

-R -R 



"S 














^2 














< 














g 






EH 


g 






< 


$ 


U 
EH 


% 








Eh 


u 


Eh 


EH 


g 






Eh 


En 


<C 


O 




Eh 


Eh 


^2 


U 


O 


^2 


tf 


^2 


O 


< 


Eh 


Eh 


<c 


^ 


<: 


O 


Eh 


U 


O 


o 


^2 


u 


En 


En 


U 


En 


o 


< 


o 


U 


Eh 


En 


<^ 


En 


o 


o 


Eh 


o 


<C 


En 


<C 


u 


g 


U 


a 


En 


<C 


O 


En 


< 


En 


u 


U 


^2 


En 


< 


o 


<n 


Eh 


<C 


<: 


^ 


o 


u 


U 


U 


Eh 


U 


^2 


g 


g 


O 


g 


Eh 


Eh 


<C 


< 


<n 


En 


< 


O 


O 


En 


EH 


g 


< 


o 


U 


O 


Eh 


< 


< 


O 


En 


O 


U 


U 


o 


o 


O 


< 


O 


Eh 


O 


g 


g 


Eh 


g 


g 


U 


En 


< 


<c 


U 


< 


<c 


En 


U 


u 


irf 


Eh 


En 


En 


O 


U 


En 


Eh 


U 


Eh 


Eh 


O 


Eh 


u 


Eh 


O 


«-H 


CN 


CN 


CO 


en 


^ 


^ 


« 


P4 


« 


Ph 


& 


Pm 


« 












^2 

S 
S 






74 



Molecular Detection of Foodborne Bacterial Pathogens 



R 
<3> 



C/5 

00 

O 

-4— > 
•i-H 

5-1 

<D 

-*— > 

O 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

O 

-*— > 



u 

Ph 
> 

+-> 

en 
<D 

5-H 









R 






R 

<3> 






V 
V 



R 



iV. 

a. 






t? *R 



CN 



00 

43 O 






fei 



O « 
bD,£> 



o 

g 

EH 

Eh 
Eh 
O 
O 
U 

O 

Eh 
Eh 
O 
Eh 

• • 

1— I 
Em 



i 

CO 



I 

■S 

X ^o 
S .R 



i 
X 

CO 






c\ 



r- 

m 








w o 



in 

bD 



T3 

O « 

bD,£> 





g 


u 






< 


Eh 






o 


u 


O 




u 


O 


u 




Eh 


U 


Eh 




u 


Eh 


Eh 




Eh 


En 


<^ 


^ 


Eh 


Eh 


^2 


<: 


U 


< 


< 


o 


U 


Eh 


Eh 


Eh 


<C 


Eh 


O 


Eh 


^2 


1^ 


En 


U 


<C 


rtj 


< 


< 


EH 


^ 


En 


U 


<^ 


EH 


O 


O 


O 


< 


g 


Eh 


U 


Ej 


<< 


U 


<^ 


< 


^2 


<c 


Eh 


U 


<c 


tf 


Eh 


< 


rf 


rtj 


Eh 


O 


rf 


Eh 


U 


U 


u 


Eh 


U 


U 


o 


<^ 


En 


Ej 


Eh 


EH 


<C 


<^ 


U 


Eh 


O 


O 


< 


< 


O 


O 


O 


«-H 


CN 


CN 


i-H 


« 


fe 


« 


Ph 



X 

CO 






a to 

-R vl 



•r^ 






t§^^' 



o 

Eh 

"s 

o u 

En U 
Eh <C 

U U 

i 

u 

Eh _ 



«-H CN CN 

rt Ph ^ 



CN 



CO CO 

Ph ffJ 



<* ^ in 

Ph P^ Ph 



oo 




of; 









o o 

En O 
< Eh 
O O 
O 
Eh 
O 
Eh 
O 

En _ 
U < 
Eh U 



in i-H 

ffj P4 



*o 


^ 






<N 


i — i 


1 — 1 




o 


O 


o 




^ 


^ 


^ 


"**H 


<^> 


•^ 


•^ 


X 


^3 


a 


a 


<^> 


•^ 


•^ 


EHEC 0157: s 
H7 






< 


< 


O 


o 


Eh 


O 


Q 


Eh 


Eh 


^2 


O 


U 


Eh 


<n 


Eh 


U 


^2 


U 


H 


Eh 


^ 


< 




Eh 


O 


U 


rt! 


U 


O 


< 


o 




O 


En 


o 


^ 


U 


< 


Eh 


En 


U 


En 


O 


<: 


U 


O 


Eh 


o 


Eh 


U 


U 


u 


Eh 


Eh 


<c 


Eh 


< 


o 


ffi 


U 


Eh 


U 


tf 


O 


U 


Eh 


rt! 


Eh 


U 


O 


o 


< 


En 


U 


u 


O 


<d 


O 


o 


Eh 



i-H CN CN 

P^ Ph tf 



05 



CO CO 

Ph PS 



5 



Multiplex PCR Detection of Foodborne Pathogens 



75 







in Z 

of; 






c\ 



< 



< 

z 



Eh 
Eh 

i-H 

Cm 






S3 £ 

Eh 

Eh O 

En O 

< Eh 

En O 

<: C) 
Eh 



O Eh 



i-H CN 
tf Cm 



"-H 



u 

Eh 
Eh 



i 



CN 



<N 



K 


X 


i^i 


^ 


>} 


^ 






Eh Eh 

° id 



o 

Eh 

o 



u _ 

Eh O O 

g o eh 

<C u u 

O Eh Eh 

Eh Eh U 

U U En 

Eh Eh < 

Ej Eh U 

< En O 

^ ^ Eh 

U Eh < 

O O O 



Cm « 



CN 



in 

o 






D 



c\ 



GO 



^3 <u 13 ^ 

o u o> 3 <£ 

bDJD O B ctf 



CO 

^— > 

O 

CO 



u 

Eh 
Eh 

S3 

u 

Eh 
O 
Eh 
U 
O 
U 
O 



h 









U 
U 
Eh 

En 
Eh 

H 

o 

Eh 
O 
O 
U 



U Eh 



"**i 



u 

Eh 

u 

Eh 
En 
U 
Eh 
Eh 
Eh 
En 
Eh 
O 



ii 



Eh 
U 
Eh 



Eh 



E" 1 g 

Ej fS 

<c g 

E" 1 g 

o g 

eh <c 






u g 

EJ Eh 
"S EJ 



Eh 

u 



Eh 
Eh 

O 
Eh 
O 
U 



«-H CN CN 

« h « 



CO 



ro ^ ^ 
tf Cm pcj 



<N 



K 


K 


**a 


i^i 


^ 


>5 






*& EJ 



O 
Eh 



Eh 
Eh 
Eh 



H 



u g 

Eh <C 
EJ & 






o u 

O Eh 

Eh O 

o g 

EJ <3 

O En 

y Eh 



in in 



in 



o 



lO 

in 




OK 






c\ 









<3 

O 
Eh 
En 



U 
O 
U 
U 
O 

o 

Eh 



O 
Eh 

S3 

• • 

Cm 






OS 



U 

w 

H 






^ 



U 
Eh 
U 
Eh 

o 

Eh 
U 
U 
U O 



Eh 


< 


Eh 


o 


Eh 


u 


Eh 


u 


% 


g 


En 


< 


Eh 


^2 


U 


< 


g 


Eh 


<$ 


u 


8 


Eh 

g 


^2 


< 


<^ 


o 


o 


Eh 


Eh 


u 


u 


o 



• • ^H 

« Cm 






■a O 



•i>«» 






C§(^ 



o 

Eh 

u 

Eh 

u 



Eh 
O 
Eh 

•• 

t-H 



"^3 






76 



Molecular Detection of Foodborne Bacterial Pathogens 



R 



CO 

bO 

O 

-4— > 
•i-H 

5-1 

<D 

-*— > 

O 

d 
5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

CO 

O 

-*— > 



u 

Ph 
> 

+-> 

en 
<D 

5-H 









R 






R 






V 
V 



R 

=0 



a. 






t? *R 
oq c^ 



u 



u <tj 



CN CN 

Ph « 






in 



U 

w 

H 

W 



Z 



Z 



o 
u 



S 

O En 
U U 



u 

Eh 

En 
Eh 
U 
Eh 
U O 
U O 
U U 
U U 



Eh 
Eh 
En 






eh <: 



«-H iH CN CN 

Ph tf Ph tf 



CO CO 

Ph « 



Ph « 



^o 



=0 

r 

bo 



^1 



< 



< 



Pm 



^ 


^ 


^ 


1=3 




^ 


>^ 


^Ka 


t~o 


^ 


=0 


^^ 


>} 


03 


or 


s 


^ 


CU 


su 










^ 










•^ 










R 










<^> 










5>0 










C> 










i«* 










^ 2 










** R 










•S3 c> 



o 

Eh 

Eh 
O 
Eh 
U 
O 
Eh 
Eh 



On 



CO 

bo 

o 

o 
o 

a 
h-i 



X ^0 



D 

CO 

O 




• • •• 

« Ph 



-R 



^1- 



^1 

•T-H 



SO 

R 

R 



bD 

o 



O o u 

O K O 
O bJO^ 





Eh 


<c 


U 





<C 


Eh 


Eh 


U 


U 


g 


^ 


<: 


<: 


u 


U 


En 


< 


< 


U 


U 


<U 


<^ 


^ 


Eh 


Eh 


U 


U 




• • •• 

« Ph 



or 



10 

CN 



=o 

I 

bo 

R 



^ 



in 

CN" 

o 
M2 



b0 



Eh 
O 

^ 

O 
Eh 
O 

S3 

U 

Eh 
U 
Eh 
O 
Eh 
U 
Eh 



-R 



en 



=o 

R 
bO 

&> 

R 



^1 



-5P 

X c5 



1 £ 

d +-» 



u 













Eh 






Eh 






Eh 






Eh 





U 


U 


^2 


Eh 


EH 


<c 


O 


< 


Eh 


U 


U 


Eh 


<^ 


Eh 


< 


EH 


Eh 


^2 


Eh 


U 


<C 


U 


<c 


En 


Q 


f^ 


<C 


<C 





O 


O 


Eh 


O 


Eh 


Eh 


U 


^2 


H 


^2 


^ 





< 


u 


En 


U 





<c 


Eh 


u 





Eh 


Eh 



u u 



^ Ph tf Pm 



oq 

^ 



R 






^i 



Multiplex PCR Detection of Foodborne Pathogens 



77 



en 



02 
<^> 

R 

bo 

& 

R 



^1 



(N 



-h X bD 
X nl 



<G ^ * a 

K <D Cd -=' 



CD 

O czf c3 c/f 

T3 It! "O ctf 



CO 



c3 

o 



cd ^ <D -* 

13 p 15 5 H 



C^DrfpH^>(^ Ph. 



o 



• • 



Cm 



3 



CN 

Cm 



00 



03 
R 

bo 

r 



^ 



<N 



X 

^1- 








CO 




(D 




CO 


<£ 







^ 


CO 


O 



a 



• • •• 

tf Cm 






U 



m 



>3 

R 

bo 

R 









^ 



-R 






02 
^> 

R 

bO 
C> 

R 



^ 



bW 










<; u 



rt Cm tf fe 



I 

Oh 

00 ci 



5-H 



o 



o 

■4— > 

5-h 00 



<N 



c\ 



a 



R 



3 



< 

z 



M 

o 

C4-T 
43 



<N 



a 






£ 



o 

1— I 
I 



<D 
O 

•i-H 

43 
O 



O 
En U 

Eh 









R 



<§ 



OO 
CN 



R 



3 



bo 
o 





fl 


a 


(D 


CD 


^ 


N 


CJ 


O 


•i-H 

4^ 


Ch 


O 



rt Pm p^ Pm p^ Pm 



O 






^3 

S 

St 



St 



78 



Molecular Detection of Foodborne Bacterial Pathogens 



R 



C/5 

bO 

O 

H— > 

Oh 

•i-H 

5-1 

<D 

-*— > 

O 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

O 

-*— > 



u 

Ph 
> 

+-> 

en 
<D 

5-H 









R 






R 






V 
V 



R 



iV. 

a. 






t? *R 
oq c^ 






R 



<3 



bD 



(N 



O « 
bD,£> 



Pm 



5-H 



R 



<3 



bJO 



C-N 



<d 
<d 



^3 

0) 



c\ 



c\ 



^3 s a 

o o g | 



bD 

GO C+-H 





Eh 






Eh 






O 




Ej 


U 




<< 


U 


o 


Eh 


O 




O 


Eh 


^ 


Eh 


U 


o 


U 


O 


u 


U 


Eh 


o 


O 


Eh 


o 


U 


O 


g 


Eh 


O 


icC 


U 


O 


^ 


O 


U 


o 


U 


<d 


u 


<c 


O 


o 


^ 


O 


u 


Eh 


Eh 


o 


U 


O 


u 


O 


^2 


o 


U 


^ 


Eh 


g 


^2 


U 


< 


< 


o 



• • •• 






CN 



^3 



•^ 
S 
O 



<§ 



bJO 



'Eh 

bJO^D O 



5-1 





Eh 




Eh 




U 


EH 


Eh 


Ej 


g 


< 


<^ 


U 


Eh 


U 


O 


U 


U 


Eh 


Q 


Eh 


rtC 


^2 


tf 


<C 


Eh 


^2 


Eh 


< 


Eh 


U 


Eh 


O 


O 


O 


O 


U 


Eh 


U 


Q 


U 


O 


Eh 


U 



• • 



tf Ph 









^3 



•^ 
S 
O 



<§ 



On 



b0 






• • •• 

tf Ph 



3 



C\ 






u 

Eh 
Eh 
Eh 
U 



ON 



Q 



Q 



5 >. 



<3 






<; 



ii 



U Eh 

Eh Eh 

PQ O 

Eh <C 

O U 

Eh Eh 

• • •• 



o 



CO 



1 

6 

o 

Eh 
U 



^D 



a 






3 



CO 

I 



Q 

CD 
>^ 

o 

CD 

GO 



X 



bX) 



^1- 






^ 



< 



< 



bD 

b/) y^r 



s 

Eh 
U 

ii 

O 
U 

u 



CO 

• i-H 

•i-H 
•i-H 

(D 



u 



5S, 

«>5 






^1- 



R 



3 



< 



< 



< u 












^ Pm ^ Pm pej Pm ^ 



Multiplex PCR Detection of Foodborne Pathogens 



79 






Q 



Co H 













Pm 






oo 



Q 



bo 



Co 






c\ 



o 



u 




Eh 




Eh 




U 


O 


U 


En 


U 


En 


O 


U 


U 


En 


< 


O 


O 


U 


g 


< 


<^ 


EH 


u 


Eh 


u 


U 


Eh 


En 


Eh 


< 


<: 


O 


U 


U 


O 


^ 


U 


< 



• • •• 

tf Ph 



O . 



c\ 



oi U Oj Oj 



O 

o 

o 

o 



i 

CO 



O O O »h ^ O 






a 



.bo 



M2 



■S2 
.bo 

CO 



V 






S3 
o 

Eh 

o 
u 

•• iH iH 

rt Ph rt 



•& 



o 

Eh 

O 
Eh 
O 
O 
Eh 

O 
Eh 

% 



8 

o 

Eh 

o 

Eh 



CN CN CO CO 
pH tf PH tf 



r- 



co W 



o 



i 
O (U 



Pm 






• • 



in 



t3 



.bp w 

co W 






r\ 



(D 
2 



cn 



O 



CO 

co G o 



^2 


<c 


<C 


Eh 


o 


O 


En 


O 


< 


Eh 


o 


U 



• • 



« Ph 






o 

Eh 
En 
Eh . 



o 

Eh 



Eh 
Eh 
U 



9 y 



«-H CN CN 

tf Ph rt 



^3 

S 

St 



St 



80 



Molecular Detection of Foodborne Bacterial Pathogens 



<0 



GO 

00 

O 

+- > 

•i-H 

5-1 

<D 

-«— > 

O 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

O 



u 

Oh 

> 

• i-H 

+-> 

en 

5-H 
























CO 



ft, 






^ so 

oq c^ 






CO W 



<N 



in ^ 

O <H 



rs 



O 
=5 



r\ 



£ ^ ^ c£ 

C3 g (D c^ 



c\ 



CO 



CD 

•I-H 

o 



2 U§ 



GO 

-«— > 

O 

5-H 

go 



u 

O 
U 

I 

u 

Eh 
U 
U 
O 
U 

O 
Eh 
Eh 
U 
U 
Eh 
Eh 
O 

• • 



&3 

.Si 



• • 



^- 



I 

5-H 

o 

Si 

tO 



X 

o 

-4— > 

o 

5-H 

<D 

■4— > 



GO 

b0 



a ^ 



00 en 






• • 



« Em 



^3 

CO 



«>3 

s 



or ^: 

CO Q 



u 

Eh 



Eh U 



Eh O 
U O 
U 



U 

En 

O < 

U U 



S3 



«-H CN CN 
« PM Ph" 



00 00 



<* ^ m 

Ph P^ fc 



^ 


^J 


^S 


^> 


•^ 


<^> 


•^ 


•^ 


sn 


«>2 


^ 


«>3 



u 

Eh 
En 



^r 



bX) 

^ ° 

^ > 

co e 



CO 

bJO O 



CD 

a 

o 

5-H 






CO 



o x 

^ o 

go ^— > 



SZ 

Q 



< 






U Eh 

O O 
O Eh 



Eh 


Eh 


O 


O 


U 


Eh 


U 


Eh 


< 


Eh 


Eh 


U 


En 


U 




LT) i-H 

« Pm 






Multiplex PCR Detection of Foodborne Pathogens 



81 



Eh 

EH 

Eh 

1 

O 
U 

u 
o 

Eh 
En 
Eh 
U 



U 
Eh 

i 

Eh 
O 
Eh 
Eh 

% 

U 
O 
Eh 



i-H CN CN CO CO 

tf Pm ^ Pm p^ 



S3 


-C> 


i~»> 


i~»> 


su 


su 






to 
3 

Si 

CO 



in 5p 

IT) O 



c\ 



M 



En 



3 

co 

en 
CN 



^ ft 



U 



«-H CN CN 
^ PH « 






o 

i-H 

Q g 



o 

5-i 



Si 



3 Q 

CD CO 






CN 



a 





^3 



GO 



T3 d 

^ 43 ^ 
co O O 



S2 



Pm 






I 

I 

o 

En 

O 
Eh 
U 
U 







Eh 


En 


O 


^2 


En 


< 


Eh 


EH 


O 


< 


U 


En 


^ 


< 


< 


Eh 


U 



S3 S 



i-H CN CN 
^ PH « 



O 

s 

R 



5-H 

O .S 

a Q 



Si 



CO 

bQ 



O 

S-H 



S « 



co SO 






CD 

£ 3 






ii 

rtj 



Pm 



u 

Eh 
O 
Eh 

u 

Eh 
O 
Eh 

O 
En 
O 



U U O U 



«-H CN CN 
Ph" PH rt 



CO CO 

ph rt 



CO 



to 



-R 

s 



^ « 



a 



CO 



EH 
EH 

Eh 

S3 

u 

Eh 

i 

8 

Eh 
En 
Eh 

• • 

Pm 



^ 



!ao 


~R 


•i>«» 


=s 


*> 


<^> 


<^» 


^S 


SO 


to 


«o 


pathogenic t 
Vibrio species 



I 

o 

Eh 
Eh 
O 
Eh 
Eh 
Eh 

• • 



^2 






82 



Molecular Detection of Foodborne Bacterial Pathogens 



R 



C/5 

00 

O 

-4— > 
•i-H 

5-1 

<D 

-*— > 

O 

5-1 

o 

O 
O 

a 
a 

o 

o 

5-1 

a 

O 

-*— > 



u 

Ph 
> 

en 
<D 

5-H 









R 






R 

<3> 






V 



R 



iV. 

a. 






^ so 
t? *R 

oq c^ 



OS 



3 



<N 



O 

a 



^ 

^ 

^ 
^ 



CN 



u 
o 

Eh 



Ph « 



3 

U 
Eh 
U 
O 
U 
U 
O 
U 

u 

En 
Eh 

i 



CN 



CN 



en 



s 

■ ?s 
So 

-R 



^ 



< 



< 



Pm 






o 



lAi 






2 






3 

s 



s 



3 






oo 



in 

CN 



m 



<N O (N 

W) rH W) 



5-1 

-*— > 

o 



5-1 

o 

-(— > 

o 



o 

Eh 
O 
O 
O 
En 
Eh 
U 
O 
Eh 
O 
Eh 

a 

Eh 
U 
Eh 
O 



-R 



-R 



s 

CO 



o 

Eh 
Eh 

I 

u 
u 

Eh 
Eh 
O 



& Pm p^ Pm rt Pm 



tin 

O 



oo 



3 






a 

o 
in 

CN 






o Eh 
g u 

<tj En 



• • •• 






(D 

ex, O 



R 



CN 



2 



< 



CO 
CO 






3 



-R 





<: 














g 














< 




^ 










Eh 




O 










EH 




EH 




U 






Eh 




< 




Eh 




o 


Eh 




o 




Eh 


O 


Eh 


Eh 


u 


O 




O 


Eh 


Eh 


U 


U 


En 


< 


g 


Eh 


a 


Eh 


U 


EH 


U 


<c 


O 


Eh 


Eh 


O 


EH 


Eh 


En 


O 


o 


Eh 


Eh 


< 


U 


Eh 


Eh 


u 


Eh 


U 


En 


Eh 


< 


O 


a 


a 


U 


o 


U 


g 


Eh 


o 


O 


Eh 


g 


O 


<C 


O 


Eh 


g 


En 


<L 


U 


^2 


U 


u 


<^ 


En 


^ 


U 


<c 


En 


g 


Eh 


Eh 


Eh 


Eh 


U 


<C 


<^ 


O 


U 


O 


En 


o 


^2 


En 


U 


U 


Eh 


En 


g 


<: 


u 


Eh 


U 


^2 


< 


<j 


O 


o 


Eh 


Eh 


•S 


U 


^ 


U 


Eh 


U 


O 


r^ 


U 


^2 


Eh 


Eh 


En 


Eh 


o 


U 


<^ 


V 


^2 


<: 


U 


u 


O 


o 


O 


^ 


U 


Eh 


o 


Eh 


u 


Eh 


• • 


t-H 


i-H 


CN 


CN 


CO 


CO 


rt 


Pm 


fH 


fe 


« 


Pm 


pc; 






^ 

"^^^i 

^ 



Multiplex PCR Detection of Foodborne Pathogens 



83 



10 



^ 



in 

o 



• • 






•• 








^3 




<o 




•^ 




^ 




"i»na 


era 


**^ 


3 


o 


^> 


<u 


^ 

•rO 


o 

^ 


-S 




s 


s 


^ 


<^> 



w 






^H 




4—) 


^ 


C/) 


S-H 


>^ 


o 


O 


a 



• • 



Pm Ph Pm 



3 

in 









Eh 
U 

I 

Eh 
Eh 



En 


<C 


Eh 


CJ 


<: 


"5 


u 


o 


o 


u 


En 


a 


<$ 


u 


En 


o 


< 


Eh 


U 


Eh 


U 


rt] 


Eh 


<] 


U 


Eh 


Eh 


En 


< 


Eh 


U 


U 


Eh 


Eh 


U 


Eh 


Eh 


Eh 


Eh 


U 


O 


Eh 


Eh 


Eh 



«-H CN CN 

« h « 



3 









in 



VI 



o 

Eh 

i 

l 

Eh 
Eh 
O 

o 

Eh 

u 

Eh 

<3 
O 

• • 

P4 



r-4 






*n 

bfl 

o 





3 




Ch 




1 o 


-o 


'O H— > 


& , 


fl r 


d ^ 


d ^ 


s-h O 


S-H O 


bO Oh 


bO Oh 



u o 

U Eh 
Eh 

o 

Eh 

<^ U 

U Eh 

U O 

U Eh 

U U 

U O 

O 
U 



• • 



Ph" Ph 






u 

Eh 
Eh 

H 

O 
Eh 

ii 

U 
Eh 






X5 

• r- 1 

o 

CD 
Ph 



43 



CO 
CO 



•Ms 

o ^ 












c^ 



0> 
T3 



o 2 

"-• Oh 

r _» ■ ■ 

Oh 






u 

Oh 

o .2 

S-H (U 

-s s 



c^ 



q t3 



S-H 

d 



o 

d 



S-H (H 

CO CO 

a a 

CO GO 



T3 

a 

c2 

5-H 

<U 
Oh 

d 
o 

n ^d 

dn ^ 

>> o 



<u 



d 
o 

• i-H 
H— » 

Ph 
CO 

O 

• r— ( 

^ 2 

S 2 

•Y d 

■g.|y 

o G 

O Ph O 



o 



03 



c^ 



a ^ a 



c2 



co r O 



£ ?■£ 



(N ro 



H (AJ H 
in ^O C-- 



84 Molecular Detection of Foodborne Bacterial Pathogens 



chromosomal flagella (fliC H1 ; flagellar structural gene of H7 serotype), Shiga 
toxins (stx v stx 2 ), and attaching and effacing (eaeA) genes for specific identifi- 
cation of E. coli 0157:H7. Similar protocols were reported for concurrent 
determination of multiple toxin genes in C. perfringens (70) and S. aureus 
(4, 10). Further, species or serotype differentiation can also be achieved via mul- 
tiplex PCR. Denis et al. (14) selected 16S rRNA, map A, and ceuE as the target 
genes for simultaneous detection of Campylobacter jejuni and Campylobacter 
coli. A similar assay was established for simultaneous identification of 
Salmonella sp., S. Enteritidis, and S. Typhimurium in one reaction (64). In multi- 
plex PCR, bacterial pathogens belonging to different genera can also be screened 
in the same amplification system. Li and Mustapha (41) and Li et al. (42) 
established a multiplex PCR for simultaneous detection of E. coli 0157:H7, 
Salmonella, and Shigella in apple cider, produce and raw and ready-to-eat meat 
products. In most situations of multiplex PCR, the optimal conditions for dif- 
ferent primer sets may be unique and interference among different primer pairs 
may occur, resulting in uneven amplification of different target sequences and 
limited sensitivity (68). Thus, adjusting the concentration of Taq DNA poly- 
merase, MgCl 2 , or dNTPs, as well as the concentration ratio among the differ- 
ent primer pairs is needed in the optimization of the amplification system. 
Although to design a robust multiplex PCR assay for foods can be challenging, 
once optimized for the specific pathogens and food products, this method has 
the advantage of being cost effective and highly efficient. Because of its selec- 
tivity, sensitivity, and efficiency, a multiplex PCR protocol is very applicable and 
suitable for comprehensive testings of specific foods. 



REVERSE TRANSCRIPTION-PCR DETECTION 
OF FOODBORNE PATHOGENS 



The use of reverse transcription PCR (RT-PCR) in foods is limited due to 
the difficulty of extracting undegraded mRNA from pathogens in complex 
food matrices. By amplifying the iap mRNA, a RT-PCR was successfully 
developed for detecting viable L. monocytogenes cells in cooked ground beef, 
artificially contaminated with ca. 3 cfu/g, following a 2-h enrichment step (34). 
Mclngvale et al. (52) established a similar protocol for Shiga-toxin-producing 
E. coli with optimal growth medium, incubation temperature, and aeration. 
The assay was validated in artificially contaminated ground beef. Viable E. coli 
0157:H7 at an initial inoculum of 1 cfu/g was detectable in the meat after a 
12-h enrichment. In addition, a RT-PCR was developed for detecting mRNA 
from the sefA gene of S. Enteritidis (66). The sensitivity of the assay depended 
on the physiological state of the cells under different temperatures and pH. 
With the RT-PCR, it was possible to detect 10 cells of S. Enteritidis PT4 in 
contaminated minced beef and whole egg samples following a 16-h enrich- 
ment step. Although not cost-effective for routine testings of pathogens in 



Conclusions 85 



foods, these reports highlight the potential of RT-PCR for the detection of 
viable bacterial pathogens in foods. 



