Skip to main content

Full text of "كتب في علم الميكروبيولوجي"

See other formats





Itofs, A.Sc[)in 




H. HtrmM 



Vol. 12 






w 

H. 




sell 






KARGER 




Concepts m 8acteHal Virulence 



Contributions to 
Microbiology 



Vol. 1 Z 



Series Editors 

/Lve/ Schmidt iVuppenal 

Heiko Hef-wald Lund 



KAKGER 



Concepts in Bacterial 
Virulence 



Volume Editors 



.TT,:: 



Wayne Russell Lund 
Heiko Herwald Lund 



27 figures, I in color, and 7 tabies> 2005 



K AKG E IV 



Basel ■ Freiburg ■ Paris ■ London ■ New York ■ 
Bangalore ^ Bangkok ■ Singapore ■ 1 okyo ■ Sydney 



Contributions to Microbtofogy 

formerly 'Concepts in Immunopathology' and 
'Contribudons to Microbiology and immunology' 



Wayne Russeff, PhD Heiko Herwald, PhD 

Lnnd University Lund University 

Dept. of CelJ and Molecular Biology DepL of Cell and Molecular Biology 

Section for Clinical and Experimeutal Section for Clinical and Experimental 

Infectious Medicine Infectioiiii Medicine 

PUmBM PlanB]4 

Tomavageii 10 Tomavagen 10 

S-221 84 Lund (Sweden) S-221 84 Lund (Sweden) 



Library of Congress Catalogme-in-PublkaHon Daia 

Concepts in bacterial viriilcnce / volume editors, Wayne Russell, 

Heiko HenvalfL 
p. ; cm. -(Conlribulioni to mkrobtology^ ISSN 1420-9519 ; 
V. 12> 

Irtcludcx bibHogi-iiphical references and index. 

ISBNf 3.fi05J^77a6'9 fhard cover: alk. p^sper) 

I. Virulence (Microbiology) 2. Molecular microbiology. 3. Endo 
lo>:ins, I. Russell, W^yr^. \\. jlerwald, Heiko. IIL Series. 

IDNLM^ I. Virulence Fatlori'-physiology. 2, Bacicria-patho- 
gcnscity. 3, Communicable DisiMscs-microbiology 
Wl <J077SUE V.I2 2004 /QW 730 C744 2004] 
QR\75.Cf>b2WA 
6l5,9'52y3^c22 

20040 1 S270 



Bibiiofiraphic Indices. Ihis publication is Jistcd in bibliographic serviccis including Cuirenl Coxilcnls*^ and 
frtife^ Medic us. 

Drug Dosage. The aushors 3f>d (he publis^her have exerted every cffon lo ensure Ibar drug select ion and 
dbsage scl forih in Lhis t^xL are in sccord \v\ih currt^nL recommend alion^^ ;md pracuc? nl ^hc limi? of publication. 
I-lovvc\'cr. in view oron^ing i^tstsrch, ehH>ngcs in government regui^tions, H>nd ihc cons^lfint flow of in forma lion 
relating lo drug therapy and drug reacljons, I he reader is urged to check iHe package insen for each drug for 
any change in indicalioni^ artd dosage and for added warnings and precauiions. This is panicularly nnponani 
xvbcn I be recommen<k:d agent is a new and^or infrequently employed drug. 

All Tighis reserved. No part of thss publication may be jransJated into other language^ repnoduced or 
utilised in any form or by any means electronic or mechanica!, including photocopying, recording, microcopying, 
or by any infortnalion storage and reineva] system, wjiinoul permission in wiinj^ from the publisher 

fe Copynghi 2OT3 by S. Karger AG. RO. Box. CH^009 Ba^el (Switzerland) 

w>vw.kafXcr.coni 

Prinlcd in Switzerland on acid-free paper by Reinhardl Druck, Basel 

rSSN 1420-9519 

ISBN 3^055-7786-9 



v.4--.-----.-7A+^ + R + -+« 



Contents 



VII Foreword 

titfS5elUW,;Herwafd, K £Lund) 

Tccons 

Bishop^ R.E. fTurur^ttF) 

ZS Bacterial Ex-otvixins 

PopofF,M.R_ {Paris) 

55 Capsular Patysaccharides andThcJr Role inViriflcnce 

Taylor, C-M-, itol^-E3. I-S. (Mancliestfir) 



Adhesin 



& 



67 FifnbrJa=e+ Pilj| FI^Lgetla and Bacterial Virulence 

90 Gram-PoiltfveAdh^iljii 

Tnfny, S.R. [Braunschwuig) 

114 Microbiil Pathogenesh -ind Dconim Development 

Rckncr^ A. (Lyngby)" Hdiby, N. (CopniFragcn)' Tulkcr-Niclscn^T.; Molin, S. (Lyngbyl 

Enz/mc& 

112 B^ctenal Peptidn^es 

Putcrtipa, J. (Krakow/Athjens, G^-); Pike, R-N. fCEaj^l4mJ 



V 



131 Bacteria] Invasins: Molecular Systems Dedicate to the Invasion of 
Host Tissues 

Cainbrormep E.U; Sciineewind, O. (Chicago, IIJ,) 
Signaling and Gene R^gulatton 



no Bacterial Iron Transport Related to Virulence 
Bsaim, V (Tubingfsn) 

234 Pathogenicity Islands andTheir Role in Bacterial Virulence and Survival 
Hoclihgt. B.; Dobrindt, U; Hacker, J. (Wiiraburg) 

255 Horizontal and Vertk&l Gene Transfer: The Life History of Pathogens 

La;vrence, J.G. (Pittsburgh. Pa,) 

272 Subject Index 



Coniems 



VI 



Foreword 



With rtie current volume of the Karger book sefies Comnbutions to 
Microbiology, we attempt to summarize some of the most important virulence 
mechiuiisms in bacterial ijifectious diseases. In many cases the disease pathway 
begins with the invasfon of the host and ends with the outbreak of physiologi- 
cal responses that may lead to severe complications and ultimately death. Over 
the years it has been shown that the interplay between pathogenic bacteria and 
the host is complex and finely balanced. The ability of successful pathogens to 
survive in an imiinmologically hostile environment is provided by a large 
armamentarium of virulence mechanisms, which includes bacterial factors thai 
evade, neutralise or counter the host defense systems, but also manipulate hosi 
homeostasis and normal cell fijnctions. In order to give a comprehensive 
update, we were able to recruit some of the most eminent scientists in ijifectious 
diseases to give an overview of the most important recent findings in their 
fields. We hope that this volume provides a thought-provoking update on these 
important medical issues. 

Limd, May 2004 Wayne Russell 

Heiko Her^'aJd 



vii 



Toxins 

Russell W, Herwald H (eds): Concepts in Bacterial Virulence! 
Contrib Microbiol. Basel, Karger, 2005, vol 12, pp 1-27 



Fundamentals of Endotoxin 
Structure and Function 



Russell E. Bishop 

Departments of Laboratory Medicine and Pathobiology, and Biochemistry, 
University of Toronto, Toronto, Canada 



In 1892, flichard Pfeiffer first defined endotoxin as a heat-stable toxic sub- 
stance that was released upon disruption of microbial envelopes [1]. The toxic- 
ity is now known to be a consequence of the host inflammatory response, which 
appears to be optimally adapted for the clearance of most local infections. 
However, when severe infections become distributed systemically, the inflam- 
matory response can lead to septic shock and death. Most of the early efforts to 
determine the signal transduction events that occur between the presentation of 
endotoxin to the myeloid cells of the immune system and the production of 
inflammatory cytokines have utilized lipopolysaccharide (LPS) from gram- 
negative bacteria [2], The bioactive lipid A component of LPS is arguably the 
most potent of the substances that fit Pfeiffer's endotoxin definition, and lipid A 
has become synonymous with endotoxin. However, many other inflammatory 
mediators derived from bacteria can also be regarded as endotoxins, including 
peptidoglycan, the diacylglycerylcysteine moiety of bacterial lipoproteins, and 
bacterial nucleic acid signatures, to name only a few. The recent discovery that 
Toll-like receptor 4 (TLR4) is the lipid A inflammatory signal transducer has 
been folJowed by the identification of signal transducers for different Inflam- 
matory mediators [3, 4]. Coincident with these developments in endotoxin 
signaling has been the revelation that pathogenic gram-negative bacteria can 
modulate the structure of lipid A in order to evade detection by the host immune 
system. This article summarizes the recently elucidated pathways for the 
biosynthesis of lipid A in enteric bacteria, which provide a framework for 
understanding lipid A structure and fijnction in all gram-negative bacteria. 
Readers are referred to the recent review of Raetz and Whitfield [5] for a more 
complete treatment of LPS structure and function that accounts for its diversity 
in more divergent organisms. 



Overview of the Gram-Negative Cell Envelope 

The cell envelope of gram-negative bacteria (fig. 1) consists of the inner 
membrane (IM), the peptidoglycan (murein) and the outer membrane (OM) [5]. 
The IM is a phospholipid bilayer, much like the plasma membrane of eukaryotic 
cells, and is permeable to lipophilic compounds. Numerous integral trans- 
membrane a-helical proteins and peripheral membrane proteins are primarily 
responsible for transport, cell signaling and metabolic fijnctions [6]. The IM pro- 
vides a topologically closed environment for the vectorial translocation of ions to 
generate a transmembrane electrochemical potential or proton-motive force that 
governs cellular energetics. Proteins synthesized with a cleavable amino -terminal 
signal peptide can be targeted for export across the TM [7]. The periplasm is the 
gelatinous material between the OM and the IM. It contains enzymes for nutrient 
breakdown as well as binding proteins to facilitate the transfer of nutrients across 
the IM. Additionally, the murein sacculus in the periplasmic space is composed 
of alternating N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid 
(MurNAc) sugars that are cross-linked by short peptide bridges [8]. The highly 
reticulated murein layer plays a crucial role in maintaining the cell's characteris- 
tic shape and in countering the effects of osmotic pressure. The murein is bridged 
to the OM by the abundant covalently bound murein lipoprotein, while numerous 
low-abundance non-covalently-bound lipoproteins are anchored to the inner 
leaflet of the OM and a few are anchored to the outer leaflet of the IM. 

The OM is unique to gram-negative bacteria, and its role is to serve as a 
protective structure. Tlie lipid arrangements of the OM are highly asymmetric. 
While phospholipids [70-80% phosphatidylethanolamine (PtdEtn), 20-30% 
phosphatidylglycerol (PtdGro) and cardiolipin] occupy the inner leaflet, LPS 
molecules pack against one another in a tight architecture in the outer leaflet of 
the OM [9]. Due to the low fluidity of lipid A hydrocarbon chains and the strong 
lateral interactions between LPS molecules, the OM bilayer is impermeable to 
lipophilic compounds and, thus, serves as an important permeability barrier for 
gram-negative bacteria [10]. To allow uptake of essential nutrients, the OM is 
studded with trimeric |3-barrel proteins, known as porins, which allow diffusion 
of solutes with a molecular weight below approximately 600 daltons. Additional 
P-barrel proteins in the OM are adapted for the uptake of particular nutrients that 
carmot gain access through porins, and a few OM |3-barrel proteins function as 
enzymes [11]. One consequence of porins is that the OM is believed to lack any 
transmembrane electrochemical potential. 

LPS is composed of three parts: the proximal, hydrophobic lipid A region, 
which anchors LPS to the outer leaflet of the OM, the distal, hydrophilic 
0-antigen repeats, which extend into the aqueous medium, and the mterconnect- 
ing core oligosaccharide (fig. 2). The 0-antigen and core sugars are not essential 



Bishop 



LPS 



GPL 




Transmembrane 
p-barrel protein 




/ 



/ 



Lipoprotein 
Murein 





Prolon 
motive 
force 




Transmembrane a-hellcal protein 



Outer 

membrane 



Periplasmic 
space 



Inner 
membrane 



Fig, L Molecular organization of the gram-negative cell envelope. The OM is an 
asymmetric bilayer with an outer leaflet of LPS and an mner leaflet of glycerophospholipids 
(GPL). The integral OM proteins are exclusively transmembrane (3-barrels. Lipoproteins 
anchored to the OM inner leaflet can link the OM to the murein exoskeleton. The energy- 
transducing IM is a phospholipid bilayer that supports the proton motive force and contains 
transmembrane ct-helical proteins. The periplasmic space is the region between the IM and 
OM and contains numerous globular proteins. 



for survival, but they provide bacterial resistance against various antimicrobial 
agents including detergents and the membrane attack complex of serum comple- 
ment [12], Wild-type cells that produce 0-antigen are termed 'smooth' due to 
their glossy colony morphology, while those that lack 0-antigen are termed 
'rough'. The term LPS formally applies only to the molecule that contains the 



Endotoxin Structure and Function 



3 




Hex 



Hex 



Hex 



O-antigen 



Outer core 








EtN-P-P-/ Hep 



EtN-P' 



Kdo 



/ 



Kdo 



P^GIcN) ( GlcNKP 



Inner core 



Lipid A 



Bishop 



0-antigen polysaccharide, while molecules that lack 0-antigen, as in the case of 
Neisseria, are more appropriately termed lipooligosaccharide or LOS, Lipid A is 
a target for the development of antibiotics and anti-inflammatory agents because 
it is both essential for survival and a potent inflammatory mediator 



TLR Signaling 

When LPS is shed from the bacterial surface durmg infection, lipid A 
recognition in mammalian cells is mediated by the TLR4 signal transduction 
pathway [13, 14]. LPS is first recognized by the circulating acute phase LPS- 
binding protein (LBP), which then interacts with the glycosylphosphatidylinositol- 
anchored CD 14 on the surface of myeloid cells. Subsequent interaction with 
TLR4 and its associated factor MD2 initiates a cascade of signahng pathways 
that, in turn, elicit the production of cationic antimicrobial peptides (CAMPs), 
a variety of cytokine and chemokine molecules, and the costimulatory mole- 
cules that are expressed on the surface of antigen-presenting cells and further 
signal the presence of an infection to the cells of the adaptive immune system 
[15]. Upon activation, TLR4 recruits to its intracellular Toll-interieukin recep- 
tor homology region (TIR), the adapter protein MyD88, which associates by a 
homotypic protein-protein interaction with its own TJR domain (fig. 3). Another 
homotypic protein-protein interaction between the death domains of MyD88 
and the mterieukin-l receptor-associated kinase lRAK-1 initiates the autophos- 
phorylation of lRAK-1, which then associates with a signal transduction way 
station known as tumor necrosis factor-a (TNF-ct) receptor-associated factor-6 
(TRAF-6). An ubiquitin-conjugating enzyme complex is bound to TRAF-6 
along with the TAK-1 kinase complex, which is anchored by the TAB adapter 
proteins [3]. The pathway impinges on the master regulator of inflammation 
known as nuclear factor kB (NFkB), which activates transcription of inflam- 
matory response genes. However, NFkB is normally sequestered in the cyto- 
plasm in complex with its inhibitory subunit IkB. Proteolytic degradation of 
IkB enables NFkB to migrate inlo the nucleus and activate inllammatory gene 



Fig. Z Structural organization of LPS, The most highly conserved region of the LPS 
molecule is the lipid A domain, which is an acylated and phosphorylated disaccharide of 
glucosamine. Assembly of lipid A is contingent upon the addition of the two 8-carbon Kdo 
sugars, which are the only essential components of the inner core. The inner core normally 
includes three 7-carbon Hep sugars and can be modified by the addition of phosphate and 
pEtN substituents. Outer core sugars provide the acceptor for 0-antigen ligation, but tend to 
be composed of hexose sugars that differ between species. The 0-antigens represent the most 
highly species-variable component of the LPS molecule. 



Endotoxin Structure and Function 



TLR4 



LPS/LBP 



MD2jp» 




MyD88 

IRAK-1 
TRAF-6 

Ubc13 _4.TAK-1 



IKK y 



i 
fi 



a 



Degradation 

t 




NFkB 



TNF-a; IL-13; costimulatory molecules 
Cationic antimicrobial peptides 





'M 



NFkB 



Fig. 3. TLR4 signal transduction pathway. LPS released from the surface of gram- 
negative bacteria is bound to the circulating LPS-binding protein (LBP) and delivered to the 
glycosylphosphatidyhnositol-anchored CD 14 on the surface of myeloid cells. The leucine- 
rich repeats (LRR) of CD14 are also shared with the extracellular domain of TLR4, which, 
in association with MD2, can transduce a signal to its intracelJular 77/?. TIR-TIR interactions 
with the adapter protein MyD88 promote mteractions between the death domains (DD) of 
MyD88 and the interleukin-1 receptor-associated kinase IRAK-l. Autophosphorylation of 
lRAK-1 promotes an association with the TNF-a receptor-associated factor TRAF-6, which 
anchors both the kinase TAK-1, by its TAB adapter proteins, and the dimeric ubiquitin- 
conjugating enzyme complex composed of UevlA and Ubcl3. Subsequent phosphorylation 
events activate the trimeric IkB kinase complex IKK, which phosphorylates the NFkB 
inhibitory subunit IkB and targets it for proteolytic degradation. The master regulator of 
inflammatory response gene expression NFkB is then released and migrates into the nucleus 
where inflammatory response genes are transcriptionally activated. 



expression. IkB is targeted for proteolysis upon phosphorylation catalyzed by 
the IkB kinase complex (IKK), which is itself phosphorylated by the TAK-1 
kinase in a manner that depends on the ubiquitin-conjugating enzyme complex 
in association with TRAF-6. TAK-1 also phosphorylates mitogen-activated 



Bishop 



protein kinases that impinge on the AP-I transcription family members Jun and 
Fos, leading to further immune activation. 

The response to LPS includes locaJ inflammation, which is highly beneficial 
in providing antibacterial defenses. If infection persists, however, the subse- 
quent systemic responses, including the overwhelming production ofTNF-a and 
interleukin-lp by the host immune system, can lead to septic shock [16]. Efforts 
to understand the lipid A signal transduction pathway were largely motivated 
by a desire to develop endotoxin antagonists for the treatment of septic shock. 
The discovery that some bacteria can evade host immune defenses by modifying 
the structure of lipid A suggested that naturally occurring lipid A structures 
may function as potent endotoxin antagonists. The microbial pathways for the 
biosynthesis of lipid A and its derivatives have been elucidated recently and pro- 
vide powerful tools for the investigation of endotoxin signaling, in addition to 
illustrating the pathogenic mechanisms utilized by gram-negative bacteria. 