REAL-TIME PCR DETECTION OF FOODBORNE 
PATHOGENS 



Recent advances in fluorescent chemistries and detection instruments allow fur- 
ther development of PCR technology as a more efficient and sensitive tool for 
"real-time" microbiological analysis of foods. The use of nonspecific fluorescent 
double-stranded DNA-binding dyes (such as SYBRGreen or SYBRGold), or 
specific fluorescence resonance energy transfer technology (such as 5'-nuclease 
assay [TaqMan], or molecular beacon) has resulted in PCR assays with quanti- 
tative capability in a real-time manner (53). A number of real-time PCR assays 
have been described for the detection and quantification of C. jejuni (59), 
enterohemorrhagic E. coli serotypes 0157, Olll, and 026 in ground beef (62), 
L. monocytogenes in cabbage (25), Salmonella serotype D in egg (61), pathogenic 
Vibrio species in oyster (7, 50, 57), and Y. enterocolitica in ground pork (29, 72). 
Further, a sensitive multiplex real-time PCR has been developed for the simulta- 
neous detection of E. coli 0157:H7, Salmonella, and Shigella in pure culture and 
in ground beef (A. Mustapha, unpublished data). In addition to maintaining all 
the advantages of conventional PCR, real-time PCR has added speed and sensi- 
tivity. This technique can quantify a target DNA with greater reproducibility, 
which is very valuable in the quantitative assessment of microbial risks and the 
execution of HACCP programs in the food industry. The current drawback for 
using real-time PCR for routine food testing is the cost involved, not only in the 
equipment but the reagents. 



CONCLUSIONS 



The PCR has come a long way since its discovery, evolving from a tool used 
mainly in forensic, medical, pharmaceutical, and plant sciences to food sci- 
ence and the food industry. It may be one of the most remarkable discoveries 
of the 20th century and has opened new doors in a wide array of fields that 
would never have been possible prior to its utilization. Although research 
have shown that PCR can be a powerful method for detection of foodborne 
pathogens in pure culture as well as in certain foods, much more work needs 
to be done to truly make it the best alternative detection technique to con- 
ventional cultural methods. Until the enrichment steps can be eliminated, the 
rapidity of PCR assays can still be argued. Foods also are so different in their 
composition, resulting in a multitude of compounds that may be inhibitory to 
the detection of some pathogens while not affecting others, thus making it 
more challenging to design a one-size-fits-all PCR assay for foods. 



86 Molecular Detection of Foodborne Bacterial Pathogens 



REFERENCES 



1. Aabo, S, O.F. Rasmussen, L. Rossen, RD. Sorensen, and J.E. Olson. 1993. Salmonella 
identification by the polymerase chain reaction. Mol. Cell. Probes 7:171-178. 

2. Aabo, S., J.K Andersen, and J.E. Olson. 1995. Detection of Salmonella in minced 
meat by the polymerase chain reaction. Lett. Applied Microbiol. 21:180-182. 

3. Amagliani, G., G. Brandi, E. Omiccioli, A. Casiere, I.J. Bruce, and M. Magnani. 
2004. Direct detection of Listeria monocytogenes from milk by magnetic based DNA 
isolation and PCR. Food Microbiol. 21:597-603. 

4. Becker, K., R. Roth, and G. Peters. 1998. Rapid and specific detection of toxigenic 
Staphylococcus aureus: Use of two multiplex PCR enzyme immunoassays for ampli- 
fication and hybridization of staphylococcal enterotoxin genes, exfoliative toxin 
genes, and toxic shock syndrome toxin 1 gene. J. Clin. Microbiol. 36:2548-2553. 

5. Bolla, J.M., E. De, A. Dorez, and J.M. Pages. 2000. Purification, characterization, 
and sequence analysis of Omp50, a new porin isolated from Campylobacter jejuni. 
Biochem. J. 352:637-643. 

6. Bubert, A., S. Kohler, and W. Goebel. 1992. The homologous and heterologous 
regions within the iap gene allow genus- and species- specific identification of Listeria 
spp. by polymerase chain reaction. Appl. Environ. Microbiol. 58:2625-2632. 

7. Campbell, M.S. and A.C. Wright. 2003. Real-time PCR analysis of Vibrio vulnificus 
from oysters. Appl. Environ. Microbiol. 69:7137-7144. 

8. Cebula, T.A., W.L. Payne, and P. Feng. 1995. Simultaneous identification of strains 
of Escherichia coli serotype 0157:H7 and their shiga-like toxin type by mismatch 
amplification mutation assay-multiplex PCR. J. Clin. Microbiol. 33:248-250. 

9. Chang, Y.H., Y.H. Shangkuan, H.C. Lin, and H.W. Liu. 2003. PCR assay of the 
groEL gene for detection and differentiation of Bacillus cereus group cells. Appl. 
Environ. Microbiol. 69:4502^-510. 

10. Chen, T.R., C.S. Chiou, and H.Y Tsen. 2004. Use of novel PCR primers specific to 
the genes of staphylococcal enterotoxin G, H, I for the survey of Staphylococcus 
aureus strains isolated form food-poisoning cases and food samples in Taiwan. Int. 
J. Food Microbiol. 92:189-197. 

11. Cocolin, L., M. Manzano, C. Cantoni, and G Comi. 1998. Use of polymerase chain 
reaction and restriction enzyme analysis to directly detect and identify Salmonella 
typhimurium in food. J. Appl. Microbiol. 85:673-677. 

12. Davis, C.R., L.C. Heller, K.K. Peak, D.L. Wingfield, C.L. Goldstein-Hart, D.W. 
Bodager, A.C. Cannons, P.T. Amuso, and J.C. Cattani. 2004. Real-time PCR detec- 
tion of the thermostable direct hemolysin and thermolabile hemolysin genes in a 
Vibrio parahaemolyticus cultured from mussels and mussel homogenate associated 
with a foodborne outbreak. J. Food Prot. 67:1005-1008. 

13. Dedieu, L., J.M. Pages, and J.M. Bolla. 2004. Use of the omp50 gene for identifica- 
tion of Campylobacter species by PCR. J. Clin. Microbiol. 42:2301-2305. 

14. Denis, M., C. Soumet, K. Rivoal, G Ermel, D. Blivet, G Salvat, and P. Colin. 1999. 
Development of an m-PCR assay for simultaneous identification of Campylobacter 
jejuni and C coli. Lett. Appl. Microbiol. 29:406-410. 

15. Ehling-Schulz, M., M. Fricker, and S. Scherer. 2004. Identification of emetic toxin 
producing Bacillus cereus strains by a novel molecular assay. FEMS Microbiol. Lett. 
232:189-195. 

16. Ercolini, D., G Blaiotta, V. Fusco, and S. Coppola. 2004. PCR-based detection of 
enterotoxigenic Staphylococcus aureus in the early stages of raw milk cheese making. 
J. Appl. Microbiol. 96:1090-1096. 



References 87 



17. Fach, P. and M.R. Popoff. 1997. Detection of enterotoxigenic Clostridium perfrin- 
gens in food and fecal samples with a duplex PCR and the slide latex agglutination 
test. Appl. Environ. Microbiol. 63:4232-4236. 

18. Fatemeh, R., H. Michael, H. Water, and C. Carl. 1995. Survival of Shigella flexneri 
on vegetables and detection by polymerase chain reaction. J. Food Prot. 58:727-732. 

19. Fluit, A.C., R. Torensma, M.J.C. Visser, C.J.M. Aarsman, J.J.G. Poppelier, B.H.I. 
Keller, P. Klapwijk, and J. Verhoef. 1993. Detection of Listeria monocytogenes in 
cheese with the magnetic immuno-polymerase chain reaction assay. Appl. Environ. 
Microbiol. 59:1289-1293. 

20. Fluit, A.C., M.N. Widjojoatmodjd, A.T.A. Box, R. Torensma, and J. Verhoef. 1993. 
Rapid detection of Salmonella in poultry with the magnetic immuno-polymerase 
chain reaction assay. Appl. Environ. Microbiol. 59:1342-1346. 

21. Fratamico, P.M., L.K. Bagi, and T. Pepe. 2000. A multiplex polymerase chain reac- 
tion of Escherichia coli 0157:H7 in foods and bovine feces. J. Food Prot. 
63:1032-1037. 

22. Fratamico, P.M. and T.P. Strobaugh. 1998. Simultaneous detection of Salmonella 
spp. and Escherichia coli 0157:H7 by multiplex PCR. J. Industrial Microbiol. 
Biotechnol. 21:92-98. 

23. Harvard, H.L., E.C. Hunter, and W. Titball. 1992. Comparison of the nucleotide 
sequence and development of a PCR test for the epsilon toxin gene of Clostridium 
perfringens type B and type D. FEMS Microbiol. Lett. 97:77-82. 

24. Hansen, B.M., T.D. Leser, and N.B. Hendriksen. 2001. Polymerase chain reaction 
assay for the detection of Bacillus cereus group cells. FEMS Microbiol. Lett. 
202:209-213. 

25. Hough, A.J., S.A. Harbison, M.G. Savill, L.D. Melton, and G Fletcher. 2002. Rapid 
enumeration of Listeria monocytogenes in artificially contaminated cabbage using 
real-time polymerase chain reaction. J. Food Prot. 65:1329-1332. 

26. Houng, H.S., O. Sethabutr, and P. Echeverria. 1997. A simple polymerase chain 
reaction technique to detect and differentiate Shigella and enteroinvasive Escherichia 
coli in human feces. Diag. Microbiol. Infect. Dis. 28:19-25. 

27. Jackson, C.J., A.J. Fox, and D.M. Jones. 1996. A novel polymerase chain reaction 
assay for the detection and specification of thermophilic Campylobacter spp. 
J. Appl. Bacteriol. 81:467-473. 

28. Jitrapakdee, S., A. Tassanakajon, V. Boonsaeng, S. Piankijagum, and S. Panyim. 
1995. A simple, rapid and sensitive detection of Salmonella in food by polymerase 
chain reaction. Mol. Cell. Probes 9:375-382. 

29. Jourdan, A.D., S.C. Johnson, and I.V. Wesley 2000. Development of a fluorogenic 
5' nuclease PCR assay for detection of the ail gene of pathogenic Yersinia enter oco- 
litica. Appl. Environ. Microbiol. 66:3750-3755. 

30. Jung, Y.S., J.F. Frank, R.E. Brackett, and J.R. Chen. 2003. Polymerase chain reac- 
tion detection of Listeria monocytogenes on frankfurters using oligonucleotide 
primers targeting the genes encoding internalin AB. J. Food Prot. 66:237-241. 

31. Karunasagar, I., G Sugumar, I. Karunasagar, and P.J. Reilly. 1996. Rapid poly- 
merase chain reaction method for detection of Kanagawa positive Vibrio para- 
haemolyticus in seafoods. Int. J. Food Microbiol. 31:317-323. 

32. Kim, S., R. G Labbe, and S. Ryu. 2000. Inhibitory effects of collagen on the PCR 
for detection of Clostridium perfringens. Appl. Environ. Microbiol. 66:1213-1215. 

33. Kim, Y.B., J. Okuda, C. Matsumoto, N. Takahashi, S. Hashimoto, and M. Nishibuchi. 
1999. Identification of Vibrio par ahaemolyticus strains at the species level by PCR tar- 
geted to the toxR gene. J. Clin. Microbiol. 37:1 173-1 177. 



88 Molecular Detection of Foodborne Bacterial Pathogens 



34. Klein, P. A. and V.K. Juneja. 1997. Sensitive detection of viable Listeria monocyto- 
genes by reverse transcription-PCR. Appl. Environ. Microbiol. 63:4441^1448. 

35. Kwang, J., E.T. Littledike, and J.E. Keen. 1996. Use of the polymerase chain reac- 
tion for Salmonella detection. Lett. Appl. Microbiol. 22:46-51. 

36. Lampel, K.A., P.A. Orlandi, and L. Kornegay 2000. Improved template preparation 
method for PCR-based assays for detection of foodborne bacterial pathogens. Appl. 
Environ. Microbiol. 66:4539^542. 

37. Lantz, P.G., R.K. Knutsson, Y. Blixt, W.A. Al-Soud, E. Borch, and P. Radstrom. 
1998. Detection of pathogenic Yersinia enter ocolitica in enrichment media and pork 
by a multiplex PCR: A study of sample preparation and PCR-inhibitory compo- 
nents. Int. J. Food Microbiol. 45:93-105. 

38. Lee, J.W., I.J. Jun, H.J. Kwun, K.L. Jang, and J. Cha. 2004. Direct identification of 
Vibrio vulnificus by PCR targeting elastase gene. J. Microbiol. Biotechnol. 14:284-289. 

39. Lee, J.Y., Y.B .Bang, J.H. Rhee, and S.H. Choi. 1999. Two stage nested PCR effec- 
tiveness for direct detection of Vibrio vulnificus in natural samples. J. Food Sci. 
64:158-162. 

40. Lett, P.W., J.P. Southworth, D.D. Jones, and A.K. Bej. 1995. Detection of patho- 
genic Escherichia coli in ground beef using multiplex PCR. Food Testing & Analysis 
1:34-38. 

41. Li, Y. and A. Mustapha. 2004. Simultaneous detection of Escherichia coli 0157:H7, 
Salmonella, and Shigella in apple cider and produce by a multiplex PCR. J. Food 
Prot. 67:27-33. 

42. Li, Y, S. Zhuang, and A. Mustapha. 2005. Application of a multiplex PCR for the 
simultaneous detection of Escherichia coli 0157:H7, Salmonella and Shigella in raw 
and ready-to-eat meat products. Meat Sci. 71:402-406. 

43. Lin, C.K., C.L. Hung, S.C. Hsu, C.C. Tsai, and H.Y Tsen. 2004. An improved PCR 
primer pair based on 1 6S rDNA for the specific detection of Salmonella serovars in 
food samples. J. Food Prot. 67:1335-1343. 

44. Lin, J.S. and H.Y. Tsen. 1999. Development and use of polymerase chain reaction 
for the specific detection of Salmonella Typhimurium in stool and food samples. 
J. Food Prot. 62:1103-1110. 

45. Lindqvist, R. 1999. Detection of Shigella spp. in food with a nested PCR 
method — Sensitivity and performance compared with a conventional culture 
method. J. Appl. Microbiol. 86:971-978. 

46. Linton, D., A.J. Lawson, R.J. Owen, and J. Stanley. 1997. PCR detection, identifi- 
cation to species level, and fingerprinting of Campylobacter jejuni and 
Campylobacter coli direct from diarrheic samples. J. Clin. Microbiol. 35:2568-2572. 

47. Liu, D., A.J. Ainsworth, F.W. Austin, and M.L. Lawrence. 2004. Use of PCR 
primers derived from a putative transcriptional regulator gene for species-specific 
determination of Listeria monocytogenes. Int. J. Food Microbiol. 91:297-304. 

48. Longhi, C, A. Maffeo, M. Penta, G. Petrone, L. Seganti, and M.P. Conte. 2003. 
Detection of Listeria monocytogenes in Italian-style soft cheeses. J. Appl. Microbiol. 
94:879-885. 

49. Luk, J.M., U. Kongmuang, PR. Reeves, and A. A. Lindberg. 1993. Selective ampli- 
fication of abequose and paratose synthase gene (rfb) by polymerase chain reaction 
of Salmonella major serotypes (A, B, C2, D). J. Clin. Microbiol. 31:2118-2123. 

50. Lyon, W.J. 2001. Taqman PCR for detection of Vibrio cholerae Ol, 0139, Non-Ol, 
and Non-Ol 39 in pure cultures, raw oysters, and synthetic seawater. Appl. Environ. 
Microbiol. 67:4685-4693. 



References 



89 



51. Manzano, M., C. Giusto, L. Lacumin, C. Cantoni, and G. Comi. 2003. A molecu- 
lar method to detect Bacillus cereus from a coffee concentrate sample used in indus- 
trial preparations. J. Appl. Microbiol. 95:1361-1366. 

52. Mclngvale, S.C., D. Elhanafi, and M.A. Drake. 2002. Optimization of reverse tran- 
scriptase PCR to detect viable Shiga-toxin-producing Escherichia coli. Appl. 
Environ. Microbiol. 68:799-806. 

53. McKillip, J.L. and M. Drake. 2004. Real time nucleic acid-based detection methods 
for pathogenic bacteria in food. J. Food Prot. 67:823-832. 

54. Meng, X, S. Zhao, M.P. Doyle, S.E. Mitchell, and S. Kresovich. 1997. A multiplex 
PCR for identifying Shiga-like toxin-producing Escherichia coli 0157:H7. Lett. 
Appl. Microbiol. 24:172-176. 

55. Miyamoto, T., N. Ichioka, C. Sasaki, H.K. Kobayashi, K. Honjoh, M. Iio, and 
S. Hatano. 2002. Polymerase chain reaction assay for detection of Escherichia coli 
0157:H7 and E. coli 0157:H-. J. Food Prot. 65:5-11. 

56. Osek, J. 2001. Multiplex polymerase chain reaction assay for identification of 
enterotoxigenic Escherichia coli strains. J. Vet. Diag. Invest. 13:308-311. 

57. Panicker, G, M.L. Myers, and A.K. Bej. 2004. Rapid detection of Vibrio vulnificus 
in shellfish and Gulf of Mexico water by real-time PCR. Appl. Environ. Microbiol. 
70:498-507. 

58. Rasmussen, H.N., J.E. Olsen, K. Jorgensen, and O.F. Rasmussen. 1996. Detection of 
Campylobacter jejuni and Campylobacter coli in chicken faecal samples by PCR. 
Lett. Appl. Microbiol. 23:363-366. 

59. Sails, A.D., A.J. Fox, F.J. Bolton, D.R. Waring, and D.L. Greenway. 2003. A real- 
time PCR assay for the detection of Campylobacter jejuni in foods after enrichment 
culture. Appl. Environ. Microbiol. 69:1383-1390. 

60. Schraft, H. and M.W. Griffiths. 1995. Specific oligonucleotide primers for detec- 
tion of lecithinase positive Bacillus spp. by PCR. Appl. Environ. Microbiol. 
61:98-102. 

61. Seo, K.H., I.E. Valentin-Bon, R.E. Brackett, and PS. Holt. 2004. Rapid, specific 
detection of Salmonella Enteritidis in pooled eggs by real-time PCR. J. Food Prot. 
67:864-869. 

62. Sharma, VK. 2002. Detection and quantitation of enterohemorrhagic Escherichia 
coli 0157, Ol 11, and 026 in beef and bovine feces by real-time polymerase chain 
reaction. J. Food Prot. 65:1371-1380. 

63. Somer, L. and Y. Kashi. 2003. A PCR method based on 16S rRNA sequence for 
simultaneous detection of the genus Listeria and the species Listeria monocytogenes 
in food products. J. Food Prot. 66:1658-1665. 

64. Soumet, C, G Ermel, N. Rose, V Drouin, G. Sal vat, and P. Colin. 1999. Evaluation 
of a multiplex PCR assay for simultaneous identification of Salmonella sp., 
Salmonella Enteritidis, and Salmonella Typhimurium from environmental swabs of 
poultry houses. Lett. Appl. Microbiol. 28:113-117. 

65. Straub, J. A., C. Hertel, and W.P. Hammes. 1999. A 23S rDNA-targeted polymerase 
chain reaction-based system for detection of Staphylococcus aureus meat starter cul- 
tures and dairy products. J. Food Prot. 62:1150-1156. 

66. Szabo, E.A., and B.M. Mackey. 1999. Detection of Salmonella Enteritidis by reverse 
transcription-polymerase chain reaction. Int. J. Food Microbiol. 51:113-122. 

67. Tamarapu, S., J.L. McKillip, and M. Drake. 2001. Development of a multiplex poly- 
merase chain reaction assay for detection and differentiation of Staphylococcus 
aureus in dairy products. J. Food Prot. 64:664-668. 



90 Molecular Detection of Foodborne Bacterial Pathogens 



68. Taylor, M.S., A. Challed-Spong, and E.A. Johnson. 1997. Co-amplification of the 
amelogenin and HLA DQ? genes: Optimization and validation. J. Forensic Sci. 
42:130-136. 

69. Tsen, H.Y., L.Z. Jian, and W.R. Chi. 1998. Use of a multiplex PCR system for the 
simultaneous detection of heat labile toxin I and heat stable toxin II genes of entero- 
toxigenic Escherichia coli in skim milk and porcine stool. J. Food Prot. 61:141-145. 

70. Uzal, FA., J.J. Plumb, L.L. Blackall, and W.R. Kelly 1997. PCR detection of 
Clostridium perfringens producing different toxins in faeces of goats. Lett. Appl. 
Microbiol. 25:339-344. 

71. Villalobo, E. and A. Torres. 1998. PCR for detection of Shigella spp. in mayonnaise. 
Appl. Environ. Microbiol. 64:1242-1245. 

72. Vishnubhatla, A., R.D. Oberst, D.Y. Fung, W. Wonglumsom, M.P. Hays, and T.G. 
Nagaraja. 2001. Evaluation of a 5' nuclease (TaqMan) assay for the detection of 
virulent strains of Yersinia enter ocolitica in raw meat and tofu samples. J. Food Prot. 
64:355-360. 

73. Wilson, I.G. 1997. Inhibition and facilitation of nucleic acid amplification. Appl. 
Environ. Microbiol. 63:3741-3751. 

74. Yeung, P.S., A. Depaola, C.A. Kaysner, and K.J. Boor. 2003. A PCR assay for spe- 
cific detection of the pandemic Vibrio par ahaemolyticus 03:K6 clone from shellfish. 
J. Food Sci. 68:1459-1466. 



CHAPTER 6 



Molecular Approaches for the Detection 

of Foodborne Viral Pathogens 

Doris H. D'Souza* and Lee-Ann Jaykus 

Department of Food Science, College of Agriculture and Life Sciences, 
North Carolina State University, Raleigh, NC 27695-7624 



Introduction 

General Detection Considerations and the Challenges 

Virus Concentration 

Virus Concentration Methods for Shellfish 

Virus Concentration Methods for Other Than Shellfish 

Nucleic Acid Extraction 

Detection 

RT-PCR Detection of Viruses in Foods 
Alternative Nucleic Acid Amplification Methods 

Confirmation 

Real-Time Detection 

Conclusions 

Acknowledgments 

References 



INTRODUCTION 



The human enteric viruses are now recognized as major causes of acute non- 
bacterial gastroenteritis throughout the world. Those of primary epidemiolog- 
ical significance include hepatitis A virus (HAV) and the noroviruses, formerly 
known as the Norwalk-like viruses (NLVs), or the small round structured 
viruses (SRSVs) (reviewed in 88). Currently, the noroviruses consist of five 
genogroups: GI (prototype Norwalk virus); Gil (prototype Snow Mountain 
Agent); GUI (prototype bovine enteric calicivirus); GIV (prototype Alphatron 
and Fort Lauderdale virus); and GV (prototype Murine norovirus) (Vinje, per- 
sonal communication). The sapoviruses (previously called Sapporo-like viruses) 
are genetically related to the noroviruses and have occasionally caused viral gas- 
troenteritis in humans. Both the noroviruses and the sapoviruses are members 
of the Calicivirideae family, an antigenically and genetically diverse group of 
gastrointestinal viruses (9, 39, 40). Other viruses that can cause food and water- 
borne disease include the adenoviruses, astroviruses, the human enteroviruses 
(polioviruses, echoviruses, groups A and B coxsackieviruses), hepatitis E virus, 



Corresponding author: Phone: (919) 513-2076, FAX: (919) 513-0014 e-mail: 
ddsouza@unity.ncsu.edu 



91 



92 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



parvoviruses, and other relatively uncharacterized small round viruses. The 
rotaviruses, which are the leading cause of infantile diarrhea worldwide, are 
transmitted primarily by contaminated water but can on occasion be foodborne. 

The human enteric viruses replicate in the intestines of infected human hosts 
and are excreted in the feces. Their primary mode of transmission is the 
fecal-oral route through contact with human fecal matter, although they may 
also be shed in vomitus. These viruses are readily spread by person-to-person 
contact, which is frequently responsible for the propagation of primary food- 
borne outbreaks. Contamination of foods may occur directly, through poor 
personal hygiene practices of infected food handlers, or indirectly via contact 
with fecally contaminated waters or soils (49, 50). Since viruses must survive the 
pH extremes and enzymes present in the human gastrointestinal tract, they are 
regarded as highly environmentally stable, allowing virtually any food to serve 
as a vehicle for their transmission (50). Although enteric viruses are unable to 
replicate in contaminated foods, they are able to withstand a wide variety of 
food processing and storage conditions. When present in contaminated food, 
their numbers are usually quite low, but since their infectious doses are also low, 
any level of contamination may pose a public health threat. 

In spite of their initial recognition decades ago, the human enteric viruses 
can be considered "emerging" agents of foodborne disease, mainly because only 
recently have scientists been able to reliably detect these pathogens. In fact, prior 
to the advent of molecular biological techniques, epidemiological criteria were 
the best means, by which cases of enteric viral illness were recognized. Over the 
last 10 years, significant advances in nucleic acid amplification methods have 
made detection of enteric viruses in human clinical samples, specifically feces, 
all but routine. 

The opposite is the case for the detection of viruses in foods. Historically, 
this has been done by infectivity assay using susceptible, live laboratory hosts. 
Host systems employed were mainly mammalian cell cultures of primate origin. 
Unfortunately, the epidemiologically important human enteric viruses, includ- 
ing the noroviruses and wild-type hepatitis A virus, cannot be propagated in 
mammalian cell culture systems, and so these are not viable detection options 
(49, 50). In the absence of in vitro virus propagation methods, nucleic acid 
amplification has been a promising alternative. In this chapter, we will discuss 
existing technologies that can be applied to the detection of viruses in foods, 
and we will address new developments and research needs for the application of 
these methods on a routine basis. 



GENERAL DETECTION CONSIDERATIONS 
AND THE CHALLENGES 



The development of effective virus detection methods from food commodities 
poses several challenges. Like many bacterial pathogens, these agents are typi- 
cally present at low levels in contaminated foods. However, unlike bacterial 
pathogens, viruses cannot replicate in foods, making the use of traditional 



Virus Concentration 93 



food microbiological techniques of cultural enrichment and selective plating 
inapplicable per se. Therefore, the first goal in developing virus detection meth- 
ods for foods is to separate and concentrate the agents from the food matrix. It 
is also necessary to sample relatively large volumes to assure adequate sample 
representation, thereby optimizing detection assay sensitivity. 



VIRUS CONCENTRATION 



Sample preparation prior to detection is of key importance when applying 
molecular methods to detect viral contamination in foods. In this regard, spe- 
cific challenges include high sample volumes in relation to small amplification 
volumes, low levels of contamination, and the presence of residual food com- 
ponents that can later compromise detection (31, 37, 86, 97). 

The main goals of virus concentration methods are to decrease sample vol- 
ume and eliminate matrix-associated interfering substances, while simultane- 
ously recovering most of the viruses present in the food sample. In order to 
achieve these goals, sample manipulations are undertaken that capitalize on the 
behavior of viruses to act as proteins in solutions, and to remain infectious at 
extremes of pH or in the presence of organic solvents. Because of their frequent 
association with viral foodborne disease outbreaks, early work on virus concen- 
tration and purification from foods focused mainly on bivalve molluscan shell- 
fish. Recent research endeavors have included a broader range of at-risk foods. 

Two major approaches to virus concentration, particularly as applied to shell- 
fish commodities, are termed extraction-concentration and adsorption- 
elution-concentration (49). Both methods utilize conditions that promote the 
separation of viruses from shellfish tissues, through the use of filtration, centrifu- 
gation, adsorption, elution, solvent extraction, precipitation, and/or organic floc- 
culation. The procedure generally begins with sample blending in a buffer, usually 
containing amino acids and an elevated pH. A common example of elution buffer 
is 0.1 M glycine-0.14 N saline, pH 9.0. A crude filtration step through a mesh 
material such as cheesecloth may be done to remove large sample particulates. 
Viruses do not sediment unaided, even at centrifugation speeds approaching 
10,000 x g. Therefore, centrifugation can be used to sediment large food parti- 
cles, with the recovery of the virus-containing supernatant. The next step usu- 
ally involves pH manipulation or the addition of precipitation agents, creating 
conditions such that viruses adsorb to the remaining shellfish tissues. Upon 
subsequent centrifugation, the adsorbed viruses sediment with the tissues and 
the supernatant is discarded. Elution, whereby the viruses are desorbed from 
the tissues by further pH and/or ionic manipulations is then carried out. On 
subsequent centrifugation, the precipitated tissue is discarded, again retaining 
the supernatant. Using sequential steps of adsorption, elution, filtration, pre- 
cipitation, and centrifugation, viruses are concentrated to small sample volumes 
and simultaneously purified, with the removal of large proportions of the food 
matrix and matrix-associated organic materials that may later compromise 
detection of the viruses. 