Re Endotoxin Biosynthesis 

The recent completion of the Raetz pathway for lipid A biosynthesis [5] 
hinged on the serendipitous discovery of lipids X and Y in a conditional 
PtdGro-deficient pg.?^ mutant o^ Escherichia coli in 1979 [17]. Lipid X was 
subsequently shown to be a diacylglucosamine i-phosphate bearing R-3- 
hydroxymyristoyl (3-OH-14:0) groups at positions 2 and 3, while lipid Y only 
differed from lipid X by the presence of a palmitoyl (16:0) group in acyloxy- 
acyl linkage at position 2 [18, 19]. Around the same time, the determination of 
the correct chemical structure of lipid A [20] revealed possible biosynthetic 
routes for the production and utilization of Mpids X and Y. The accumulation of 
these glucosamine-based phospholipids in the PtdGro-deficient mutant proved 
to be a consequence of a second unlinked conditional mutation in the gene 
pgsB(lpxB) [21, 22], which was later shown to encode the lipid A disaccharide 
synthase. LpxB generates the P-1 ',6-glycosidic bond that is a characteristic fea- 
ture of lipid A [23]. Both lipids X and Y couJd activate macrophages in a simi- 
lar manner as lipid A [24], but only lipid X proved to be a substrate for LpxB 
[23], raising doubts about the physiological significance of lipid Y. Lipid A 
biosynthesis is now known to occur in four separate cellular compartments, 
namely, the cytoplasm, the cytoplasmic face of the IM, the periplasmic face of 
the IM, and ui the OM (where the origin of lipid Y was recently found). 
Conceptually, it is helpful to recognize that lipid A and the core oligosaccharide 
are assembled together as a single unit starting in the cytoplasm and moving to 
the cytoplasmic face of the IM, but the subsequent lipid A modifications and 
en bloc ligation of O-antigen occur in the extracellular compartments. 



Endotoxin Structure and Function 7 



OH 



^fi 



O 



.OH 
Q=i UDP 




!pxA 



UDP-GlcNAc 




o 




Nl 



O 



^ 



UDP 



Acetate 



In 



2+ 



IpxC (envA) 




UDP 
3-OH-C1.-ACP 



OH 

0=^ nhI 

HOl< O 
HO 

(2X) 



IpxD {firA) 



HOli'^OH 

o 



HO 
HO 



9ho \ 




^^'^^ 



o 



"03PO 




o^ nh| 

HO 

/pxM /pxL 

(msbS) {htrB) 



Ci4-ACPCt2-ACP 



KdOj-lipid A 
(Re endotoxin) 



O 

o=<. nhI 

HOK 0=^ OPO3' 
HO I 



UDP 



OH 

o=<, nh] 

0< 0P03= 



HO 



UMP 



HOI 



/pxH 



fpxB 




UDP 



Lipid X 




OH 

o< o^ nhI 

HO I 




"O3PO 




/(dtA (waa>A) 
;2x)CMP-Kdo 



ATP 

0=^ nhI 

HO< ^^ OP^S^ 
HOI 



Lipid IV^ 



F/^. 4, The Raetz pathway for synthesis of Kd02-iipid A, LpxA catalyzes the addition 
of 3-OH-14:0 to position 3 of UDP-GlcNAc. LpxC then removes the acetamido group at 
position 2, which allows LpxD to add a second 3-OH-]4:0 group. LpxH cleaves the 
nucleotide to generate lipid X, which is condensed with UDP-diacyl-GlcN to generate the 
disaccharide 1 -phosphate. The 4'-kiiiase LpxK then generates lipid IV^, which is converted 
into Kdo2-lipid TV^ by a bifiinctional Kdo transferase KdtA. Kdo2-lipid IV^ is a substrate for 
the LpxL and LpxM acyltransferases, which generate the acyloxyacyl linkages at positions 
2' and 3', respectively. 



The moJecular genetics and enzymology of the conserved steps of Jipid A 
biosynthesis are best characterized in E. coli, as shown in figure 4. The Raetz 
pathway begins with the key precursor molecule UDP-GJcNAc, which is also the 
first substrate for peptidoglycan biosynthesis. The first enzyme in lipid A 
biosynthesis is a cytoplasmic acyltransferase LpxA, which selectively transfers 
thiol ester-activated 3-OH-14:0 from acyl carrier protein (ACP) to the 3-OH of 
UDP-GlcNAc [25]. The crystal structure of LpxA revealed a homotrimeric 



Bishop 



8 



molecule that self-associates by a distinctive left-handed parallel p-helix 
motif [26]. E, coli LpxA is extraordinarily selective for 3-OH-14:0-ACP as 
the acyl donor substrate while the Pseudomonas aeruginosa LpxA prefers 
3-OH-10:0-ACP, However, the specificity could be modulated by mutating cer- 
tain key residues lining the active site cleft [27]. For example, the specificity for 
the G173M mutant oi E. coli LpxA was shifted to 3-OH-10:0-ACP. In contrast^ 
the specificity of P. aeruginosa LpxA could be extended to accommodate 
3-OH-l4;0-ACP in the corresponding M169G mutant. These findings demon- 
strated the existence of precise hydrocarbon rulers in LpxAs, which can explain 
variations in lipid A acylation that are observed between different organisms. 

The acylation of UDP-GlcNAc by LpxA is thermodynamically unfavorable 
[25], and the first committed step in lipid A biosynthesis is the subsequent 
deacetylation catalyzed by LpxC (EnvA) [28, 29]. LpxC is a Zn^^-dependent 
enzyme that is an established target for antibiotic development [30, 31]. The 
recent crystal and NMR structures of Aquifex LpxC revealed tu^o slightly differ- 
ent models for the mechanism of catalysis [32, 33], but both include a critical 
role for Zn^^. Most LpxC inhibitors are hydroxamate compounds that interact 
with the catalytic Zn^^ ion. Current challenges are aimed at the development of 
inhibitors with the ability to evade efflux pumps that provide resistance, particu- 
larly in pseudomonads [34, 35]. 

Following deacetylation, an N-linked 3-OH-14:0 moiety is incorporated 
from ACP by LpxD (FirA) to generate UDP-2,3-diacylglucosamine [36]. A 
highly selective pyrophosphatase LpxH then cleaves UDP-2,3-diacylglucosamme 
to form lipid X [37, 38]. Next the disaccharide synthase, LpxB, condenses 
UDP-2,3-diacylglucosamine and lipid X to generate the |3-T,6-linkage found 
in all lipid A molecules [23]. The membrane-bound 4' kinase LpxK then phos- 
phorylates the disaccharide 1 -phosphate to produce lipid IV^ [39, 40], which is 
an important pharmacological agent because it functions as an endotoxin anta- 
gonist in human cell lines [41, 42]. Next, two 3-deoxy-Z)-manno-2-octulosonic 
acid (Kdo) sugars are incorporated by a Kdo transferase, which is encoded by 
the kdtA (waaA) gene, using the labile nucleotide CMP-Kdo as the Kdo donor 
[43]. The final lipid A biosynthetic steps that occur on the cytoplasmic side 
of the IM depend on the prior addition of the Kdo sugars and involve the trans- 
fer of lauroyl (12:0) and myristoyl (14:0) groups from ACP to the distal glucos- 
amine unit to produce acyloxyacyl Linkages; the reactions are catalyzed at the 
2'-position by LpxL (HtrB) and at the 3'-position by LpxM (MsbB), respect- 
ively [44-46]. Under conditions of cold growth at 12''C, LpxL is replaced by 
LpxP, which has a preference for palmitoleate (16:lcisA^) in ACP [47, 48]. The 
incorporation of an unsaturated acyl chain into lipid A likely increases mem- 
brane fluidity under cold growth conditions. Viable mutants that lack acyloxy- 
acyl linkages in lipid A are attenuated for virulence and reveal the importance 



Endotoxin Structure and Function 



of the lipid A acylation pattern in inflammation [49, 50]. All other enzymatic 
steps of the Raetz pathway, and those for the biosynthesis of CMP-Kdo, are 
essential for cell viability and, thus, provide potential targets for antibiotic 
development 



Assembly of LPS 

Kd02-lipid A, also known as Re endotoxin, can be regarded as the simplest 
chemotypeof LPS [5], Completion of thecore-Kdo2-lipid A molecule involves the 
subsequent addition of core sugars to the nascent Kdo2-lipid A anchored on the 
cytoplasmic side of the IM [51]. The two essential 8-carbon Kdo sugars are 
regarded as pait of the inner core, which is normally extended to include three 
7-carbon Z.-glycero-Z)-manno-heptose (Hep) sugars (fig. 2). Core oligosaccharide 
synthesis is contingent upon modification with phosphate at position 4 of the first 
Hep, which can be followed by the addition of phosphoethanolamine (pEtN) at the 
same position. Phosphate also normally occurs at position 4 of the second Hep, 
and pEtN modification at position 7 of the second Kdo can occur under Ca^^-rich 
growth conditions [52]. The so-called 'deep-rough' mutants have defects in the 
inner core heptose sugars and are sensitive to detergents and hydrophobic anti- 
biotics [53], The outer core sugars are predominantly hexoses and exhibit a greater 
degree of structural diversity than is seen in the inner core and lipid A regions [54]. 
The outer core sugars provide the acceptor residue for 0-antigen ligation. 

The 0-antigen is synthesized and anchored to a carrier lipid, undecaprenyl 
phosphate, in the IM. The remarkable diversity in 0-antigen structures reflects the 
multitude of glycosyl transferases that utilize various sugars and create diverse 
glycosidic linkages, combined with the occasional presence of substoichiometric 
sugar modifications [5, 55]. However, biosynthesis of all 0-antigens is initiated 
by the formation of a common diphosphate linkage between the first sugar and 
undecaprenyl phosphate. 0-antigen units are then completed in the cytoplasm 
and transported to the periplasmic face of the IM by one of three distinct pathways 
termed Wzy-dependent, ATP binding cassette (ABC) transporter-dependent, and 
synthase-dependent. The most common of these is the Wzy-dependent pathway, 
which is characteristic of £". coli and is followed by polymerization of 0-antigen 
units on the periplasmic face of the IM. Recent studies have implicated an 
essential ABC transporter MsbA in translocating the core-Kdo2-lipid A mole- 
cule to the periplasmic side of the IM [56-60]. Core-Kdo2-lipid A and poly- 
merized 0-antigens from the various pathways are then linked together by a 
common ligation mechanism at the periplasmic surface of the IM. 

The completed LPS is transported for assembly in the OM by a poorly under- 
stood process. Interestingly, certain integral membrane proteins can passively 



Bishop 



10 



promote the translocation of phospholipids across the IM [61], but MsbA is 
required for the transport of both LPS and phospholipids to the OM [56, 57], 
Jt has been known for more than 25 years that phospholipids freely exchange 
between the IM and OM, while LPS transport appears to be unidirectional [62, 
63], The mechanism by which LPS is translocated to the outer leaflet of the OM 
is unknown, but it may depend on the highly conserved OM protein OMP85 
[64], which is also implicated in the assembly of OM proteins [65]. 



The OM Permeability Barrier 

LPS contains phosphate and acidic sugars and is therefore negatively 
charged. In order to reduce the electrostatic repulsion between LPS molecules at 
the cell surface, the bacterial OM sequesters divalent cations, mainly Mg^^ [66, 
67], which neutralize the negative charges and maintain the integrity of the OM. 
The presence of hydrogen-bond donors and acceptors in the lipid A molecule 
allows for additional lateral interactions that cannot occur between phospholipid 
molecules [67], Moreover, the six or seven saturated acyl chaijis of lipid A serve 
to reduce the fluidity of the OM bilayer compared with the IM. The tight lateral 
interactions between LPS, combined with low membrane fluidity, provides a 
permeability barrier in the OM to lipophilic solutes and detergents [10]. 



Mechanism of Action of CAMPs 

The requirement for Mg^"^ ions to bridge LPS molecules at the cell surface 
is an Achilles' heel for the OM. Numerous CAMPs are produced in nature, but 
the main types produced by the immune system are the small a-helical proteolytic 
digestion products that are released from precursors known as the cathelicidins, 
and the disulfide-bonded p-sheet peptides known as the defensins. CAMPs can 
navigate through the OM by a nonporin pathway termed the 'self-promoted 
uptake pathway' [68]. They are ijiitially unstiojctured in aqueous medium, and 
their initial electrostatic interactions with the bacterial surface serves to displace 
some Mg^"^ ions. The reduced dielectric constant at the membrane interface 
induces dehydration of peptide bonds that become hydrogen-bonded in ct-helical 
or p-sheet structures. The induced structure reveals the amphipathic nature of 
CAMPs, which may promote changes in phase and/or motion in the OM bilayer 
and, in turn, facilitates their translocation through the hydrocarbon layer, TTiese 
peptides are then thought to target the IM bilayer and to produce a detergent-like 
disruption of permeability. Some possible consequences of IM permeation 
include the fatal depolarization of the transmembrane potential across the IM, 



Endotoxin Structure and Function 1 1 



leakage of cytoplasmic contents, cell lysis and cell death. The actions of CAIVfPs 
are thought to selectively target bacterial membranes [69]. The outer leaflet of the 
bacterial IM is negatively charged because it contains anionic phospholipids, 
whereas eukaryotes tend to sequester anionic lipids internally. Moreover, choles- 
terol molecules, which are embedded only in the eukaryotic plasma membrane, 
could stabilize the lipid bilayer and, thus, reduce the activity of CAMPs. 



Lipid A Modifications 

Considering the importance of Mg^^ in maintaining the OM permeability 
barrier, it is not surprising that Mg^"^ limitation can regulate the covalent structure 
of lipid A. Mg^^ limitation is also believed to signal the presence of an intra- 
cellular environment [70]. For example, in the phagocytic vacuoles of macro- 
phages, the natural resistance-associated macrophage protein 1 (Nrampl) serves 
to pump divalent cations into the cytosol, thereby withholding Mg-^^ required for 
bacterial growth [71], Figure 5 outlines several covalentmodificationsof lipid A 
found under Mg-^^- limited growth conditions that have been characterized in 
E. coli and Salmonella enterica [72 75]. Three enzymes function to modify the 
acylation pattern of lipid A. LpxO is a hydroxylase that generates 5'-2-hydroxy- 
myristate (2-OH-14:0) at position 3' [76]. PagP is a transacylase that incorpo- 
rates a palmitate chain at position 2 [77], while PagL is a deacylase that removes 
the 0-linked 3-OH-14:0 chain at position 3 [78]. Moreover, the phosphate 
groups at positions 1 and 4' of the lipid A disaccharide backbone can be modi- 
fied with 4-amino-4-deoxy-L-arabinose (L-Ara4N) and/or pEtN, which serve to 
reduce the overall negative charge of lipid A [79, 80]. 



Roles in Counteracting CAMPs 

Lipid A modifications provide a dual protective mechanism against 
CAMPs. First, substituting the phosphate groups on lipid A with L-Ara4N and 
pEtN could effectively weaken the electrostatic attraction between the nega- 
tively charged cell surface and CAMPs. In fact, the resultant neutralization 
of the negatively charged bacterial surface is associated with resistance to 
polymyxin B, a lipid A-binding cationic cyclic peptide antibiotic, in E. coli and 
S. enterica [81, 82]. Moreover, lipid A acylation may block the hydrophobic 
interaction between CAMPs and the membrane bilayer. Lipid A pahnitoylation 
by PagP has been shown to provide bacterial resistance against CAMPs [83]. 
Possibly, the resultant hepta-acylated lipid A could further reduce OM fluidity 
and, thus, prevent CAMP insertion. The pattern of lipid A acylation is also 



Bishop 



12 



L-Ara4N 

(pmrE/ 
pmrHFIJKL) 




S-2-0H 
(IpxO) 



pEtN 
(pmrC) 



G-O-Deacylation 



PalmitatefpagPj 



Fig. 5. Regulated covalent lipid A modifications. The conserved lipid A nucleus can be 
modified by the addition of L-Ara4N and pEtN to the phosphate groups, by the 5'-2-hydroxy- 
lation of the secondary myristoyl group at position 3\ by the removal of the 3-OH- 14:0 group 
at position 3, and by the addition of a pahnitate chain at position 2. Modifications to the acy- 
lation of Hpid A are under the direct control ofPhoP/PhoQ, while the phosphate modifications 
are controlled indirectly by PhoP/PhoQ through the downstream effectors PmrA/PmrB. 



known to be critical in mediating its endotoxic activity through interactions 
with the TLR4 signal transduction pathway [50, 84], Hepta-acylated lipid A 
bearing a palmitate chain can function as an endotoxin antagonist, which blocks 
the inflammatory effects of the hexa-acylated lipid A in human cell lines [85, 
86]. Consequently, modifications to the acylation pattern of lipid A may^ 
remarkably, block both direct interactions between CAMPs and the bacterial 
cell, and the induction of CAMP synthesis in the eukaryotic host. The enzymes 
responsible for S-2-hydroxylation and 3-0-deacylation are absent from E. coli 
and then- roles are less clear, but they may serve to stabilize lateraJ LPS inter- 
actions by introducing new hydrogen-bond donors [67]. 



The PhoP/PhoQ and PmrA/PmrB Two-Component 
Regulatory Systenns 

Gram-negative bacteria use the PhoP/PhoQ two-component signal transduc- 
tion pathway to respond to Mg^"^-limited environments that can be encountered 
during infection [87]. PhoQ is a membrane-bound sensor kinase that detects 



Endotoxin Structure and Function 



13 



Mg^"^ and can phosphorylate and activate the transcriptional regulatory protein 
PhoP [88]. Mutants altered in the PhoP/PhoQ system display greatly reduced 
virulence. PhoP controls the expression of over 40 different genes, many of 
which are involved in Mg^"^ transport and in lipid A modification. For example, 
transcription of pagP, pagL and IpxO, which are involved in the modification 
of lipid A acyl chains, are under the direct influence of PhoP/PhoQ [76-78]. 

The PmrA/PmrB two-component regulatory system is one of the down- 
stream effectors of the PhoP/PhoQ system, and is required for the modification 
of lipid A with pEtN and L-Ara4N [80], PmrA is the transcriptional response 
regulator and PmrB is the membrane-bound sensor kinase. While PmrA can be 
activated by PhoP/PhoQ via a mediating protein PmrD [89], the PmrA-induced 
genes can also be activated independently of PhoP/PhoQ by exposure of PmrB 
to Fe^"^ or mild acidic conditions [90]. PmrA/PmrB activation has also been 
shown to repress PmrD expression [91], which thereby creates a negative feed- 
back loop. Interestingly, CAMPs themselves have been reported to activate 
PhoP/PhoQ in Salmonella and PmrA/PmrB in Pseudomonas [92, 93], 



L-Ara4N Cluster 

PmrA/PmrB is onJy one of several clusters of pmr genes that were origi- 
nally identified in polymyxin-resistant mutants of £^. coli [94, 80]. The pmrF 
(pbgP) locus encodes an operon of 7 open reading frames pmrHFIJKLM, of 
which the first 6, together with the unlinked pmrE (ugd), are required for 
L-Ara4N synthesis. The proposed biosynthesis and attachment of L-Ara4N to 
lipid A is shown in figure 6. The first step involves the conversion of UDP- 
glucose into UDP-glucuronic acid catalyzed by a dehydrogenase encoded by 
pmrE. Complex regulation of dehydrogenase gene expression reflects the fact 
that UDP-glucuronic acid is a precursor for both colanic acid-containing cap- 
sules and L-Ara4N [95]. Next, Pmrl (ArnA) catalyzes the oxidative decarboxy- 
lation of UDP-glucuronic acid to generate a novel UDP-4-keto-pyranose 
iiitennediate [96]. PmrH (AniB) then catalyzes a transamination reaction using 
glutamate as the amine donor to generate UDP-L-Ara4N [97]. The crystal 
structure of PmrH has verified that a pyridoxal phosphate cofactor contributes 
to the catalytic mechanism [98]. Interestingly, Pmrl contains a second domain 
that formylates the 4-amine of UDP-L-Ara4N. The resultant UDP-L-Ara4- 
formyl-N is transferred by PmrF (AmC) to the membrane-anchored unde- 
caprenyl phosphate, forming undecaprenyl phosphate-Z.-Ara4-formyl-N [97]. 
The formylation step may drive forward the equilibrium of the transamination 
step, which is thermodynamically unfavorable. Formylation may also facilitate 
translocation across the IM by neutralizing positive charge. It is speculated that 



Bishop 



14 



UDP-glucose 

.OH 



UDP-/.-Ara4N 



o 



.T^o 2NAD+ Ho^V^'^v NAD'" CO. ^^ 



UDP UCP 

Colanic acid capsule 




Q Glutamate 



NH, N-10-formyl ^-^ 



OHi 



™^^ 



UDP 

a-Ketoglutarate 
arnB (pmrH) 



THF HO 

UCP 

am^ (pmr/J 



'^ 



UDP 



P-O 



f^),>' 



arnC (pmrF) 
UDP 








NH 



J/ 



o 



HO- 




PO 



arnT (pmrK) 



o-p-o- Lipid A core ^'> 

>'^ OH 




Transport? 




n 



\^ 



P-of-^) 



Deformylation? 



1 1 



Fig. 6. Pathway for attachment of L-Ara4N to lipid A. The Ugd dehydrogenase con- 
verts UDP-glucose into UDP-glucuronic acid, which is a precursor for both colanic acid cap- 
sular polysaccharides and L-Ara4N. The first committed step of L-Ara4N biosynthesis is the 
AmA-catalyzed oxidative decarboxylation, which generates a novel UDP-4-keto-pyranose 
mtermediate.Transainination catalyzed by ArnB is followed by formylation due to a second 
catalytic domaiji in AmA. Transfer of the fonnylated monosaccharide to undecaprenyl phos- 
phate by ArnC is presumably followed by translocation to the periplasmic side of the IM for 
deformylation. Undecaprenyl phosphate Z.-Ara4N is the substrate for ArnT, which transfers 
L-Ara4N to the lipid A acceptor. 



a putative transporter may be specific for the formylated compound and that 
deformylation may then occur at the periplasmic surface [97], These steps 
would ensure the vectorial translocation of the lipid across the IM and avoid 
futile cycling. The necessity of the deformylation step is dictated by the fact that 
undecaprenyl phosphate-L-Ara4N is the substrate for PmrK (ArnT), which cata- 
lyzes the final transfer of L-Ara4N to lipid A at the peripJasmic surface of the 
IM [99, 100]. Roles for the remaining /?mr genes in the transport and periplas- 
mic deformylation reactions are suspected, but remain to be established. 



EptA 



The putative pEtN adding enzyme EptA has recently been cloned from 
E, coll [101], and a homologous gene from Neisseria has been associated with 
the addition of pEtN to lipid A [1 02], The EptA-encoding gene is the upstream 



Endotoxin Structure and Function 



15 



-OCcte 



OH 



'■lO^O 



o- 



o 



eplA (pmrC) 



,OC<He 




o o- 



NH3 




iNH. 



'1 10.^0 



0= 



o 



Diacylglycerol 



V 



OH 






.OCcre 



pagP 



-\- 



PtdEtn 



sn-1-lyso-RdEln 




Fig. 7. Modification of lipid A with pEtN and palmitate. EptA at the periplasmic side 
of the IM uses PtdEtn as the pEtN donor to generate diacylglycerol and pEtN-modified lipid 
A, PagP also uses PtdEtn (or another glycerophospholipid) as the palmitoyl donor in the OM 
to generate sn-1-lyso-PtdEtn and hpid A modified by the addition of a pahnitoyi group. 



open reading frame that is part of the pmrAB operon, and is also known as 
pmrC (pagB) [103, 104]. PtdEtn is the reported pEtN donor (fig. 7) and several 
EptA homoJogues are likely responsible for pEtN addition to other cell enve- 
lope components including the inner core sugars of LPS. It is noteworthy that 
roughly one third of E, coli lipid A carries a diphosphate moiety instead of the 
monophosphate at position 1 [56], and that the putative phosphorylating 
enzyme shares with EptA the ability to generate a phosphodiester bond at the 
same position in lipid A. 



PagP 



PagP is encoded by a PhoP/PhoQ-activated gene and functions to transfer 
a palmitate chain from a phospholipid to the hydroxyl group of the N-linked 
3-OH-14:0 chain on the proximal glucosamine unit of lipid A [77, 83]. PagP 
was the first enzyme of lipid A biosynthesis shown to be localized in the OM 



Bishop 



16 



[77]. Since thiolester-containing substrates are not available in the extracellular 
compartments, PagP uses a phospholipid as the palmitoyl donor instead (fig. 7). 
PagP appears to be responsible for the production of lipid Y as a side reaction 
in IpxB mutants. It was first identified in the salmonellae due to its role in pro- 
viding resistance to CAMPs [83], and was subsequently purified from E. coll 
[77]. In addition to these enteric pathogens, PagP homologues are present in the 
respiratory pathogens Legionella pneumophila and Bordetella bronchlseptlca, 
where PagP has been shown to be necessary for disease causation in animal 
models of infection [105, 106]. In B. bronchlseptlca, PagP is controlled by a 
different two-component virulence signal transduction pathway known as 
BvgA/BvgS, and palmitoylation occurs at the 0-linked 3-OH-14:0 chain on the 
distal glucosamine sugar [106]. PagP homologues are also found in Yersinia, 
Photorhahdus and Erwlnia species, which adopt pathogenic lifestyles in ani- 
mals, insects, and plants, respectively. Current efforts to understand the struc- 
ture and function of PagP are aimed at developing a treatment for infections 
caused by this important group of pathogens. The structure and dynamics of 
PagP in detergent micelles have been determined by both NMR spectroscopy 
[107] and X-ray crystallography [Bishop and Prive, unpubl. data]. 

PagP is an 8-stranded antiparallel (3-barrel preceded by an N-terminal 
amphipathic a-helix. The ^-barrel is well defined in the structure while the 
extracellular loops are not. Unlike other |3-barrel membrane proteins, proline 
residues at two sites between (3-strands disrupt the continuity of hydrogen 
bonding in the outer leaflet half of the PagP p-barrel. These non-hydrogen- 
bonded regions are located between strands p-1 and p-2, generating a (3-bulge, 
and between strands (3-6 and (3-7. The (3-bulge is largely responsible for the 
highly dynamic nature of the extracellular loop L1 [107]. Additional features 
not seen in any other (3-barrel membrane protein include a tilting of the PagP 
barrel axis by 30^ with respect to the membrane normal and the presence of an 
interior hydrophobic pocket in the upper half of the |3-barrel [Bishop and Prive, 
unpubl. data]. The hydrophobic pocket harbors a single detergent molecule and 
functions as a hydrocarbon ruler that allows the enzyme to distinguish pahni- 
tate from otlier acyl chains present in phospholipids. Mutation of Gly 88 lining 
the bottom of the hydrophobic pocket can modulate the acyl chain length speci- 
ficity of PagP [Bishop and Prive, unpubl. data]. Internalization of phospholipid 
palmitoyl groups within the barrel interior likely occurs by lateral diffusion 
through the non-hydrogen-bonded regions between the |3-strands in the upper 
half of the molecule. 

Three putative catalytic residues were identified by site-directed mutagene- 
sis and mapped to the extracellular loops Li and L2, indicating that the active 
site is localized at the cell surface in the most dynamic region of the molecule 
[1 07]. The putative catalytic residues project their side chains toward the barrel 



Endotoxin Structure and Function 17 



interior and are positioned above the hydrocarbon ruler [Bishop and Prive, 
unpubl. data]. The requirement of invariant His 33, Asp 76, and Ser 77 for cataly- 
sis might suggest that PagP utilizes an acyl-enzyme mechanism characteristic 
of known serine esterases. However, the putative active site residues are not 
organized into a catalytic triad that could enhance the nucleophiJic character 
of Ser 77 [107]. The presence of two non-hydrogen-bonded regions that could 
provide shnultaneous access for both substrates to the |3-barrel interior raises 
the distinct possibility that PagP catalysis proceeds through the formation of a 
ternary complex. Such a mechanism could promote the direct transfer of the 
palmitoyi group from the phospholipid donor to the lipid A acceptor without the 
formation of an acyl-enzyme intermediate, but the detailed mechanism of PagP 
catalysis remains to be elucidated. 

The clear alignment of the PagP active site with the OM outer leaflet cre- 
ates an important topological problem for the enzyme. How does PagP access 
phospholipids if OM lipid asymmetry is maintained? Chelating agents such as 
EDTA can strip a fraction of LPS from the bacterial surface [108]. A large body 
of evidence indicates that EDTA promotes the migration of phospholipids into 
the OM outer leaflet [10]. Indeed, brief treatment of cells with EDTA rapidly 
induces lipid A palmitoylation through a process that is independent of both 
pagP gene regulation and de novo protein synthesis [Bishop, unpubl. data]. 
Lipid A palmitoylation induced by EDTA in vivo also requires functional MsbA 
[Bishop, unpubl. data], which is presumably needed to replenish phospholipids 
lost from the OM inner leaflet. These findings suggest that PagP may function 
to maintain the OM permeability barrier under Mg^"^-limited growth condi- 
tions, in addition to providing CAMP resistance and converting lipid A into an 
endotoxin antagonist. 



LpxO 

An Fe^"^/a-ketoglutarate-dependent dioxygenase homologue in Salmonella 
has recently been shown to catalyze the hydroxylation of lipid A and is 
expressed in a PhoP/PhoQ-dependent manner [76]. Under aerobic conditions, 
LpxO uses molecular oxygen to hydroxylate the 3' secondary acyl chain to gen- 
erate 2-OH-14:0-modified lipid A (fig. 8). Homologues are found in other 
gram-negative bacteria that similarly incorporate S-2-0H groups into their lipid A. 
The function of S-2-hydroxylation is unknown, but the authors speculate that 
the action of leukocyte acyloxyacyl hydrolase, an enzyme that releases sec- 
ondary acyl chains from the lipid A of phagocytosed bacteria, would release 
2-OH-14:0, which is possibly converted into 2-OH-14:0-CoA, a known 
inhibitor of protein N-myristoylation needed for cell signaling functions. 



Bishop 



18 



.0-Cofo 



O2 + 

a-keloglutarate 

IpxO Jte2+ 
^^ CO2 + 

succinate 





HoO^ 



.OCOTfa 



pagL 




Fig. 8. S-2-hydroxylation and 3-0-deacylation of lipid A, LpxO is an LM Fe^"^/a- 
ketoglutarate-dependent dioxygenase homologue that uses molecular oxygen to incorporate 
a hydroxyl group into the secondary myristoyl group at position 3', PagL is an OM lipase 
that removes the 3-OH-l^:0 group at position 3, 



S-2-hydroxylation may also function to provide an additional hydrogen-bond 
donor that could stabilize the lateral interactions between LPS molecules in 
the OM [67]. Given that S-2-hydroxylation is contingent upon lipid A acylation 
by LpxM, the LpxO reaction could occur on either side of the IM without inter- 
fering with the sequential steps of the Raetz pathway. However^ LpxO is pre- 
dicted to be anchored on the periplasmic face of the IM. 



PagL and Rhizobium Lipid A 

Lipid A 3-0-deacylase activity was observed in Salmonella during investi- 
gations of PagP in membranes from a PhoP-constitutive mutant [77]- The 
responsible enzyme was subsequently identified as the PagL gene product, 
which proved to be the second enzyme of lipid A metabolism that is located 
in the OM [78]. PagL functions to deacylate the 0-linked 3-OH-14:0 chain at 
the proximal glucosamine unit of lipid A (fig. 8). By exposing the 3-OH group 
in lipid A, PagL may provide a new hydrogen-bond donor to stabilize the lat- 
eral interactions between LPS molecules in the OM [67]. Although a similar 



Endotoxin Structure and Function 



19 



reaction had been described in Rhizobium legiiminosarum membranes [109], 
PagL homologues are only found in the various serovars of Salmonella. 

Lipid A recovered from Rhizobium species is structurally quite different from 
£". coli lipid A, a fact that may reflect the symbiotic relationship between nitrogen- 
fixing rhizobia and leguminous plants, which normally mount an innate immune 
response to endotoxin. Rhizobium lipid A biosynthesis proceeds according to the 
Raetz pathway, but the molecule is subsequently remodeled by numerous modify- 
ing enzymes. Besides the absence of phosphate groups at positions I and 4' [I 10], 
due to the presence of specific phosphatases [11 1, 112], the distal glucosamine 
sugar exhibits a 27-OH-28:0 acyl chain as part of a characteristic acyloxyacyl moi- 
ety at position 2' and a galacturonic acid residue at position 4' [113, 1 14]. LpxQ 
is the third OM enzyme found to be involved in lipid A modification [115, 1 16], 
and catalyzes the oxidation of the proximal 1-dephospho sugar to generate an 
acylated 2-ammogluconate moiety. 

Rhizobium lipid A serves to illustrate a fundamental point that is supported 
by functional genomics; namely^ that the essential enzymes of the Raetz pathway 
are highly conserved in gram-negative bacteria and that the observed variations 
in lipid A structure are a consequence of the presence of additional modifying 
enzymes. Aside fi^om variations in lipid A structure due to cytoplasmic ACP- 
dependent acyl transferases [1 17-1 19] and Kdo transferases [ 1 20, 121] with dis- 
tinct substrate specificities, it appears that most modifying enzymes act on the 
lipid A nucleus in the extracytoplasmic compartments. These observations may 
reflect a need to avoid futile cycling and to maintain a sequential order of Raetz 
pathway reactions. These principles should faithfully guide future discoveries of 
new enzymes that are employed to generate novel lipid A structures in diverse 
organisms. 



Perspectives 

LPS structure and function are unique to gram-negative bacteria, but some 
intriguing parallels are seen with the cholesterol and glycosphingolipid-rich 
lipid rafts, and N-luiked protein glycosylation pathways of eukaryotic cells. 
Both lipid A and eukaryotic glycolipids differ from phospholipids by the pres- 
ence of hydrogen-bonded lateral interactions that tend to exclude phospholipids 
leading to the formation of detergent resistant lipid domains [67, 122]. 
Additionally, the undecaprenyl phosphate-dependent pathways for the synthe- 
sis and mcorporation of 0-antigens into the core-Kdo2-lipid A molecule at the 
IM mirrors the dolichol phosphate-dependent pathway m the endoplasmic 
reticulum, where Glc3-Man9-GlcNAc2 is incorporated into targeted protein Asn 
residues [1 23]. Finally, it now appears that many of the Raetz pathway enzymes 



Bishop 



20 



are conserved in the genomes of plants, perhaps reflecting the presence of lipid 
A-like molecules in plastids [5]. 

Lipid A and its regulated covalent modifications exhibit profound effects on 
bacterial and human physiology. Novel endotoxin antagonists and immune adju- 
vants have already been developed from modified lipid A structures [124, 125], 
By revealing the biochemical details of lipid A structure and function we hope to 
understand its role in bacterial pathogenesis and to intervene with novel treat- 
ments for infection. However, we must remind ourselves that multiple molecular 
subtypes of lipid A are acting in concert in the bacterial cell. The need to unravel 
the interactions between individual lipid A modifications will provide fertile 
ground for future research. 



Acknowledgments 

Work in the author's laboratory was supported by the Canadian [nstitutes of Health 
Research. Eileen K Lo is acknowledged for her assistance with the initial drafts of this 
manuscript. 



References 

1 Beutler B, Rietschel ET: Innate immune sensing and its roots: The story of endotoxin. Nat Rev 
Immunol 2003;3:169-176. 

2 Janeway CA Jr: Approaching the asymptote? Evolution and revolution in immunology. Cold 
Spring Harb Symp Quant Biol 1989;54:1-13. 

3 Akira S: Toll-like receptor signaling. J Biol Chem 2003;278:38105-38108. 

4 Inohara N, Nunez G: NODs: Intracellular proteins involved in inflammation and apoptosis. Nat 
Rev Immunol 2003;3:371-382. 

5 Raetz CR^ Whitfield C: Lipopolysaccharide endotoxins. Annu Rev Biochem 2002;71:635-700. 

6 Harald FM: Gleanings of a chemiosmotic eye. Bioessays 2001;23:848-855. 

7 Duong F, Eichler J, Price A, Leonard MR, Wickner W: Biogenesis of the Gram-negative bacterial 
envelope. Cell 1997;91:567-573. 

8 Holtje JV: Growth of the stress-bearing and shape-maintaining murein sacculus of Escherichia 
coli, Microbiol Mol Biol Rev 1998;62:181-203. 

9 Kamio Y, Nikaido H: Outer membrane of Salmonella typhimnnum: Accessibility of phospholipid 
head groups to phospholipase C and cyanogen bromide activated dextran in the external medium. 
Biochemistry 1976;15:2561-2570. 