94 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



Precipitation of viruses can be achieved by lowering pH, so called acid precip- 
itation, or by the addition of polyethylene glycol (PEG). Both methods capitalize 
on the property that viruses behave as proteins in solution. The viruses, along with 
some of the matrix-associated proteins, will precipitate when the pH is lowered to 
that approximating the virus isoelectric point. Polyethylene glycol causes removal 
of water, allowing proteins to fall out of solution. Another method similar to 
precipitation is organic flocculation. Flocculating agents interact with organic 
material in the matrix, causing the formation of a gelatinous "floe," to which 
the viruses absorb (49, 50). In the case of acid and PEG precipitation, and in 
organic flocculation, the virus-containing solid materials can be readily harvested 
by centrifugation, usually done at fairly low speeds (e.g., <5000 x g). 

Further removal of matrix-associated organic materials can be done using a 
variety of agents. Since viruses remain infectious even after exposure these 
organic solvents such as chloroform, trichloro trifluoroethane (Freon) and 
more environmentally friendly solvents such as Vertrel (Dupont), these chemi- 
cals can be used to remove polar food components such as lipids. Alternative 
commercial virus purification agents such as ProCipitate and Viraffinity 
(LigoChem, Inc., Fairfield, NJ 07004) (31, 51, 66) are known to eliminate poly- 
saccharides, an important matrix-associated inhibitor in shellfish and produce. 
The cationic detergent cetyltrimethylammonium bromide (CTAB) (6, 7, 52) 
also aids in the removal of polysaccharides. The use of Sephadex (23), cellulose 
(110), or Chelex (98) is helpful in the elimination of salts and small proteins. 
Ultrafiltration, a method that is frequently applied at the latter stages of a 
virus concentration scheme, provides reduction in volume while simultaneously 
purifying the sample. 



VIRUS CONCENTRATION METHODS FOR SHELLFISH 



The early molecular work aimed at detecting viruses in the food matrix focused 
almost exclusively on shellfish. The most common method utilizes some sort of 
virion concentration step prior to release or extraction of nucleic acid, followed 
by amplification. A second, less commonly used method resorts to direct 
nucleic acid extraction of a previously untreated food matrix, which circum- 
vents the need for virus concentration. 

The objective of the virion concentration approach is to concentrate viruses 
and remove inhibitors prior to nucleic acid amplification, with or without prior 
nucleic acid extraction. In early work by Atmar et al. (6, 7), investigators 
processed artificially contaminated shellfish samples using an initial concentra- 
tion and utilized a purification scheme that consisted of solvent extraction 
and PEG precipitation steps. This was followed by nucleic acid extraction and 
subsequent amplification. Cetyltrimethylammonium bromide (CTAB) was 
added to remove residual inhibitors from crude nucleic acid extracts, and the 
resulting eluant was amplified using reverse transcriptase-polymerase chain 
reaction (RT-PCR) (6, 7). Dissecting the oysters, discarding the muscle tissue 
and processing only the digestive diverticula improved the PCR's detection 



Virus Concentration Methods for Shellfish 95 



limits. This sampling approach is now the method of choice (11, 65, 67, 69, 90, 92). 
Figure 6.1 illustrates a representative sample-preparation protocol for shellfish. 
Note that second generation protocols frequently employ sequential PEG 
precipitation in addition to adsorption, elution, and solvent extraction steps 
(8, 16, 19, 20, 25, 26, 30, 59, 60, 63, 64). 

Some investigators apply an antibody capture step to further concentrate 
and purify viruses from shellfish extracts prior to detection using RT-PCR. 



25 g OYSTER MEAT 

t 

HOMOGENIZATION 

Add 175 ml of sterile cold deionized water (ratio of meat: water is 1:7) 

t 
VIRUS ADSORPTION 

Adjust pH to 4.8 and with conductivity of < 2000 jiS using water 

t 

CENTRIFUGATION 

Collect pellets at 2,000 x g for 20 min 

t 
VIRUS ELUTION 

Add 0.75 M glycine-0.15 M NaCl, pH 7.6 (1:7 ratio of meat: eluant buffer) 

pH adjustment to 7.5 to 7.6, Vortex at room temp for 15 mins 

Centrifuge at 5,000 x g for 20 min at 4 C 

Collect supernatant (A) 

t 
RE-ELUTE VIRUS FROM PELLET 

Add 0.5 M threonine-0.15 M NaCl, pH 7.6 (1:7 ratio) 

Collect supernatant (B) 

Combine/Pool supernatant A and B 

t 
PEG PRECIPITATION 

Add 8% PEG 8000-0.3 M NaCl 

Incubate at 4 C for 4 h or overnight 

Centrifuge 6,700 x g for 30 min 

Collect pellets and resuspend in 10 ml of Phosphate-buffered saline 

t 

SOLVENT EXTRACTION OF VIRUS 

Add 10 ml of chloroform (ratio of eluant: chloroform is 1:1) 

Centrifuge at 1, 700 x g for 30 min 

Collect supernatant 1 

t 
Reextract from bottom layer with half volume of 0.5 M threonine 

Collect supernatant 2 

Combine/pool supernatant 1 and 2 

t 
PEG PRECIPITATION 

Add 8% PEG-0.3 M NaCl at 4 C for 2-4 h 
Centrifuge at 14,000 x g for 15 min, Collect pellets 

t 
RNA EXTRACTION 



Figure 6.1. Concentration of viruses from oysters using virus adsorption, glycine- 
saline buffer and threonine-saline extraction, PEG precipitation, chloroform extrac 
tion, and PEG concentration as modified by Shieh et al. (95). 



96 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



Desenclos et al. (29) were the first to implicate hepatitis A virus in oyster out- 
break specimens by immunocapture of the virus, heat release of viral nucleic 
acids, and subsequent RT-PCR detection. Capitalizing on the work of Jansen 
et al. (48), other investigators have coated paramagnetic beads with anti-HAV 
IgG and used these to capture HAV from oyster extracts initially processed for 
virus concentration using a combination of elution, poly electrolyte floccula- 
tion, filtration and/or ultrafiltration (28, 71). Sunen et al. (100) and Schwab 
et al. (94) also used antibody capture as a final virus concentration step. More 
recently, Kobayashi et al. (61) used magnetic beads coated with the antibody to 
the baculovirus-expressed recombinant capsid proteins of the Chiba virus 
(rCV) to capture noroviruses from food items implicated in an outbreak of 
acute gastroenteritis in Aichi Prefecture, Japan, detecting the virus in these 
foods by RT-PCR. Abd El Galil et al. (1) have used immunomagnetic separa- 
tion for the detection of hepatitis A virus from environmental samples using 
real-time nucleic acid amplification methods. 

The alternative approach of direct nucleic acid extraction and RT-PCR 
applied to unprocessed food sample, involves extraction of total RNA from the 
sample without any prior sample manipulations. This method is best suited for 
simple sample matrices such as the surfaces of fresh fruits and vegetables. 
However, Legeay et al. (65) recently reported a method that involved enzymatic 
liquefaction of shellfish digestive tissues, followed by clarification using 
dichloromethane extraction. In this case, the investigator reported that the sam- 
ple could be directly processed for nucleic acid isolation and subsequent virus 
detection by RT-PCR. 



VIRUS CONCENTRATION METHODS FOR FOODS 
OTHER THAN SHELLFISH 



Gouvea et al. (36) first reported a systematic method for the detection of 
norovirus and rotavirus from representative food commodities other than shell- 
fish, including orange juice, milk, lettuce, and melon. The method involved 
blending or washing, clarification by centrifugation, and removal of inhibitors 
by Freon extraction followed by RNA extraction. Leggitt and Jaykus (66) devel- 
oped a prototype method for the concentration of poliovirus, hepatitis A virus, 
and Norwalk virus from 50 g samples of artificially contaminated hamburger 
and lettuce. The steps used included homogenization, filtration through cheese- 
cloth, Freon extraction (hamburger only) and two sequential PEG precipita- 
tions. The sequential precipitations, which used increasing PEG concentrations, 
resulted in a 10- to 20-fold sample volume reduction from 50 g to approximately 
2.5 ml (66). The resuspended PEG precipitate could be assayed for virus recov- 
ery by mammalian cell culture infectivity assay, when applicable, which allows 
for direct comparison between virus infectivity and molecular detection (104). 
Subsequent nucleic acid extraction resulted in an additional 100-fold sample 
volume reduction with detection at initial inoculum levels of >10 2 infectious 
units per 50-g food sample (66). A schematic overview of this procedure is 



Virus Concentration Methods for Foods Other than Shellfish 97 



provided in Figure 6.2. Schwab et al. (91) used TRIzol, a proprietary RNA 
extraction method, as a surface wash for deli meats, including samples artifi- 
cially contaminated with norovirus and ones implicated in an outbreak of 
norovirus gastroenteritis. Although simple, the main drawback of the TRIzol 
surface wash method was that nucleic acid amplification inhibition persisted 
unless sample concentrates were diluted 10- to 100-fold. A flow diagram of this 
protocol is depicted in Figure 6.3. 

Bidawid et al. (12) reported an immunocapture method for the concentration 
of hepatitis A virus from lettuce and strawberries. After surface washing to elute 
the viruses, the wash solution was passed through a positively charged filter, 
eluted, and concentrated by immunocapture. As few as 10 PFU of cell culture- 
adapted hepatitis A virus per piece of lettuce or strawberry could be detected by 
RT-PCR using this sample preparation method. However, this method too had 
problems with residual matrix-associated amplification inhibition. 



50 g FOOD SAMPLE 

(Complex food such as hamburger) 
Add 350 ml of 0.05 M Glycine/0.14 N Saline, pH 9.0 

HOMOGENIZE (350-400 ml) 

I 

FILTER 

Cheesecloth 



I 



SOLVENT EXTRACT 

(Vortex gently with 60% Freon or Chloroform: Isobutanol (1:1) as needed, ~70 ml) 

Centrifuge at 2,500 x g for 10 min. 

I 

1° PEG PRECIPITATION and ELUTION 

To eluant add 6% PEG, pH to 7.2, incubate 4°C, 2 H. 

Centrifuge 5000 xg for 15 min. 
Resuspend pellet in 25 ml of 50mM Tris, 0.2% Tween 20, pH 9.0 

Elute at room temperature for 1 H. 
Centrifuge at 3500 x g for 15 min. 

(30-35 ml) 



I 



2°PEG PRECIPITATION and RESUSPENSION 

To eluant add 12% PEG; incubate 4°C, 2 H. 

Centrifuge at 5000 x g for 15 min. 

Resuspend pellet in 50mM Tris, 0.2% Tween 20, pH 8.0 

(3-5 ml) 

I I 

RNA EXTRACTION and RT-PCR OR CELL-CULTURE ASSAY 

(25-40 ul) 



Figure 6.2. Virus concentration from hamburger/complex foods using glycine-saline 
buffer, chloroform extraction and two PEG extractions as followed by Leggitt and 
Jaykus (66) 



98 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



25-50 g PRODUCE SAMPLE 

TRIZOL® SURFACE WASH OR BUFFERED SURFACE WASH 

(4 mis of TRIzol® Reagent, twice) 0.05 M Glycine-0.14M Saline buffer, pH 9.0 

(4 ml of buffer, twice) 

ELUTE AND CLARIFY SUPERNATANT 

Centrifuge 8ml at 5000 rpm for 15 mins at 4°C 

EXTRACT RNA 

Add 1.6 ml of Chloroform to 8 ml of TRIzol/Glycine extract 
Shake and incubate at room temperature for 2-3 min. 

t 
Centrifuge 9000 rpm for 15 min. at 4°C 

Collect aqueous phase (~ 4.8 ml) 

t 
Add 4 ml of isopropanol and incubate at room temperature for 10 min. 

t 
Centrifuge at 9000 rpm for 10 min. at 4°C 

Wash pellet with 8ml of 75% ethanol 

Centrifuge 7000 rpm for 5 min. at 4°C 

i 

Air-dry for 5 min. 
Dissolve pellet in 100 \il of sterile water and store at -70°C to -80°C 



Figure 6.3. Virus concentration and extraction from produce using TRIzol® or 
glycine-saline buffer (modifications of protocol by Schwab et al. (91)). 



As a general rule, virus concentration methods result in sample volume 
reductions ranging from 10- to 1000-fold. This means that a 25-g sample theo- 
retically can be reduced to 25 |il-2.5 |Lil volumes with recovery of infectious 
virus (50). The yields after virus concentration in a food matrix have ranged 
from as low as 1-2% to as high as 90%. Recovery efficiency is almost always 
virus-specific, usually hepatitis A virus recovery is quite low when compared to 
recovery of other viruses such as poliovirus (reviewed in 50, 88). 

Although virus concentration and sample purification steps prior to nucleic 
acid amplification can achieve significant sample volume reduction with rela- 
tively efficient virus recovery, there are some additional considerations when 
selecting a virus concentration approach. The antibody capture methods are 
often simpler than others as they may require fewer sample manipulations. 
There is also speculation that antigen-associated viral nucleic acid is more 
highly associated with infectious virus. However, these methods may be limited 
by reagent availability and high specificity, which means that only a single virus 
type is detected in a single assay. Other concentration and purification methods 
that rely on steps such as PEG precipitation and solvent extraction usually 
require manipulations, which may result in substantial virus loss during extrac- 
tion. The direct nucleic acid extraction methods almost always result in residual 
RT-PCR inhibitors and often times do not provide adequate sample volume 
reduction. 



Nucleic Acid Extraction 99 



In short, there are many virus extraction and concentration approaches that 
have been applied in a variety of instances. All of these methods have limita- 
tions that ultimately hinder the ability to detect the relatively low levels of virus 
that might be anticipated in naturally contaminated foods. 



NUCLEIC ACID EXTRACTION 



The earliest and simplest protocol for amplifying nucleic acids from shellfish 
concentrates was direct heat release followed by RT-PCR, in which case, an 
RNA extraction step was not applied (31, 51, 52). It soon became apparent, 
however, that in many instances the additional volume reductions and sample 
clean up provided by RNA extraction was critical to improving detection lim- 
its and circumventing the effects of residual matrix-associated amplification 
inhibition. 

Before applying nucleic acid amplification, an efficient nucleic acid extraction 
step is critical in most instances. This is important because the amplification effi- 
ciency is dependent on both the purity of the target template and the quantity 
of target molecules obtained from the sample. Accordingly, the main goals of 
nucleic acid extraction are: (1) to extract and purify the nucleic acids, (2) to 
provide additional sample concentration, and (3) to remove residual matrix- 
associated inhibitory substances that could remain after the initial concen- 
tration steps are completed. The components that can hinder molecular 
amplification are diverse and include compounds such as divalent cations, 
matrix-associated components such as proteoglycans (42), polysaccharides, (6, 7, 
27), glycogen, (6, 7, 51, 52), and lipids (84, 87, 91), among others. In many cases, 
inhibitory compounds have been largely uncharacterized. Matrix-associated 
inhibitors usually act by degrading the target and/or primer nucleic acids, and/or 
inactivating or inhibiting enzymes (86, 109, 111). Unfortunately, these inhibitory 
substances are frequently coextracted during virus concentration protocols, 
adding to the challenge of identifying reliable nucleic acid extraction processes. 

Early studies used SDS-proteinase K digestion to release nucleic acids from 
shellfish concentrates, followed by phenol chloroform extraction with, or with- 
out the addition of cetyltrimethylammonium bromide (CTAB) to remove resi- 
dual inhibitors (6, 7, 16, 90). In the last decade or so, guanidinium thiocyanate 
(GuSCN)-based methods became the RNA extraction method of choice largely 
because they are effective at deproteinization of nucleic acids while providing 
ample protection of RNA against native RNases. Many commercial guani- 
dinium based kits have been used in more recent studies (4, 12, 22, 24, 25, 32, 
59, 60, 75, 87, 90, 91). 

Combinations of multiple extraction methods can also be used to purify 
nucleic acids. A simple and rapid protocol for the purification of nucleic acid 
that utilizes a combination of the chaotropic agent GuSCN and silica particles 
was first described by Boom et al. (13), and later used by others (41, 53). Other 
methods include a GuSCN method followed by RNA binding to glass powder, 
instead of silica, to provide further nucleic acid purification (63, 64). 



100 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



Several investigators have compared various RNA extraction approaches 
specifically aimed at preparing samples for the detection of human enteric 
viruses. Hale et al. (44) compared four different RNA extraction methods for 
RT-PCR detection of noroviruses in fecal specimens, finding that the 
GuSCN/silica (13) method was the best at removal of inhibitors. Likewise, 
another study compared seven RNA extraction methods to purify hepatitis 
A virus RNA from stool and shellfish concentrates for RT-PCR detection (4); 
again, the GuSCN-silica methods were found to be the most suitable from the 
standpoint of speed, ease, and cost. Gouvea et al. (36) used deproteinization 
with GuSCN followed by adsorption of RNA to hydroxyapatite and sequential 
precipitation with CTAB and ethanol to purify RNA from shellfish and other 
selected foods. Sair et al. (87) compared multiple RNA extraction methods after 
concentration of noroviruses from model food commodities (hamburger sand- 
wiches and lettuce). These included GuSCN, commercial microspin columns, 
the QIAshredder Homogenizer and TRIzol alone and in their various combi- 
nations. These investigators found that the use of TRIzol followed by further 
sample preparation using the QIAshredder Homogenizer yielded the best detec- 
tion limits (< 1 RT-PCR amplifiable units/reaction) for Norwalk virus precon- 
centrated from food samples. Similar studies by Svensson (101) demonstrated 
that the use of the metal chelating agent Chelex-100, or alternatively, Sephadex 
G200 column chromatography, during RNA extraction, provided the best RT- 
PCR detection limits. Others have had success using phenol-chloroform-based 
methods followed by further selection for viral RNA using magnetic poly (dT) 
beads (30, 35, 59, 60). 

Despite all the efforts in identification of efficacious RNA extraction proto- 
cols, food-related amplification inhibitors frequently remain. Multiple sample 
manipulation steps can result in incomplete recovery and/or degradation of 
RNA during the extraction procedure, the consequence of which is less than 
optimal RNA yields (49, 50). A major problem with RNA extraction is the 
necessity to destroy virion integrity, thereby losing the ability to directly corre- 
late infectivity to RT-PCR detection limits, at least when effective cell culture- 
based methods are available. The two main areas of active research in RNA 
purification are increasing yield and improving the purity of the resultant prod- 
uct for detection by PCR. 



DETECTION 



The progress in clinical detection of pathogens has always been ahead of detec- 
tion in foods and many of our food methods rely on protocols initially devel- 
oped for clinical samples. However, whether clinical, food, or environmental 
sample, the sensitivity and the specificity of molecular amplification methods is 
largely dependent on the choice of primers. 

The genetic diversity in the Calicivirideae family makes primer design for the 
detection of the noroviruses quite challenging. Initial studies used extremely spe- 
cific primers such as NV 573' and NV 36/35, which were based on sequences in 



RT-PCR Detection of Viruses in Foods 101 



the prototype Norwalk virus genome (23, 74). With the availability of sequence 
information from related viruses, more broadly reactive primers have been 
designed. (2, 38, 57, 108). Most of the primer sequences reported are based on 
the highly conserved RNA-dependent RNA polymerase region of the 
noroviruses; occasionally the capsid region has been targeted (39, 40, 62, 81, 105). 

Initially, the primer sets developed by Ando et al. (2) for the genogroup I 
(GI) and genogroup II (Gil) Noroviruses were used as the "gold-standard" for 
the detection of noroviruses in clinical (fecal) and food samples (47, 80, 112). 
Later on, degenerate primers, or a mixture of oligonucleotide primers that vary 
in nucleotide sequence but have the same number of nucleotides, were devel- 
oped and used for the detection of the noroviruses (38, 68, 69). The use of 
degenerate primers is advantageous in that all combinations of nucleic acid 
sequence that code for the amino acid are used in the PCR amplification. 
Combinations of the Ando et al. (2) GI and Gil primers and various degener- 
ate sets are routinely used in norovirus epidemiological investigations as applied 
to the detection of virus in fecal samples. 

As with RNA extraction methods, investigators have compared the per- 
formance of various primers for the detection of a broad range of noroviruses, 
largely in fecal specimens. For instance, the NV110/NV36 primer set was the 
found to be the most efficient of the nine primer sets tested in a comprehensive 
study, even though it could not detect 100% of the norovirus strains tested (46). 
When five laboratories in five countries evaluated different RT-PCR methods 
on a panel of 91 fecal samples (106), no single assay was superior based on the 
criteria of sensitivity, detection limit, assay format, and successful implementa- 
tion. However, the Boom extraction method and the use of the JV12/JV13 
primer set were recommended for norovirus diagnostics. 

Nonetheless, there is some lack of consensus regarding the optimal primer 
pair(s) to detect the noroviruses and different laboratories tend to use various 
methods that were developed, or are optimally suited for their purposes (112). 
Primers used for the detection of hepatitis A virus usually target the VP1/2A 
junction sequence. In this case, the issue of diversity is not relevant as it is for 
the noroviruses (48). 



RT-PCR DETECTION OF VIRUSES IN FOODS 



The choice of primers is even more critical when attempting to detect viral con- 
tamination in foods. This is because the levels of contamination are typically 
much lower in foods when compared to clinical samples, and even with optimal 
concentration and nucleic acid extraction methods, residual inhibitors often 
persist. Furthermore, the matrix can be responsible for nonspecific amplifica- 
tion and false positive results. The primers selected for the detection of viral 
nucleic acids derived from the food matrix should therefore have the following 
criteria: (1) a reasonably high annealing temperature, (2) relative nondege- 
neracy, and (3) broad reactivity. High stringency and primer specificity (hence 
the relative absence of degeneracy) are necessary to prevent nonspecific 



102 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



amplification. For the genetically diverse norovirus group, the use of primers 
that are broadly reactive and can detect as many genetically distinct strains as 
possible in a single assay is essential. 

The various primers used in the RT-PCR detection of noroviruses from dif- 
ferent food matrices are summarized in Table 6.1. As with primers used in the 
clinical realm, these sequences correspond to mainly the viral RNA dependent 
RNA polymerase or the capsid protein genome regions. Primer sets targeting 
both the RNA polymerase and capsid genes, Mon38 1/383 and SR33/46, respec- 
tively, were used by Shieh et al. (96, 97) to identify a Gil norovirus in oyster 
samples implicated in a California outbreak. For the detection of the Gil 
noroviruses in shellfish, the NI/E3 primer set has also been used (33, 37, 63). 
Dubois et al. (32) used a newer primer pair to detect both noroviruses and 
sapoviruses in artificially contaminated produce. In a systematic comparison of 
four primer pairs, as applied to the detection of noroviruses in hamburger sand- 
wiches and lettuce, Sair et al. (87) found the best detection limits using the 
NVpl 10/NVp36 primer combination. Honma et al. (46) reported that this same 
primer pair was broadly reactive for a range of noroviruses, eliminating the 
need for separate amplifications for the two norovirus genogroups (GI and 
Gil). These primers have also been used together or in combination with NI or 
NVp69 for the detection of norovirus contamination in shellfish (59, 67, 69). 

"Nested" RT-PCR or double amplification has been used for the detection 
of noroviruses (36, 63, 91, 99) and hepatitis A virus (18, 19, 20, 43, 59) in foods. 
This approach can improve assay sensitivity and also provide another method 
for the confirmation of amplified product. However, a major disadvantage is 
that these nested reactions are prone to carryover contamination. Novel single- 
tube nested RT-PCR methods may help circumvent these issues. Ratcliff et al. 
(81) pooled the reagents required for the nested amplification in a "hanging 
drop" that could be introduced by centrifugation after the first RT-PCR ampli- 
fication while Burkhardt et al. (14) compartmentalized the nested RT-PCR 
cocktail in a "tube-within-a-tube" device by using inexpensive materials such as 
a pipette tip and a microcentrifuge tube. Primers for the detection of hepatitis 
A virus by RT-PCR are summarized in Table 6.2. 



ALTERNATIVE NUCLEIC ACID AMPLIFICATION 
METHODS 



Nucleic acid sequence-based amplification (NASBA) is an amplification method 
that specifically detects RNA to the exclusion of DNA. The transcription-driven 
NASBA reaction is carried out at a single temperature (41 °C) and theoretically 
amplifies the RNA target more than 10 12 -fold within 90 min (34, 53). The 
final product of the amplification is single-stranded RNA, which can be readily 
detected by hybridization. The system utilizes three enzymes, (1) a reverse 
transcriptase (AMV-RT), (2) an RNase H, and (3) a T7 RNA polymerase, all of 
which act in a stepwise (sequential) manner with two oligonucleotide primers 
specific to the target (34, 53). One of the primers (PI) contains the T7 RNA 



Alternative Nucleic Acid Amplification Methods 



103 



u 

Oh 

GO 

O 



CO 

<D 

CO 

•i-H 

> 

O 

o 

o 
o 

•I-H 

o 

<d 

D 

43 



*T3 
en 

go 

5-i 



5-i 









R 



^3 









R 

T«H 






<? 



<"o 

T 

V 

■n 



R 

CO 



«>2 

s 









oo 



o 

O 

-4— » 

CO 

13 
o 

•I-H 

U 



CN 
CN 

oo 



U 
U 
Eh 
Eh 
CJ 



U 



> 

z 



<3 

o 

Eh 



C-2 En 

<C O 

En En 

Eh Eh 

O CJ 

Eh Eh 

O CJ 

En O 

U O 



Eh 
Eh 



^h CN 

I I 

13 13 

u u 



oo 



43 

GO 

s 

43 
co 



CN 

co 



H 

c-> eh 
ej u 

o H 



i 






V CJ 

O < Eh U 



< Eh 






OO 



43 

CO 

S 

13 

43 

CO 



oo 

CN 



u 

U 
Eh 

o 
o 

Eh 



^ 



o 

Eh 

o 

Eh 






Eh 
Eh 



C3 g 
CO 
CO 



U 
U 
Eh 

ii 



Eh 






> 

Z 



CN 




bJO 



On 

a 

o 









«N 




■ 


^sD 


OO 


^1- 


^1- 


P^ 


p^ 


C/3 


CO 


> 


> 


<L> 


CD 


& 


^ 



EH 

Eh 



<d eh 

O CJ 
O Eh 



Eh 
Eh 



O O 
Eh Eh 

CJ CJ 



<N 



c> 






o 

(D 



CO 



CO 


•O 


<N 


^ 


o 


^o 


<N 


^H 


i— i 


r- 


t> 


G\ 


^o 


^O 


<N 


o 


lO 


OO 


in 


t> 


m 


T 


LO 


T 


T 


T 


T 


in 


m 


t> 


en 


0\ 


oo 


^o 


<N 


o\ 


a\ 


lO 


lO 


^o 


oo 


^f- 


y—{ 


a\ 


IT) 


OO 


•o 


r- 


m 


^t 


^r 


^1- 


^1- 


^1- 


^i- 


in 










o 

Eh 
Eh 
Eh 



CJ 
CJ 
CJ 

O 
Eh 
U 



fe P^ 

co CO 



< 

I 



Eh 
En 






C\ 






r\ 






CO 

S 

13 

CO 



ro 



io 



+ 



Eh 
CJ 

O 
Eh 

O CJ 
CJ 

o 

cj d 

<C Eh 
CJ 

cj o 






CO 

in 

oo 

I 

On 

OO 

^1- 



cj 

3 

cj 

Eh 
CJ 

% 



o a 



& 









T3 co 



GO 



5-h JD 



0\ 



O ^O 
On OO 
lO OO 



OO in 

lO OO 
^1- ^1- 



+ 



CJ 
° EJ 

cj <; 

Eh CJ 

y y 

O U 

O Ej 

u 

Eh 



Eh 

a 



CJ Eh 

cj o 

Eh 

|, 

E" 1 CJ 

Ej ^ 

O Eh 



o3 > 



GO 

<U 
GO 

5-H 



GO 



Z co 



O On 
On OO 
CN CN 

Oh Oh 



in 

M2 



4^ 

GO 

s 

13 
co 



in 



CN 

in 



TT T 



On 
OO 

^1- 



Eh 
Eh 

CJ 

Eh 



O 
Eh 
O 



CJ O 

En O 

CJ CJ 

En Eh 



Eh 
Eh 



3 



CJ CJ 
Eh Eh 







CN 

ID 



^i- 



> 

z 



< 



o 

CJ 
CJ 

o 

CJ 
CJ 

o 



o 



lO 



4^ 

GO 

S 

13 
co 



IT) 



CO 



in 



wo 



cj 

Eh 

u + 

Eh 

cj o 

Eh 

. U 

o cj 

— CJ 

CJ CJ 

Eh < 

o cj 

CJ 





CO 

•i-H 

> 

o 

5-H 

o 



CO 
CN 

On oo 
co ^ 
oo 



h-1 kJ 
> > 

z z 



CN 

h-l 

oo 
co 



> 

z 



CN 
CN 

o 

•I-H 

CO 

Q 



CN 
CN 

co 



co 
oo 
co 
in 

CN 

CO 

in 



+ 

Eh 

O CJ 

— O 

^ Eh 

<C O 

CJ O 

CJ Eh 
CJ 
Eh 

U CJ 

CJ <C 

CJ Eh 

O O 

CJ Eh 

cj <c 

o cj 

Eh cj 



O 

CO 

•i-H 

> 

O 

5-H 

o 



OO 
CO 

Ch 

o 







R 
CJ 



104 



Molecular Approaches for the Detection of Foodborne Viral Pathogens 






u 

43 
en 

O 



CO 

(D 

CO 

U 

o 
o 

o 
o 

• 1— I 

-4— > 

o 

-*— > 
<D 

<L> 

43 



T3 
en 

en 

5-H 



5-H 









R 



^3 









R 






<? 



T 

V 



R 
<^> 

R 
C<3 



03 
R 









in 

Ov 



43 
en 

s 

43 



en 



U 



oo 
en 

43 

•i-H 

^— > 

CO 



On r-> 
lO oo 



oo 

On 



O 



< 



CO 

CD 

CO 

U 
•i-H 

> 

O 

5-i 

o 



en 


(N 


o 




oo 


oo 


i— 1 


M2 


CO 


ro 


i-H 


en 


rt 


S3 


a 


Oh 


o 


O 


> 


> 


§ 


§ 


Z 


£ 











o 


CO 


o 


a 


2 


03 


-4- » 




<D 


U 


J 



CO 



CO 

(D 

CO 

•i-H 

> 

O 

5-H 

o 



43 

s 

13 

43 
<Z3 



m 


^t 


^t 


1—1 


<N 


OO 


oo 


oo 


oo 


o 


i> 


t> 


VO 


U^) 


oo 


io 


in 


oo 


*T) 


*T> 


T 


T 


T 


T 


^H 


o\ 


IA) 


t> 


(N 


oo 


vo 


IT) 


M2 


oo 


IT) 


IT) 


vo 


IT) 


oo 


^r 


IT) 


OO 


IT) 


^n 


^- 


^- 


^t 


^t 



+ I 



g 3 

H En 

<C ^ 

O U 

^ U 

< kc 

Eh U 

U Eh 

<J <J 

U U 

U Eh 

< Eh 



> > 



CO 
CO CO 



CO 


5-H 



> 

O 



> 

o 

5-H 

o 



£ 






O 



OO 



(N 



Alternative Nucleic Acid Amplification Methods 



105 



P^ 

u 

PL 

GO 

O 



CO 



> 

GO 



43 

o 
o 

^— > 

o 

43 



T3 

CO 
CO 






H 












CO 












T 

V 



CO 












CO 

(D 

CO 

H— > 

5-H 

PL, 



CN 






E 



CN 



c\ 



CO 

CO 
CO 

2 




^- 


CN 


i-H 


a\ 


^T 


i—i 


<N 


<N 


O 


t> 


On 


^O 


co 


^^ 


CN 


<N 



+ 



o 

Eh < 

Eh Eh 

Eh U 

U O 

Eh Eh 
Eh 
Eh 
U 



6 

_ Eh 
Eh Eh 



Eh 
Eh 



U Eh 
Eh U 
U U 
Eh Eh 
O U 
O 






CN 



t> 



Cn 



CN 



CN 

co 






C\ 



in 



ON 



U 
5-i 

D 
43 

CO 



c\ 



O 



43 

CO 

s 

43 

C/3 



CO 

CD 

^— > 

CO 

o 



CO 
CO 

o 

Q 



CO £> 



Q 



IT) 



m 



c\ 



c\ 



WO 



5-1 
GO 

o 



M2 

CN 
<N 

CN 

oo 



CN 
CN 



oo 



c\ 






OO 



On 

IT) 



IT) 

m 



3 

s 



X 



O 

-*— » 
s— > 

h-l 



^1- 



CN 

IT) 

CO 



CN 



U U 



< < 



CO 
CO 



: h 



CO 
CO 

=3 



CO 




c3 





43 

CO 

S 

<u 
o 

o 

5-1 

CL 



U 

Oh 

-4— > 



^i- o 

CO 

co co 

OO OO 
OS CO 
CN CO 



^1- 



1^ 
in 

CN 



OO 



CN 



r- 



uo 

oo 



CN 



CO 

oo 

wo 

oo 



+ 



Eh 

O Eh 

a g 

Eh <! 

u 
u 

Eh 



Eh 

Eh 
U 
Eh 



o 

Eh 

u 
S3 

U 

Eh 
Eh 
Eh 

O 
Eh 
U 



+ 






+ 



!>■ OO 

oo co 

CN CO 







c^J 


a 


t> 


oo 


t> 


oo 














PQ 


PQ 


PQ 


PQ 



OO 
IT) 



C\ 



CO 

S 

13 

43 
CO 



CN 


^r- 


^o 


^r 


(^ 


0"\ 


CO 


t> 


^sO 


M^ 


^^ 


in 


CN 


CN 


CN 



CO 
OS 

CN 

OS 

^t 

OS 

CN 



+ 



OS 

OS 
CN 



P4 

u 

PL 

D 



in 

m 
oo 

m 




< 


<C 


Eh 


<] 


U 


En 


>H 


Eh 


^ 


Eh 


O 


O 


U 


^ 


En 


Eh 


^2 


^2 


<d 


g 


Eh 


<c 


< 


g 


^ 


U 


o 


g 


< 


U 


U 


Eh 


< 


u 


g 


Eh 


U 


EH 


Eh 




U 


^2 


O 


o 


rt] 


Eh 


< 


Eh 


Eh 


o 


O 


u 


Eh 


u 


o 


O 


Eh 


U 


Eh 


Eh 


O 


En 


U 


o 


o 


Eh 


Eh 


Eh 


Eh 


^2 


U 


Q 


U 


Eh 


<c 


Eh 


O 


Eh 


En 


o 


U 


Eh 


O 


< 


^2 


U 


U 


O 


Eh 


<c 



CN 

OS 
CO 



M2 



en 

5-1 

o 



CN 



co 
co 



^r 



CN 

oo 



co co 



oo 

OS 
CN 



u u 



^1- 


wo 


CN 


CN 


< 


< 


M 


M 


^ 


^ 



O OS 
CN ^h 



P4 

u 

PL 

o 

■4— > 
CO 

O 



CN 



43 
co 

S 

13 

43 
CO 



io 

oo 

CO 



oo 

CN 



+ 



p^ 
u 

PL 

D 

-4— > 
GO 



CO 

CO 
CO 

2 




^1- 



CO 



CN 



0) 




s 


B 


•i-H 


•y—i 


^H 


^H 


PL, 


CL 



^3 






106 



Molecular Approaches for the Detection of Foodborne Viral Pathogens 






u 

o 



CO 


•H 

> 

< 

CO 



43 

o 
o 

•!-H 

-4— > 

o 

-t— > 
<D 

<L> 

43 



T3 
en 

en 



S-H 

Oh 






R 

J*. 









■a 

CO 






R 
Q 






V 

t 

V 



R 

<^> 

CO 






J*. 



m 



m ^|- 



oo 



P4 

u 

a* 

+-> 

CO 

o 

o 



5 



+ 



<N 





as 




^1- 






o 




oo 




t> 


i— 1 


»o 


o 


lO 


Os 


*-H 


oo 


*-H 


(N 


o 


<N 


^sD 


CO 


OO 


o 


as 


Os 


^O 


r- 


oo 


o 


SO 


o 


o 


r- 


^^ 


MD 


^H 


oo 



Eh 


u 


< 


Eh 


Eh 


O 


Eh 


U 


Eh 


O 


Ej 


^2 


Q 


< 


< 


< 


Eh 


U 


EH 


O 


O 


En 


O 


Eh 


Eh 


Ej 


U 


< 


< 


■3 


U 


Eh 


O 


O 


O 


^2 


O 


En 


< 


O 


< 


En 


^ 


EH 


< 


< 


<n 


O 


U 


O 


Eh 



oo 



+ 



i 

Eh 
Eh 
U 



U 
O 
Eh 
Eh 
Eh 
Eh 












s 


s 






• 1— 1 


*— i cn m ^t 


dn 


PLh 


CLh Ph Ph CLh 



oo 



co 
O 

GO 

=5 




PQ 



t> 



cn 

<N 

Os 



cn 



Tf oo 
> > 

X X 






u 

Oh 
<D 

■4— > 
CO 

CO 

s 

43 
CO 



oo 



cn 
cn 



+ 



t> 



oo 



O 



> > 

X X 



as 

<N 



as 
cn 






+ 






^1- 



X X 



Confirmation 107 



polymerase promoter sequence at 5' terminal and the other primer (P2) can be 
designed with a generic sequence that facilitates probe capture for amplicon 
detection by liquid hybridization (described in the Confirmation section 
below). 

NASBA assays have been developed for the detection of foodborne enteric 
viruses such as hepatitis A virus, rotavirus and noroviruses. For instance, 
Greene et al. (41) applied the NucliSens Basic Kit NASBA protocol for the 
detection of norovirus RNA in stool using primers targeting the RNA poly- 
merase region of the viral genome. Jean et al. (54, 55, 56) developed a NASBA- 
based method to detect hepatitis A virus on artificially contaminated lettuce 
and blueberry samples and also for the detection of human rotavirus. More 
recently, Jean et al. (53) developed a multiplex NASBA method for the simulta- 
neous detection of hepatitis A and noroviruses (GI and Gil) from lettuce and 
sliced turkey (deli meats). 

The NASBA method is isothermal, as rapid (if not faster than) as RT-PCR, 
and demonstrates detection limits equal to if not better than RT-PCR (41, 53, 56). 
However, NASBA technology has many of the same limitations as RT-PCR (e.g., 
contamination control, sample volume considerations, matrix-associated reaction 
inhibitors). Nonetheless, it remains an important alternative method for the 
detection of foodborne viruses. 



CONFIRMATION 



Since nonspecific products of amplification are a major issue when food and 
environmental samples are tested, it is critical to confirm that the nucleic acid 
amplification products obtained are specific to the target. Most often, the con- 
firmation step also improves the sensitivity of the assay. The most common 
confirmatory tool is Southern hybridization using specific oligoprobes internal 
to the amplicon. These probes are usually enzyme labeled for colorimetric, 
luminescent or fluorescent endpoints. When RNA products (for NASBA) are 
tested, Northern hybridization with labeled internal oligonucleotide probes may 
be used (45, 46). An oligonucleotide array dot-blot format for the simultaneous 
confirmation of norovirus amplicons and strain genotyping has recently been 
reported, offering the promise of providing both detection and strain typing in 
a single test (107). 

DNA enzyme immunoassay (DEIA) methods provide an alternative to 
Southern hybridization. In these assays, a capture probe is immobilized to a 
microtiter plate well and a labeled amplicon can then be detected directly, or 
alternatively, an unlabeled amplicon can be hybridized to a second labeled detec- 
tor probe followed by detection after the addition of an enzyme-conjugate and 
appropriate substrate (sandwich assay). For colorimeteric, luminescent, or fluo- 
rescent endpoints, absorbance is read using a conventional microtiter plate spec- 
trophotometer or fluorescent plate reader. The intensity of the signal obtained 
may be approximately proportional to the concentration of amplicon. The sensi- 
tivity of the microtiter plate assay is generally equal to or better than Southern 



108 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



hybridization and this approach has advantages including ease of interpretation, 
rapid (4 h) amplicon confirmation, and the potential for automation (54, 55, 90). 

A liquid electrochemiluminescence (ECL) hybridization technology has 
been used commercially for the detection of NASBA amplicons. This technol- 
ogy utilizes two specific oligoprobes; a capture probe complementary to the 
sequence on primer P2, immobilized to streptavidin-labeled magnetic beads and 
a detector probe complexed to a ruthenium chelate. The hybridized magnetic 
particles are trapped on an electrode, and application of a voltage trigger to the 
electrode induces the ECL reaction such that the amount of emitted light is 
directly proportional to the amount of the amplicon. The signals are reported 
as ECL units by the NucliSens Reader and associated software. The NASBA- 
ECL system typically generates confirmed detection results in a day and has 
been used by Fox et al. (34) for the detection and confirmation of enterovirus 
from clinical samples and by Greene et al. (41) and Jean et al. (53) for the detec- 
tion Norwalk virus in stool and food samples. 

Other confirmation methods include specific "nested" PCR reactions (36, 
43, 63, 99), which use a second pair of primers internal to the first amplicon 
sequence; and restriction endonuclease digestion of RT-PCR products (36, 43). 
Direct sequencing of the amplicon for the confirmation of RT-PCR products is 
another method of choice, and is frequently applied in the clinical realm, and 
more recently when amplicons are obtained from foods implicated in outbreaks 
(69, 91). 



REAL-TIME DETECTION 



Real-time detection refers to the simultaneous detection and confirmation of 
amplicon identity as the amplification reaction is progressing, thereby linking 
nucleic acid amplification with hybridization. There are currently five main 
chemistries used for real-time amplification and detection. One of the earliest 
and simplest approaches to real-time PCR, called DNA binding fluorophores, 
uses ethidium bromide or SYBR green I compounds that fluoresce when asso- 
ciated with double stranded DNA and exposed to a suitable wavelength of 
light. These methods tend to lack specificity but this has been addressed 
recently by coupling the assay with melting curve analysis. The 5' endonucle- 
ase assay (e.g., TaqMan oligoprobes, Applied Biosystems, Foster City, CA), 
adjacent linear probes (e.g., HybProbes, Roche Molecular Biochemicals, 
Germany) and hairpin oligoprobes (e.g. molecular beacons, Molecular Probes, 
Eugene, Oregon) have received considerable attention of late. Self-fluorescing 
amplicons (e.g., Sunrise primers, Amplifluor hairpin primers, Intergen Co., 
Purchase, NY) incorporated into the PCR product as the priming continues 
have a 3' end complimentary to the target strand and Scorpion primers have 5' 
end complementary to the target strand) (reviewed in 72). Recently, investiga- 
tors have developed real-time PCR systems for the detection of a wide array of 
bacterial pathogens in foods. Prototype real-time RT-PCR amplification tech- 
nologies have been developed for the detection of hepatitis A virus (17) and 



Acknowledgments 109 



noroviruses (79) using the TaqMan format, and the norovirus detection uses 
the SYBR Green melting curve format (73). Beuret et al. (10) have used multi- 
plex real-time PCR for the simultaneous detection of a panel of enteric viruses. 
Research efforts are currently underway to apply these methods to the detec- 
tion of viruses in food matrices. Indeed, Myrmel et al. (78) recently reported 
the detection of viral contamination in shellfish using a commercial 
SYBRGreen PCR kit, while Narayanan et al (79) used their TaqMan assay to 
detect noroviruses in shellfish. 



CONCLUSIONS 



It should be clear from the preceding discussion that the current methodology 
for the detection of enteric viral contamination in foods is less than ideal and 
that research is necessary to improve these methods. Indeed, these protocols are 
applied infrequently and usually only in response to known or suspected food- 
borne disease outbreaks. The most important reasons for their limited use 
include: (1) the inability of molecular amplification methods to discriminate 
between infectious and inactivated virus, (2) the lack of widely accepted, col- 
laboratively tested methods, (3) the requirement that most methods be product 
specific, meaning that universal approaches do not exist, and (4) the cost and 
need for highly trained personnel (83). When taken together, detection limits 
ranging from approximately 1-100 infectious units/g food have been obtained 
using various RT-PCR methods. The use of internal amplification standards to 
simultaneously evaluate RT-PCR inhibition and/or to provide a semiquantita- 
tive assay is also frequently done (5, 6, 67, 69, 89, 93, 103). 

The failure to discriminate between infectious and inactivated virus is of 
critical importance because the inactivated forms of these pathogens pose no 
real public health threat. There is also a need to develop more universal sam- 
ple extraction methods. For the most part, virus concentration from foods is 
likely to remain product dependent but research is needed to develop and 
refine the prototype methods into collaboratively tested protocols. 
Researchers continue to seek efficacious methods to concentrate the 
pathogens from the food matrix with the simultaneous removal of matrix- 
associated inhibitors. Even so, the methods will probably never be perfect and 
will always require a high degree of sample manipulation by the laboratory 
personnel (83). There is hope, however, that over time these rapid methods to 
detect human enteric viruses in foods may become more widely available 
to the food safety community. 



ACKNOWLEDGMENTS 



This effort was supported in part by a grant from the USDA National Research 
Initiative, Competitive Grants Program: Ensuring Food Safety, 2002-35201- 
11610. The use of trade names in this chapter does not imply endorsement by 



110 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



the North Carolina Agricultural Research Service nor criticism of similar ones 
not mentioned. 



REFERENCES 



1. Abd El Galil, K.H., M.A. El Sokkary, S.M. Kheira, A.M. Salazar, M.V. Yates, 
W. Chen, and A. Mulchandani. 2004. Combined immunomagnetic separation- 
molecular beacon-reverse-transcription-PCR assay for detection of hepatitis A virus 
from environmental samples. Appl. Environ. Microbiol. 70:4371-4374. 

2. Ando, T., S.S. Monroe, J.R. Gentsch, Q. Jin, D.C. Lewis, and R. I. Glass. 1995. 
Detection and differentiation of antigenically distinct small round- structured 
viruses (Norwalk-like viruses) by reverse transcription-PCR and Southern 
hybridization. J. Clin. Microbiol. 33:64-71. 

3. Arnal, C, V. Ferre-Aubineau, B. Besse, B. Mignotte, L. Schwartzbrod, and 
S. Billaudel. 1999. Comparison of 7 RNA extraction methods on stool and shellfish 
samples prior to hepatitis A virus amplification. J. Virol. Methods 77:17-26. 

4. Arnal, C, V. Ferre-Aubineau, B. Mignotte, B.M. Imbert-Marcille, and S. Billaudel. 
1999. Quantification of hepatitis A virus in shellfish by competitive reverse tran- 
scription-PCR with coextraction of standard RNA. Appl. Environ. Microbiol. 
65:322-326. 

5. Atmar, R.L., F.H. Neill, CM. Woodley, R. Manger, GS. Fout, W. Burkhardt, 
L. Leja, E.R. McGovern, F. LeGuyader, T.G Metcalf, and M.K. Estes. 1996. 
Collaborative evaluation of a method for the detection of Norwalk virus in shellfish 
tissues by PCR. Appl. Environ. Microbiol. 62:254-258. 

6. Atmar, R.L., F.H. Neill, J.L. Romalde, F. Le Guyader, CM. Woodley, T.G. Metcalf, 
and M.K. Estes. 1995. Detection of Norwalk virus and hepatitis A virus in shellfish 
tissues with the PCR. Appl. Environ. Microbiol. 61:3014-3018. 

7. Atmar, R.L., T.G. Metcalf, F.H. Neill, and M.K. Estes. 1993. Detection of enteric 
viruses in oysters by using polymerase chain reaction. Appl. Environ. Microbiol. 
59:631-635. 

8. Barardi, C.R., M.H. Yip, K.R. Emslie, G Vesey, S.R. Shanker, and K.L. Williams. 
1999. Flow cytometry and RT-PCR for rotavirus detection in artificially seeded oys- 
ter meat. Int. J. Food Microbiol. 49:9-18. 

9. Berke, T, B. Golding, X. Jiang, W.D. Cubitt, M. Wolfaardt, A.W. Smith, and D.O. 
Matson. 1997. Phylogenetic analysis of the Caliciviruses. J Med Virol 52:419-424. 

10. Beuret, C. 2004. Simultaneous detection of enteric viruses by multiplex real-time 
RT-PCR. J. Virol. Methods 115:1-8. 

11. Beuret, C, A. Baumgartner, and J. Schluep. 2003. Virus-contaminated oysters: A 
three-month monitoring of oysters imported to Switzerland. Appl. Environ. 
Microbiol. 69:2292-2297. 

12. Bidawid, S., J.M. Farber, and S.A. Sattar. 2000. Rapid concentration of detection of 
hepatitis A virus form lettuce and strawberries. J. Virol. Methods 88:175-185. 

13. Boom, R., C.J. A. Sol, M.M.M. Salimans, C.L. Jansen, P.M.E. Wertheim-Van 
Dillen, and J. Van Der Noordaa. 1990. Rapid and simple method for purification of 
nucleic acids. J. Clin. Microbiol. 28:495-503. 

14. Burkhardt, W., G Blackstone, D. Skilling, and A. Smith. 2002. Applied technique 
for increasing calicivirus detection in shellfish extracts. J. Appl. Microbiol. 
3:235-240. 



References 111 



15. Chung, H., L-A Jaykus, and M.D. Sobsey. 1996. Detection of human enteric viruses 
in oysters by in vivo and in vitro amplification of nucleic acids. Appl. Environ. 
Microbiol. 62:3772-3778. 

16. Coelho, C, A.P. Heinert, C.M.O. Simoes, and C.R.M. Barardi. 2003. Hepatitis A 
virus detection in oysters (Crassostrea gigas) in Santa Caratina State, Brazil, by 
reverse transcription-polymerase chain reaction. J. Food Prot. 66:507-511. 

17. Costa-Mattioli, M, S. Monpoeho, E. Nicand, M-H Aleman, S. Billaudel, and V. Ferre. 
2002. Quantification and duratin of viraemia during hepatitis A infection as deter- 
mined by real-time RT-PCR. J. Viral Hepatitis 9:101-106. 

18. Croci, L., D. De Medici, G. Morace, A. Fiore, C. Scalfaro, F. Beneduce, and L. Toti. 
2002. The survival of hepatitis A virus in fresh produce. Int. J. Food Microbiol. 
73:29-34. 

19. Croci, L., D. De Medici, C. Scalfaro, A. Fiore, M. Divizia, D. Donia, A.M. 
Cosentino, P. Moretti, and G. Costantini. 2000. Deternination of enteroviruses, hep- 
atitis A virus, bacteriophages and Escherichia coli in Adriatic Sea mussels. J. Appl. 
Microbiol. 88:293-298. 

20. Croci, L., D. DeMedici, G. Morace, A. Fiore, C. Scalfaro, F. Beneduce, and L. Toti. 
1999. Detection of hepatitis A virus by nested reverse transcription-PCR. Int. 
J. Food Microbiol. 70:67-71. 

21. Cromeans T.L., O.V. Nainan, and H.S. Margolis. 1997. Detection of hepatitis A virus 
RNA in oyster meat. Appl. Environ. Microbiol. 63:2460-2463. 

22. Daniels, N.A., D.A. Bergmire-Sweat, K.J. Schwab, K.A. Hendricks, S. Reddy, S.M. 
Rowe, R.L. Fankhauser, S.S. Monroe, R.L. Atmar, R.I. Glass, and P. Mead. 2000. A 
foodborne outbreak of gastroenteritis associated with Norwalk-like viruses: First 
molecular traceback to deli sandwiches contaminated during preparation. J. Infect. 
Dis. 181:1467-1470. 

23. De Leon, R., S.M. Matsui, R.S. Baric, IE. Herrmann, N.R. Blacklow, H.B. 
Greenberg, and M.D. Sobsey 1992. Detection of Norwalk virus in stool specimens 
by reverse transcriptase-polymerase chain reaction and nonradioactive oligoprobes. 
J. Clin. Microbiol. 30:3151-3157. 

24. De Medici, D., Croci, L., E. Suffredini, and L. Toti. 2004. Reverse transcription- 
booster PCR for detection of noroviruses in shellfish. Appl. Environ. Microbiol. 
70:6329-6332. 

25. De Medici, D., L. Croci, S. Di Pasquale, A. Fiore, and L. Toti. 2001. Detecting the 
presence of infectious hepatitis A virus in molluscs positive to RT-nested-PCR. Lett. 
Appl. Microbiol. 33:362-366. 

26. De Medici, D., F. Beneduce, A. Fiore, C. Scalfaro, and L. Croci. 1998. Application 
of reverse transcriptase-nested-PCR for detection of poliovirus in mussels. Int. 
J. Food Microbiol. 40:51-56. 

27. Demeke, T. and R.P. Adams. 1992. The effects of plant polysaccharides and buffer 
additives on PCR. Biotechniques 12:332-333. 

28. Deng M.Y., S.P. Day, and D.O. Cliver. 1994. Detection of hepatitis A virus in envi- 
ronmental samples by antigen-capture PCR. Appl. Environ. Microbiol. 
60:1927-1933. 

29. Desenclos, J.C., K.C. Klontz, M.H. Wilder, O.V. Nainan, H.S. Margolis, and R.A. 
Gunn. 1991. A multistate outbreak of hepatitis A caused by the consumption of raw 
oysters. Am. J. Public Health 81:1268-1272. 

30. DiPinto, A., V.T. Forte, GM. Tantillo, V. Terio, and C. Buonavoglia. 2003. 
Detection of hepatitis A virus in shellfish (Mytilus galloprovincialis) with RT-PCR. 
J. Food Prot. 66:1681-1685. 



112 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



31. Dix, A.B. and L.-A Jaykus. 1998. Virion concentration method for the detection of 
human enteric viruses in extracts of hard-shelled clams. J. Food Protect. 61:458^465. 

32. Dubois, E., C. Agier, O. Traore, C. Hennechart, G. Merle, C. Cruciere, and 
H. Laveran. 2002. Modified concentration method for the detection of enteric 
viruses on fruits and vegetables by reverse transcriptase polymerase chain reaction 
or cell culture. J. Food Prot. 65:1962-1969. 

33. Formiga-Cruz, M., G. Tofino-Quesada, S. Bofill-Mas, D. N. Lees, K. Henshilwood, 
A. K. Allard, A-C Conden-Hansson, B. E. Hernroth, A. Vantarakis, A. Tsibouxi, 
M. Papapetropoulou, M. D. Furones, and R. Girones. 2002. Distribution of human 
virus contamination in shellfish from different growing areas in Greece, Spain, 
Sweden, and the United Kingdom. Appl. Environ. Microbiol. 68:5990-5998. 

34. Fox, J.D., S. Han, A. Samuelson, Y. Zhang, M.L. Neale, and D. Westmoreland. 
2002. Development and evaluation of nucleic acid sequence based amplification 
(NASBA) for diagnosis of enterovirus infections using the NucliSens® Basic Kit. 
J. Clin. Virol. 24:117-130. 

35. Goswami, B.B., M. Kulka, D. Ngo, P. Istafanos, and T.A. Cebula. 2002. A polymerase 
chain reaction-based method for the detection of hepatitis A virus in produce and 
shellfish. J. Food Prot. 65:393-402. 

36. Gouvea, V., N. Santos, M. Carmo-Timenetsky, and M.K. Estes. 1994. Identification 
of Norwalk virus in artificially seeded shellfish and selected foods. J. Virol Methods 
48:177-187. 

37. Green, X, K. Henshilwood, C.I. Gallimore, D.W.G Brown, and D.N. Lees. 1998. 
A nested reverse transciptase PCR assay for detection of small round-structured 
viruses in environmentally contaminated molluscan shellfish. Appl. Environ. 
Microbiol. 64:858-863. 

38. Green, J., C.I. Gallimore, J.P. Norcott, D. Lewis, and D.W.G. Brown. 1995. Broadly 
reactive reverse transcriptase polymerase chain reaction (RT-PCR) for the diagnosis 
of SRSV-associated gastroenteritidis. J. Med. Virol. 47:392-398. 

39. Green, S.M., PR. Lambden, O. Caul, and I.N. Clarke. 1997. Capsid sequence diver- 
sity in small structured viruses from recent UK outbreaks of gastroenteritis. J. Med. 
Virol. 52:14-19. 

40. Green, S.M., PR. Lambden, E.O. Caul, C.R. Ashley, and I.N. Clarke. 1995. Capsid 
diversity in small round- structured viruses: Molecular characterization of an anti- 
genically distinct human enteric calicivirus. Virus Res. 37:271-283. 

41. Greene, S.R., C.L. Moe, L-A Jaykus, M. Cronin, L. Grosso, and P. van Aarle. 2003. 
Evaluation of the NucliSens® Basic Kit assay for the detection of Norwalk virus 
RNA in stool specimens. J. Virol. Methods 108:123-131. 

42. Groppe, J.C. and D.E. Morse. 1993. Isolation of full-length RNA templates for 
reverse transcription from tissues rich in RNase and proteoglycans. Anal. Biochem. 
210:337-343. 

43. Hafliger, D., M. Gilgen, J. Luthy, and P. Hubner. 1997. Seminested RT-PCR systems 
for small round-structured viruses in faecal specimens. J. Virol. Methods 
57:195-201. 