10 Nikaido H, Vaara M: Molecular basis of bacterial outer membrane permeabilit)'. Microbiol Rev 

1985;49:1-32. 
I I Schuiz GE: The structure of bacterial outer membrane proteins. Biochim Biophys Acta 2002; 

1565:308-317. 

12 Vaara M: Antibiolic-supersusceplible mutants of Escherichia coll and Salmonella (yphimiirium. 
Antimicrob Agents Chemother 1993;37:2255-2260. 

13 Poltorak A, He X, Smu-nova I, Liu MY, Van HutTel C, Du X, Birdwell D, AJejos E, Silva M, 
Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, Beutler B: Defective LPS signaling 
in C3H/HeJ and C57BL/10ScCr mice: Mutations inTlr4 gene. Science 1998;282:2085-2088. 

14 Qureshi ST, Lariviere L, Leveque G, Clermont S, Moore KJ, Gros P, Malo D: Endotoxin-tolerant 
mice have mutations in Toll-like receptor 4 (Tlr4). J Exp Med 1999;189:615-625. 



Endotoxin Structure and Function 21 



15 HofTmarm JA, Kafatos FC, Janeway CA, Ezekowitz RA: Phylogenetic perspectives in innate 
immunity. Science 1999;284:1313-1318. 

16 Aderem A, Uleviich RJ: Toll-like receptors in the induction of the innate immune response. Nature 
2000;406:782-787. 

17 Nishijima M, Raetz CR: Membrane lipid biogenesis in Escherichia coli: Identification of genetic 
loci for phosphatidylgJycerophosphate synthetase and construction of mutants lacking phosphatidyl- 
glycerol. J Biol Chem 1979;254:7837-7844. 

18 TaJcayama K, Qureshi N, Mascagni P, Nashed MA, Anderson L, Raetz CR: Fatty acyl derivatives 
of glucosamine 1 -phosphate in Escherichia coli and their relation to lipid A. Complete snucture 
of A diacyl GlcN-l-P found in a phosphatidylglycerol-deficient mutant. J Biol Chem I983;258: 
7379-7385. 

19 TakayamaK, Qureshi N, Mascagni P, Anderson L, Raetz CR: Glucosamine-derived phospholipids 
in Escherichia coli. Structure and chemical modification of a triacyl glucosamine l-phosphate 
found in a phosphatidylglycerol-deficient mutant. J Biol Chem 1983;258:14245-14252, 

20 Rietschel ET, BradeH,Brade L, Kaca W, Kawahara K, Lindner B, LuderitzT, TomitaT, Schade U, 
Seydel U, Zahringer U: Newer aspects of the chemical structure and biological activity of bacterial 
endotoxins. Prog Clin Biol Res 1985;189:31-51. 

21 Nishijima M, Bulawa CE, Raetz CR: Two interacting mutations causing temperature-sensitive 
phosphatidylglycerol synthesis in Escherichia coli membranes. J Bacteriol 198 1; 145:1 13-121. 

22 Nishijima M, Raetz CR: Characterization of two membrane-associated glycolipids from an 
Escheiichia coli mutant deficient in phosphatidylglycerol. J Biol Chem 1981;256:10690-10696. 

23 Ray BL, Painter G, Raetz CR: The biosynthesis of Gram-negative endotoxin. Formation of lipid 
A disaccharides from monosaccharide precursors in extracts of Escherichia coli. J Biol Chem 
1984:259:4852^859. 

24 Nishijima M, Amano F, AkamalsuY, Akagawa K, Tokunaga T, Raetz CR: Macrophage activation by 
monosaccharide precursors oi Escherichia coli lipid A. Proc Natl Acad Sci USA 1985;82:282-286. 

25 Anderson MS, Bull HG, Galloway SM, Kelly TM, Mohan S, Radika K, Raetz CR: UDP-N- 
acetylglucosamjne acyltransferase of Escherichia coli. The first step of endotoxin biosynthesis is 
thermodynamically unfavorable. J Biol Chem 1993;268:19858-19865. 

26 Raetz CR, Roderick SL: A left-handed parallel beta helix in the structure of UDP-N-aceiyl- 
glucosamine acyltransferase. Science 1995;270:997-1000, 

27 Wyckoff TJ, Lin S, Cotter RJ, Dotson GD, Raetz CR: Hydrocarbon rulers in UDP-N-acetyl- 
glucosamine acyltransferases. J Biol Chem 1998;273:32369-32372. 

28 Young K, Silver LL, Bramhill D, Cameron P, JEveland SS, Raetz CR, Hyland SA, Anderson MS: 
The envA permeability/cell division gene of Escherichia coli encodes the second enzyme of lipid 
A biosynthesis. UDP-3-0-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase. J Biol Chem 
1995;270:30384-30391. 

29 Sorensen PG, Lutkenhaus J, Young K, Eveland SS, Anderson MS, Raetz CR: Regulation of UDP- 
3-0-[R-3-hydroxymyristoyl]-N-acelylglucosamine deacetylase in Escherichia coli. The second 
enzymatic step of lipid a biosynthesis. J Biol Chem 1996;271:25898-25905. 

30 Onishi HR, Pelak BA, Gerckens LS, Silver LL, Kahan FM, Chen MH, Patchett AA, Galloway SM, 
Hyland SA, Anderson MS, Raetz CR: Antibacterial agents that inhibit lipid A biosynthesis. 
Science 1996;274:980-982. 

31 Jackman JE, Raetz CR, Fierke CA: Site-directed mutagenesis of the bacterial metalloamidase 
UDP-(3-0-acyl)-N-acetylglucosamine deacetylase (LpxC). Identification of the zinc binding site. 
Biochemistry 200 1 ;40:5 1 4-523. 

32 Coggins BE, Li X, McClerren AL, Hindsgaul O, Raetz CR, Zhou P: Structure of the LpxC 
deacetylase with a bound substrate -analog inhibitor. Nat Struct Biol 2003;10:645-651. 

33 Whittington DA, Rusche KM, Shin H, Fierke CA, Christianson DW; Crystal structure of LpxC, a 
zinc-dependent deacetylase essential for endotoxin biosynthesis. Proc Natl Acad Sci USA 
2003;100:8146-8150. 

34 Jackman JE, Fierke CA, T\imey LN, Pirrung M, Uchiyama T, Tahir SH, Hindsgaul O, Raetz CR: 
Agents that target lipid A biosynthesis in gram-negative bacteria. Inhibition of diverse UDP-3-0- 
(r-3-hydroxymyristoyl)-n-acetylglucosamine deacetylases by substrate analogs containing zinc 
binding motifs, J Biol Chem 2000;275:1 1002-1 1009. 



Bishop 



22 



35 Pirrung MC, Tumey LN, McClerren AL, Raetz CR: High-throughput catch-and-release synthesis 
of oxazoline hydroxamates. Structure-activity relationships in novel inhibitors of Escherichia coli 
LpxC: In vitro enzyme inhibition and antibacterial properties. J Am Chem Soc 2003; 1 25: 
1575-1586. 

36 Kelly TM, Stachula SA, Raetz CR, Anderson MS: The firA gene oi Escherichia coli encodes 
UDP-3-0-(R-3-hydroxymyristoyl)-glucosamine N-acy I transferase. The third step of endotoxin 
biosynthesis. J Biol Chem 1993;268:19866-19874, 

37 Babinski KJ, Ribeiro AA, Raetz CR: The Escherichia coli gene encoding the UDP-23-diacyl- 
glucosamine pyrophosphatase of lipid A biosynthesis. J Biol Chem 2002;277:25937-25946. 

38 Babinski KJ, Kanjilal SJ, Raetz CR: Accumulation of the lipid A precursor UDP-2,3-diacyl- 
glucosamine in an Escherichia coli mutant lacking the IpxH gene. J Biol Chem 2002;277: 
25947-25956. 

39 Garrett TA, Kadrmas JL, Raetz CR: Identification of the gene encoding the Escherichia coli lipid 
A 4'-kinase, Facile phosphorylation of endotoxin analogs with recombinant LpxK. J Biol Chem 
1997;272:21855-21864, 

40 Garrett TA, Que NL, Raet2 CR: Accumulation of a lipid A precursor lacking the 4'-phosphate 
following inactivation of the Escherichia coli IpxK gene. J Biol Chem 1998;273:12457-12465. 

41 Lien E, Means TK, Heine H, Yoshimura A, Kusumoto S, Fukase K, Fenton MJ, Oikawa M, 
Qureshi N, Monks B, Finberg RW, Ingalls RR, Golenbock DT: Toll-like receptor 4 imparts ligand- 
specific recognition of bacterial lipopolysaccharide. J Clin Invest 2000;105:497-504. 

42 Poltorak A, Ricciardi-Castagnoli P, Citterio S, Beutler B: Physical contact between lipopolysaccha- 
ride and toll-like receptor 4 revealed by genetic complementation, Proc Natl Acad Sci USA 2000;97; 
2163-2167. 

43 Clementz T, Raetz CR: A gene coding for 3-deoxy-D-manno-octulosonic-acid transferase in 
Escherichia coli. Identification, mapping, cloning, and sequencing. J Biol Chem I99l;266: 
9687-9696. 

44 Brozek KLA, Raetz CR: Biosynthesis of lipid A in Escherichia coli, Acyl carrier protein-dependent 
incorporation of laiirate and myristate. J Biol Chem 1990;265:15410— 15417. 

45 Clementz T, Bednarski JJ, Raetz CR: Function of the htrB high temperature requirement gene 
of Escherchia coli in the acylation of lipid A: HtrB catalyzed incorporation of laurate. J Biol 
Chem 1996;271:12095-12102. 

46 Clementz T, Zhou Z, Raetz CR: Function of the Escherichia coli msbB gene, a multicopy sup- 
pressor of htrB knockouts, in the acylation of lipid A. Acylation by MsbB follows laurate incor- 
poration by HtrB. J Biol Chem 1997;272:10353-10360. 

47 Carty SM, Sreekumar KR, Raetz CR: Effect of cold shock on lipid A biosynthesis in Escherichia 
coli. Induction at 12 degrees C of an acyl transferase specific for palmitoleoyl-acyl carrier protein. 
J Biol Chem 1999;274:9677-9685. 

48 Vorachek-Warren MK, Carty SM, Lin S, Cotter RJ, Raetz CR: An Escherichia coli mutant lacking 
the cold shock-induced palmitoleoyltransferase of lipid A biosynthesis: Absence of unsaturated 
acyl chains and antibiotic hypersensitivity at 12 degrees C. J Biol Chem 2002;277: 14186^14193. 

49 Vorachek-Warren MK, Ramirez S, Cotter RJ, Raetz CR: A triple mutant of Escherichia coli lack- 
ing secondary acyl chains on lipid A. J Biol Chem 2002;277:14194-14205. 

50 Somer^'ille JE Jr, Cassiano L, Bainbridge B, Cunningham MD, Darveau RP: A novel Escherichia 
coli lipid A mutant that produces an anti-inflammatory lipopolysaccharide. J Clin Invest 1996;97: 
359-365. 

51 Heinrichs DE, Yethon JA, Whitfield C: Molecular basis for structural diversity in the core regions 
of the lipopolysaccharides of Escherichia coli and Salmonella enfen'ca. Mol Microbiol 1998;30: 
221-232. 

52 Kanipes MI, Lin S, Cotter RJ, Raetz CR: Ca^^-induced phosphoethanolamine transfer to 
the outer 3-deoxy-Z)-manno-octuIosonic acid moiety of Escherichia coli lipopolysaccharide, 
A novel membrane enzyme dependent upon phosphatidylethanolamine. J Biol Chem 2001 ;276: 
1156-1163. 

53 Yethon JA, Heinrichs DE, Monteiro MA, Perry MB, Whitfield C: Involvement of waaY, waaQ, 
and waaP in the modification of Escherichia coli lipopolysaccharide and their role in the forma- 
tion of a stable outer membrane. J Biol Chem 1998;273:26310-26316. 



Endotoxin Structure and Function 23 



54 Heinrichs DE, Yethon JA, Amor PA, Whitfield C: The assembly system for the outer core por- 
tion of Rl- and R4-type lipopolysaccharides of Escherichia coli. The RI core-specific beta- 
glucosyltransferase provides a novel attachment site for O-polysaccharides. J Bio) Chem 1998; 
273:29497-29505. 

55 Valvano MA: Export of 0-specific lipopolysaccharide. Front Biosci 2003;8:s452-s47l. 

56 Zhou Z, White KA, Polissi A, Georgopoulos C, Raetz CR: Function o^ Escherichia coli MsbA, an 
essential ABC family transporter, in lipid A and phospholipid biosynthesis. J Biol Chem 1998; 
273:12466-12475. 

57 Doerrler WT, Reedy MC, Raetz CR: An Escherichia coli mutant defective in lipid export. J Biol 
Chem 200l;276:l 1 461-1 1464, 

58 Chang G, Roth CB: Structure of MsbA from E, coli: A homolog of the multidrug resistance ATP 
binding cassette (ABC) transporters. Science 200 1 ;293: 1793-1800. 

59 Doerrler WT, Raetz CR: ATPase activity of the MsbA lipid flippase oi Escherichia coli, J Biol 
Chem 2002;277:36697-36705. 

60 Chang G: Structure of MsbA from Vibrio cholera'. A multidrug resistance ABC transporter 
homolog in a closed conformation. J Mol Biol 2003;330:4I9^30. 

61 Kol MA, van Dalen A, de Kroon AJ, de KruijiTB: Translocation of phospholipids is facilitated by a 
subset of membrane-spanning proteins of the bacterial cytoplasmic membrane. J Biol Chem 2003; 
278:24586-24593. 

62 Jones NC, Osborn MJ: Interaction of Salmonella typhimurium W\\\\ phospholipid vesicles. 
Incorporation of exogenous lipids into intact cells. J Biol Chem 1977;252:7398-7404. 

63 Jones NC, Osborn MJ: Translocation of phospholipids between the outer and inner membranes of 
Salmonella typhimurium. J Biol Chem 1977;252:7405-7412. 

64 Genevrois S, Steeghs L, Roholl P, Letesson JJ, van der Ley P: The Omp85 protein of Neisseria 
meningitidis is required for lipid export to the outer membrane. EMBO J 2003;22: 1780-1789. 

65 Voulhoux R, Bos MP, Geurlsen J, Mols M, Tommassen J: Role of a highly conserved bacterial 
protein in outer membrane protein assembly. Science 2003;299:262-265, 

66 CoughJin RT, Tonsager S, McGroarty EJ: Quantitation of metal cations bound to membranes and 
extracted lipopolysaccharide of £5<:/renc/?/i7 co//. Biochemistry 1983;22:2002-2007. 

67 Niakido H: Molecular basis of bacterial outer membrane permeability revisited. Microbiol Mol 
Biol Rev 2003;67:593-656. 

68 Hancock RE, Falla T, Brown M: Cationic bactericidal peptides. Adv Microb Physiol 1995;37: 
135-175, 

69 Zasloff M: Antimicrobial peptides of multicellular organisms. Nature 2002;415:389-395. 

70 Groisman EA:The ins and outs of vimlence gene expression: Mg^^ as a regulatory signal. Bioessays 
1998;20:96-101. 

71 Forbes J R-, Gros P: Divalent-metal transport by NRAMP proteins at the interface of host-pathogen 
interactions. Trends Microbiol 2001;9:397^03. 

72 Guo L, Lim KB, Gunn JS, Bainbridge B, Darveau RP, Hackett M, Miller SI: Regulation of 
W^'id A mo^\r\z^\\owsby Salmonella typhimurium virulence genes phoP-phoQ. Science 1997;276: 
250-253. 

73 Zhou Z, Lin S, Cotter RJ, Raetz CR: Lipid A modifications characteristic of Salmonella 
typhimurium are induced by NH^V03 in Escherichia coli K12. Detection of 4-amino-4-deoxy-L- 
arabinose, phosphoethanolamine and palmitate. J Biol Chem 1999;274:18503-18514. 

74 Zhou Z, Ribeiro AA, Raetz CR: High-resolution NMR spectroscopy of lipid A molecules contain- 
ing 4-amino-4-deoxy-/--arabinose and phosphoethanolamine substituents. Different attachment sites 
on lipid A molecules from NHl4V03-treated Escherichia coli versus kdsA mutants of Salmonella 
typhimurium. J Biol Chem 2000;275:13542-13551. 

75 Zhou Z, Ribeiro AA, Lin S, Cotter RJ, Miller SI, Raetz CR: Lipid A modifications in polymyxin- 
resistant Salmonella typhimurium: PMRA-dependent 4-amino-4-deoxy-/.-arabinose, and phospho- 
ethanolamine incorporation. J Biol Chem 2001;276:43l 1 1^3121. 

76 Gibbons HS, Lin S, Cotter RJ, Raetz CR: Oxygen requirement for the biosynthesis of 
the S-2-hydroxymyristate moiety in Salmonella typhimurium lipid A. Function of LpxO, a new 
Fe^"^/alpha-ketoglutarate-dependent di oxygenase homologue. J Biol Chem 2000;275: 
32940-32949. 



Bishop 



24 



77 Bishop RE, Gibbons HS, GuinaT, Trent MS, Miller SI, Raetz CR: Transfer of palmitate from phos- 
pholipids to lipid A in outer membranes of Gram-negative bacteria. EMBO J 2000;19:5071-5080, 

78 Trent MS, Pabich W, Raetz CR, Miller SJ: A PhoP/PhoQ-induced lipase (PagL) that catalyzes 
3-0-deacylation of lipid A precursors in membranes of Salmonella typhimurinm. J Biol Chem 
2001;276:9083-9092. 

79 Helander IM, KJIpelainen I, Vaara M: Increased substitution of phosphate groups in lipopolysaccha- 
rides and lipid A of the polymyxin-resistant pmrA mutants of Salmonella typhimtirmm\ A ^'P-NMR 
study. Mol Microbiol I994;l l:48J^87. 

80 Gunn JS, Lim [CB, Krueger J, Kim K, Guo L, Hackett M, Miller SI: PmrA-PmrB-regulated genes 
necessary for 4-ammoarabinose lipid A modification and polymyxjn resistance. Mol Microbiol 
1998;27:1171-1 182. 

81 Bruch MD, Cajal Y, Koh TT, Jain MK: Higher-order structure of polymyxin B:The functional sig- 
nificant of topological specificity. J Am Chem Soc I999;I21:I 1993-12004. 