44. Hale, A.D., J. Green, and D.W.G. Brown. 1996. Comparison of 4 RNA extraction 
methods for the detection of small round structured viruses in faecal specimens. 
J. Virol. Methods 57:195-201. 

45. Hardy, M.E., S.F. Kramer, J.J. Treanor, and M.K. Estes. 1997. Human calicivirus 
genogroup II capsid sequence diversity revealed by analyses of the prototype Snow 
Mountain agent. Arch. Virol. 142:197-202. 



References 113 



46. Honma, A., S. Nakata, K. Kinoshita-Numata, K. Kogawa, and S. Chiba. 2000. 
Evaluation of 9 sets of PCR primers in the RNA dependent RNA polymerase 
region for detection and differentiation of members of the family Caliciviridae, 
Norwalk virus and Sapporo virus. Microbiol. Immunol. 44:411^119. 

47. Iritani, N., Y. Seto, H. Kubo, K. Haruki, M. Ayata, and H. Ogura. 2002. Prevalence 
of "Norwalk-like virus" infections in outbreaks of acute nonbacterial gastroenteri- 
tis observed during the 1999-2000 season in Osaka city, Japan. J. Med. Virol. 
66:131-138. 

48. Jansen, R.W., G. Siegl, and S.M. Lemon. 1990. Molecular epidemiology of human 
hepatitis A virus defined by antigen-capture polymerase chain reaction. Proc. Natl. 
Acad. Sci. USA 87:2867-2871. 

49. Jaykus, L., R. DeLeon, and G.A. Toranzos. 2001. Detection of bacteria, viruses and 
parasitic protozoa in shellfish, In C. J. Hurst (ed.), Manual of Environmental 
Microbiology, 2nd edn, pp. 264-276, ASM Press, American Society for 
Microbiology, Washington, DC. 

50. Jaykus, L. 2000. Detection of human enteric viruses in foods. In S. Sattar (ed.), 
Foodborne Diseases Handbook, Vol. 2: Viruses, Parasites, Pathogens and HACCP, 
2nd edn., pp. 137-163. Marcel Dekker, New York. 

51. Jaykus, L.A., R. De Leon, and M.D. Sobsey 1996. A virion concentration method 
for detection of human enteric viruses in oysters by PCR and oligoprobe hybridiza- 
tion. Appl. Environ. Microbiol. 62:2074-80. 

52. Jaykus L.A., R. De Leon, and M.D. Sobsey 1995. Development of a molecular 
method for the detection of human enteric viruses in oysters. J. Food Protect. 
58:1357-1362. 

53. Jean, J., D.H. D'Souza, and L. Jaykus. 2004. Multiplex nucleic acid sequence-based 
amplification (NASBA) for the simultaneous detection of enteric viruses in ready- 
to -eat food. Appl. Environ. Microbiol. 70:6603-6610. 

54. Jean, J., B. Blais, A. Darveau, and I. Fliss. 2002. Simultaneous detection and identi- 
fication of hepatitis A virus and rotavirus by multiplex nucleic acid sequence-based 
amplification (NASBA) and micro titer plate hybridization system. J. Virol. Methods 
105:123-132. 

55. Jean, J., B. Blais, A. Darveau, and I. Fliss. 2002. Rapid detection of human rotavirus 
using colorimetric nucleic acid sequence-based amplification (NASBA) -enzyme 
linked immunosorbent assay in sewage treatment effluent. FEMS Microbiol. Lett. 
210:143-147. 

56. Jean, J., B. Blais, A. Darveau, and I. Fliss. 2001. Detection of hepatitis A virus by 
the nucleic acid sequence-based amplification (NASBA) technique and comparison 
with RT-PCR. Appl. Environ. Microbiol. 67:5593-5600. 

57. Jiang, X., P.W. Huang, W.M. Zhong, T. Farkas, D.W. Cubitt, and D.O. Matson. 
1999. Design and evaluation of a primer pair that detects both Norwalk- and 
Sapporo-like caliciviruses by RT-PCR. J. Virol. Methods 83:145-154. 

58. Kingsley, D.H. and G.P. Richards. 2003. Persistence of hepatitis A virus in oysters. 
J. Food Protect. 66:331-334. 

59. Kingsley, D.H., G.K. Meade, and G.P. Richards. 2002. Detection of both hepatitis 
A virus and Norwalk-like virus in imported clams associated with food-borne illness. 
Appl. Environ. Microbiol. 68:3914-3918. 

60. Kingsley, D.H. and G.P. Richards. 2001. Rapid and efficient extraction method for 
reverse transcription-PCR detection of hepatitis A and Norwalk-like viruses in 
shellfish. Appl. Environ. Microbiol. 67:4152-4157. 



114 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



61. Kobayashi, S., K. Natori, N. Takeda, and K. Sakae. 2004. Immunomagnetic capture 
RT-PCR for detection of norovirus from foods implicated in a foodborne outbreak. 
Microbiol. Immunol. 48:201-204. 

62. Kojima, S., T. Kageyama, S. Fukushi, F.B. Hoshino, M. Shinohara, K. Uchida, 
K. Natori, N. Takeda, and K. Katayama. 2002. Genogroup-specific PCR primers 
for detection of Norwalk-like viruses. J. Virol. Methods 100:107-114. 

63. Lees, D. N., K. Henshilwood, J. Green, C. I. Gallimore, and D. W. Brown. 1995. 
Detection of small round structured viruses in shellfish by reverse-transcription- 
PCR. Appl. Environ. Microbiol. 61:4418-4424. 

64. Lees, D.N., K. Henshilwood, and W.J. Dore. 1994. Development of a method for 
detection of enteroviruses in shellfish by PCR with poliovirus as a model. Appl. 
Environ. Microbiol. 60:2999-3005. 

65. Legeay, O., Y. Caudrelier, C. Cordevant, L. Rigottier-Gois, and M. Lange. 2000. 
Simplified procedure for detection of enteric pathogenic viruses in shellfish by RT- 
PCR. J. Virol. Methods 90:1-14. 

66. Leggitt, PR. and L.-A Jaykus. 2000. Detection methods for human enteric viruses 
in representative foods. J. Food Protect. 63:1738-1744. 

67. LeGuyader, F, L. Haugarreau, L. Miossec, E. Dubois, and M. Pommepuy. 2000. 
Three-y study to assess human enteric viruses in shellfish. Appl. Environ. Microbiol. 
66:3241-3248. 

68. LeGuyader, F, M.K. Estes, M.E. Hardy, FH. Neill, J. Green, D.W.G. Brown, and 
R.L. Atmar. 1996. Evaluation of a degenerated primer for the PCR detection of 
human caliciviurses. Arch. Virol. 141:2225-2235. 

69. LeGuyader, F, FH. Neill, M.K. Estes, S.S. Monroe, T. Ando, R.L. Atmar. 1996. 
Detection and analysis of a small round- structured virus strain in oysters implicated 
in an outbreak of acute gastroenteritis. Appl. Environ. Microbiol. 62:4268-4272. 

70. Le Guyader, F, E. Dubois, D. Menard, and M. Pommepuy 1994. Detection of 
hepatitis A virus, rotavirus, and enterovirus in naturally contaminated shellfish and 
sediment by reverse transcription-seminested PCR. Appl. Environ. Microbiol. 
60:3665-3671. 

71. Lopez- S abater, E.I., M.Y. Deng, and D.O. Cliver. 1997. Magnetic immunosepa- 
ration PCR assay (MIPA) for detection of hepatitis A virus (HAV) in American 
oyster (Crassostrea virginica). Lett. Appl. Microbiol. 24:101-104. 

72. Mackay, I.M., K.E. Arden, and A. Nitsche. 2002. Real-time PCR in virology. Nucl. 
Acids Res. 30:1292-1305. 

73. Miller, I., R. Gunson, and W.F Carman. 2002. Norwalk like virus by light cycler 
PCR. J. Clin. Virol. 25:231-232. 

74. Moe, C, J. Gentsch, T. Ando, G Grohmann, S.S. Monroe, X. Jiang, J. Wang, M.K. 
Estes, Y. Seto, C. Humphrey, S. Stine, and R.I. Glass. 1994. Application of PCR to 
detect Norwalk virus in fecal specimens from outbreaks of gastroenteritidis. J. Clin. 
Microbiol. 31:2866-2872. 

75. Mullendore, J.L., M.D. Sobsey, and Y.S. C. Sheih. 2001. Improved method for the 
recovery of hepatitis A virus from oysters. J. Virol. Methods 94:25-35. 

76. Muniain-Mujika, I.,M. Calvo, F Lucena, and R. Girones. 2003. Comparative analy- 
sis of viral pathogens and potential indicators in shellfish. Int. J. Food Microbiol. 
83:75-85. 

77. Muniain-Mujika, I., R. Girones, and F Lucena. 2000. Viral contamination of shell- 
fish: Evaluation of methods and analysis of bacteriophages and human viruses. 
J. Virol. Methods 89:109-118. 



References 115 



78. Myrmel, M., E.M. Berg, E. Rimstad, and B. Grinde. 2004. Detection of enteric 
viruses in shellfish from the Norwegian coast. Appl. Environ. Microbiol. 
70:2678-2684. 

79. Narayanan, J., J. Lowther, K. Henshilwood, D.N. Lees, V.R. Hill, and J. Vinje. 2005. 
Rapid and sensitive detection on noroviruses using Taq-Man-based one-step RT- 
PCR assays and application to clinical and shellfish samples. Appl. Environ. 
Microbiol. (In Press). 

80. Noel, IS., T. Ando, IP. Leite, K.Y. Green, K.E. Dingle, M.K. Estes, Y. Seto, S.S. 
Monroe, and R.I. Glass. 1997. Correlation of patient immune responses with geneti- 
cally characterized small round-structured viruses involved in outbreaks of nonbacte- 
rial acute gastroenteritidis in the United States, 1990-1995. 1 Med. Virol. 53:372-383. 

81. Ratcliff, R.M., IC. Doherty, and GD. Higgins. 2002. Sensitive detection of RNA 
viruses associated with gastroenteritis by a hanging-drop single-tube nested reverse 
transcription-PCR method. I. Clin. Microbiol. 40:4091-4099. 

82. Ribao, C, I. Torrado, M.L. Vilarino, and XL. Romalde. 2004. Assessment of differ- 
ent commercial RNA-extraction and RT-PCR kits for detection of hepatitis A virus 
in mussel tissues. I Virol. Methods 115:177-182. 

83. Richards, G. P. 1999. Limitations of molecular biological techniques for assessing 
the virological safety of foods. I Food Protect. 62:691-697. 

84. Rohayem, I, S. Berger, T. luretzek, O. Herchenroder, M. Mogel, M. Poppe, 
I Henker, A. Rethwilm. 2004. A simple and rapid single-step multiplex RT-PCR to 
detect Norovirus, Astrovirus and Adenovirus in clinical stool samples. I Virol. 
Methods 118:49-59. 

85. Romalde, J.L., E. Area, G Sanchez, C. Ribao, I. Torrado, X. Abad, R.M. Pinto, XL. 
Barja, and A. Bosch. 2002. Prevalence of enterovirus and hepatitis A virus in bivalve 
molluscs from Galicia (NW Spain): Inadequacy of the EU standards of microbio- 
logical quality. Int I Food Microbiol 74:1 19-130. 

86. Rossen, L., P. Norskov, K. Holmstrom, and O.F. Rasmussen. 1992. Inhibition of 
PCR by components of food samples, microbial diagnostic assays and DNA extrac- 
tion solutions. Int. I Food Microbiol. 17:37^45. 

87. Sair, A.I., D. D'Souza, C.L. Moe, and L-A Jaykus. 2002. Improved detection of 
human enteric viruses in foods by RT-PCR. I Virol. Methods 100:57-69. 

88. Sair, A.I., D.H. D'Souza, and L. laykus. 2002. Human enteric viruses as causes of 
foodborne disease. Comp. Rev. Food Sci. Safety 1:73-89. 

89. Schvoerer, E., M. Ventura, O. Dubos, G Cazaux, R. Serceau, N Gournier, V Dubois, 
P. Caminade, H.I Fleury, and M.E. Lafon. 2001. Qualitative and quantitative molec- 
ular detection of enteroviruses in water from bathing areas and from a sewage treat- 
ment plant. Res. Microbiol. 152:179-186. 

90. Schwab, K.I, F.H. Neill, F. Le Guyader, M.K. Estes, and R.L. Atmar. 2001. 
Development of a reverse transcription-PCR-DNA enzyme immunoassay for detec- 
tion of "Norwalk-like" viruses and hepatitis A virus in stool and shellfish. Appl. 
Environ. Microbiol. 67:742-749. 

91. Schwab, K.I, F.H. Neill, R.L. Fankhauser, N.A. Daniels, S.S. Monroe, D.A. 
Bergmire-Sweat, M.K. Estes, and R.L. Atmar. 2000. Development of methods to 
detect "Norwalk-like Viruses" (NLVs) and hepatitis A virus in delicatessen foods: 
Application to a foodborne NLV outbreak. Appl. Environ. Microbiol. 66:213-218. 

92. Schwab, K.I, F.H. Neill, M.K. Estes, T.G. Metcalf, and R.L. Atmar. 1998. 
Distribution of Norwalk virus within shellfish following bioaccumulation and sub- 
sequent depuration by detection using RT-PCR. I Food Prot. 61:1674-1680. 



116 Molecular Approaches for the Detection of Foodborne Viral Pathogens 



93. Schwab, K.J., M.K. Estes, F.H. Neill, and R.L. Atmar. 1997. Use of heat release 
and an internal RNA standard control in reverse transcription-PCR detection of 
Norwalk virus from stool samples. J. Clin. Microbiol. 35:511-514. 

94. Schwab, K.J., R. De Leon, and M.D. Sobsey. 1996. Immuno affinity concentration 
and purification of waterborne enteric viruses for detection by reverse transcrip- 
tase PCR. Appl. Environ. Microbiol. 62:2086-2094. 

95. Shieh, Y.C., R.S. Baric, J.W. Woods, and K.R. Calci. 2003. Molecular surveillance 
of enterovirus and Norwalk-like virus in oysters relocated to a municipal- sewage- 
impacted gulf estuary. Appl. Environ. Microbiol. 69:7130-7136. 

96. Shieh, Y.C., S.S. Monroe, R.L. Frankhauser, G.W. Langlois, W. Burkhardt, III, 
and R.S. Baric. 2000. Detection of Norwalk-like virus in shellfish implicated in 
illness. J. Infect. Dis. 181(S2):S360-366. 

97. Shieh, Y.C., K.R. Calci, and R. S. Baric. 1999. A method to detect low levels of 
enteric viruses in contaminated oysters. Appl. Environ. Microbiol. 65:4709^4714. 

98. Straub, T.M., I.L. Pepper, and C.R Gerba. 1994. Detection of naturally occurring 
enteroviruses and hepatitis A in undigested and anaerobically digested sludge 
using the polymerase chain reaction. Can. J. Microbiol. 40:884-888. 

99. Sugieda, M., K. Nakajima, and S. Nakajima. 1996. Outbreaks of Norwalk-like 
virus-associated gastroenteritis traced to shellfish: Coexistence of two genotypes in 
one specimen. Epidemiol. Infect. 116:339-346. 

100. Sunen, E. and M.D. Sobsey. 1999. Recovery and detection of enterovirus, hepati- 
tis A virus and Norwalk virus in hardshell clams (Mercenaria mercenaria) by RT- 
PCR methods. J. Virol. Methods 77:179-187. 

101. Svensson, L. 2000. Diagnosis of foodborne viral infections in patients. Int. J. Food 
Microbiol. 59:117-126. 

102. Traore, O., C. Arnal, B. Mignotte, A. Maul, H. Laveran, S. Billaudel, and 
L. Schwartzbrod. 1998. Reverse transcriptase PCR detection of astrovirus, hepatitis 
A virus, and poliovirus in experimentally contaminated mussels: Comparison of sev- 
eral extraction and concentration methods. Appl. Environ. Microbiol. 64:3118-3122. 

103. Tsai, Y.L. and S.L. Parker. 1998. Quantification of poliovirus in seawater and sewage 
by competitive reverse transcriptase — Polymerase chain reaction. Can. J. Microbiol. 
44:35^-1. 

104. Tsai, Y-L., B. Tran, and C.J. Palmer. 1995. Analysis of viral RNA persistence in 
seawater by reverse-transcriptase-PCR. Appl. Environ. Microbiol. 61:363-366. 

105. Vinje, J., R.A. Hamidjaja, and M.D. Sobsey. 2004. Development and application 
of a capsid VP1 (region D) based reverse transcription PCR assay for genotyping 
of genogroup I and II noroviruses. J. Virol. Methods 116:109-117. 

106. Vinje, I, H. Vennema, L. Maunula, L., C-H van Bonsdorff, M. Hoehne, E. Schreier, 
A. Richards, J. Green, D. Brown, S.S. Beard, S.S. Monroe, E. de Bruin, L. Svensson, 
and M P.G Koopmans. 2003. International collaborative study to compare reverse 
transcriptase PCR assays for detection and genotyping of noroviruses. J. Clin. 
Microbiol. 41:1423-1433. 

107. Vinje, J. and M.P. Koopmans. 2000. Simultaneous detection and genotyping of 
"Norwalk-like viruses" by oligonucleotide array in a reverse line blot hybridization 
format. J. Clin. Microbiol. 38:2595-2601. 

108. Wang, X, X. Jiang, H.P. Madore, J. Gray, U. Desselberger, T. Ando, Y Seto, 
I. Oishi, IF. Lew, K.Y Green, and M.K. Estes. 1994. Sequence diversity of small, 
round-structured viruses in Norwalk virus group. J. Virol. 68:5982-5990. 

109. Wiedbrauk, D.L. and A.M. Devron. 1995. Nucleic acid detection methods. In 
D.L. Wiedbrauk and D.H. Farkas (eds.), Molecular Methods for Virus Detection, 
pp. 1-24. Academic Press Inc., San Diego, CA. 



References 117 



110. Wilde, X, J. Eiden, and R. Yolken. 1990. Removal of inhibitory substances from 
human fecal specimens for detection of group A rotaviruses by reverse transcrip- 
tase and polymerase chain reactions. J. Clin. Microbiol. 28:1300-1307. 

111. Wilson, J. G. 1997. Inhibition and facilitation of nucleic acid amplification. Appl. 
Environ. Microbiol. 63:3741-3751. 

112. Wolfaardt, M., M.B. Taylor, H.F. Booysen, L. Engelbrecht, WO. Grabow, and 
X. Jiang. 1997. Incidence of human calicivirus and rotavirus infection in patients 
with gastroenteritis in South Africa. J. Med. Virol. 51:290-296. 

113. Yan, H., F. Yagyu, S. Okitsu, O. Nishio, and H. Ushijima. 2003. Detection of 
norovirus (GI, Gil), sapovirus and astrovirus in fecal samples using reverse tran- 
scription single-round multiplex PCR. J. Virol. Methods 114:37^44. 



CHAPTER 7 



Molecular Tools for the Identification 

of Foodborne Parasites 

Ynes Ortega*, Ph.D. 

Center for Food Safety, College of Agriculture and Environmental Sciences, The University 

of Georgia, Griffin, GA 30223 



Introduction 

DNA Extraction Procedures 
Protozoal Infections 
Cryptosporidium parvum 

Parasite Description and Identification 

Molecular Detection 
Cyclospora cayetanensis 

Parasite Description and Identification 

Molecular Detection 
Giardia intestinalis 

Parasite Description and Identification 

Molecular Detection 
Toxoplasma gondii 

Parasite Description and Identification 

Molecular Detection 
Microsporidia 

Parasite Description and Identification 

Molecular Detection 

Helminth Infections 

Viability Assays 

Conclusions 

References 



INTRODUCTION 



Parasites have long been associated with food and waterborne outbreaks. 
Although parasites have been consistently reported in developing and endemic 
countries, the number of parasites present in the food supply of Americans has 
multiplied by more than a factor of 8 during the past 1 5 years. This is partly due 
to the increase of international travel and population migration. Rapid and 
refrigerated food transportation from foreign countries facilitates consumer 
contact with emerging parasites. Cultural habits have also changed towards the 



* 



Corresponding author. Phone: (770) 233-5587; e-mail: ortega@uga.edu 



119 



120 Molecular Tools for the Identification of Foodborne Parasites 



consumption of (raw or undercooked) fresh produce. These conditions have 
increased the probability that parasites infect naive populations and cause gas- 
trointestinal illness (91). 

Most parasites are obligate intracellular organisms. In contrast with bac- 
teria, parasites are inert and do not multiply in the environment. Any isola- 
tion and detection procedures are crucial because an enrichment process for 
parasites is not available. Molecular assays overcome these difficulties and 
specific limitations per organism will be discussed. 

Based on their morphological attributes, parasites are classified into two 
groups: protozoa and helminths. The protozoa are single-celled organisms and 
the helminths are metazoans with a rudimentary digestive and reproductive 
tract. The helminths are grouped as the nematode or roundworms, the cestoda 
or tapeworm, and the trematoda or flukes. The primary objectives in develop- 
ing diagnostic molecular assays have been focused on protozoa because of the 
limitations of conventional parasitological methods to identify them in foods 
and the environment. 



DNA EXTRACTION PROCEDURES 



Many protocols have been described for the isolation of parasite DNA. Some 
are protocols prepared at individual laboratories. The trend is now to use 
DNA extraction kits, as this will reduce the possible variables among proce- 
dures and laboratories. The recovery efficiency of the extraction procedures, 
particularly with parasites, is important because of the potentially low num- 
ber of parasites in environmental samples and food matrices. The sensitivity 
of the PCR or any molecular assay is dependent on the DNA extraction 
methodology. Enzyme digestion of the protozoal oocysts has been done using 
proteinase K in lysis buffer (10 mg proteinase K/ml, 120 mM NaCl 2 , 10 mM 
Tris and 0.1% SDS) followed by phenol chloroform-isoamyl alcohol (25:24:1) 
separation, and DNA precipitation using salts such as 0.3 M sodium acetate 
with 10 |ig glycogen, a DNA carrier, and 2 volumes of 100% ethanol. This 
method extracts DNA efficiently from parasites, but the most important step 
in this process is to break open the oocysts to release the protozoal DNA. Not 
all protozoal cysts or oocysts will be effectively digested; therefore other 
means of oocyst rupture have been described. One of the commonly used 
methods for cyst/oocyst breakage is the freeze/thaw method with cycles of 
freeze/thaw that vary from 3 to 12 cycles. The freezing is done in dry 
ice/ethanol slurry and thawed at 55 °C. This process may change accordingly 
to the parasite and the laboratory (114). Oocysts can also be disrupted using 
a bath sonicator (101), keeping under consideration the possible denaturation 
of the DNA. Based on forensic studies, an extraction free, filter based prepa- 
ration of DNA has been used and can successfully rupture the parasite cysts 
or oocysts. This method uses an FTA filter, which is impregnated with denat- 
urants, chelating agents, and free radical traps. The sample is placed on the 
filter and cut with a hole puncher. The membrane is then rinsed and used 



Cryptosporidium Parvum 121 



directly for PCR amplification (92). The use of this methodology needs to be 
evaluated for the processing and evaluation of large sample sizes and the 
potential for cross-contamination. 



PROTOZOAL INFECTIONS 



Protozoan parasites relevant to public health and associated with foodborne 
infections include the ciliates (i.e., Balantidium coli), amoeba (i.e., Entamoeba his- 
tolytica), flagellates (i.e., Giardia lamblia), and coccidia (i.e., Toxoplasma gondii, 
Cryptosporidium parvum and Cyclospora cayetanensis). Taxonomical placement 
of some protozoan pathogens has been changing as we learn more about the 
genetic makeup of these organisms. Such is the case with the Microsporidia, 
which are now considered to be more closely related to fungi than to the proto- 
zoa. Cryptosporidium, originally from the phylum Apicomplexa, has more char- 
acteristics associated to the gregarines than to coccidia. Its taxonomical 
classification is uncertain and as more evidence is published, these parasites may 
be reclassified to more appropriate groups. 



CRYPTOSPORIDIUM PARVUM 



Parasite Description and Identification. Cryptosporidium sp. was first recognized 
by Tyzzer in 1907, from the stomach of a mouse (78). It is currently identified 
as C muris. Subsequently, other species of Cryptosporidium were described and 
renamed. Cryptosporidium species that were morphologically similar were 
named Cryptosporidium parvum, but recent molecular analysis have lead to 
reclassification into several species: C hominis (C. parvum genotype 1 or the 
human genotype) in humans, C andersoni (C muris-like or C muris bovine 
genotype) and C bovis {Cryptosporidium bovine genotype B) in calves and adult 
cattle, C canis, (C parvum dog genotype) in dogs, and C suis {Cryptosporidium 
pig genotype I) in pigs. To date, there are 15 established Cryptosporidium species 
in fish, reptiles, birds, and mammals and 8 have been reported in humans 
(C hominis, C bovis, C canis, C felis, C meleagridis, C muris, C suis, and 
Cryptosporidium cervine genotype). Cryptosporidium has been associated with 
gastrointestinal illness in humans and it is acquired by ingestion of fecally 
contaminated water or foods. It can also be acquired via person to person. 

The life cycle of Cryptosporidium starts when mature and infectious oocysts 
are excreted in the feces of an infected host. The oocysts are ingested along with 
contaminated water and/or foods and excyst in the gastrointestinal tract. 
Sporozoites are released and infect the epithelial cells of the small intestine, par- 
ticularly the ileum. Parasites multiply asexually, producing type I and II 
meronts containing 8 and 4 merozoites, respectively. Asexual multiplication can 
continue or differentiate to produce the sexual stages of the parasites. 
Microgametocytes (male) fertilize the macrogametocyte (female) producing the 
zygote, which in turn becomes the oocyst. If a thin-walled oocyst is formed, the 



122 Molecular Tools for the Identification of Foodborne Parasites 



life cycle can initiate again. If a thick-walled oocyst (which is environmentally 
resistant) is formed, it is excreted in the feces (78). 

Individuals at risk of acquiring cryptosporidiosis include children in daycare 
centers, individuals caring for animals, the elderly, immunocompromised indi- 
viduals, and travelers. Since Cryptosporidium is highly resistant to common dis- 
infectants, including chlorine, water parks and fountains have been implicated 
in numerous outbreaks. 

A large waterborne outbreak occurred in 1993 in Milwaukee (76, 146), 
where more than 400,000 people suffered gastrointestinal illness. Initially, 
C parvum was considered to be the agent responsible for this outbreak, how- 
ever, molecular analysis of clinical specimens and water samples demonstrated 
that it was actually C hominis. Contamination occurred when tap water was 
contaminated with sewer water back-flow (76, 146). 

Two main target antigens of 15-17 and 23 kDa molecular weights have been 
used for detection of the humoral immune response of individuals with cryp- 
tosporidiosis (102). These antigens have been used in ELISA, western blot, or 
multiplex bead assay for the detection of Cryptosporidium antibodies in sera 
and oral fluids (85, 99, 148). 

Stool specimens are the most common clinical samples examined for diag- 
nosis of Cryptosporidium. Oocysts are usually concentrated using ethyl acetate 
concentration methodologies (139). The sample is then examined using 
microscopy, immunoassays, and molecular techniques. The modified Ziehl- 
Neelsen acid-fast stain and modified Kinyoun's acid-fast stain are more com- 
monly used in the microscopic identification of Cryptosporidium (87, 103). 

Immunofluorescence assays (IFA) are more sensitive and specific than the 
modified acid fast stains and are now being used more frequently in clinical lab- 
oratories. They are the gold standard when examining new diagnostic assays 
(9, 40), but cannot differentiate among the Cryptosporidium species (46). Some of 
these commercial kits include the Merifluor Cryptosporidium! Giardia kit 
(Meridian Bioscience; Cincinatti, OH), Giardia/ Crypto IF kit (TechLab; 
Blacksburg, VA), Monofluo Cryptosporidium kit (Sanofi Diagnostics Pasteur), 
Crypto/ 'Giardia Cel kit (TCS Biosciences; Buckingham, UK), and Aqua-Glo G/C 
kit (Waterborne; New Orleans, LA). Commercial antigen-capture-based enzyme 
immunoassays (EIA) available are the Alexon-Trend ProSpecT Cryptosporidium 
Microplate Assay (Alexon-Trend- Seradyn; Ramsey, MN) and Meridian Premier 
Cryptosporidium kit. These assays may not react to Cryptosporidium species that 
are genetically distant from C parvum, such as C muris, C andersoni, C serpen- 
tis and C bailey i (46). Lateral flow immunochromatographic assays have also 
been commercialized for use with stool samples (60) 

Parasites can be recovered from foods by washing the samples in 0.025M 
phosphate buffered saline, pH 7.25 (95). Detergents (1% sodium dodecyl sulfate 
and 0.1% Tween 80, or the membrane filter elution buffer from EPA method 
1623) and sonication (3-10 min) are also used to facilitate the elution of para- 
sites from the food matrices (13, 105). Cryptosporidium can then be concen- 
trated by centrifugation and examined by immunofluorescence staining 
(13, 95). A sucrose flotation step may be included producing a cleaner sample 



Cryptosporidium Parvum 123 



but at the cost of losing parasites. Moderate recovery rates of 18.2-25.2% were 
reported for a variety of fresh produce. Immunomagnetic separation has been 
included in the Cryptosporidium recovery procedures from lettuce, Chinese 
leaves, and strawberries to 42% for Cryptosporidium and 67% for Giardia 
(105, 106). Cryptosporidium oocysts can be detected by IFA in shellfish gills, 
gastric glands, and hemocytes from the hemolymph (32, 49). 