82 PrisLovsek P, Kidric J: Solution structure of polymyxins B and E and effect of binding to lipopoly- 
saccharide: An NMR and molecular modeling study. J Med Chem I999;42: 4604—4613. 

83 Guo L, Lim K, Poduje C, Daniel M, Gunn J, Hackett J, Miller SJ: Lipid A acylation and bacterial 
resistance against vertebrate anti-microbial peptides. Cell 1998;95:189-198. 

84 Hajjar AM, Ernst RK, Tsai JH, Wilson CB, Miller SI: Human Toll-like receptor 4 recognizes host- 
specific LPS modifications. Nat Immunol 2002;3:354-359. 

85 Tanamoto K, Azumi S: Salmonella-type heptaacylated lipid A is inactive and acts as an antagonist 
of lipopolysaccharide action on human line cells. J Immunol 2000;164:3149-3156. 

86 Muroi M, Ohnishi T, Tanamoto K; MD-2, a novel accessory molecule, is involved in species-specific 
actions of Salmonella lipid A. Intect bnmun 2002;70:3546-3550. 

87 Groisman EA: The pleiotropic two-component regulatory system PhoP-PhoQ. J Bacteriol 2001; 
183:1835-1842. 

88 Garcia Vescovi E, Soncini FC, Groisman EA: Mg^"^ as an extracellular signal: Environmental 
regulation of Salmonella virulence. Cell 1996;84:165-174. 

89 Kjox LF, Wosten MM, Groisman EA: A small protein that mediates the activation of a two- 
component system by another two-component system. EMBO J 2000;19: 1861-1872. 

90 Wosten MM, Kox LF, Chamnongpol S, Soncini FC, Groisman EA: A signal transduction system 
that responds to extracellular iron. Cell 2000;103:1 13-125. 

91 Kato A, Latifi T, Groisman EA: Closing the loop: The PmrA/PmrB two-component system nega- 
tively controls expression of its posttranscriptional activator PmrD. Proc Natl Acad Sci USA 2003; 
100:4706^71). 

92 Bader MW, Navarre WW, Shiau W, Nikaido H, Frye JG, McClelland M, Fang FC, Miller SI: 
Regulation of Salmonella typhimnrium virulence gene expression by cationic antimicrobial pep- 
tides. Mol Microbiol 2003;50:219-230. 

93 McPhee JB, Lewenza S, Hancock RE: Cationic antimicrobial peptides activate a two-component 
regulatory system, PmrA-PmrB, that regulates resistance to polymyxin B and cationic anti- 
microbial peptides in Pseiidomonas aefuginosa. Mol Microbiol 2003;50:205-217. 

94 Vaara M, Vaara T, Jensen M, Helander 1, Nurminen M, Riet5chel ET, Makela PH: Characterization 
of the lipopolysaccharide from the polymyxin-resistant pmrA mutants of Salmonella 
typhimurium, YEBSL^n 1981;129:145-149. 

95 Mouslim C, Groisman EA: Control of the Salmonella ugd gene by three two-component regula- 
tory systems. Mol Microbiol 2003;47:335-344. 

96 Breazeale SD, Ribeiro AA, Raetz CR: Oxidative decarboxylation of UDP-glucuronic acid in 
extracts of polymyxin-resistant Escherichia coli. Origin of lipid A species modified with 4-amino- 
4-deoxy-L-arabinose. J Biol Chem 2002;277:2886-2896. 

97 Breazeale SD, Ribeiro AA, Raetz CR: Origin of lipid A species modified with 4-amino-4-deoxy- 
L-arabinose in polymyxin resistant mutants of Escherichia coli: An aminotransferase (ArnB) that 
generates UDP-4-amino-4-deoxy-L-arabinose. J Biol Chem 2003;278: 24731-24739. 

98 Noland BW, Newonan JM, Hendle J, Badger J, Christopher J A, Tresser J, Buchanan MD, Wright TA, 
RuUer ME, Sanderson WE, Muller-Dieckmann HJ, Gajiwala KS, Buchanan SG: Structural studies 
of Salmonella typhimwiitm ArnB (PmrH) aminotransferase: A 4-amino-4-deoxy-L-arabinose 
lipopolysaccharide -modifying enzyme. Structure 2002;10:1569-1580. 



Endotoxin Structure and Function 25 



99 Trent MS, Ribeiro AA, Lin S, Cotter RJ, Raetz CR: An inner membrane enzyme in Salmonella 
and Escherichia coli that transfers 4-amino-4-deoxy-I-arabinose to lipid A: Induction on 
polymyxin-resistant mutants and role of a novel lipid-linked donor. J Biol Chem 200l;276: 
43122^3131. 

100 Trent MS, Ribeiro AA, Doerrler WT, Lin S, Cotter RJ, Raetz CR: AccumuJation of a polyisoprene- 
linked amino sugar in polymyxin-resistant Salmonella typhimuhum and Escherichia coli: Structural 
characterization and transfer to lipid A in the periplasm. J Biol Chem 2001 ;276:43] 32^3 144. 

101 Trent MS, Raetz CRH: Cloning of EptA, the lipid A phosphoethanolamine transferase associated 
with polymyxin resistance. J Endotoxin Res 2002;8:I58. 

102 Cox AD, Wright JC, Li J, Hood DW, Moxon ER, Richards JC: Phosphorylation of the lipid a 
region of meningococcal lipopolysaccharide: Identification of a family of transferases that add 
phosphoethanolamine to lipopolysaccharide. J Bacteriol 2003;185:3270-3277. 

103 Gunn JS, Miller SI: PhoP-PhoQ activates transcription of pmrAB, encoding a two-component reg- 
ulatory system involved in Salmonella typhimitrium antimicrobial peptide resistance. J Bacteriol 
1996;178:6857-6864. 

104 Soncini FC, Groisman EA: Two-component regulatory systems can interact to process multiple 
environmental signals. J Bacteriol 1 996; 1 78:6796-6801. 

105 Robey M, O'Connell W, Cianciotto NP: Identification o^ Legionella pneumophila rep, a pagP-like 
gene that confers resistance to cationic antimicrobial peptides and promotes intracellular infec- 
tion. Infect Immun 200l;69:4276-^286. 

106 Preston A, Maxim E, Toland E, Pishko EJ, Hai'vill ET, Caroff M, Maskell DJ: Bordetella 
bwnchi^eptica PagP is a Bvg-regulated lipid A palmiioyi transferase that is required for persistent 
colonization of the mouse respiratory tract. Mol Microbiol 2003;48:725-736, 

107 Hwang PM, Choy W Y, Lo EI, Chen L, Forman-Kay JD, Raetz CR, Prive GG, Bishop RE, Kay LE: 
Solution structure and dynamics of the outer membrane enzyme PagP by NMR. Proc Natl Acad 
Sci USA 2002;99:13560-13565. 

108 Leive L: Release of lipopolysaccharide by EDTA treatment of £. coli. Biochem Biophys Res 
Commun 1965;21:290-296. 

109 Basu SS, White ICA, Que NL, Raetz CR: A deacylase in Rhizobliim legttminosamm membranes 
that cleaves the 3-0-linked beta-hydroxymyristoyl moiety of lipid A precursors. J Biol Chem 
1999;274:11150-11158. 

1 10 Brozek KA, Kadrmas JL, Raetz CR: Lipopolysaccharide biosynthesis in Rhizobium leguminosanm. 
Novel enzymes that process precursors containing 3-deoxy-D-manno-octulosonic acid. J Biol Chem 
1996;271:32) 12-321 18. 

1 1 1 Price NP, Jeyaretnam B, Carlson RW, Kadrmas JL, Raetz CR, Brozek KA: Lipid A biosynthesis 
in Rhizobium leguminosamm: Role of a 2-keto-3-deoxyoctuIosonate-activated 4' phosphatase. 
Proc Natl Acad Sci USA 1995;92:7352-7356. 

1 12 Karbarz MJ, Kalb SR, Cotter RJ, Raetz CR: Expression cloning and biochemical character- 
ization of a Rhizobium leguminosanim lipid A I -phosphatase. J Biol Chem 2003;278: 
39269-39279. 

1 13 Que NL, Lin S, Cotter RJ, Raetz CR: Purification and mass spectrometry of six lipid A species 
from the bacterial endosymbiont Rhizobium etli. Demonstration of a conserved distal unit and a 
variable proximal portion. J Biol Chem 2000;275:28006-28016. 

1 14 Que NL, Ribeiro AA, Raetz CR: Two-dimensional NMR spectroscopy and structures of six lipid 
A species from Rhizobium etli CE3. Detection of an acyloxyacyl residue in each component and 
origin of the aminogluconate moiety. J Biol Chem 2000;275:28017-28027. 

1 15 Que-Gewirth NL, Lin S, Colter RJ, Raetz CR: An outer membrane enzyme that generates the 
2-amino-2-deoxy-gluconate moiety o^ Rhizobium leguminosaimm lipid A. J Biol Chem 2003;278: 
12109-12119. 

1 16 Que-Gewirth NL, Karbarz MJ, Kalb SR, Cotter RJ, Raetz CR: Origin of the 2-amino-2-deoxy- 
gluconate unit in Rhizobium leguminosanim lipid A. Expression cloning of the outer membrane 
oxidase LpxQ. J Biol Chem 2003;278:12120-12129. 

1 17 Sweet CR, Lin S, Cotter RJ, Raetz CR: A Chlamydia trachomatis UDP-N-acetylglucosamine acyl- 
transferase selective for myrisioyl-acyl carrier protein. Expression in Escherichia coli and forma- 
tion of hybrid lipid A species. J Biol Chem 2001;276:19565-19574. 



Bishop 



26 



1 18 Sweet CR, Preston A, Toland E, Ramirez SM, Cotter RJ, Maskell DJ, Raetz CR: Relaxed acyl 
chain specificity of Bordetella UDP-N-acetylglucosamine acyltransferases, J Biol Chem 2002; 
277:18281-18290. 

1 19 Basu SS, Karbarz N4J, Raetz CR: Expression cloning and characterization of the C28 acyllrans- 
ferase of lipid A biosynthesis in Rhizobium leguminoaarum. J Biol Chem 2002;277;28959-2897r 

120 White ICA, Kaltashov I A, Cotter RJ, Raetz CR: A mono-functional 3-deoxy-D-manno-octulosonic 
acid (Kdo) transferase and a Kdo kinase in extracts oi Haemophilus influenzae, J Biol Chem 1997; 
272:16555-16563. 

121 Belunis CJ, Mdluli KE, Raetz CR, Nano FE: A novel 3-deoxy-D-manno-octulosonic acid trans- 
ferase from Chlamydia trachomatis required for expression of the genus-specific epitope. J Biol 
Chem 1992;267:18702-18707. 

122 MunroS: Lipid rafts: Elusive or illusive? Cell 2003;! 15:377-388. 

123 Drickamer K, Taylor ME: Evolving views of protein glycosylation. Trends Biochem Sci 1998; 
23:321-324. 

124 Christ WJ, Asano O, Robidoux AL, Perez M, Wang YA, Dubuc GR, Gavin WE, Hawkins LD, 
McGuinness PD, Mullarkey MA, Lewis MD, Kishi Y, KawalaT, Bristol JR, Rose JR, Rossignol DP, 
Kobayashi S, Hishinuma L, ICimura A, Asakawa N, Katayama K, Yamatsu I: E553I, a pure endo- 
toxin antagonist of high potency. Science 1995;268:80-83. 

125 Ulrich JT, Myers KR: Monophosphoryl lipid A as an adjuvant; in Powell MF, Newman MJ (eds): 
Vaccine Design: The Subunit and Adjuvant Approach, New York, Plenum Press, 1995, pp 495-524, 



Russell E. Bishop 

6213 Medical Sciences Building, I King's College Circle 

Toronto, Ont. M5S I A8 (Canada) 

Tel. +1 416 946 7103, Fax +1 416 978 5959, E-Mail russell.bishop@utoronto.ca 



Endotoxin Structure and Function 27 



Toxins 



Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kaiger, 2005, vol 12, pp 28 54 



Bacterial Exotoxins 



Michel R. Popoff 

Unite dcs Bactcrics anacrobics ctToxincs, Fnstitut Pasteur, Paris, France 



Amongst the various mechanisms developed by pathogenic bacteria to 
cause disease, toxins play an important role, since they are responsible for the 
majority of symptoms and lesions during infection. Exotoxins act at a distance 
from the infectious site and can diffuse through the organism. While some 
cytotoxins can cause disruption of cells permitting the pathogens access to nutri- 
ents, other toxins are only active on specific cells, for example intestinal cells, 
neuronal cells, or leukocytes. This is achieved by the recognition of specific cell 
surface receptors. When bound to the receptor, toxins can unleash their toxic 
program at the cell membrane by interfering with signal transduction pathways, 
pore formation, or enzymatic activities towards membrane compounds. In con- 
trast, other toxins enter the cytosol, and recognize and modify specific intracel- 
lular targets. According to the nature of the target and the type of modification, 
intracellular active toxins cause a dramatic alteration of cellular functions such 
as protein synthesis, cell homeostasis, cell cycle progression, vesicular traffic, 
and actin cytoskeletal rearrangements. Alternatively, invasive bacteria can 
directly inject toxins or virulence factors into target cells. This chapter is a com- 
parative overview of the molecular mechanisms of the main bacterial exotoxins. 



Toxins Active at the Cell Surface 

Toxins Modulating Signal Transduction Pathways 

Some enterotoxigenic Escherichia coli and other gram-negative entero- 
pathogens (Yersinia enterocolitica, Vibrio cholerae) secrete heat-stable entero- 
toxins (STs) that can cause acute diarrhea in humans and animals. These toxins 
are small peptides which fall into two subgroups: methanol-soluble (STa or ST-I) 
and methanol-insoluble (STb or ST-II) toxins. Analysis of STs shows they possess 
a similar structure, containing 3 segments joined by 3 disulfide bridges. Alal3 in 



Hormone-like 
toxins 



CI", H2O 



Adenylcyclase 




Pore-forming 
toxins 



Fig. L Toxins that alter cell homeostasis. Some of the mechanisms used by bacteria to 
modify cell homeostasis are depicted. E. coli heat-stable enterotoxin (STa) binds to the extra- 
cellular domain of transmembrane guanylate cyclase, resulting in an increase in cyclic GMP, and 
secretion of CI" and H2O, PFT inserted into the membrane cause leakage of ions and HjO, CT 
and E. colt heat-labile toxins enter the cell cytosol and ADP-ribosylate the Gsu: subunit of het- 
erotrimericG proteins, leading to a permanent active molecule by inhibition of its GTPase activ- 
ity and subsequent stimulation of adenylcyclase. The resulting increase in cyclic AMP induces 
the secretion of Cl~ and HjO. PT inactivates the inhibitory heterotrimeric G protein Gia, lead- 
ing to a upregulation of adenylcyclase activity. Bacterial adenylcyclases, such as EF from 
anthrax toxin and Bordetella adenylcyclase (Cya), can also modulate cAMP levels in the celJs. 



the flexible central segment plays a key role in the toxin's activity. This residue 
is probably involved in the interaction of the toxin with its receptor. In the case 
of STa, the secreted protein encompasses 18-19 amino acids, including 6 cys- 
teines, and is capable of forming 3 disulfide bridges to create a highly stable 
molecule. The carboxy-terminal segment of STs shares similarities with ionu- 
phores and is therefore expected to interact with metal ions. Enteroaggregative 
E. coli (EAggEC) strains also produce a heat-stable enterotoxin related to STa 
with similar pathological effects, 

STa induces watei^ diarrhea without causing obvious histological morpho- 
logical damage. The toxin binds to the extracellular domain of guanylate cyclase 
(GC-C) localized on the apical membrane of enterocytes. GC-C consists of 
4 domains: an extracellular domain, a transmembrane segment, a kinase-like 
domain and an enzymatic domam, which catalyzes the formation of cyclic GMP 
(cGMP) (fig. I), The kinase-like domain has an inhibitory effect on the catalytic 



Bacterial Exotoxins 



29 



activity. Binding of STa to the extracellular domain of GC-C has been suggested 
to induce a conformational change in the protein kinase-liJce domain resulting in 
an uncontrolled increase of GC-C activity. Elevation of intracellular cGMP 
activates protein kinase II (cGKIl), which in turn stimulates the cystic fibrosis 
transmembrane conductance regulator (CFTR) C\~ channels. This results in a 
net fluid secretion through activation of apical CI" channels in parallel with the 
inhibition of coupled NaCl transporters. Hiese findings have been confirmed in 
GC-C knockout mice, which have a lower intestinal GC-C activity and do not 
exhibit a secretory response to STa treatment [reviewed in 1]. 

STa was the first ligand found to bind GC-C and later studies demonstrated 
that the hormones guanylin and uroguanylin are the natural ligands for this 
receptor. These hormones have been shown to be involved in the regulation of 
fluid and electrolyte transport in many tissues. Guanylin and uroguanylin con- 
sist of 15 amino acids and are highly homologous to STa. 

Toxins with Enzymatic Activity at the Cell Surface 

That Alters Cell Signaling 

Phospholipases 

The first toxin that was recognized to possess an enzymatic activity was 
the Clostridium perfringens a-toxin. This protein is a zinc-dependent phospho- 
lipase C, which degrades phosphatidylcholine and sphingomyelin. Both 
in vitro and in vivo studies have shown that it has cytolytic, dermonecrotic, and 
hemolytic activities, and is lethal to animals at low doses. The toxin causes 
membrane damage to a variety of different human and animal cell types includ- 
ing platelets, leukocytes, and fibroblasts, as well as erythrocytes. It is the major 
toxin involved in gangrene, which is characterized by extensive local tissue 
destruction and necrosis progressing to profound shock and death. The secreted 
protein consists of 370 amino acids (43 kD), and contains 2 domains, an 
a-helical amino-terminal domain (residues 1-246) harboring the active site, 
and a p-sandwich carboxy-terminal domain (residues 256-370), which medi- 
ates membrane binding. The carboxy-terminal domain is structurally similar to 
eukaryoLic calcium-binding C2 domains, which are involved in Ca^"*" -dependent 
phospholipid binding. a-Toxin preferentially binds to phospholipids in the 
intact membrane, opening the active site of the toxin and resulting in cleavage 
of phospholipids [2]. In the activated state, the active site contains two tightly 
bound zinc ions and one loosely bound zinc ion and is accessible for substrate 
binding, whereas in the closed or inactive conformation, the active site is 
occluded and one zinc ion binding site is lost [2-A]. 

In addition to its lytic activity, a-toxin is also involved in intracellular sig- 
naling and the activation of endogenous metabolism cascades. Diacylglycerol and 
ceramide generated from limited hydrolysis of phospholipids and sphingomyelin, 



Popoff 



30 



respectively, activate endogenous phospholipases A2, C and D, and protein 
kinase C. This in turn stimulates membrane phospholipases and initiates the 
arachidonic acid pathway leading to the production of proinflammatory molecules 
(prostaglandins, thromboxanes, and leukotrienes responsible for vasodilatation, 
bronchostriction), and platelet aggregation [4]. 

Other bacterial phospholipases include phospholipase C from Pseudomonas, 
Listeria, and various Clostridium species, phospholipase A from Helicobacter 
pylori, phosphatidyl inositol phospholipase C from Bacillus, Clostridium, and 
phospholipase D from Corynebacterium. 

Bacteroides fragilis Enterotox in 

B. fragilis enterotoxin (BFT) induces morphological changes in cultured 
intestinal and renal cells, including cell rounding, increase in volume, and efface- 
ment of microvilli and apical junctional complexes. BFT has zinc-dependent 
protease activity, which has been shown to cleave the extracellular domain of 
E-cadherin, the primary protein of the zonula adherens. Experimental studies 
have led to the proposed two-step hypothesis, whereby the extracellular domain 
of E-cadherin is cleaved by BFT, followed by intracellular degradation by as yet 
unidentified protease(s). As a consequence, nuclear signaling and actin rearrange- 
ment occur, which leads to the production of proinflammatory cytokines, 
diminished epithelial barrier function, and activation of apical membrane ion 
transporters. These cytotoxic effects are reversible, since 2-3 days after toxin 
treatment cells appear normal [reviewed in 5]. 

Pore- Forming Toxins 

So far more than 80 toxins have been identified that act by forming a trans- 
membrane pore in the target cell. The general mechanism of pore-forming 
toxins (PFT) is to bind to cell surface receptors where they then oligomerize. 
The insertion of the oligomer into the cell membrane results in the formation of 
a channel, which impairs the osmotic balance of the cell and causes cytolysis. 
Most of the PFTs are cytolytic and/or hemolytic and they have been classified 
into several families [for review see 6-8]. 

RTX toxins (repeats in toxin) are synthesized by many gram-negative 
pathogens (Escherichia, Proteus, Pasteurella) . Members of the RTX toxin fam- 
ily, including cytolytic toxins, meta'lloproteases and lipases, share a common 
gene organization and distinctive structural features. They are secreted by 
the type I secretion system which is mediated by the Sec machinery. At the 
carboxy-terminal end, RTX contains 10^0 repeats of glycine- and aspartate-rich 
nonapeptide domains. Most RTX toxins are posttranslationally activated by 
acylation. The prototype of this family is the a-hemolysin (1 lOkD) of £. coli 
and its target receptors on leukocytes have been identified as members of the |32 



Bacterial ExotoxJns 



31 



integrin family. Insertion of a-hemolysin into the membrane, probably mediated 
by four predicted hydrophobic a-hehces in the amino-terminal region, leads to 
the formation of a hydrophilic- and cation-selective pore of at least 1 nm in 
diameter [9], A related family of hemolysins consists of streptolysin S and 
streptolysin S-like cytolysins expressed in streptococci. 

Cholesterol-binding cytolysins are produced by a wide variety of bacterial 
species including Streptococcus, Bacillus^ Clostridium^ and Listeria. Perfringo- 
lysin O (PFO) is one of the best-studied toxins from this family. PFOs are 
secreted as water-soluble monomers, which contain 4 domains rich in [3-strands. 
A short hydrophobic loop in domain 4 is involved in the binding to cholesterol 
[10], After cholesterol binding, PFO undergoes a conformational change result- 
ing in the unfolding of domain 3 a-helices and the formation of two amphipathic 
p-hairpins in each monomer. This leads to an association of neighboring 
monomers and the subsequent formation of a large p-barrel, which then inserts 
into the membrane forming the pore. In general, cholesterol-bindmg cytolysins 
form large pores (300 A) containing about 50 monomers [1 1]. 

Staphylococcus aureus a-hemolysin, aerolysin and the binary staphylo- 
coccal leukocidins, such as LukF, are also synthesized as monomers consisting 
of a very hydrophilic sequence essentially arranged in ^-sheets. Binding of 
monomers to an as yet unidentified cell receptor triggers the heptamerization of 
the toxin, which adopts a mushroom shape with cap, rim and stem domains. The 
amino-terminus detaches from the core monomer unmasking a small hydro- 
phobic surface and assembles with the corresponding domains of the neighboring 
monomers to form the cap. In contrast to PFO, only one antiparallel [S-hairpin 
loop of each monomer unfolds and contributes to the stem formation, which 
consists of l4-stranded p-barrels and results in pores with a small diameter 
(15^5 A) [11, 12]. Aerolysin is secreted as an inactive precursor, which binds 
to aglycosylphosphatidylinositol (GPI)-anchored protein. The toxin is activated 
by cleavage of a carboxy-terminal peptide (40 amino acids) by soluble proteases 
(trypsin or chymotrypsin) or furin. The localization of the aerolysin receptor on 
lipid rafts probably facilitates toxin oligomerization [13]. Clostridium septicum 
a-toxin, which is responsible for gangrene, shares a similar mode of activation 
and pore formation with aerolysin [14]. 

The multicomponent leukocidins and ^-hemolysin from S. aureus also 
assemble in hexamers (1:1 stoichiometry), which form transmembrane pores 
[7]. One component (class S) is involved in the recognition of a cell suiface 
receptor and allows the binding of the other component (class F). The p-toxin 
from C perfiingens, which is involved hi necrotic enteritis, is related to S. aureus 
a- and 7-hemolysin, and triggers pore formation [15]. 

C perfringens enterotoxin is a toxin that causes food poisoning via the 
specific binding of the enterotoxin to receptor(s) from the claudin family, 



Popoff 



32 



present on enterocytes. This complex is then able to associate with additional 
membrane proteins, including occludin, to form larger complexes. It has been 
suggested that these complexes form pores in the plasma membrane, which 
alters the permeability of the plasma membrane for small molecules and 
ultimately causes cell death by lysis or metabolic shut-down [16], 

Superantigens 

A particular class of bacterial toxins referred to as superantigens (entero- 
toxins, toxic shock syndrome toxms from Staphylococcus and Streptococcus) 
are characterized by their ability to bind both MHC class 11 molecules and T cell 
receptors. UnJike conventional antigens that are presented to the T cell receptor 
in complex with the MHC class II molecule, superantigens bind to the T cell 
receptors and MHC class 11 molecules outside the classical antigen-binding 
groove. This results in a massive antigen-independent proliferation of the targeted 
T lymphocytes, leading to the release of various cytokines and inflammatory 
factors [6], 



Intracellularly Active Toxins 

Inhibition of Protein Synthesis 

Diphtheria Toxin - Inactivation of Elongation Factor 2 

Corynebacterium diphtheriae is a human pathogen that normally colonizes 
the throat The bacterium secretes a potent toxin, also known as diphtheria toxin 
(DT), which is one of the most extensively studied and well-understood bacterial 
toxins. Once DT has entered the bloodstream it can affect various organs, caus- 
ing serious complications such as nephritis and cardiac dysfunction associated 
with high mortality rates. DT is a single-chain protein of 58kD encompassing 
three structural and functional domains: a carboxy-terminal domain rich in 
|3-sheets (domain R), which binds to cell surface receptors, a central transloca- 
tion domain containing 9 a-helices (domain T), and the amino-terminal catalytic 
domain consisting of a mixLure of a- and (3-sLrucLures wilh a cleft forming the 
active site (domain C). The toxin is activated by proteolysis at a furin cleavage 
site located in an exposed loop between Cysl86 and Cys201. The amino- 
terminal fragment corresponds to the catalytic domain and remains Imked by a 
disulfide bridge to the rest of the molecule. 

The receptor for DT has been identified as heparin-bhiding epidermal 
growth factor- 1 ike growth factor precursor which forms complexes with other 
membrane components, including CD9, heparin sulfate proteoglycans and inte- 
grins. Epidermal growth factors are synthesized as transmembrane proteins, 
which are subsequently cleaved close to the transmembrane segment to release 



Bacterial ExotoxJns 



33 



DT 



ER 



Golgi 




ADP-ribosylation 




Pore-forming toxin 
CPE 



ST, VT 



Hydrolysis of 
N-glycosidic bond 
in ribosomal RNA 



Leakage of 

nucleotides and 

amino acids 



Fig, 2. Toxins that inhibit proLein synthesis, DT enters the cytosol via the early endosomes 
(EE) and inactivates EF2 by ADP-ribosylation, which results in impaired protein synthesis. 
Pseudomonas exotoxin A (ExoA), Shiga toxm (ST) and E. coli verotoxin (VT) enter cells via 
the Golgi apparatus and ER. While ExoA inactivates EF2, ST and VT impair ribosomal RNA 
function by cleaving an N-glycosidic bond in the 60S subunit. PFT such as C perfhngens 
enterotoxin (CPE) inhibit protein synthesis by inducing leakage of nucleotides, amino acids, 
and other small molecules. 



the active growth factor Once bound to the receptor complex, DT is proteo- 
lytically cleaved by furin and internalized into cells by receptor-mediated endo- 
cytosis via clathrin-coated vesicles. DT is then transported to late endosomes 
and lysozomes where further degradation occurs. An acidification of the early 
endocytic vesicles (pH less than 6) triggers a conformational change in the 
T domain, to form a molten globule structure exposing hydrophobic sites (in 
particular TH5-7 and TH8-9) that insert into the membrane forming cation- 
selective channels. The ammo-terminal fragment of DT is then translocated 
in an unfolded state from the endosomes into the cytosol where it inhibits pro- 
Lein synthesis by ADP-ribosylaLion of elongation factor 2 (EF2) [see details in 
17-21]. 

The DT catalytic domain belongs to a family of mono-ADP ribosyltrans- 
ferases, which bind to NAD and transfer the ADP-ribose group to a specific 
residue on the target protein. The active site is conserved among the bacterial 
ADP-ribosylating toxins. It consists of an a-helix bent over a p-strand, which 
forms the NAD-binding cavity that is flanked by two residues (His and Glu) 
that have a major role in catalytic activity. The ADP-ribosylation of diphtamide 
715 by DT prevents the binding of EF2 to tRNA, resulting in the mhibition of 
protein synthesis (fig. 2). 



Popoff 



34 



Pseudomonas Exotoxin A - Inactivation of EF2 

Pseiidomonas exotoxin A (ExoA) is a 66-kD single-chain protein, which 
shares the same mechanism of action as DT. ExoA is the major virulence factor 
of the opportunistic pathogen Pseudomonas aeruginosa, which often infects 
immunocompromised patients. The toxin is synthesized as a precursor, con- 
taining an amino-terminal signal peptide that directs the polypeptide into the 
type II secretion pathway. The crystal structure reveals three distinct domains: 
an amino-terminal domain consisting of 17 antiparallel (^-strands that recog- 
nizes the cell surface receptor, a central domain composed of 6 oc-helices form- 
ing the translocation domain, and a carboxy-terminal domain containing the 
catalytic site. ExoA binds to Jipoprotein-receptor-related protein (LRP), which 
is a multifunctional scavenger receptor that is expressed by many cell types. 
Upon binding to LRP, ExoA is internalized into the cell by receptor-mediated 
endocytosis. Inside the endosome, the toxin is cleaved by furin, which results in 
two fragments. The enzymatic domain is transported from the Golgi to the 
endoplasmic reticulum (ER), where it is then translocated to the cytosol. In the 
cytosol the enzymatic domain of ExoA catalyzes the ADP-ribosylation of EF2, 
resulting in an inhibition of protein synthesis and ultimately leading to cellular 
death [22, 23] (fig. 2). 

Shiga Toxin - Inactivation of Ribosomal RNA 

Another family of toxins consists of Shiga toxin, Shiga-like toxins, vero- 
toxins, and verocytotoxins which are expressed by several enteric pathogens, 
including Shigella dysenteriae and enterohemorrhagic E, coli. This group of 
toxins plays an important role in the disease pathogenesis of a number of severe 
complications, such as hemorrhagic colitis and the hemolytic uremic syndrome. 

Shiga toxins are composed of a catalytically active subunit (A subunit) and 
a receptor recognition subunit (B subunit). The B subunit that recognizes the 
cell surface receptor globotriosyl ceramide Gb3 consists of 5 B fragments that 
form a symmetrical ring-like structure in solution. The catalytic domain is 
located in the A subunit, which is activated by proteolytic cleavage leading to 
two fragments (Al and A2) that are linked together by a disulfide bridge. 

Several studies have previously shown that Shiga toxin enters the cell by the 
clathrin-dependent pathway and is then transported directly from early/recycling 
endosomes to the Golgi apparatus and then to the ER [24], However, a clathrin- 
independent mechanism has also been described involving lipid rafts [25]. 

Activation of the catalytic domain probably occurs in the trans-Golgi 
network and/or in endosomes by the action of furin^ and to a lesser extent by 
other cellular proteases. The Al fragment is released into the cytosol and inacti- 
vates the 60S subunit of host cell ribosomes by cleaving the N-glycosidic bond 
of adenosine 4324 of the 28S ribosomal RNA of the 60S subunit. This induces 



Bacterial Exotoxins 



35 



a dramatic inhibition of cellular protein synthesis (fig. 2). It has been reported that 
Shiga toxin and verotoxins also cause apoptosis characterized by DNA degrada- 
tion and subsequent cell lysis by an independent mitochondrial pathway [26]. 

Alteration of Cell Homeostasis 

Alteration of Heterotrimeric G Protein Signaling 

Cholera Toxin. Cholera is a serious epidemic disease characterized by 
severe diarrhea and dehydratation, caused principally by the cholera toxin (CT). 
Other members of the CT family are the E. coli heat-labile enterotoxins LT-1 
and LT-1 1. The CT gene is localized to filamentous bacteriophage DNA and can 
be chromosomally integrated or replicated as a plasmid [27]. Similarly, the 
heat-labile enterotoxin genes are located on plasmids (LT-l) or are integrated 
into the chromosome (LT-II) [28]. CT and lethal toxin (LT) subunits are 
exported across the bacterial membrane by Sec proteins and assemble in the 
periplasm. In V. cholerae, CT is actively secreted through the outer membrane, 
while the release of LT-I depends on cell lysis [for a review, see 29]. 

Like Shiga toxin, CT and LTs consist of an A subunit (28 kD) and 5 B 
subunits (1 1 kD each) assembled in a pentamer (AB5 structure). The A subunit 
is proteolytically activated by a ff cholerae endopeptidase into two com- 
ponents Al (approximately 22kJI)) and A2 (approximately 5.5 kD) which 
remain linked by a disulfide bridge. The carboxy-terminal part of A2 extends 
through the central pore of the B pentamer and is linked noncovalently to the 
B subunits. 

CT is internalized into noncoated vesicles after binding of the B subunits 
to ganglioside GMl, which is located at the epithelial cell surface. GMl directs 
the toxin into lipid rafts from where it enters the Golgi via early and late endo- 
somes in a Rab9-dependent pathway [30]. In the perinuclear region of the 
Golgi, the A subunit dissociates from the B subunits and enters the ER via 
coatomer 1-coated vesicles. The carboxy-terminal sequence of the A2 fragment 
contains an ER retention sequence (KDEL), which recognizes the receptor 
Erd2p and directs the Golgi-ER trafficking of CT [31]. B subunits lacking an 
ER retention signal are also LransporLed to the ER, via an unknown niechanism, 
and translocate into the cytosol via the Sec61 complex [32]. The Al fragment 
is responsible for the enzymatic activities of the toxin in the presence of the 
membrane factor Arf. This activity includes NAD hydrolysis of ADP-ribose and 
nicotinamide, and transfer of ADP-ribose to Argl87 of the ot-subunit of stimu- 
latory protein (Gsct), leading to stimulation of adenylcyclase and elevated 
intracellular cAMP. The increased cAMP levels lead to an activation of protein 
kinase A, which subsequently phophorylates numerous substrates in the 
cell [33]. This results in an increase of Cl~ secretion by intestinal crypt cells 
(fig. I) and a decrease of NaCl-coupled absorption by villus cells. 



Popoff 



36 



Pertussis Toxin. Pertussis toxin (PT) is an important virulence factor of 
Bordetella pertussis, the causative agent of whooping cough in humans. PT is 
a hexameric protein consisting of an enzymatic A domain (subunit SI) and five 
binding B domains (subunits S2-S5). Whereas the 5 B subunits of CT are iden- 
tical and arranged in a regular pentamer, the correspondmg PT subunits are 
distinct (n-26kD) and organized as an oligomer (S5-S2-S4-S3-S4). This 
structure forms a disc-like base upon which the pyramid-shaped enzymatic A 
subunit (SI) rests. The different B subunits form a pentameric domain in the 
center of the B oligomer, consisting of a ring of 30 antiparallel p-strands, which 
is surrounded by a barrel of five a-helices. The pore of the barrel is partially 
penetrated by the carboxy-terminus of SI. 

Glycoproteins and glycolipids found on many types of eukaryotic cells 
have been shown to act as a receptor for the B oligomer of PT, seemingly via 
carbohydrate-recognizing domains on subunits S2 and S3. The interaction of 
the toxin with cells of the immune system leads to the induction of lympho- 
cytosis, inhibition of macrophage migration, adjuvant activity, and T cell mito- 
genicity. The T cell mitogenic effect is mediated by the B oligomer and is 
thought to be independent of the S 1 subunit of the toxin. PT possibly undergoes 
a retrograde transport to the ER to deliver SI into the cytosol, although SI does 
not contain an ER retention sequence [34]. 

Internalization of PT is mediated by endocytosis through coated pits, and 
seems to be routed to the late endosome and to the Golgi apparatus. 

The SI subunit of PT, which shares high homology with the enzymatic 
domains of CT and LT, catalyzes the ADP-ribosylation of the inhibitory 
a-subunits of the heterotrimeric GTP-binding proteins (G proteins) involved in 
a variety of signaling pathways. This results in the prevention of the a-subunit 
coupling with the corresponding (3/7-subunits, an increase of adenylcyclase 
activity, which is no longer negatively regulated, and the impairment of several 
second-messenger pathways including an increase in cAMP (fig. 1). 