Identification of Cryptosporidium oocysts in water samples is achieved 
by using IFA after concentration processes (EPA ICR method, EPA 
methodl622/1623, United Kingdom SCA method, and United Kingdom regu- 
latory method) (72). Oocysts are recovered by filtration of 10-100 L or more 
water, concentrated and stained with FITC-labeled Cryptosporidium antibodies. 
The ICR or SCA methods use nominal 1 |im 10" cartridge filters and washes 
concentrated by flotation (using Percoll, sucrose, or potassium citrate). The 
EPA method 1622 and the United Kingdom regulatory method use capsule fil- 
ters for filtration followed by immunomagnetic separation (IMS). To determine 
oocyst viability, 4', 6-diamidoino-2-phenylindole (DAPI) vital dye is used. The 
recovery rates of the EPA method 1622/1623 for Cryptosporidium oocysts are 
between 10 and 75% for surface water (67, 115, 137). One of the limitations of 
these procedures is the cross-reactivity of the monoclonal antibodies used in the 
IMS and IFA kits. Dinoflagellates (120) and algae (111) may not provide accu- 
rate detection and quantification of Cryptosporidium oocysts, and may require 
confirmation by differential interference contrast microscopy. 

Surface water samples may contain Cryptosporidium oocysts, which are a non- 
pathogenic species or genotypes for humans. Therefore, identification of oocysts 
to the species/genotype level is of significant public health importance. The PCR 
has become a useful tool for determining the genotype Cryptosporidium oocysts 
found in shellfish (32, 44, 45). 

Analysis of environmental samples presents several limitations to these 
assays. The number of parasites in foods is usually small. Other structures mor- 
phologically similar may nonspecifically react with antibody-based assays for 
Cryptosporidium and make them less reliable. In addition, the presence of 
Cryptosporidium oocysts, which are not infectious to humans, may be present 
and could be confused with those of public health relevance. When examining 
environmental samples (soil, water, or foods), density gradients or antibody- 
based concentration procedures have been described (i.e., IMS concentration). 
Concentrates are then used for direct-fluorescent antibody (DFA) or for PCR 
detection. 



Molecular Detection. One of the advantages of using molecular assays on envi- 
ronmental samples is the capability of identifying low numbers of parasites and 
being able to determine their species and genotype. This allows for parasite fin- 
gerprinting in waterborne and foodborne outbreaks, particularly when deter- 
mining if the parasite is anthroponotic or zoonotic. It also aids in determining 
the risk factors associated with transmission of cryptosporidiosis in a particu- 
lar setting (4, 5, 43, 66). Subtyping tools have been useful in the investigation of 
foodborne and waterborne outbreaks of cryptosporidiosis (43, 71, 124). 



124 Molecular Tools for the Identification of Foodborne Parasites 



Earlier PCR methods (22, 65, 140) have been used only for the identification 
of Cryptosporidium spp. Several PCR-Restriction Fragment Length 
Polymorphism (RFLP)-based genotyping tools have been developed for the 
detection and differentiation of Cryptosporidium at the species level (6, 62, 70, 
74, 86, 120, 146). Most of these techniques are based on the SSU rRNA gene. 
However, some of the SSU rRNA-based techniques (62, 70) used conserved 
sequences of eukaryotic organisms, which amplify DNA from organisms other 
than Cryptosporidium (127). Nucleotide sequencing-based approaches have also 
been developed for the differentiation of various Cryptosporidium spp. (82, 84, 
125, 126, 136). These techniques use long PCR amplicons, and some amplify 
other Apicomplexan parasites and dinoflagellates. Because of this lack of speci- 
ficity and sensitivity they cannot be used for diagnostic purposes (125, 126, 136). 

Other genotyping techniques are used specifically to differentiate C parvum 
from C hominis (15, 83, 97, 118, 141). Out of ten commonly used genotyping 
tools for Cryptosporidium species/genotypes, only the SSU rRNA-based PCR 
tools can detect all seven Cryptosporidium species/genotypes (57). 

Microsatellite analysis has been used to characterize diversity between 
C parvum or C hominis and their subtypes (19, 20, 35, 142). High-sequence 
polymorphism in the gene of 60 kDa glycoprotein precursor has also been used 
for subtype analysis (98, 124). Other subtyping tools include sequence analysis 
of HSP70 (98, 124), heteroduplex analysis and nucleotide sequencing of the 
double-stranded RNA (71, 146), and single-strand conformation polymor- 
phism (SSCP)-based analysis of the internal transcribed spacer (ITS-2) (41-42). 

The SSU rRNA-based nested PCR-RFLP method has been successfully 
used in conjunction with IMS in the detection and differentiation of 
Cryptosporidium oocysts present in storm water, raw surface water, and waste- 
water (147, 148). 

Two SSU rRNA-based PCR-sequencing tools and one other SSU-based 
PCR-RFLP tool can differentiate Cryptosporidium oocysts in surface and waste- 
water samples (86, 136), suggesting that humans, farm animals, and wildlife con- 
tribute to Cryptosporidium oocyst contamination in water. Other genes have also 
been used for genotyping of Cryptosporidium. HSP70 and TRAP-C2-genes have 
limited use as they do not amplify DNA of Cryptosporidium species distant from 
C parvum(51). 

A few PCR related techniques have also been used to quantify and deter- 
mine viability of Cryptosporidium oocysts. Excy station followed by DNA 
extraction and PCR has been developed to detect viable C parvum oocysts 
(36, 134). Tissue culture and PCR (26, 67, 108) or reverse transcription-PCR 
(RT-PCR) (108, 109) has been used to detect viable Cryptosporidium oocysts. 
RT-PCR techniques have been described for the detection of viable oocysts (48, 
56, 143), but it may overestimate the viability of oocysts (37). A new integrated 
detection assay combining capture of double-stranded RNA with probe-coated 
beads, RT-PCR, and lateral flow chromatography has also been developed, 
which should shorten detection time (64). 

Other molecular tools, such as fluorescence in situ hybridization (FISH), or 
colorimetric in situ hybridization of probes to the SSU rRNA have been used in 



Cyclospora Cayetanensis 125 



the detection or viability evaluation of C parvum oocysts (72, 116). Nucleic acid 
sequence-based amplification (NASBA) has been used in the detection of viable 
C parvum oocysts (10). More recently, a biosensor technique for the detection 
of viable C parvum oocysts has also been described (11), and a microarray 
technique based on HSP70 sequence polymorphism has been developed to 
differentiate Cryptosporidium genotypes (119). 



CYCLOSPORA CAYETANENSIS 



Parasite Description and Identification. Cyclospora cayetanensis was initially 
identified as a cyanobacteria-like organism. The first reports of gastrointestinal 
infections in humans go back to 1988, when it was described as a blue-green 
algae or a large Cryptosporidium. It was not until 1992 when Ortega et al. 
reported a complete description of this parasite as belonging to the coccidian 
(93, 96). Cyclospora has been identified in insectivores, snakes, and rodents. In 
1997, three species were identified in nonhuman primates (30, 73). These are 
morphologically similar to C cayetanensis, but examination of the 18S rDNA 
demonstrated that they are phylogenetically different. A challenge when work- 
ing with Cyclospora is that is there is no animal model suitable to propagate it 
in laboratory conditions. The same limitation applies to the nonhuman primate 
Cyclospora species. 

Cyclosporiasis is characterized by prolonged watery diarrhea. 
Nondifferentiated oocysts are excreted in the feces of the infected individual 
into the environment. Oocysts differentiate after 7-15 days in the environment, 
becoming fully sporulated and infectious. When a susceptible individual ingests 
oocysts from contaminated water or foods, the oocyst will excyst and release the 
sporocysts, which also will undergo excystation (96). Each contains two sporo- 
zoites that will infect the epithelial cells of the small intestine (94). Cyclospora 
preferentially colonizes the ileum; however, there are few reports suggesting 
extraintestinal colonization such as the biliary and respiratory tracts. 

Even though there have been few reports of Cyclospora associated with 
drinking or swimming in contaminated water, most of the reported outbreaks 
in the developed world have been associated with contaminated fresh produce 
and berries. Lettuce, basil, and raspberries have been the most frequently impli- 
cated products (50). In 2004, the first documented outbreak of cyclosporiasis 
was linked to Guatemalan snow peas (113). Some patients reported consump- 
tion of untreated water or reconstituted milk. 

Diagnosis of Cyclospora in clinical specimens is performed using a modified 
acid fast stain, or by direct microscopical examination. A procedure to isolate 
Cyclospora oocysts from produce was described by Robertson (107). For mush- 
rooms, lettuce, and raspberries, recovery was 12%; while recovery from bean 
sprouts was 4% using lectin-coated paramagnetic beads to concentrate the par- 
asite. Although the lectin-coated paramagnetic beads did not significantly 
improve recovery of the oocysts, it did produce a cleaner and smaller final vol- 
ume for easier identification of the protozoan under the microscope. 



126 Molecular Tools for the Identification of Foodborne Parasites 



Molecular Detection. The first PCR for Cyclospora was developed for clinical 
specimens. Fecal samples containing oocysts were disrupted using a bath soni- 
cator at 120 W. The product of the nested PCR was a 304 bp amplicon (101). 
However, this PCR produced amplicons for Eimeria; a coccidian that infects a 
wide range of animals commonly found in environmental samples and is non- 
pathogenic to humans. This nested PCR was modified using the same primers 
but without the leader sequence. The template was prepared using 6 freeze/thaw 
cycles (2 min in liquid nitrogen and heating at 98°C). To address the issue of 
PCR inhibitors, Instagene Matrix (Bio-Rad; Hercules, CA) was added during 
the DNA extraction. Nonfat milk (50 mg/ml) was used in the PCR reaction to 
overcome the effect of inhibitory effects of the food matrix extracts, soil, and 
plant matrices. An RFLP PCR was also developed to differentiate between 
Eimeria and Cyclospora (58). 

An extraction free filter based template preparation was evaluated using 
Cyclospora oocysts from fecal and food matrix samples. Pieces of the FTA fil- 
ters (Whatman; Florham Park, NJ) were added directly to the PCR mixture 
after washing with 10 mM Tris (pH8.0) containing 0.1 mM EDTA and heat 
treatment at 56°C. A sensitivity of 10 to 30 Cyclospora oocyst could be detected 
in 100 g of fresh raspberries (92). 

Quantitative real-time PCR was developed for the identification of 
Cyclospora, targeting a 83-bp region of the 18s rRNA gene. This analysis was 
based on one Cyclospora isolate. The sensitivity and specificity of the assay 
will need to be confirmed when examining a larger number of environmental 
samples (132). 

Since other nonhuman primate Cyclospora also produced 294 bp amplicon 
with the SSU-rRNA nested PCR primers, a multiplex PCR was developed. 
Briefly, PCR is performed first using external, nested PCR primers described by 
Jinneman, followed with PCR using a series of specific primers that differenti- 
ate Cyclospora species and Eimeria species (90). 

A restriction fragment length polymorphism (RFLP) assay was developed 
using the endonuclease Mnl I. RFLP DNA patterns are different between 
Cyclospora and Eimeria (58). To simplify the methodology for use at inspection 
sites, an oligo-ligation assay (OLA) was developed. The target amplicon was 
detected with an antibody-enzyme conjugate that could be read as a colorimetric 
assay (59). 

Methodologies that could be used for fingerprinting analysis and genotype 
discrimination are not yet available. The intervening transcribed spacer 1(ITS1) 
was examined as a potential sequence that will allow discrimination among 
Cyclospora genotypes. When compared, the sequences of 5 isolates from a 
Cyclospora foodborne outbreak were identical. One of two Guatemalan isolates 
and 2 out of 4 Peruvian isolates were also identical (2). Thirty-six Cyclospora 
samples were examined using ITS1 specific primers. Sequence homology of 
460-465 bp from various isolates varied between oocyst samples from 1 to 6.5% 
and between samples and 5.7%. Cryptosporidium ITS is also highly variable 
(1.1-1.3% within and 0.8-1.6% between oocyst samples). The lack of animal 
models to obtain a clonal population of Cyclospora limits the possibility to 



Giardia Intestinalis 127 



determine if there are different ITS sequence types within one oocyst or if there 
are infections with various Cyclospora strains (88). If intraisolate variation 
occurs, as with many coccidian parasites, ITS1 may not be a suitable target for 
genotyping of Cyclospora. 



GIARDIA INTESTINALIS 



Parasite Description and Identification. Giardia was initially described by 
Leeuwenhoek in 1681 (1). Giardia infecting humans was renamed in the early 
1990s: as G intestinalis, G lamblia, and G duodenalis. The name G lamblia was 
well-recognized in the 1970s, but encouraged by other investigators it was 
changed to G duodenalis and G intestinalis in the 1990s. Over 40 species names 
have been proposed on the basis of hosts of origin. Now, several species are rec- 
ognized: G muris from rodents, G agilis from amphibians, G psittaci from 
parakeets, G ardae from herons, and G microti from voles and muskrats. 
Giardia is the most common cause of waterborne outbreaks; however, there 
have also been reports of foodborne outbreaks. In developing countries, chronic 
giardiasis has been associated with long-term growth retardation. Giardia can 
be asymptomatic or cause intermittent or chronic diarrheal complaints. 
Symptomatic giardiasis is characterized by prolonged and intermittent diar- 
rhea, anorexia, flatulency, weight loss, and malabsorption. Whether Giardia 
lamblia infects humans and other mammals, whether it is a single species, and if 
so, whether it is a zoonotic infection has been questioned. Foods implicated 
with giardiasis are fresh produce or foods contaminated by food handlers (1). 

The infective stage of Giardia is the environmentally resistant cyst and the 
vegetative stage is the trophozoite. The trophozoite is pear shaped and has 8 fla- 
gella and a ventral adhesive disk. When a susceptible individual ingests con- 
taminated water or foods containing the cysts, these cysts will excyst in the 
intestine aided by bile salts and gastric acids. The trophozoite will multiply 
asexually. Giardia does not invade tissues and propagation occurs on the epithe- 
lial surface. The trophozoites attach to the epithelial surface. Some of the 
trophozoites will encyst in the jejunum and are passed in the feces. 

Giardia is the most commonly isolated gastrointestinal parasite worldwide 
and the U.S., (61) with large waterborne outbreaks having been reported. The 
cysts are environmentally resistant and survive long periods of time in water at 
cold temperatures. Giardia has been detected in 81% of raw water samples and 
17% of filtered water samples (68). Children attending day care centers are at 
higher risk of acquiring Giardiasis. Waterborne Giardiasis has been associated 
with unfiltered water; recreational water, such as swimming pools; water foun- 
tains; and travel (78). 

Molecular classification tools have been used with various Giardia isolates 
and have determined various assemblages or genotypes based on the sequence 
comparisons of the SS rRNA, triosephosphate isomerase, and glutamate dehy- 
drogenase genes. Genotype A groups 1 and 2 have been isolated from humans 
and animals. Genotype B has also being isolated from humans and some 



128 Molecular Tools for the Identification of Foodborne Parasites 



animals. Assemblages C and D have been isolated from dogs, F from cats, 
G from rats, and E from cows, sheep alpaca, goat, and pigs. The significance of 
these assemblages in the human infections is being examined (1). In the 
Netherlands, patients with assemblage A isolates presented with intermittent 
diarrheal complaints, while assemblage B was present in individuals with per- 
sistent diarrheal complaints (51). 

Diagnosis of Giardia in clinical samples is performed using bright field 
microscopy. Immunofluorescent assay and EIA are commercially available. 
Merifluor Cryptosporidium! Giardia kit (Meridian Bioscience), Giardia/ Crypto 
IF kit (TechLab), Crypto/ Giardia Cel kit (TCS Biosciences), and Aqua-Glo 
G/C kit (Waterborne) are some of the commercially available kits. Detection of 
Giardia cysts in environmental samples presents a significant challenge. 
Recovery of Giardia cysts from surface water to be used by drinking water treat- 
ment plants is achieved using the United States EPA Method 1623. Several of 
these methods have been adapted for detecting parasites in food matrices. 

Molecular Detection. However, the low number of cysts typically present in 
these types of samples requires the use of molecular tools. In clinical specimens 
Giardia and Cryptosporidium were detected 22 times more often by PCR than 
by conventional microscopy (7). 

Analysis of nucleotide sequences of glutamate dehydrogenase (GDH), elon- 
gation factor la (EFla), SSU rRNA, and triosephosphate isomerase (TPI), 
and ADP-ribosylating factor genes can discriminate five to seven defined line- 
ages and assemblages of G intestinalis (80, 81, 121). Thus far, the TPI gene has 
the highest polymorphism in G intestinalis at both intergenotype as well as 
intragenotype levels, and TPI genotyping has proven very useful in epidemio- 
logical investigations of human Giardiasis (121, 123). 

Molecular characterization of Giardia species in wastewater has been used 
for community wide surveillance of human Giardiasis (130, 135). The distribu- 
tion of the Giardia species in environmental samples correlates directly with 
human, agricultural, and wildlife activities. SSU rRNA-based PCR-RFLP 
(130) and beta-giardin-based PCR-RFLP methods were used to identify and 
differentiate between Giardia assemblages A and B from water samples. 

The phylogenetic distance between G intestinalis assemblages A and B is 
greater than typically used to differentiate two protozoan species (79, 81, 121, 
131), suggesting that G intestinalis may be a species complex. Phenotypic dif- 
ferences have also been observed. Assemblage B is more likely found in patients 
with persistent diarrhea, whereas intermittent diarrhea is mostly observed with 
assemblage A (51). 



TOXOPLASMA GONDII 



Parasite Description and Identification. Toxoplasma is a coccidian parasite that 
can cause severe complications for individuals with the infection. It can be 
asymptomatic or can cause abortion in humans if an acute infection develops 



Toxoplasma Gondii 129 



during pregnancy. Healthy individuals may develop encephalitis or be asymp- 
tomatic. Toxoplasma can be found worldwide and serologically it can be identi- 
fied in as high as 85% of the population in some European countries. 
Toxoplasma is responsible for 20.7% of foodborne deaths due to known infec- 
tious agents. Waterborne outbreaks in Canada and Brazil (12, 54) have been 
reported as well. 

Infection occurs when water or foods are contaminated with cat feces con- 
taining Toxoplasma oocysts. The oocysts excyst and the sporozoites migrate and 
preferentially localize in muscle and the brain. The parasites will encyst and 
form cysts. These contain bradyzoites, which are slow multiplying parasites. 
Once they become active they are called tachyzoites and multiply quickly. The 
parasite can cross the placenta to infect the fetal tissues (29). 

A large variety of animals can acquire toxoplasmosis, but only felines 
(domestic and wild) are the definitive hosts. When infected tissues are ingested, 
the parasites will be released from the tissues and develop to the asexual and 
sexual stages. Once fertilization occurs, the oocysts are formed and then 
excreted in the environment. The oocysts are highly resistant even to desiccation 
and survive on dry surfaces for weeks or even months. Toxoplasma causes fatal 
meningoencephalitis in a variety of marine mammals. Shellfish has been stud- 
ied during the past several years as indicators of or vectors for transmission of 
protozoal agents (29). Shellfish can concentrate large volumes of water and it 
has been demonstrated experimentally that viable Toxoplasma oocysts can also 
be concentrated. Toxoplasma has been classified into three different lineages I, 
II, and III (53). Toxoplasma belonging to type I lineage are highly virulent in 
laboratory animals, whereas the type II and III lineages are nonvirulent. In 
humans, the type II lineage predominates among Toxoplasma associated with 
infections in AIDS and non-AIDS immunocompromised patients (75-80%), as 
well as congenital Toxoplasma infections (encephalitis, pneumonitis, or dissem- 
inated infections). It appears that the type I lineage is more prevalent in con- 
genital Toxoplasma infections in Spain (38). Ocular toxoplasmosis is a common 
sequela of congenital toxoplasmosis, but can be dormant for years and emerge 
at adulthood; causing severe retinochoriditis. PCR analysis of clinical samples 
from patients with these conditions determined that most Toxoplasma isolates 
were type I, type IV, or novel types (69). Outbreaks in Canada and Brazil char- 
acterized by severe ocular toxoplasmosis were caused by Toxoplasma type I 
strains (16). 

Toxoplasma infections can be diagnosed by serological assays examining the 
antibody response towards the infection. Identification of Toxoplasma oocysts 
can be identified in the environment using conventional microscopy; however, 
one must consider the limitation to this approach: (1) the small number of par- 
asites present in environmental samples; and (2) the oocysts are indistinguish- 
able morphologically from other coccidians. Toxoplasma oocysts have been 
isolated from mussels, which serve as paratenic hosts assimilating and concen- 
trating oocysts. Toxoplasma oocysts can be identified from water samples using 
the current USEPA method for concentration of Cryptosporidium (54). 
Centrifugation and flocculation procedures using aluminum sulfate and ferric 



130 Molecular Tools for the Identification of Foodborne Parasites 



sulfate can also concentrate Toxoplasma oocysts. Sporulated oocysts were 
recovered more efficiently using aluminum sulfate and unsporulated oocysts 
could be better recovered using ferric sulfate (63). A TaqMan PCR assay was 
developed to detect the ssrRNA. Infectious Toxoplasma oocysts were detected 
up to 21 days in mussels as confirmed using the mouse bioassay (8). 

Molecular Detection. Most PCR assays used for Toxoplasma identification use 
primers targeting the Bl gene. It is a 3 5 -fold repetitive gene that is highly spe- 
cific and conserved among strains of Toxoplasma (18). A PCR-enzyme 
immunoassay oligoprobe was developed to detect Toxoplasma oocysts. The 
PCR was directed towards the amplification of the Bl gene. Avidin coated 
plates were used to capture the biotin-labeled PCR amplicons, and an internal, 
FITC labeled oligoprobe was allowed to hybridized to the denatured and bound 
amplicon. The bound FITC-tagged oligoprobe was detected using anti-FITC 
antibodies tagged with horse radish peroxidase. This assay could detect 50 
oocysts in a clean preparation (114). Other assays have focused on the sensitiv- 
ity of the PCR assay. Jalal designed primers for Bl gene PCR amplification 
with a sensitivity of 2 parasites/sample (55). 

The freeze thaw procedure in Tris-EDTA buffer and proteinase K digestion 
has been used to break open Toxoplasma oocysts. This is followed by DNA 
extraction using the QIAamp DNA minikit. Again, the Bl gene was used in a 
real-time PCR using an Icycler device (Bio Rad). Sensitivity of the real-time 
PCR using experimentally spiked deionized and public drinking water samples 
was of 1 and 10 oocysts. However, the sensitivity of the PCR was reduced for 
raw surface waters as only 20%, and 50% were positive by real-time PCR for 
samples spiked with 100 and 1000 oocysts, respectively (133). 

Oocyst heating at 100°C for 40 min in TE buffer followed with 9 freeze-thaw 
cycles and proteinase K digestion at 56°C overnight has been used to break 
open the oocysts and provide a template for PCR. DNA was subsequently 
extracted from the broken oocysts using the phenol: chloroform: isoamyl alco- 
hol procedure. DNA amplification using thel8S-rRNA gene designed by 
McPherson and Gajadhar (77) had a theoretical detection limit of 0.1 oocyst if 
the preparation included oocyst concentration using aluminum sulfate floccu- 
lation (63). TaqMan PCR assays were done using Bl and ssrRNA genes to 
study experimentally inoculated mussels (8). Real-time PCR was more sensitive 
to nested PCR using Bl and bradyzoites specific genes. LC-PCR also had the 
advantage to quantify parasites present in serum and peripheral blood mononu- 
clear cells (23). Other targets were used for DNA amplification. These included 
a 529 bp sequence present at 300 copies in the parasite genome. Using this tar- 
get sequence, real-time PCR had a tenfold higher sensitivity compared to PCR 
targeting the 35 copy Bl gene (52). Mobile genetic elements (MGE), which has 
100-500 copies/cell was also used for Toxoplasma identification followed by 
RFLP analysis (128). Other single copy genes SAG1-4 and GRA4 genes have 
been used as targets for Toxoplasma characterization and identification. 

Characterization of Toxoplasma isolates was achieved using PCR-amplified 
products digested with 13 restriction enzymes and determined the genetic 



MlCROSPORIDIA 131 



relationship among Toxoplasma isolates and other coccidia (17). RFLP-PCR, 
random-amplified polymorphic DNA (RAPD), PCR, and sequencing has 
allowed for genotyping analysis (3, 14, 47). Sequencing of DNA polymerase 
and the gra6 genes have also been used to determine strain types (33). 



MlCROSPORIDIA 



Parasite Description and Identification. Microsporidia belongs to the phylum 
Microspora. It contains more than 1000 species and infects a wide range of 
hosts. Five genera have been implicated in human illness: Encephalitozoon, 
Enterocitozoon, Septata, Pleistophora, and Vitaforma. Originally called 
S. intestinalis, it has been reclassified as Encephalitozoon and Nosema cornea as 
Vitaforma cornea. E. bieneusi causes persistant diarrhea in the immunocompro- 
mised, where it is found frequently in the feces. 

Microsporidia are obligate intracellular organisms that form highly resistant 
spores of pirifiorm or ovid shape ranging from 1 to 2 |im in diameter. Spores 
are ingested along with contaminated water or foods, or inhaled. The spore 
extrudes its polar filament and injects the sporoplasm (infectious spore mate- 
rial). The sporoplasm undergoes merogony forming multiple primordial forms. 
Sporogony follows, forming dividing sporonts, which form the sporoblast. They 
then develop into mature spores. Spores are excreted along with the feces, urine, 
or respiratory secretions. E. bieneusi has been reported in mixed infections with 
Cryptosporidium. 

Microsporidial spores can be identified microscopically using Calcofluor 
white, or modified trichrome with Chromotrope 2R stains. The use of other 
stains has been reported. Identification of the spores is difficult because of the 
small size of the spores, which require a well-trained microscopist. Molecular 
assays have played a significant role, particularly in the identification of particu- 
lar Microsporidia (78). The various species of Microsporidia can be acquired via 
various routes. The species acquired by ingestion of contaminated foods or water 
are E. bieneusi and E. intestinalis. The latter can be successfully grown in vitro. 

Molecular Detection. Primers were designed to amplify the SSU rRNA gene of 
E. bieneusi in fecal and biopsy samples, either by PCR or in situ hybridization 
(21). PCR protocols call for rupture of Microsporidia spores using glass beads 
and overnight digestion with proteinase K to release the template. This is fol- 
lowed by DNA extraction using commercial kits and PCR amplification using 
SSU rRNA gene (24). PCR protocols seem to work with fresh as well as for- 
malized specimens. Other PCR primers have been developed for detecting the 
various Microsporidia species. 

To confirm the identity of the amplified PCR products from the SSU rRNA 
gene, restriction endoucleases Haelll and Pst I have been used to distinguish 
between E. bieneusi and E. intestinalis (34); however, these restriction enzymes 
do not differentiate E. intestinalis from E. cuniculi. Primers were also designed 
to amplify conserved segments of the SSU rRNA of these two Microsporidia. 



132 Molecular Tools for the Identification of Foodborne Parasites 



Results were confirmed by standard staining methods and immunofluorescence 
assay specific for E. intestinalis (89). Orlandi evaluated the filter based protocol 
described for Cyclospora and Cryptosporidium in PCR detection of E. intesti- 
nalis. The PCR assay could identify as few as 10-50 E. intestinalis spores (92). 
PCR amplification followed by Hinfl endonuclease restriction could identify E. 
intestinalis from clinical specimens (100). Examination of a fragment of the ITS 
region suggests the presence of genetically distinct strains of E. bieneusi (104). 
Detection of Microsporidia spores in environmental samples was evaluated 
using immunomagnetic separation followed by PCR and with the concentration 
of the Microsporidia spores by immunomagnetic separation, PCR could detect 
as few as 10 spores per 100 L of tap water (117). Real-time quantitative PCR 
has been developed using the polar tube protein gene 2 of E. intestinalis, which 
could be used to quantify the number of spores produced in vitro, or to deter- 
mine the effect of inactivation procedures (138). Real-time PCR using com- 
mercial DNA isolation kits and an automated MagNA Pure LC instrument 
could identify microsporidia. The sensitivity of the PCR was 100-10,000 
spores/ml of feces (145). PCR followed by sequencing of the ITS fragment of 
various Microsporidia isolates from human and animal origin has demon- 
strated high variability among isolates, and that few of the isolates from animal 
origin may be of public health relevance (122). 



HELMINTH INFECTIONS 



There are three groups of helminthic parasites and each group is significantly 
relevant to public health. Cestodes or flatworms associated with foodborne 
outbreaks include Taenia solium and T. saginata, Diphylobrotrium latum, 
Echinococcus granulosus, and E. multilocularis. Identification of the parasites 
can be done by observation of the larval (or cystic) stages of the parasite in 
meat. Nematodes or round worms have also been identified in meats as larval 
forms (Trichinella spiralis, Anisakis) and the eggs can be present in fresh pro- 
duce or in contaminated water (Ascaris lumbricoides, Toxocara canis, Capillaria, 
Gnatostoma, and Angiostrongilus). Flukes or trematodes can also be acquired 
by ingestion of the cystic forms in fish, crabs, or shellfish (Paragonimus wester- 
mani, Heterophyes, and Nanophyetus). In most instances, these parasites are 
ingested when foods are eaten fresh or raw (not frozen). Another fluke (Fasciola 
or Fasciolopsis) can be acquired by ingestion of raw vegetables containing the 
metacercaria stages. These parasites can be isolated from meats and produce 
(39, 75, 1 12, 129, 144). Molecular assays for detection of these parasites are cur- 
rently not done as routine procedure; however, laboratories that do molecular 
epidemiology have developed assays to describe the genotypes most commonly 
isolated in certain animal species. Most of these parasites have been reported in 
developing areas and countries, where Good Agricultural Practices (GAPs) are 
not established. Some of these parasites can be acquired by ingestion of game 
meats, raw fish, or shellfish. Most of these parasites can be inactivated when 
frozen. 



Conclusions 133 



VIABILITY ASSAYS 



The viability of cysts or oocysts has been examined by using vital dyes. In vitro 
cultivation has been successful for some genotypes and species of 
Cryptosporidium and Toxoplasma. Most of the microsporidian spores can be 
propagated in vitro except for E. bieneusi, which is the most commonly identi- 
fied microsporidia in humans and associated with gastrointestinal illness 
(25, 27, 28). To date, there is no effective in vitro cultivation assay for Cyclospora. 
Giardia can be excysted and grown in TYI-S-33 media; however, a large number 
of cysts are required for successful propagation. To date, some Giardia assem- 
blages or genotypes cannot be propagated in vitro. Animal models that could be 
used to determine infectivity and viability are limited to Cryptosporidium and 
Toxoplasma (108). Cryptosporidium can be propagated using neonate calves or 
mice (110). However, C hominis, which is anthroponotic, does not infect these 
animals. Gnotobiotic pigs have been used to propagate C hominis, but they are 
not a practical animal model for inactivation and viability studies. No animal 
model is currently available for Cyclospora (31), nor has the disease been repro- 
duced with this agent in healthy, human volunteers. Toxoplasma tachyzoites can 
be propagated using the MRC-5 cell line and most other fibroblast cell lines. It 
can infect cats, mice, and chickens. Whether this infectivity is selective to certain 
genotypes is currently not known. 

As evident from this discussion, there are several viability assays available 
for some but not all foodborne parasites. However, how practical and cost- 
effective are these assays in assessing effectiveness of certain processes for elim- 
inating these pathogens, or reducing their load in food? Obviously, there is a 
need for developing methods to discern the effectiveness of certain food 
processes for eradicating or reducing these parasites. Reverse transcriptase PCR 
developed as "real-time" detection of these foodborne parasites might prove to 
be an important tool, in the future, for this endeavor. 



CONCLUSIONS 



Parasites and the intestinal diseases they cause are more frequently being asso- 
ciated with consumption of raw vegetables, fruits, and nonfiltered water. These 
outbreaks are most often associated with produce imported from areas, where 
these parasites are endemic. Importation of foods is now necessary in order to 
satisfy the consumer demands for certain commodities, especially fresh fruits 
and vegetables. Incentives for importation of foods include the cost of produc- 
tion for particular crops throughout the year. The fast transportation of fresh 
produce from the farm to the consumers has favored the survival of these 
pathogens on these commodities. Due to advances in medicine, the U.S. demog- 
raphy has also changed. The elderly, immunocompromised individuals and 
children are at higher risks of acquiring and having a more severe illness. Most 
foodborne outbreaks are considered to be bacterial or viral. To determine the 
etiology of an outbreak may take many days, by which time samples of the 



134 Molecular Tools for the Identification of Foodborne Parasites 



implicated product may not be available for investigation. Because of this fac- 
tor, food scientists and the medical community need to be aware that parasites 
are significant agents for foodborne outbreaks and routine examination of clin- 
ical samples may miss the identification of parasites, particularly with 
Cyclospora and Cryptosporidium. Molecular assays have been very helpful in 
foodborne outbreak investigations, but they have also demonstrated the current 
limitations in food parasitology. Parasite isolation and recovery procedures are 
crucial to have a very accurate and specific diagnosis. This is particularly impor- 
tant, since parasites are generally inert in the environment and enrichment pro- 
cedures necessary for bacterial contaminants are not an option in the detection 
of these pathogens in foods. With further development and refinement, PCR 
will prove to be an important tool in surveillance of foods for these emerging 
human parasites. 



REFERENCES 



1. Adam, R.D. 2001. Biology of Giardia lamblia. Clin. Microbiol. Rev. 14:447^475. 

2. Adam, R.D., Y.R. Ortega, R.H. Gilman, and C.R. Sterling. 2000. Intervening tran- 
scribed spacer region 1 variability in Cyclospora cayetanensis. J. Clin. Microbiol. 
38:2339-2343. 

3. Ajzenberg, D., A.L. Banuls, M. Tibayrenc, and M.L. Darde. 2002. Microsatellite 
analysis of Toxoplasma gondii shows considerable polymorphism structured into 
two main clonal groups. Int. J. Parasitol. 32:27-38. 

4. Alves, M., O. Matos, and F. Antunes. 2003. Microsatellite analysis of Cryptosporidium 
hominis and C parvum in Portugal: A preliminary study. J. Eukaryot. Microbiol. 50 
Suppl:529-530. 

5. Alves, M., L. Xiao, I. Sulaiman, A.A. Lai, O. Matos, and F. Antunes. 2003. 
Subgenotype analysis of Cryptosporidium isolates from humans, cattle, and zoo 
ruminants in Portugal. J. Clin. Microbiol. 41:2744-2747. 

6. Amar, C.F., P.H. Dear, and J. McLauchlin. 2004. Detection and identification by 
real-time PCR/RFLP analyses of Cryptosporidium species from human faeces. Lett. 
Appl. Microbiol. 38:217-222. 

7. Amar, C.F., C. East, E. Maclure, J. McLauchlin, C. Jenkins, P. Duncanson, and 
D.R.A. Wareing. 2004. Blinded application of microscopy, bacteriological culture, 
immunoassays and PCR to detect gastrointestinal pathogens from faecal samples of 
patients with community-acquired diarrhoea. Eur. J. Clin. Microbiol. Infect. Dis. 
23:529-534. 

8. Arkush, K.D., M.A. Miller, CM. Leutenegger, LA. Gardner, A.E. Packham, A.R. 
Heckeroth, A.M. Tenter, B.C. Barr, and PA. Conrad. 2003. Molecular and bioassay- 
based detection of Toxoplasma gondii oocyst uptake by mussels (Mytilus gallo- 
provincialis). Int. J. Parasit. 33:1087-1097. 

9. Arrowood, M.J. and C.R. Sterling. 1989. Comparison of conventional staining 
methods and monoclonal antibody-based methods for Cryptosporidium oocyst 
detection. J. Clin. Microbiol. 27:1490-1495. 

10. Baeumner, A.J., M.C. Humiston, R.A. Montagna, and R.A. Durst. 2001. Detection 
of viable oocysts of Cryptosporidium parvum following nucleic acid sequence based 
amplification. Anal. Chem. 73:1176-1180. 



References 135 



11. Baeumner, A. J., J. Pretz, and S. Fang. 2004. A universal nucleic acid sequence 
biosensor with nanomolar detection limits. Anal. Chem. 76:888-894. 

12. Bahia-Oliveira, L.M., J.L. Jones, J. zevedo-Silva, C.C. Alves, F. Orefice, and D.G. 
Addiss. 2003. Highly endemic, waterborne toxoplasmosis in north Rio de Janeiro 
state, Brazil. Emerg. Infect. Dis. 9:55-62. 

13. Bier, J.W. 1991. Isolation of parasites on fruits and vegetables. Southeast Asian 
J. Trop. Med. Public Health 22 Suppl: 144-145. 

14. Bohne, W., U. Gross, D.J. Ferguson, and J. Heesemann. 1995. Cloning and charac- 
terization of a bradyzoite-specifically expressed gene (hsp30lbagl) of Toxoplasma 
gondii, related to genes encoding small heat-shock proteins of plants. Mol. 
Microbiol. 16:1221-1230. 

15. Bonnin, A., M.N. Fourmaux, J.F. Dubremetz, R.G. Nelson, P. Gobet, G. Harly, 
M. Buisson, D. Puygauthier-Toubas, G. Gabriel-Pospisil, M. Naciri, and P. Camerlynck. 
1996. Genotyping human and bovine isolates of Cryptosporidium parvum by poly- 
merase chain reaction-restriction fragment length polymorphism analysis of a repeti- 
tive DNA sequence. FEMS Microbiol. Lett. 137:207-211. 

16. Boothroyd, J.C. and M.E. Grigg. 2002. Population biology of Toxoplasma gondii 
and its relevance to human infection: Do different strains cause different disease? 
Curr. Opin. Microbiol. 5:438^442. 

17. Brindley, P. J., R.T. Gazzinelli, E.Y. Denkers, S.W. Davis, J.P. Dubey, R. Belfort, Jr., 
M.C. Martins, C. Silveira, L. Jamra, A. P. Waters and A. Sher. 1993. Differentiation 
of Toxoplasma gondii from closely related coccidia by riboprint analysis and a sur- 
face antigen gene polymerase chain reaction. Am. J. Trop. Med. Hyg. 48:447-456. 

18. Burg, J. L., C. M. Grover, P. Pouletty, and J. C. Boothroyd. 1989. Direct and sensi- 
tive detection of a pathogenic protozoan, Toxoplasma gondii, by polymerase chain 
reaction. J. Clin. Microbiol. 27:1787-1792. 

19. Caccio, S., W. Homan, R. Camilli, G Traldi, T. Kortbeek, and E. Pozio. 2000. 
A microsatellite marker reveals population heterogeneity within human and animal 
genotypes of Cryptosporidium parvum. Parasitol. 120:237-244. 

20. Caccio, S., F. Spano, and E. Pozio. 2001. Large sequence variation at two microsatel- 
lite loci among zoonotic (genotype C) isolates of Cryptosporidium parvum. Int. 
J. Parasitol. 31:1082-1086. 

21. Carville, A., K. Mansfield, G Widmer, A. Lackner, D. Kotler, P. Wiest, T. Gumbo, 
S. Sarbah, and S. Tzipori. 1997. Development and application of genetic probes for 
detection of Enter ocytozo on bieneusi in formalin-fixed stools and in intestinal biopsy 
specimens from infected patients. Clin. Diagn. Lab Immunol. 4:405^408. 

22. Chrisp, C.E. and M. LeGendre. 1994. Similarities and differences between DNA of 
Cryptosporidium parvum and C. wrairi detected by the polymerase chain reaction. 
Folia Parasitol. (Praha) 41:97-100. 

23. Contini, C, S. Seraceni, R. Cultrera, C. Incorvaia, A. Sebastiani, and S. Picot. 2005. 
Evaluation of a Real-time PCR-based assay using the lightcycler system for detec- 
tion of Toxoplasma gondii bradyzoite genes in blood specimens from patients with 
toxoplasmic retinochoroiditis. Int. J. Parasitol. 35:275-283. 

24. Da Silva, A.J., F.J. Bornay-Llinares, del Aguila de la Puente Cdel, H. Moura, J.M. 
Peralta, I. Sobottka, D.A. Schwartz, GS. Visvesvara, S.B. Slemenda, and N.J. 
Pieniazek. 1997. Diagnosis of Enter ocytozoon bieneusi (microsporidia) infections by 
polymerase chain reaction in stool samples using primers based on the region cod- 
ing for small-subunit ribosomal RNA. Arch. Pathol. Lab Med. 121:874-879. 

25. Del, A.C., H. Moura, S. Fenoy, R. Navajas, R. Lopez- Velez, L. Li, L. Xiao, 
G. J. Leitch, S.A. Da, N.J. Pieniazek, A.A. Lai, and GS. Visvesvara. 2001. In vitro cul- 



136 Molecular Tools for the Identification of Foodborne Parasites 



ture, ultra structure, antigenic, and molecular characterization of Encephalitozoon cuni- 
culi isolated from urine and sputum samples from a Spanish patient with AIDS. J. Clin. 
Microbiol. 39:1105-1108. 

26. Di Giovanni, G.D. and M.W. Lechevallier. 2005. Quantitative-PCR assessment of 
Cryptosporidium parvum cell culture infection. Appl. Environ. Microbiol. 71:1495-1500. 

27. Didier, E.S., L.B. Rogers, J.M. Orenstein, M.D. Baker, C.R. Vossbrinck, G.T. van, 
R. Hartskeerl, R. Soave, and L.M. Beaudet. 1996. Characterization of 
Encephalitozoon (Septata) intestinalis isolates cultured from nasal mucosa and bron- 
choalveolar lavage fluids of two AIDS patients. J. Eukaryot. Microbiol. 43:34^3. 

28. Doultree, J.C., A.L. Maerz, N.J. Ryan, R.W. Baird, E. Wright, S.M. Crowe, and J.A. 
Marshall. 1995. In vitro growth of the microsporidian Septata intestinalis from an 
AIDS patient with disseminated illness. J. Clin. Microbiol. 33:463-470. 

29. Dubey, J.R 2004. Toxoplasmosis -a waterborne zoonosis. Vet. Parasitol. 126:57-72. 

30. Eberhard, M.L., A.J. da Silva, B.G Lilley, and N.J. Pieniazek. 1999. Morphologic and 
molecular characterization of new Cyclospora species from Ethiopian monkeys: C. 
cercopitheci sp.n., C. colobi sp.n., and C. papionis sp.n. Emerg. Infect. Dis. 5:651-658. 

31. Eberhard, M.L., Y.R. Ortega, D.E. Hanes, E.K. Nace, R.Q. Do, M.G. Robl, K.Y 
Won, C. Gavidia, N.L. Sass, K. Mansfield, A. Gozalo, J. Griffiths, R. Gilman, C.R. 
Sterling, and M.J. Arrowood. 2000. Attempts to establish experimental Cyclospora 
cayetanensis infection in laboratory animals. J. Parasitol. 86:577-582. 

32. Fayer, R., J.M. Trout, E.J. Lewis, M. Santin, L. Zhou, A.A. Lai, and L. Xiao. 2003. 
Contamination of Atlantic coast commercial shellfish with Cryptosporidium. 
Parasitol. Res. 89:141-145. 

33. Fazaeli, A., P.E. Carter, M.L. Darde, and T.H. Pennington. 2000. Molecular typing 
of Toxoplasma gondii strains by GRA6 gene sequence analysis. Int. J. Parasitol. 
30:637-642. 

34. Fedorko, D.P., N.A. Nelson, and C.R Cartwright. 1995. Identification of 
microsporidia in stool specimens by using PCR and restriction en do nucleases. 
J. Clin. Microbiol. 33:1739-1741. 

35. Feng, X., S.M. Rich, D. Akiyoshi, J.K. Tumwine, A. Kekitiinwa, N. Nabukeera, 
S. Tzipori, and G Widmer. 2000. Extensive polymorphism in Cryptosporidium 
parvum identified by multilocus micro satellite analysis. Appl. Environ. Microbiol. 
66:3344-3349. 

36. Filkorn, R., A. Wiedenmann, and K. Botzenhart. 1994. Selective detection of viable 
Cryptosporidium oocysts by PCR. Zentralbl. Hyg. Umweltmed. 195:489^94. 

37. Fontaine, M. and E. Guillot. 2003. Study of 18S rRNA and rDNA stability by real- 
time RT-PCR in heat-inactivated Cryptosporidium parvum oocysts. FEMS 
Microbiol. Lett. 226:237-243. 

38. Fuentes, I., J.M. Rubio, C. Ramirez, and J. Alvar. 2001. Genotypic characterization 
of Toxoplasma gondii strains associated with human toxoplasmosis in Spain: Direct 
analysis from clinical samples. J. Clin. Microbiol. 39:1566-1570. 

39. Gajadhar, A.A. and H.R. Gamble. 2000. Historical perspectives and current global 
challenges of Trichinella and trichinellosis. Vet. Parasitol. 93:183-189. 

40. Garcia, L.S. and WL. Current. 1989. Cryptosporidiosis: Clinical features and diag- 
nosis. Crit Rev. Clin. Lab Sci. 27:439^-60. 

41. Gasser, R.B., Y.G.A. El-Osta, S. Prepens, and R.M. Chalmers. 2004. An improved 
'cold SSCP' method for the genotypic and subgenotypic characterization of 
Cryptosporidium. Mol. Cell. Probes 18:329-332. 

42. Gasser, R.B., X.Q. Zhu, S. Caccio, R. Chalmers, G Widmer, U.M. Morgan, R.C.A. 
Thompson, E. Pozio, and GF. Browning. 2001. Genotyping Cryptosporidium 



References 137 



parvum by single-strand conformation polymorphism analysis of ribosomal and 
heat shock gene regions. Electrophor. 22:433^137. 

43. Glaberman, S., J.E. Moore, C.J. Lowery, R.M. Chalmers, I. Sulaiman, K. Elwin, P.J. 
Rooney, B.C. Millar, IS. Dooley, A.A. Lai, and L. Xiao. 2002. Three drinking- 
water-associated cryptosporidiosis outbreaks, Northern Ireland. Emerg. Infect. Dis. 
8:631-633. 

44. Gomez-Couso, H., F. Freire- Santos, C.F.L. Amar, K.A. Grant, K. Williamson, 
M.E. res-Mazas, and J. McLauchlin. 2004. Detection of Cryptosporidium and 
Giardia in molluscan shellfish by multiplexed nested-PCR. Int. J. Food Microbiol. 
91:279-288. 

45. Gomez-Couso, H., F. Freire-Santos, J. Martinez-Urtaza, O. Garcia-Martin, and 
M.E. res-Mazas. 2003. Contamination of bivalve molluscs by Cryptosporidium 
oocysts: The need for new quality control standards. Int. J. Food Microbiol. 
87:97-105. 

46. Graczyk, T.K., M.R. Cranfield, and R. Fayer. 1996. Evaluation of commercial 
enzyme immunoassay (EIA) and immunofluorescent antibody (FA) test kits for 
detection of Cryptosporidium oocysts of species other than Cryptosporidium parvum. 
Am. J. Trop. Med. Hyg. 54:274-279. 

47. Guo, Z.G, U. Gross, and A.M. Johnson. 1997. Toxoplasma gondii virulence mark- 
ers identified by random amplified polymorphic DNA polymerase chain reaction. 
Parasitol. Res. 83:458^63. 

48. Hallier-Soulier, S. and E. Guillot. 2003. An immunomagnetic separation-reverse 
transcription polymerase chain reaction (IMS-RT-PCR) test for sensitive and rapid 
detection of viable waterborne Cryptosporidium parvum. Environ. Microbiol. 
5:592-598. 

49. Higgins, J.A., R. Fayer, J.M. Trout, L.H. Xiao, A.A. Lai, S. Kerby, and M.C. 
Jenkins. 2001. Real-time PCR for the detection of Cryptosporidium parvum. 
J. Microbiol. Methods 47:323-337. 

50. Ho, A.Y., A.S. Lopez, M.G Eberhart, R. Levenson, B.S. Finkel, A.J. Da Silva, J. M. 
Roberts, P.A. Orlandi, C.C. Johnson, and B.L. Herwaldt. 2002. Outbreak of 
cyclosporiasis associated with imported raspberries, Philadelphia, Pennsylvania, 
2000. Emerg. Infect. Dis. 8:783-788. 

51. Homan, W.L. and T.G Mank. 2001. Human giardiasis: Genotype linked differences 
in clinical symptomatology. Int. J. Parasitol. 31:822-826. 

52. Homan, W.L., M. Vercammen, B.J. De, and H. Verschueren. 2000. Identification of 
a 200- to 300-fold repetitive 529 bp DNA fragment in Toxoplasma gondii, and its use 
for diagnostic and quantitative PCR. Int. J. Parasitol. 30:69-75. 

53. Howe, D.K. and L.D. Sibley. 1995. Toxoplasma gondii comprises three clonal line- 
ages: Correlation of parasite genotype with human disease. J. Infect. Dis. 
172:1561-1566. 

54. Isaac-Renton, J., W.R. Bowie, A. King, GS. Irwin, C.S. Ong, C.P. Fung, M.O. 
Shokeir, and J.P. Dubey. 1998. Detection of Toxoplasma gondii oocysts in drinking 
water. Appl. Environ. Microbiol. 64:2278-2280. 

55. Jalal, S., C.E. Nord, M. Lappalainen, and B. Evengard. 2004. Rapid and sensitive 
diagnosis of Toxoplasma gondii infections by PCR. Clin. Microbiol. Infect. 
10:937-939. 

56. Jenkins, M.C, J. Trout, M.S. Abrahamsen, C.A. Lancto, J. Higgins, and R. Fayer. 
2000. Estimating viability of Cryptosporidium parvum oocysts using reverse 
transcriptase-polymerase chain reaction (RT-PCR) directed at mRNA encoding 
amyloglucosidase. J. Microbiol. Methods 43:97-106. 



138 Molecular Tools for the Identification of Foodborne Parasites 



57. Jiang, XL. and L.H. Xiao. 2003. An evaluation of molecular diagnostic tools for the 
detection and differentiation of human-pathogenic Cryptosporidium spp. J. Euk. 
Microbiol. 50:542-547. 

58. Jinneman, K.C., J.H. Wetherington, W.E. Hill, A.M. Adams, J.M. Johnson, B.J. 
Tenge, N.L. Dang, R.L. Manger, and M.M. Wekell. 1998. Template preparation for 
PCR and RFLP of amplification products for the detection and identification of 
Cyclospora spp. and Eimeria spp. oocysts directly from raspberries. J. Food Prot. 
61:1497-1503. 

59. Jinneman, K.C., J.H. Wetherington, W.E. Hill, C.J. Omiescinski, A.M. Adams, J.M. 
Johnson, B.J. Tenge, N.L. Dang, and M.M. Wekell. 1999. An oligonucleotide-liga- 
tion assay for the differentiation between Cyclospora and Eimeria spp. polymerase 
chain reaction amplification products. J. Food Prot. 62:682-685. 

60. Johnston, S.P., M.M. Ballard, M.J Beach, L. Causer, and P.P. Wilkins. 2003. 
Evaluation of three commercial assays for detection of Giardia and Cryptosporidium 
organisms in fecal specimens. J. Clin. Microbiol. 41:623-626. 

61. Kappus, K.D., R.G. Lundgren, Jr., D.D. Juranek, J.M. Roberts, and H.C. Spencer. 
1994. Intestinal parasitism in the United States: Update on a continuing problem. 
Am. J. Trop. Med. Hyg. 50:705-713. 

62. Kimbell, L.M., III, D.L. Miller, W Chavez, and N. Altman. 1999. Molecular analy- 
sis of the 18S rRNA gene of Cryptosporidium serpentis in a wild-caught corn snake 
(Elaphe guttata guttata) and a five-species restriction fragment length polymor- 
phism-based assay that can additionally discern C parvum from C wrairi. Appl. 
Environ. Microbiol. 65:5345-5349. 

63. Kourenti, C, A. Heckeroth, A. Tenter, and P. Karanis. 2003. Development and 
application of different methods for the detection of Toxoplasma gondii in water. 
Appl. Environ. Microbiol. 69:102-106. 

64. Kozwich, D., K.A. Johansen, K. Landau, C.A. Roehl, S. Woronoff, and P.A. Roehl. 
2000. Development of a novel, rapid integrated Cryptosporidium parvum detection 
assay. Appl. Environ. Microbiol. 66:2711-2717. 

65. Laxer, M.A., B.K. Timblin, and R.J. Patel. 1991. DNA sequences for the specific 
detection of Cryptosporidium parvum by the polymerase chain reaction. Am. J. Trop. 
Med. Hyg. 45:688-694. 

66. Learmonth, J.J, G. Ionas, A.B. Pita, and R.S. Cowie. 2003. Identification and 
genetic characterisation of Giardia and Cryptosporidium strains in humans and 
dairy cattle in the Waikato Region of New Zealand. Water Sci. Technol. 47:21-26. 

67. Lechevallier, M.W, G.D. Di Giovanni, JL. Clancy, Z. Bukhari, S. Bukhari, J.S. 
Rosen, J. Sobrinho, and M.M. Frey 2003. Comparison of method 1623 and cell cul- 
ture-PCR for detection of Cryptosporidium spp. in source waters. Appl. Environ. 
Microbiol. 69:971-979. 

68. Lechevallier, M.W, WD. Norton, and R.G. Lee. 1991. Giardia and Cryptosporidium 
spp. in filtered drinking water supplies. Appl. Environ. Microbiol. 57:2617-2621. 

69. Lehmann, T., C.R. Blackston, S.F. Parmley, J.S. Remington, and J. P. Dubey 2000. 
Strain typing of Toxoplasma gondii: Comparison of antigen-coding and housekeep- 
ing genes. J. Parasitol. 86:960-971. 

70. Leng, X., D.A. Mosier, and R.D. Oberst. 1996. Differentiation of Cryptosporidium 
parvum, C muris, and C baileyi by PCR-RFLP analysis of the 18S rRNA gene. Vet. 
Parasitol. 62:1-7. 

71. Leoni, F, C.I. Gallimore, J. Green, and J. McLauchlin. 2003. A rapid method for 
identifying diversity within PCR amplicons using a heteroduplex mobility assay and 



References 139 



synthetic polynucleotides: Application to characterisation of dsRNA elements asso- 
ciated with Cryptosporidium. J. Microbiol. Methods 54:95-103. 

72. Lindquist, H.D., M. Ware, R.E. Stetler, L. Wymer, and F.W. Schaefer, III. 2001. 
A comparison of four fluorescent antibody-based methods for purifying, detecting, 
and confirming Cryptosporidium parvum in surface waters. J. Parasitol. 
87:1124-1131. 

73. Lopez, F.A., J. Manglicmot, T.M. Schmidt, C. Yeh, H.V. Smith, and D.A. Relman. 
1999. Molecular characterization of Cyclospora-like organisms from baboons. 
J. Infect. Dis. 179:670-676. 

74. Lowery, C.J., IE. Moore, B.C. Millar, D.R Burke, K.A. McCorry, E. Crothers, and 
J.S. Dooley. 2000. Detection and speciation of Cryptosporidium spp. in environmen- 
tal water samples by immunomagnetic separation, PCR and endonuclease restric- 
tion. J. Med. Microbiol. 49:779-785. 

75. Lun, Z.R., R.B. Gasser, D.H. Lai, A.X. Li, X.Q. Zhu, X.B. Yu, and YY Fang. 2005. 
Clonorchiasis: A key foodborne zoonosis in China. Lancet Infect. Dis. 5:31-41. 

76. MacKenzie, W.R., W.L. Schell, K.A. Blair, D.G. Addiss, D.E. Peterson, N.J. Hoxie, 
J.J. Kazmierczak, and J.P. Davis. 1995. Massive outbreak of waterborne Cryp- 
tosporidium infection in Milwaukee, Wisconsin: Recurrence of illness and risk of 
secondary transmission. Clin. Infect. Dis. 21:57-62. 

77. MacPherson, J.M., and A. A. Gajadhar. 1993. Sensitive and specific polymerase 
chain reaction detection of Toxoplasma gondii for veterinary and medical diagnosis. 
Can. J. Vet. Res. 57:45-48. 