Adenylcyclase Activity 

Bordelella Adenylcyclase 

The adenylate cyclase toxin (Cya) of 5. pertussis, the whooping cough 
agent, is a major virulence factor required for the early phases of lung coloni- 
zation. Cya is a single-chain 177-kD protein consisting of two domains. The 
toxin is activated after posttranslational palmitoylation of the protein at Lys856 
and Lys963. The enzymatic activity of Cya is located within the proximal 400 
amino acids at the amino-terminus. The carboxy-terminal part, also referred 
to as the hemolysin domain, contains several glycine and aspartate-rich non- 
apeptide repeats that are related to those found in RTX toxins and represent the 
main Ca^"^-binding site of the protein. In addition to its intrinsic hemolytic 



Bacterial Exotoxins 



37 



activity, this domain mediates the binding to and internalization of the toxin into 
eukaryotic cells. CyaA can penetrate a wide range of cell types, including erythro- 
cytes and immune cells. In macrophages, neutrophils and dendritic cells, CyaA 
has been demonstrated to bind specifically a^^2 integrin (CDUb/CDl 8) [35]. 
After internaJization, possibly directly through the plasma membrane, Cya is 
cleaved and the catalytic domain is released into the cytosol, where it increases 
the cAMP levels in a cahnodulin-dependent fashion (fig. 1). The toxin allows 
the pathogen to escape the host immune response by intoxicating neutrophils 
and macrophages, causing phagocyte impotence, and inducing apoptosis [36]. 

Anthrax Edema Toxin 

Anthrax toxin is a tripartite toxin consisting of the protective antigen (PA), 
edema factor (EF), and lethal factor (LP). PA is the binding component, which 
permits the entry of either EF or LF into the cell. The combination PA and EF 
is termed anthrax edema toxin, while PA and LF is termed lethal toxin (a fur- 
ther description of LT can be found in the section Apoptosis below). The genes 
of the three components are localized on a large plasmid (pXOI) present in 
virulent Bacillus anthracis strains. The proteins are secreted by means of a 
signal peptide [reviewed in 37]. 

PA is secreted as an inactive protein (83 kD), which is activated after 
removal of a 20-kD amino-terminal peptide. The cleavage site contains the 
RKKR motif, which is sensitive to proteases such as trypsin or furin. The active 
protein (PA63) has four domains [38]: an amino-terminal domain (domain 1) 
that is relatively hydrophobic and which is involved in the binding of EF orLF, 
a heptamerization domain (domain 2) containing a large amphipathic flexible 
loop implicated in membrane insertion, a small domain of unknown function 
(domain 3), and a carboxy-terminal receptor-bindmg domain which is rich in 
P-strands (domain 4) [39, 40]. The cell surface receptor for PA has been iden- 
tified as a membrane protein containing a von Willebrand factor A domain that 
is located in lipid rafts [41, 42]. Receptor-bound PA is activated at the cell sur- 
face and clusters in lipid rafts, resulting in the formation of PA63 heptamers 
that bind EF or LF. The complex is endocyLosed and transported to endosonial 
compartments where the low pH induces a conformational change in the PA63 
heptamers, leading to its insertion into the membrane and the formation of 
water-filled channels. The translocation of EF and LF into the cell occurs by 
different strategies. While LF is ftilly translocated into the cell cytoplasm, EF 
remains membrane bound, exposing its catalytic domains to the cytosoiic com- 
partment [43]. EF is an adenylcyclase, which is only active when associated 
with calmodulin (fig. 1). Ca^^-bound calmodulin is much more efficient at acti- 
vating EF than the Ca^"^-free form. The catalytic domain of EF is homologous 
with B. pertussis adenylcyclase, and contains the consensus ATP binding motif 



Popoff 



38 



(GxxxxGKS). The conversion of ATP by EF leads to an increase in intracellular 
cAMP levels. These effects are reversible and transient, since EF is instable in 
the cytosol. In human monocytes, EF enhances lL-6 production and inhibits 
LPS-dependent tumor necrosis factor (TNF) synthesis. It has been speculated 
that the main role of anthrax edema toxin is to impair the function of phago- 
cytosing cells such as polymorphonuclear cells and macrophages, which may 
facilitate the early stages of bacterial infection [37]. 

Arrest of Cell Cycle 

Cytolethal distending toxins (CDTs) belong to a recently discovered family 
of toxins, which cause irreversible cell cycle arrest and ultimately death of the 
target cells. CDT was first described in 1987 when certain strains of E. coli 
were found to cause cytopathic effects that were distinct from those induced by 
E. coli toxins such as LT, ST, verotoxin, and hemolysm. Cells that are sensitive 
to CDT first increase in size (3- to 5-fold), followed by a slowly developing cell 
distention, that finally leads to cell death. Apart from E. coli, CDTs are produced 
by a wide variety of gram-negative bacteria including Shigella, Hemophilus 
ducreyi, Actinobacillus actinomycetemcomitans, H. pylori, and Campylobacter 
[44]. Jn E. coli, it has been shown that CDT is encoded by three adjacent or 
slightly overlapping genes, cdtA, cdtB, and cdtC, all of which are required for 
the activity of the toxin. While CdtB contains the enzymatic activity, CdtA and 
CdtC are required for the translocation of CdtB into the target cell. Internali- 
zation of CDT from H. ducreyi occurs via endocytosis mediated by clathrin- 
coated pits. The toxin has been shown to traffic through the Golgi apparatus 
into the cytosol and the nucleus. The proposed mechanisms of action of CDTs 
are not yet fully elucidated; however, it has been reported that the toxin blocks 
cells in the G2 phase of the cell cycle by preventing dephosphorylation of the 
inactive form of cdc2. In addition CdtBs possess DNase I activity that causes 
double-strand DNA breaks (fig. 3) [45]. 

Apoptosis 

Vacuolating Cy to toxin 

The vacuolating cytotoxin (VacA) is one of the most important virulence 
factors produced by H. pylori, a causative agent of severe gastric diseases such 
as ulcers and cancer. VacA has been shown to induce large cytoplasmic vacuoles 
in cultured cells and apoptosis in gastric epithelial and parietal cells. Cleavage 
of the secreted VacA protein (95 kD) results in an amino-terminal 34- to 37-kD 
(p37) and a carboxy-terminal 58-kD (p58) fragment that remain associated with 
each other. The p58 fragment mediates VacA monomer binding to the target cell 
via a GPI-anchored protein, which leads to VacA oligomerization in the membrane 
and the formation of anion-selective channels that release bicarbonate, chloride 



Bacterial Exotoxins 



39 



ADP- 
ribosylation 

C3 
EDIN 



Glucosylation Deamidation Proteolysis 

Transglutamination 

^3® CNF 

clostridial ^^^ YopT 

toxins 



± 




r 



^ 




Rho. Rac, Cdc42 



GDP 
bound 



Bacterial GAPs 

SptP 

YopE 

ExoS 




ADP-ribosylation 

Clostridial binary toxins 

SpvB 



\ 



Bacterial GEF 
SopE 




GTP 
bound 



Glucosylation 




Actin 

filaments 



Actin 
monomers 



/ 



RhoK 



LT 




Ras 



Raf 



Proteolysis 
LF 

J. 

MAPKK 



CDT 



DNase 



Nuclear factors 

Proliferation, differentiation 

— ^ Cell cycle arrest 



Phosphatase 



Cdc25C 



> Cdc2-P 



Fig. J< Bacterial toxins that modify intracellular signaling, actin cytoskeleton 
rearrangement and cell cycle progression. Clostridial binary toxins and other toxins injected 
by the type tit secretion system (SpvB) depolymerize actin filaments by ADP-ribosylation 
of actin monomers. While the large clostridial toxins and C3 inactivate Rho-GTPases and 
YopT impairs the translocation of Rho-GTPases to the membrane, CNF and DNT induce an 
activation of Rho-GTPases. In contrast, SopE and YopE activate Rho-GTPases via a GEF 
activity, or inactivate these molecules through a GAP activity, respectively. These factors are 
involved in the coordinated remodeling of the actin cytoskeleton permitting the bacterial 
invasion and the subsequent restitution of the normal cell architecture after bacterial entry, 
C. sordelUi LT and anthrax LT (LF) downregulate the Ras signaling pathway by glucosylation 
of Ras molecules (LT) or proteolysis of MAPK kinase, whose subsequent molecular mecha- 
nisms and cell effects are still unclear. CDT interfere with the cell cycle through DNase 
activity, which induces DNA damage and subsequent cell cycle arrest. CDT probably also 
acts on the regulation of cyclin-dependent kinase (Cdc2) by converting this molecule to its 
phosphorylated inactive form. 



Popoff 



40 



and urea from the cell cytosol [46, 47]. VacA toxin channels are then internal- 
ized and transported to the late endosomal compartments where they change the 
anion permeability, leading to an enhancement of the vacuolar ATPase proton 
pump activity [40, 41]. It has also been reported that the p34 fragment of VacA 
targets mitochondria leading to the release of cytochrome c, activation of 
caspase 3 and cell apoptosis [48]. 

Anthrax Lethal Toxin 

B. anthracis LT is a zinc metaJ loprotease that causes hyperinflammatory 
conditions in macrophages, the release of reactive oxygen intermediates, and 
secretion of proinflammatory cytokines, such asTNF-a and Lnterleukin- 1 p [49]. 
LF (90 kD) is composed of 4 domains. As discussed for EF, domain 1 (amino 
acids 1-254) consisting of a 12-helix bundle, is involved in the interaction 
with PA. Interestingly, the structure of domain 2 is similar to that of the 
catalytic domain o^ Bacillus cereus VIP2 (vegetative insecticidal protein) and 
C. perfringens iota toxui (see below). However, LF is devoid of ADP-ribosylating 
activity. Domain 3 forms a small helical bundle, which is required for the substrate 
recognition and domain 4 (residues 552-776), consisting of a nine-helix bundle 
packed against a four-stranded (S-sheet, contains the metal loprotease active site 
(HExxH). Analysis of the ci7Stal stinjctui^e revealed that domains 2, 3 and 4 form 
a long deep groove that holds the 16-residue amino-terminal tail of mitogen- 
activated protein kinase kinase 2 (MAPKK-2) [50]. 

Subsequent studies have shown that MAPKK-2 is not the only target for LF, 
since MAPKK-1 to 7 (except MAPKK-5) are also cleaved and inactivated by 
this enzyme [51, 52]. In macrophages, LF also uihibits the extracellular signal- 
regulated kinase (ERK), c-Jun N-terminal kinase (INK), and p38 MAPKs 
pathways (fig. 3). While high concentrations of LF cause cell necrosis, low con- 
centrations (200ng/ml) induce apoptosis m macrophages. However, in order to 
trigger apoptosis, cells have to be activated, for mstance by LPS or other inflam- 
matory mediators. Apoptosis of activated macrophages was found to be dependent 
on p38 inactivation, however, the mechanism is not fully elucidated [53]. 

Alteration of Vesicular Traffic, Blockade of Neuroexocytosis, 

Clostridial Neurotoxins 

The mode of action of botulinum (BoNT) and tetanus (TeTx) neurotoxins 
consists of four steps: binding, internalization, translocation and intracellular 
activation [see also reviews 54—58], BoNT and TeTx recognize specific recep- 
tors on unmyelinated areas of the presynaptic membrane. The precise identity 
of neurotoxm receptors has still to be determined; however, gangliosides from 
the Gib series and synaptic vesicle-associated proteins known as synaptotagmins 
(a family of membrane-trafficking proteins) seem to be involved [59]. 



Bacterial ExotoxJns 



BoNT/B 
TeTx 




Proteolysis 



> 



Inhibition of 
exocytosis 



BoNT/D, F, G 



SNAP25 



VAMP 



Synaptic 
iicle 



.** .... B0NT/CI 
: / \ \ 

_* ■ X 




SynlaxJn 



BoNT/E Bo NT/A 





Assembly of 
SNARE complex 



Fig. 4. Toxins that interfere with vesicular traffic. BoNTs and TeTx are zinc-dependent 
proteases, which cleave SNARE proteins (VAMP, SNAP25 and syntaxin) and result in 
SNARE complexes with a reduced stability. This prevents synaptic vesicles from frjsing with 
the presynaptic membrane. 

Neurotoxin bound to its receptor is internalized by receptor-mediated 
endocytosis. An essential difference between BoNTs and TeTxs is that the BoNTs 
are directly endocytosed in clathrin-coated vesicles, resulting in a translocation 
of the light chain into the cytosol. In the peripheral nervous system, the BoNT 
light chain blocks the release of acetylcholine at the neuromuscular junctions, 
leading to a flaccid paralysis. In contrast, TeTx is sorted to the fast axonal retro- 
grade transport route, and delivered to the motoneurons, which are located in 
the spinal cord. TeTx enters inhibitory interneurons probably via coated vesi- 
cles, permitting the delivery of light chain into the cytosol where it inhibits the 
release of glycine and GABA. 

The light chains of clostridial neurotoxins contain a conserved zinc- 
dependent proteolytic site (His-Glu-x-x-His) with endopeptidase activity [60, 
61 ], It has been shown that the different neurotoxins preferentially target proteins 
belonging to the SNARE (soluble N-ethylmaleirrude-sensitive flision protein 
attachment protein receptors) family, comprising the three membrane-associated 
proteins VAMP/synaptobrevin, SNAP-25, and syntaxin. While TeTx, BoNT/B, D, 
F and G cleave VAMP/synaptobrevin and BoNT/A and E cleave SNAP25, 
BoNT/Cl utilizes both SNAP25 and syntaxm as substrates (fig. 4). Each neuro- 
toxm recognizes its substrate at specific bindmg sites termed SNARE motifs (two 
in VAMP and syntaxin, and four in SNAP25), resulting in a cleavage pattern 
which is characteristic for each toxin. It should be noted that TeTx and BoNT/B 
cleave VAMP at the same site. While SNARE proteins are unstructured in solu- 
tion, when they lie parallel to the membrane surface, they assemble in a ternary 
complex (SNARE complex) consisting of four tightly packed a-helices. 



Popoff 



42 



The SNARE complex is able to recruit a number of soluble cytosolic proteins 
such as NSF (N-ethymaleimide-sensitive factor) and SNAPs (soluble NSF 
accessory proteins). The resulting 20S SNARE complex has been recognized as 
essential in vesicle targeting and fusion. It has been shown that this complex is 
rapidly disassembled by NSF-dependent hydrolysis of ATR Assembly and disas- 
sembly of SNARE proteins within the complex are thought to be essential in the 
exocytosis process. Importantly, clostridial neurotoxins can only cleave SNARE 
proteins when they are disassembled. The cleavage of SNARE proteins by 
clostridial neurotoxins results in a reduction of SNARE complex stability and 
impaired neurotransmitter release. Even though VAMP, SNAP25 and syntaxin 
have different physiological properties at neuromuscular junctions, all clostridial 
neurotoxins cause similar symptoms. However, the intensity and duration of 
neurotransmission inhibition vary depending on the neurotoxin [56^ 57]. 

Alteration ofActin Cytoskeleton and Small G Protein Signaling 

Toxins Active on Actin 

Actin ADP-Ribosylating Toxins. Actin ADP-ribosylating toxins are binary 
toxins which share a common structure, composed of two individual pro- 
teins, a binding/translocation component and an enzyme component, which are 
norJinked and assemble on the target cell. So far three families have been iden- 
tified. The iota family, which encompasses iota toxin, produced by C, perfringens 
type E, Clostridium spiroforme toxin and an ADP-ribosyltransferase synthe- 
sized by some strains of Clostridium difficile. The second family (C2 family) 
contains the C2 toxins expressed by Clostridium botulimim type C and D, which 
have been shown to cause necrotizing enteritis and diarrhea. The third family 
concerns the insecticidal binary toxins or VIP produced by B. cereus and Bacillus 
thuringiensis [62]. 

The binding component binds to the surface of the target cell and is essen- 
tial for the import of the toxin into the cell. For this, the binding component 
has to be activated by protease cleavage. In solution, the binding components 
of iota and C2 toxins (lb and C2-1I, respectively) can be processed by trypsin 
or a-chymotrypsLii. However, unprocessed lb and C2-II can also bind to the 
cell surface receptor, but do not mediate the entry of the enzymatic compo- 
nent. The processed binding component recognizes specific cell membrane 
receptors, heptamerizes and forms small ion-permeable channels that trap the 
enzymatic component into endocytic vesicles. The enzymatic component is 
subsequently translocated into the cytosol [63-66], 

The enzymatic component catalyzes the ADP-ribosylation of actin 
monomers at Argl77 but not of polymerized F-actin, since Argl77 is located in 
the actin-actin binding site. The cumbersome ADP-ribose at the actin-binding 
site prevents the nucleation and polymerization of ADP-ribosylated actin 



Bacterial ExotoxJns 



43 



monomers. Moreover, ADP-ribosylated actin acts as a capping protein, it binds to 
the barbed end of the actin filament and inhibits the further addition of unmodi- 
fied actin monomers. Actin filaments depolymerize at the pointed end and the 
released actin monomers are immediately ADP-ribosylated (fig. 3), In addition, 
ADP-ribosylation inhibits the intrinsic ATPase activity of actin. Cell micro- 
injection of ADP-ribosylated actin monomers induces the same effect as C2 or iota 
toxin. This results in a complete disassembly of the actin filament and accumu- 
lation of actin monomers [67, 68]. While the microtubules are unaffected, the 
intermediate filaments are disorganized. As a consequence cells become rounded, 
detach from the surface, and die [reviewed in 68^ 69]. Studies with epithelial and 
endotheJial cells have shown that clostridial ADP-ribosylating toxins alter the 
tight and adherens junctions resulting in a loss of cell barrier function [70, 71]. 
While toxins of the iota family modify aJl actm isoforms^ including cellular and 
muscular isoforms, C2 toxins only interact with cytoplasmic and smooth muscle 
7-actin. Substrates for VIP have not yet been reported. 