78. Marshall, M.M., D. Naumovitz, Y Ortega, and C.R. Sterling. 1997. Waterborne 
protozoan pathogens. Clin. Microbiol. Rev. 10:67-85. 

79. Monis, P.T., and R.H. Andrews. 1998. Molecular epidemiology: Assumptions and 
limitations of commonly applied methods. Int. J. Parasitol. 28:981-987. 

80. Monis, P.T., R.H. Andrews, G. Mayrhofer, and PL. Ey. 1999. Molecular systemat- 
ics of the parasitic protozoan Giardia intestinalis. Mol. Biol. Evol. 16:1135-1144. 

81. Monis, P.T., G. Mayrhofer, R.H. Andrews, W.L. Homan, L. Limper, and PL. Ey. 
1996. Molecular genetic analysis of Giardia intestinalis isolates at the glutamate 
dehydrogenase locus. Parasitol 112:1-12. 

82. Morgan, U.M., C.C. Constantine, D.A. Forbes, and R.C. Thompson. 1997. 
Differentiation between human and animal isolates of Cryptosporidium parvum 
using rDNA sequencing and direct PCR analysis. J. Parasitol. 83:825-830. 

83. Morgan, U.M., C.C. Constantine, P. O'Donoghue, B.P Meloni, PA. O'Brien, and 
R.C. Thompson. 1995. Molecular characterization of Cryptosporidium isolates from 
humans and other animals using random amplified polymorphic DNA analysis. 
Am. J. Trop. Med. Hyg. 52:559-564. 

84. Morgan, U.M., P. Deplazes, D.A. Forbes, F. Spano, H. Hertzberg, K.D. Sargent, 
A. Elliot, and R.C. Thompson. 1999. Sequence and PCR-RFLP analysis of the 
internal transcribed spacers of the rDNA repeat unit in isolates of Cryptosporidium 
from different hosts. Parasitol. 118:49-58. 

85. Moss, D.M. and P.J. Lammie. 1993. Proliferative responsiveness of lymphocytes 
from Cryptosporidium parvum-exposed mice to two separate antigen fractions from 
oocysts. Am. J. Trop. Med. Hyg. 49:393-401. 

86. Nichols, R.A., B.M. Campbell, and H.V. Smith. 2003. Identification of 
Cryptosporidium spp. oocysts in United Kingdom noncarbonated natural mineral 
waters and drinking waters by using a modified nested PCR-restriction fragment 
length polymorphism assay. Appl. Environ. Microbiol. 69:4183^4189. 



140 Molecular Tools for the Identification of Foodborne Parasites 



87. Nielsen, C.K. and L.A. Ward. 1999. Enhanced detection of Cryptosporidium 
parvum in the acid-fast stain. J. Vet. Diagn. Invest 11:567-569. 

88. Olivier, C, PS. van De, P.W. Lepp, K. Yoder, and D.A. Relman. 2001. Sequence vari- 
ability in the first internal transcribed spacer region within and among Cyclospora 
species is consistent with polyparasitism. Int. J. Parasitol. 31:1475-1487. 

89. Ombrouck, C, L. Ciceron, S. Biligui, S. Brown, P. Marechal, G.T. Van, A. Datry, 
M. Danis, and I. sportes-Livage. 1997. Specific PCR assay for direct detection of 
intestinal microsporidia Enterocytozoon bieneusi and Encephalitozoon intestinalis 
in fecal specimens from human immunodeficiency virus-infected patients. J. Clin. 
Microbiol. 35:652-655. 

90. Orlandi, P.A., L. Carter, A.M. Brinker, A.J. Da Silva, D.M. Chu, K.A. Lampel, 
and S.R. Monday. 2003. Targeting single-nucleotide polymorphisms in the 18S 
rRNA gene to differentiate Cyclospora species from Eimeria species by multiplex 
PCR. Appl. Environ. Microbiol. 69:4806^813. 

91. Orlandi, P.A., D.M. Chu, J.W. Bier, and G.J. Jackson. 2002. Parasites and the Food 
Supply. Food Technol. 56:72-81. 

92. Orlandi, P.A. and K.A. Lampel. 2000. Extraction-free, filter-based template prepa- 
ration for rapid and sensitive PCR detection of pathogenic parasitic protozoa. 
J. Clin. Microbiol. 38:2271-2277. 

93. Ortega, Y.R., R.H. Gilman, and C.R. Sterling. 1994. A new coccidian parasite 
(Apicomplexa: Eimeriidae) from humans. J. Parasitol. 80:625-629. 

94. Ortega, Y.R., R. Nagle, R.H. Gilman, J. Watanabe, J. Miyagui, H. Quispe, 
P. Kanagusuku, C. Roxas, and C.R. Sterling. 1997. Pathologic and clinical findings 
in patients with cyclosporiasis and a description of intracellular parasite life-cycle 
stages. J. Infect. Dis. 176:1584-1589. 

95. Ortega, Y.R., C.R. Roxas, R.H. Gilman, N.J. Miller, L. Cabrera, C. Taquiri, and 
C.R. Sterling. 1997. Isolation of Cryptosporidium parvum and Cyclospora cayeta- 
nensis from vegetables collected in markets of an endemic region in Peru. Am. 
J. Trop. Med. Hyg. 57:683-686. 

96. Ortega, Y.R., C.R. Sterling, R.H. Gilman, V.A. Cama, and F. Diaz. 1993. 
Cyclospora species — a new protozoan pathogen of humans. N. Engl. J. Med. 
328:1308-1312. 

97. Patel, S., S. Pedraza-Diaz, and J. McLauchlin. 1999. The identification of 
Cryptosporidium species and Cryptosporidium parvum directly from whole faeces 
by analysis of a multiplex PCR of the 18S rRNA gene and by PCR/RFLP of the 
Cryptosporidium outer wall protein (COWP) gene. Int. J. Parasitol. 29:1241-1247. 

98. Peng, M.M., S.R. Meshnick, N.A. Cunliffe, B.D. Thindwa, C.A. Hart, R.L. 
Broadhead, and L. Xiao. 2003. Molecular epidemiology of cryptosporidiosis in 
children in Malawi. J. Eukaryot. Microbiol. 50 Suppl:557-559. 

99. Priest, J.W., J.P. Kwon, M.J. Arrowood, and P.J. Lammie. 2000. Cloning of the 
immunodominant 17-kDa antigen from Cryptosporidium parvum. Mol. Biochem. 
Parasitol. 106:261-271. 

100. Raynaud, L., F. Delbac, V. Broussolle, M. Rabodonirina, V. Girault, M. Wallon, 
G Cozon, C. P. Vivares, and F. Peyron. 1998. Identification of Encephalitozoon 
intestinalis in travelers with chronic diarrhea by specific PCR amplification. J. Clin. 
Microbiol. 36:37-40. 

101. Relman, D.A., T.M. Schmidt, A. Gajadhar, M. Sogin, J. Cross, K. Yoder, 
O. Sethabutr, and P. Echeverria. 1996. Molecular phylogenetic analysis of Cyclospora, 
the human intestinal pathogen, suggests that it is closely related to Eimeria species. 
J. Infect. Dis. 173:440-445. 



References 141 



102. Reperant, J.M., M. Naciri, S. Iochmann, M. Tilley, and D.T. Bout. 1994. Major 
antigens of Cryptosporidium parvum recognised by serum antibodies from differ- 
ent infected animal species and man. Vet. Parasitol. 55:1-13. 

103. Rigo, C.R. and R.M. Franco. 2002. [Comparison between the modified Ziehl- 
Neelsen and Acid-Fast-Trichrome methods for fecal screening of Cryptosporidium 
parvum and Isospora belli]. Rev. Soc. Bras. Med. Trop. 35:209-214. 

104. Rinder, H., S. Katzwinkel-Wladarsch, and T. Loscher. 1997. Evidence for the exis- 
tence of genetically distinct strains of Enter ocytozo on bieneusi. Parasitol. Res. 
83:670-672. 

105. Robertson, L.J. and B. Gjerde. 2001. Factors affecting recovery efficiency in isola- 
tion of Cryptosporidium oocysts and Giardia cysts from vegetables for standard 
method development. J. Food Prot. 64:1799-1805. 

106. Robertson, L.J. and B. Gjerde. 2000. Isolation and enumeration of Giardia cysts, 
Cryptosporidium oocysts, and Ascaris eggs from fruits and vegetables. J. Food Prot. 
63:775-778. 

107. Robertson, L.J., B. Gjerde, and A.T. Campbell. 2000. Isolation of Cyclospora 
oocysts from fruits and vegetables using lectin-coated paramagnetic beads. J. Food 
Prot. 63:1410-1414. 

108. Rochelle, P. A., D.M. Ferguson, T. J. Handojo, L.R. De, M.H. Stewart, and R.L. 
Wolfe. 1997. An assay combining cell culture with reverse transcriptase PCR to 
detect and determine the infectivity of waterborne Cryptosporidium parvum. Appl. 
Environ. Microbiol. 63:2029-2037. 

109. Rochelle, P.A., E.M. Jutras, E.R. Atwill, L.R. De, and M.H. Stewart. 1999. 
Polymorphisms in the beta-tubulin gene of Cryptosporidium parvum differentiate 
between isolates based on animal host but not geographic origin. J. Parasitol. 
85:986-989. 

110. Rochelle, P.A., M.M. Marshall, J.R. Mead, A.M. Johnson, D.G. Korich, J. S. 
Rosen, and L.R. De. 2002. Comparison of in vitro cell culture and a mouse assay 
for measuring infectivity of Cryptosporidium parvum. Appl. Environ. Microbiol. 
68:3809-3817. 

111. Rodgers, M.R., D.J. Flanigan, and W. Jakubowski. 1995. Identification of algae 
which interfere with the detection of Giardia cysts and Cryptosporidium oocysts 
and a method for alleviating this interference. Appl. Environ. Microbiol. 
61:3759-3763. 

112. Rodriguez-Canul, R., F. rgaez-Rodriguez, G. D. De, S. Villegas-Perez, A. Fraser, 
PS. Craig, L. Cob-Galera, and J.L. Dominguez-Alpizar. 2002. Taenia solium 
metacestode viability in infected pork after preparation with salt pickling or cook- 
ing methods common in Yucatan, Mexico. J. Food Prot. 65:666-669. 

113. Rose, J.B. and T.R. Slifko. 1999. Giardia, Cryptosporidium, and Cyclospora and 
their impact on foods: A review J. Food Prot. 62:1059-1070. 

114. Schwab, K.J. and J.J. McDevitt. 2003. Development of a PCR-enzyme immunoas- 
say oligoprobe detection method for Toxoplasma gondii oocysts, incorporating 
PCR controls. Appl. Environ. Microbiol. 69:5819-5825. 

115. Simmons, O.D., M.D. Sobsey, F.W. Schaefer, D.S. Francy, R.A. Nally, and CD. 
Heaney. 2001. Evaluation of USEPA method 1622 for detection of 
Cryptosporidium oocysts in stream waters. J. Amer. Water Works Assoc. 93:78-87. 

116. Smith, H.V., B.M. Campbell, C.A. Paton, and R.A. Nichols. 2002. Significance of 
enhanced morphological detection of Cryptosporidium sp. oocysts in water con- 
centrates determined by using 4 , ,6 , -diamidino-2-phenylindole and immunofluores- 
cence microscopy. Appl. Environ. Microbiol. 68:5198-5201. 



142 Molecular Tools for the Identification of Foodborne Parasites 



117. Sorel, N., E. Guillot, M. Thellier, I. Accoceberry, A. Datry, L. Mesnard-Rouiller, 
and M. Miegeville. 2003. Development of an immunomagnetic separation- 
polymerase chain reaction (IMS-PCR) assay specific for Enter ocytozo on bieneusi in 
water samples. J. Appl. Microbiol. 94:273-279. 

118. Spano, E, L. Putignani, A. Crisanti, P. Sallicandro, U.M. Morgan, S.M. Le 
Blancq, L. Tchack, S. Tzipori, and G. Widmer. 1998. Multilocus genotypic analy- 
sis of Cryptosporidium parvum isolates from different hosts and geographical ori- 
gins. J. Clin. Microbiol. 36:3255-3259. 

119. Straub, T.M., D.S. Daly, S. Wunshel, P.A. Rochelle, R. DeLeon, and D.P. 
Chandler. 2002. Geno typing Cryptosporidium parvum with an hsp70 single- 
nucleotide polymorphism microarray. Appl. Environ. Microbiol. 68:1817-1826. 

120. Sturbaum, G.D., C. Reed, P.J. Hoover, B.H. Jost, M.M. Marshall, and C.R. 
Sterling. 2001. Species-specific, nested PCR-restriction fragment length polymor- 
phism detection of single Cryptosporidium parvum oocysts. Appl. Environ. 
Microbiol. 67:2665-2668. 

121. Sulaiman, I.M., R. Fayer, C. Bern, R.H. Gilman, J.M. Trout, P.M. Schantz, P. Das, 

A. A. Lai, and L. Xiao. 2003. Triosephosphate isomerase gene characterization and 

potential zoonotic transmission of Giardia duodenalis. Emerg. Infect. Dis. 
9:1444-1452. 

122. Sulaiman, I.M., R. Fayer, C. Yang, M. Santin, O. Matos, and L. Xiao. 2004. 
Molecular characterization of Enterocytozoon bieneusi in cattle indicates that only 
some isolates have zoonotic potential. Parasitol. Res. 92:328-334. 

123. Sulaiman, I.M., J. Jiang, A. Singh, and L. Xiao. 2004. Distribution of Giardia duo- 
denalis genotypes and subgenotypes in raw urban wastewater in Milwaukee, 
Wisconsin. Appl. Environ. Microbiol. 70:3776-3780. 

124. Sulaiman, I.M., A. A. Lai, and L. Xiao. 2001. A population genetic study of the 
Cryptosporidium parvum human genotype parasites. J. Eukaryot. Microbiol. 
Suppl:24S-27S. 

125. Sulaiman, I.M., A. A. Lai, and L. Xiao. 2002. Molecular phylogeny and evolu- 
tionary relationships of Cryptosporidium parasites at the actin locus. J. Parasitol. 
88:388-394. 

126. Sulaiman, I.M., U.M. Morgan, R.C. Thompson, A. A. Lai, and L. Xiao. 2000. 
Phylogenetic relationships of Cryptosporidium parasites based on the 70-kilodal- 
ton heat shock protein (HSP70) gene. Appl. Environ. Microbiol. 66:2385-2391. 

127. Sulaiman, I.M., L. Xiao, and A. A. Lai. 1999. Evaluation of Cryptosporidium 
parvum genotyping techniques. Appl. Environ. Microbiol. 65:4431^435. 

128. Terry, R.S., IE. Smith, P. Duncanson, and G Hide. 2001. MGE-PCR: A novel 
approach to the analysis of Toxoplasma gondii strain differentiation using mobile 
genetic elements. Int. J. Parasitol. 31:155-161. 

129. van, H.L., D. Blair, and T. Agatsuma. 1999. Genetic diversity in parthenogenetic 
triploid Paragonimus westermani. Int. J. Parasitol. 29:1477-1482. 

130. van, K.H., P.T. Macechko, S. Wade, S. Schaaf, P.M. Wallis, and S.L. Erlandsen. 2002. 
Presence of human Giardia in domestic, farm and wild animals, and environmental 
samples suggests a zoonotic potential for giardiasis. Vet. Parasitol. 108:97-107. 

131. van, K.H., P.A. Steimle, D.A. Bulik, R.K. Borowiak, and E.L. Jarroll. 1998. 
Cloning of two putative Giardia lamblia glucosamine 6-phosphate isomerase genes 
only one of which is transcriptionally activated during encystment. J. Eukaryot. 
Microbiol. 45:637-642. 

132. Varma, M., ID. Hester, F.W. Schaefer, M.W. Ware, and H.D.A. Lindquist. 2003. 
Detection of Cyclospora cayetanensis using a quantitative real-time PCR assay. 
J. Microbiol. Methods 53:27-36. 



References 143 



133. Villena, I., D. Aubert, P. Gomis, H. Ferte, J.C. Inglard, H. is-Bisiaux, J.M. 
Dondon, E. Pisano, N. Ortis, and J.M. Pinon. 2004. Evaluation of a strategy for 
Toxoplasma gondii oocyst detection in water. Appl. Environ. Microbiology 
70:4035^039. 

134. Wagner- Wiening, C. and P. Kimmig. 1995. Detection of viable Cryptosporidium 
parvum oocysts by PCR. Appl. Environ. Microbiol. 61:4514^4516. 

135. Wallis, P.M., D. Matson, M. Jones, and J. Jamieson. 2001. Application of moni- 
toring data for Giardia and Cryptosporidium to boil water advisories. Risk Anal. 
21:1077-1085. 

136. Ward, P.I., P. Deplazes, W Regli, H. Rinder, and A. Mathis. 2002. Detection of 
eight Cryptosporidium genotypes in surface and waste waters in Europe. Parasitol. 
124:359-368. 

137. Ware, M.W, L. Wymer, H.D.A. Lindquist, and F.W Schaefer. 2003. Evaluation of 
an alternative IMS dissociation procedure for use with Method 1622: Detection of 
Cryptosporidium in water. J. Microbiol. Methods 55:575-583. 

138. Wasson, K. and PA. Barry. 2003. Molecular characterization of Encephalitozoon 
intestinalis (Microspora) replication kinetics in a murine intestinal cell line. 
J. Eukaryot. Microbiol. 50:169-174. 

139. Weber, R., R.T. Bryan, and D.D. Juranek. 1992. Improved stool concentration 
procedure for detection of Cryptosporidium oocysts in fecal specimens. J. Clin. 
Microbiol. 30:2869-2873. 

140. Webster, K.A., J.D. Pow, M. Giles, J. Catchpole, and M.J. Woodward. 1993. 
Detection of Cryptosporidium parvum using a specific polymerase chain reaction. 
Vet. Parasitol. 50:35-44. 

141. Widmer, G 1998. Genetic heterogeneity and PCR detection of Cryptosporidium 
parvum. Adv. Parasitol. 40:223-239. 

142. Widmer, G, X. Feng, and S. Tanriverdi. 2004. Genotyping of Cryptosporidium 
parvum with micro satellite markers. Methods Mol. Biol. 268:177-187. 

143. Widmer, G, E.A. Orbacz, and S. Tzipori. 1999. beta-tubulin mRNA as a marker 
of Cryptosporidium parvum oocyst viability. Appl. Environ. Microbiol. 
65:1584-1588. 

144. Wilson, M.E., C.A. Lorente, J.E. Allen, and M.L. Eberhard. 2001. Gongylonema 
infection of the mouth in a resident of Cambridge, Massachusetts. Clin. Infect. 
Dis. 32:1378-1380. 

145. Wolk, D.M., S.K. Schneider, N.L. Wengenack, L.M. Sloan, and J.E. Rosenblatt. 
2002. Real-time PCR method for detection of Encephalitozoon intestinalis from 
stool specimens. J. Clin. Microbiol. 40:3922-3928. 

146. Xiao, L., A. Singh, J. Limor, T.K. Graczyk, S. Gradus, and A. Lai. 2001. 
Molecular characterization of Cryptosporidium oocysts in samples of raw surface 
water and wastewater. Appl. Environ. Microbiol. 67:1097-1101. 

147. Xiao, L., I.M. Sulaiman, U.M. Ryan, L. Zhou, E.R. Atwill, M.L. Tischler, 
X. Zhang, R. Fayer, and A. A. Lai. 2002. Host adaptation and host-parasite co- 
evolution in Cryptosporidium: Implications for taxonomy and public health. Int. 
J. Parasitol. 32:1773-1785. 

148. Zu, S.X., GD. Fang, R. Fayer, and R.L. Guerrant. 1992. Cryptosporidiosis: 
Pathogenesis and immunology. Parasitol. Today 8:24-27. 



144 



Molecular Tools for the Identification of Foodborne Parasites 



CZ3 

<D 

H— » 

•i-H 

C/5 

5-H 

oj 

O 

N 

O 

+- > 

o 

5-H 

P 

o 

S3 

o 

•i-H 

-4— » 

•i-H 

53 

(D 
=5 

5-1 



5-1 

PL. 



1^ 



H 






^ 
-o 



•^ 



CO 









B 

•1— I 

Td 

•i-H 

o 

CO 

o 

Ph 

u 



<N 


NO 


OO 


<N 


<N 


<N 


(N 


(N 


ON 


^t 


^r 


ON 


On 


On 


On 


On 



en 



O 

U 

En 
En 

< 

O 
O 

Eh 
O 

<3 
Eh 
O 
U 
Eh 
U 
O 



z 

5-H 

CO 

OO 



co 



oo 
oo 



<N 

o 



ON 



CO 
M2 



On 






CO 



r-> 


On 


r-> 


o 


^t 


^r 


^o 


m 


in 



u 

Eh 
Eh 
U 
U 
Eh 
En 
Eh 

< 
U 
U 
O 



U 



o 



u 

Eh 

o 

Eh 
Eh 
O 



Eh 
O 

o 

Eh 

<: 

En 
Eh 
U 
U 
O 
Eh 

<: 

EH 

< 

U 
Eh 
U 
U 
Eh 
U 
O 

<: 

EH 



CN 

Eh 
O 
O 
O 
Eh 
U 
O 
U 

Eh 
U 
O 
O 
O 
U 
U 

Eh 
U 
O 



CO 

u o 

U Eh 

U U 

O U 

H O 

U Eh 

O U 



CO 



ro ro ro ro 



Eh 
U 
Eh 
Eh 
O 
U 



Eh 
Eh 

<: 

o 

Eh 
Eh 
O 



U O 

Eh U 

O U 

U U 



O 
U 

<: 

En 
Eh 
U 
U 
Eh 
U 

u 

O 

Eh 
U 




Eh 
Eh 

u 

O 
O 

U 
U 
Eh 
U 
< 

u 
o 



o 

Eh 
O 
Eh 
O 
Eh 
O 
Eh 

< 
O 
Eh 
O 




PHrthpHh^h^PH^pHpHh^pH^pHpHhpHpH^pH 



I 

CO 
CO 



a 

o 

O 






I I 



O 

5-H 

O 



OD 



< 

in 

oo 



oo 



z 

p4 

oo 



z 

p4 

oo 



< 

z 

p4 

oo 



GO 

•i-H 
CO 

a 

H-> 

CD 

s? 

cd 

5-i 

o 

o 

U 



CO 

i-H 
CO 



o 
o 

N 
O 

•H 

15 

4^ 

Ph 

O 

O 



•i-H 

o 

p. 

CO 

O 

o 




CO 

CD 
CD 

4^ 

fl 
O 

O 

N 

O 

o 

5-H 

D 

H— > 



=5 
O 

•I-H 

O 

o 
o 

N 

o 

-t— ' 

•i-H 
15 

43 

P 

<D 

O 

W 



Z 
p^ 

5-1 

CO 

OO 



13 

43 

fl 
O 

o 

N 
O 



43 
P 

o 

S3 

W 



References 



145 



CO 



CO 

oo 



< 

in 

oo 



•i-H 

o 



Eh 
Eh 



oo 



<N 



Eh 

Eh Eh 
O < 



U 
Eh 



U O 

U O 

<: <c 

En U 

<: eh 

o u 

U Eh 

< o 

Eh O 

U Eh 

U O 

<; o 

u u 

En U 



p4 

C/3 



o 

bo 

S 

CO 

o 

x 

e2 



OO 






U 

Eh 
U 
O 
Eh 

< 
O 
<3 
O 
Eh 
U 
O 



• • •• 



& h p^; fe # h h 



PQ 



^r 



OS 



ir> 



o 

Eh 

< 
U 
Eh 
Eh 
O 
U 
U 
Eh 

< 
U 
O 
Eh 
U 



• • •• 



PQ 



o 

Eh 
U 

<: 
o 

EH 

o 
< 

u 

Eh 
O 
U 
En 
I 

U 
En 



b (^ b h 



CO 


CO 

S-H 


CD 


<D 


a 

• i-H 


a 


*H 


J-h 


Ph 


Ph 


t— -H 


^^ 


Ctf 


ctf 


G 


a 


I-H 


Vh 


<D 


a> 


H— > 


H— » 


0> 






u u 


PLh 


PLh 


T3 


X5 


<d 


a> 


^— > 


H— > 


CO 


CO 


<D 


<L> 


££ 



s 

o 



B 

*-H 

H— > 



X 

o 

?-H 

o 

I 

P4 



< ^2 

.9 xs 

co ,0 

o ^ 

2 «-> 
V S 

^SO 5-H 

co . . O 

^H 



X 



^ r- 



H Oh 

^h (N CO ^t ^ 



INDEX 



16SrDNA, 11-12 
Amplicons, non-specific, 28-29 
Commercial PCR 

Campylobacter, 63-64 

Escherichia coli, 0157:H7, 63-64 
pathogenic, 63-64 
Listeria monocytogenes, 63-64 

Salmonella, 63-64, 65 
Controls, 28, 32, 46, 59-60 
DNA databases, 1 1 

NCBI, 14-19 

searches, 14-19 
False-negatives, PCR inhibitors, 34-35 
False-positives 

live vs. dead cells, 33-34 

non-specific amplicons, 29 

primer-dimers, 29 

viable but nonculturable (VBNC), 34 
Interpretation of PCR results, 28-3 1 
Laboratory 

equipment, 57-58 

personnel, 55 

reagents and disposables, 58-59, 60 

set-up, 52-55 

standard operating procedures, 59-60 

standardization, 62 
Micro arrays, 12 
Multiplex PCR, 10, 70, 84 
Multiplex PCR, 70, 84 
Nucleic acid extraction 

parasites, 120-121 

viruses, 99-100 
Nucleic acid sequence-based amplification 

(NASBA), viruses, 102, 107 
parvum, 124-125 
PCR contamination, 52-55 
PCR design, 12-19 
PCR detection methods, 6-8 

agarose gels, 6 

ELISA, 6-8, 56 

fluorescence, 8-10, 56-57 
PCR detection of 

Bacillus cereus, 71 

Campylobacter, 63-64, 71-72, 85 

Clostridium perfringens, 72-73 

Cryptosporidium parvum, 123-124, 144 

Cyclospora cayetanensis, 126-127, 144 

Escherichia coli 0157:H7, 63-64, 85 



Escherichia coli, pathogenic, 63-64, 

73-76, 84-85 

Giardia intestinalis, 128, 145 

hepatitis A virus, 102, 105-106 

Listeria monocytogenes, 63-64, 76-77, 

85 

Micro sporidia, 131-132, 144 

noroviruses, 100-101, 103-104 

Salmonella, 63-64, 85, 77-79, 84-85 

Shigella, 79-80 

Staphylococcus aureus, 80-81 

Toxoplasma gondii, 130-131, 145 

Vibrio, pathogenic, 81-83, 85 

Yersinia enter ocolitica, 83, 85 
PCR inhibitors, 34-35, 46^7 
PCR theory, 2-4 
Quality control, 59-60 
Real-time PCR 

Campylobacter, 63-64, 85 

concept, 8-10 

Escherichia coli 0157:H7, 63-64, 85 
pathogenic, 63-64, 85 

false-positives, 10 

hepatitis A virus, 108-109 

Listeria monocytogenes, 63-64, 85 

noroviruses, 108-109 

Salmonella, 63-64, 85 

TaqMan, 8 

Vibrio, pathogenic, 85, 

Yersinia enter ocolitica, 85 
Reverse-transcriptase PCR, detection of 

bacteria, 33-34, 84-85 

viruses, 101-102 
Sample preparation, 41^49 

concentrating microbes, 47-49, 93-99, 

122-123, 

dairy, 43, 96-99 

enrichment, 47^9 

fruits and produce, 45, 96-99 

meat, 43^4 

seafood, 44-45, 94-96 
Sensitivity 

false-negatives, 31 

limit of detection, 34-35, 47^9, 70 

PCR inhibitors, 34-35, 46-47 

false-positives, 31 
Terminal Restriction Fragment Length 

Polymorphisms, 10-12 



147 



148 



Index 



Thermocycler 

conventional, 4, 57-58 
gradient, 5 

hot-air capillary, 4-5, 57-58 
real-time PCR, 5-6, 57-58 

Thermocyclers, 4-6, 57-58 

Trouble- shoo ting 

false-negatives, 34-35, 46-47 
false-positives, 32-33, 45 



real-time PCR, 10, 29 

sensitivity, 47-49 
Validation of PCR, 31, 60-62 
Viability 

bacteria, 33-34 

parasites, 133 
Virus, concentration, 93-99