Type III Secretion System-Dependent A DP-Ribosylating Toxins, Nontyphoid 
Salmonella strains that are commonly associated with severe systemic infections 
carry a large plasmid harboring spv genes, which are required for bacterial growth 
in macrophages and monocytes. Among the four-gene operon (spvABCD), it has 
been demonstrated that the spvB gene, encoding a 65,6-kD protein, is essential 
for the virulence phenotype. Based on database searches it has been proposed that 
SpvB has two iunctional domains, an amino-terminal domain related to the insecti- 
cidal toxin Teal from Photorhabdus himinescens with an as yet unknown mecha- 
nism of action, and a carboxy-terminal domain that shares homology with the 
ADP-ribosylating part of iota, C2 and VIP Recombinant SpvB ADP-ribosylates 
nonmuscle actin and microinjection of SpvB into CHO cells causes a breakdov/n 
of actin filaments (fig. 3). In vivo studies have shown that SpvB is crucial for the 
vLnjlence in mice while a mutant strain lacking the spvB gene shows marked 
attenuation of virulence [72]. Evidence has been provided demonstrating that 
SpvB is injected into host cells by a type HI secretion system. Once bacteria have 
entered epithelial cells and macrophages, SpvB is expressed after 6h, and in 
infected macrophages SpvB-dependent cytotoxicity is evident after 10-12h. Like 
SptP, SpvB reverses the actin cytoskeleton reorganization mediating bacterial 
entry, and permits the infected cells to regain their normal architecture after inva- 
sion. Another ADP-ribosyJtransferase toxin that also targets actin and which is 
secreted into the target cell by a type III system has been found in Aeromonas 
salmonicida (AexT) [73]. 

Toxins Activating Small G Proteins 

Enzymatic Modification of the GTPase Site, Some E, coli strains have been 
shown to produce cytotoxic necrotizing factors (CNFs). To date, two variants 



Popoff 



44 



termed CNFl and CNF2 have been characterized. CNFl is synthesized by 
strains mainly isolated from human urinary infections and neonatal meningitis, 
whereas CNF2 is produced by strains that infect animals [74]. 

Both factors are highly homologous at the amino acid level (86% identity) 
and are produced as single-chain proteins with a molecular weight of about 
llOkD. CNFs are related to the dermonecrotic factor (DNT) from Bordetella, 
and homologous sequences to the cnfJ gene have been found in the genomes of 
Yersinia pestis and Yersinia pseudotuberculosis. CNF toxins consist of three 
functional domains: an amino-terminal domain (amino acids 1-299), which is 
involved in the recognition of a cell surface receptor, a central domain (amino 
acids 299-720) containing two hydrophobic regions which have been proposed 
to translocate the toxin across the cell membrane, and a carboxy-terminal 
(720-1,014) catalytic domain. TTie carboxy-terminal domain of CNFl has a 
novel protein fold as determined by crystal structure analysis. This unusual 
compact domain is formed by a central p-sandwich, that is composed of two 
mixed |3-sheets, and surrounded by helices and extensive loop regions [75]. 

CNFl catalyzes the deamidation of Ghi63 in Rho and Gln61 in Rac and 
Cdc42 to glutamic acid. Gln63/Gln61 are located in the switch II region of the 
Rho protein. This region has an important function in the turn-off mechanism 
of RhoGTPases and is essential for GTP hydrolysis by this family of proteins 
[76, 77]. Thereby, CNFl blocks the RhoGTPases in their active form linked to 
GTP. Studies with fibroblasts (Vero cells) have shown that CNFl causes dense 
actin stress fibers and focal contact point formations, whereas in epithelial cells 
(Hep2) the formation of lamellipodia and filopodia predominates. In both cell 
types, CNFl leads to cell spreading resulting from the increase in actin filament 
formation at the leading edge and anchorage of actomyosin filaments to focal 
contact points. This is followed by contraction of these filaments in a similar 
way to that seen in actin-based motility. These findings suggest that in epithe- 
lial cells CNFl first activates Cdc42 and Rac followed by the activation of Rho, 
whereas in fibroblasts activation of Rho is predominant [78]. 

Activation of RhoGTPases by CNFs is only transient and it has been shown 
that deactivation of Rac correlates with an increase in the susceptibility of 
its deamidated form to ubiquitin/proteasome-mediated degradation. During 
the first phase of CNF intoxication, which corresponds to the activation of 
RhoGTPases, uroepithelial cells begin spreading followed by intense membrane 
ruffling. In the next phase of intoxication, lamellipodia are replaced by filopodia, 
cells become highly motile, and there is an alteration in cellularjunction dynam- 
ics. This probably favors bacterial internalization, which requires coordinated 
RhoGTPase activation and inactivation for a maximal efficiency [79]. 

Type III Toxin-Activating RhoGTPases by Guanine Nucleotide Exchange 
Factor Activity. Salmonella enters the cell by a trigger mechanism that induces the 



Bacterial Exotoxins 



45 



formation of large membrane ruffles, which engulf the bacteria. The subsequent 
rearrangements of the actin cytoskeleton and the plasma membrane are reminis- 
cent of lameJIipodia and filopodia responses stimulated by various agonists such 
as growth factors, hormones, or activated oncogenes. It has been demonstrated 
that Cdc42 and to a lesser extent Rac are involved in the Salmon el la-dependent 
cytoskeletal rearrangements. These effects are mediated by SopE, which is 
delivered into the cell by a type III secretion system. Like guanine nucleotide 
exchange factors (GEFs), SopE activates RacI, Rac2, Cdc42, RhoG, and also 
to a lesser extent RhoA by catalyzing the exchange of GDP for GTP [80]. 
Interestingly, SopE2, an isoform of SopE, interacts with Cdc42 but not with 
Racl [81]. 

SopE binds to the switch 1 and switch II regions of Cdc42 and promotes 
guanine nucleotide release. This mechanism is similar to that used by the eukaryo- 
tic Dbl-like exchange factor Tiaml in complex with Racl (fig. 3). However, the 
catalytic domain of SopE has a different structure to that of Tiaml and interacts 
with the switch regions via a GAGA motif [82]. SopE also acts as a GEF for Rab5 
and mediates the recruitment of Rab5 in its GTP form to phagosomes containing 
Salmonella. This promotes the fiision of these phagosomes with early endosomes, 
preventing their transport to lysozomes and subsequent destruction [83]. In addi- 
tion, activation of Cdc42 and Rac by SopE leads to stimulation of p2 1 -activated 
kinase (PAK) and subsequent activation of JNK, the N4AP kinase pathway and a 
number of transcriptional factors [80]. 

Toxins Inactivating Small G Proteins 

ADP-Ribosylating C3 Exoenzyme. C. botulinum C3 exoenzyme belongs 
to the family of Rho-ADP-ribosylating toxins. Other C3-like ADP- 
ribosyltransferases have been identified in S. aureus and B. cereus and are 
termed EDIN (epithelial differentiation inhibitor) and B. cereus exoenzyme, 
respectively. It should be noted that genes encoding EDIN have a higher preva- 
lence in S. aureus strains isolated from infection sites than in strains isolated 
from nasal carriers [84]. 

The C3-like exoenzymes ADP-ribosylaLe Asn41, which is located within 
the (3-strand, align next to the switch 1 region of the Rho-GTPases [85]. However, 
the Asn41 residue is not accessible when Rho is associated with GDI (guanine 
nucleotide dissociation inhibitor), resulting in a protein that is resistant to C3 
exoenzyme ADP-ribosylation. Studies have shown that ADP-ribosylation of 
Rho-Asn41 by C3 exoenzyme does not affect the activity of the protein [86, 87], 
but prevents Rho translocation to the membrane that is required for its activation 
and subsequent interaction with effector molecules [83]. While C3 exoenzyme 
recognizes RhoA, B and C, but not RhoE, EDIN ribosylates all four proteins. 
This results in the disassembly of actin filaments (fig. 3). 



Popoff 



46 



Glucosylating Toxins. Glucosylating toxins, also referred to as large 
clostridial toxins, are proteins with a molecular weight of approximately 
250-300 kD. The family consists of C. difficile toxin A and B (ToxA, ToxB), 
Clostridium sordellii LT and hemorrhagic toxin, and Clostridium novyi a toxin 
(a-novyi). In Clostridium isolates that cause intestinal disease and myonecro- 
sis, the toxins are considered to be the main virulence factors. 

The glucosylating toxins are single chain proteins containing thi^ee func- 
tional domains. In ToxA and ToxB, the carboxy-terminal domains contain mul- 
tiple repeated sequences and are involved in cell surface receptor recognition. 
A trisaccharide (Gal-al 3Gal-pl^GlcNac) has been found to be the motif 
recognized by ToxA. The central domain contains hydrophobic sequences that 
are thought to mediate the translocation of the toxin across the membrane and 
the enzymatic and cytotoxic activity (DxD motif) of the toxins is found at the 
amino-terminus. Sequence analysis has revealed that ToxB and LT are highly 
homologous (76% amino acid sequence identity) and are more distantly related 
to ToxA and a-novyi (48-60% identity) [88]. 

The large clostridial toxins enter cells by receptor-mediated endocytosis. 
The cytotoxic effects are blocked by endosomal and lysosomal acidification 
inhibitors (monensin, bafilomycin Al, ammonium chloride) and the inhibiting 
effects can be bypassed by an extracellular acidic pulse. This indicates that the 
large clostridial toxins translocate from early endosomes upon an acidification 
step. At low pH, ToxB and LT induce channel formation in cell membranes and 
artificial lipid bi layers, and show an increase in hydrophobicity [89, 90]. This 
is thought to involve a conformational change and insertion of the toxin into the 
membrane mediated by the hydrophobic segment of the central domain. 

Large clostridial toxins catalyze the glucosylation of 21-kD G proteins 
using UDP-glucose as the sugar donor (with the exception of a-novyi that prefer- 
entially uses UDP-N-acetylglucosamine) (fig. 3). The toxins transfer the glucose 
or N-acetylglucosamine moiety to the acceptor amino acid Thr37 of Rho or 
Thr35 of Rac, Cdc42 and Ras proteins [91, 92]. Rho complexed to GDI is not a 
substrate for glucosylation, and modified Rho does not bind to GDI [93]. 

The conserved glucosylated Tin* (Thr37/35) is located in switch I of 
Rho/Ras GTPases. Thr37/35 is involved in the coordination of Mg^"^ and sub- 
sequently to the binding of the (3 and -y phosphates of GTR The hydroxy! group 
of Thr37/35 is exposed at the surface of the molecule in its GDP-bound form, 
which is the only accessible substrate for glucosylating toxins. Crystal structure 
analysis of Ras modified by LT shows that glucosylation prevents the forma- 
tion of the GTP conformation in the effector loop of Ras, which is required for 
the interaction with the effector Raf [94]. Similar results were found when 
RhoA glucosylation by ToxB was studied [91]. It has been shown that glucosyl- 
ation of GTPase by the toxins reduces the intrinsic GTPase activity, completely 



Bacterial Exotoxins 



47 



inhibits GTPase-activating protein (GAP)-stimulated GTP hydrolysis, and leads 
to accumulation of the GTP-bound form of Rho at the membrane [93, 95]. 

The modification of Rho proteins by the large clostridial toxins induces 
cell rounding, the loss of actin stress fibers, reorganization of cortical actin, and 
disruption of the intercellular junctions. ToxB and ToxA have been reported to 
trigger apoptosis as a consequence of Rho glucosylation. In addition to the 
effects on the cytoskeleton, the inactivation of Rho proteins impairs other 
cellular functions such as endocytosis, exocytosis, NADPH oxidase regulation, 
and transcriptional activation mediated by JNK and/or p38 [88]. 

Proteolytic Toxins. YopT is one of the six Yop effector proteins which are 
injected into host cells by the Yersinia type HI secretion system. This protein 
inactivates Rho-GTPases leading to the disruption of actin filaments and the 
accumulation of inactive RhoA in the cytosol (fig. 3). Recently, it has been 
reported that YopT is a cysteine protease that cleaves prenylated Rho-GTPases 
near their carboxy-termini and results in the release of these proteins from the 
membrane [96]. 

Rho-GTPases Inactivating Toxins by GAP Activity. As discussed earlier, 
Salmonella enters nonphagocytic cells by delivering effector proteins, such as 
SopE, into the host cell cytosol by the type ill secretion system that directly 
modulates host actin dynamics to facilitate bacterial uptake. Importantly, the 
infected cells quickly recover from the above-mentioned cytoskeletal rearrange- 
ments. It has been shown that the reversal of actin cytoskeleton rearrangements 
is promoted by SptP, another type Ill-secreted protein, which acts as a GAP for 
Cdc42 and Rac. 

SptP is a modular molecule that consists of an amino-terminal domain 
that shares sequence similarity with YopE of Yersinia spp. and ExoS of 
P. aeruginosa and binds to Rac and Cdc42 but not Rho in the GTP-bound 
form. The carboxy-terminal domain is related to YopH and several eukaryotic 
tyrosine phosphatases [97]. Crystal structure analysis revealed that SptP binds 
Racl exclusively through an amino-terminal four-helix bundle domain that 
targets the nucleotide and both the switch I and switch II regions of the 
GTPase. 

Interestingly, eukaryotic GAPs show a larger surface of interaction with 
Rho-GTPases than SptP. This outlines the minimal structure involved in the 
GAP activity and argues for a convergent evolution of eukaryotic and bacterial 
GAPs. SptP binding to Rac does not change the conformation of the carboxy- 
terminal domain (tyrosine phosphatase domain). It is possible that the GAP 
domain targets the tyrosine phosphatase to its relevant substrate(s) [98, 99] and 
it has been speculated that the tyrosine phosphatase activity of SptP is involved 
in the downregulation of the subsequent nuclear response to Cdc42 and Rac 
stimulation [ 1 00]. While SopE is rapidly degraded by the proteasome pathway, the 



Popoff 



48 



degradation kinetics of SptP is much slower, permitting the transient reorgani- 
zation of the actin cytoskeleton involved during bacterial invasion [101]. 

YopE from Yersinia, E\oS and ExoT from F aei^ginosa are secreted into 
macrophages by the type III secretion system and display a GAP activity towards 
Rho-GTPases. Despite the fact that the amino acid sequences are not highly 
conserved among these proteins, the GAP domains of YopE and ExoS show a 
similar structure to that of SptP with a conserved Arg finger that is essential for 
activity [102]. These factors induce actin cytoskeleton disorganization and cell 
rounding, and support the antiphagocytic activity permitting the survival of the 
bacterium [103, 104]. In addition, ExoS exerts an ADP-ribosyltransferase activity 
towards several proteins including Ras [105], 



Concluding Remarks 

Whilst most of the bacterial toxjns form pores that act on cell membranes, 
many of them have the ability to enter host cells and enzymatically modify 
intracellular targets. As discussed in the present review, while some toxins con- 
tain specific translocation domains that attach to the cell membrane forming 
small pores, others lack such domains and are directly injected into cells by a 
type III secretion system. 

Over the last years, evidence has accumulated showing that many bacter- 
ial toxins interfere with physiological processes by modulating host effector 
systems. In contrast to the host, bacteria, however, are not able to regulate these 
cascades, since toxins often act in an uncontrolled manner. This may trigger a 
noxious amplification of the signal and lead to severe systemic complications 
from the infection. Among the numerous potential cellular targets, bacterial 
toxins have only selected some key physiological pathways, such as the inacti- 
vation of EF and ribosomal RNA, leading to the inhibition of protein synthesis, 
as well as interfering with cell homeostasis by stimulating the overproduction 
of secondary messengers. It is also interesting to note that even though the regu- 
lation of acLin polymerization requires a large number of proteins, bacterial 
toxins only act on two essential targets, namely monomeric actin and Rho- 
GTPases. 

Many toxins target the same host effector systems; however, the physio- 
logical effects can differ from species to species. For instance, Clostridium 
secretes actin-modifying exotoxins, which act at a distance from the bacterium 
and disrupt cell barriers and tissues, permitting massive bacterial colonization 
of necrotic tissues. In contrast, some bacteria use specific toxins at the site of 
infection which interfere with the cytoskeleton, facilitatmg the invasion into 
target cells or preventing phagocytosis. 



Bacterial Exotoxins 



49 



Finally, the specificity of bacterial toxins makes these molecules highly 
attractive as potential therapeutic agents (for instance, botulinum neurotoxins 
and ijnmunotoxinsX valuable tools in cell biology and the vectorization of 
molecules into cells. 



References 

Hirayama T, Wada A: Heat-stable enterotoxin of Escherichia coli\ in Aktories K, Just ] (eds): 
Bacterial Prolei a Toxins. Berlin, Springer, 2000, pp 577-593. 

2 Eaton JT, Naylor CE, Howells AM, Moss DS, Titball RW, Basak AK; Crystal structure of the 
C. perfhngens alpha-toxin with the active site closed by a flexible loop region. J Mol Biol 
2002;319:275-28K 

3 Naylor CE, Eaton JT, Howells A, Justin N, Moss DS, Titball RW, Basak AK; Structure of the key 
toxin in gas gangrene. Nat Struct Biol 1998;5:738-746. 

4 Titball RW, Naylor CE, Basak AK: The Clostridium perfringens a-toxin. Anaerobe 1999;5:5 1-64. 

5 Sears CL: The toxins of Bacteroidesfragilis. Toxicon 2001;39:1737-1746. 

6 Alouf JE, MCiller-Alouf H: Staphylococcal and streptococcal superantigens: Molecular, biological 
and clinical aspects. Int J Med Microbiol 2003;292:429-440. 

7 Alouf JE: Moleculai' features of the cytolytic pore forming bacterial protein toxins. Folia Microbiol 
2003;48:5-16. 

8 Menestiina G, Semjen BV: Biophysical methods and model membranes for the study of bacterial 
pore-forming toxins; in Alouf JE, Freer JE (eds): The Comprehensive Sourcebook of Bacterial 
Protein Toxins. London, Academic Press, 1999, pp 287-309. 

9 Ludwig A, Goebel W; The family of the multigenic encoded RTX toxins; in Alouf JE, Freer JE 
(eds): The Comprehensive Sourcebook of Bacterial Protein Toxins, London, Academic Press, 
1999, pp 330-348. 

10 Ramachandran R, Heuck AP, Tweten RK, Johnson AE: Structural insights into the membrane- 
anchoring mechanism of a cholesterol-dependent cyiolysin. Nat Struct Biol 2002;9:823-827. 

1 1 Heuck AP, Tweten RK, Johnson AE: 3-Barrel pore-forming toxins: Intriguing dimorphic proteins. 
Biochemistry 2001;40:9065-9073, 

12 Bhakdi S, Walev NI, Palmer M, Valeva A: Staphylococcal ot-toxin; in Aktories K, Just 1 (eds): 
Bacterial Protein Toxins. Berlin, Springer, 2000, pp 509-527, 

13 Fivaz M, Abrami L, Tsitrin Y, van der Goot FG: Not as simple as just punching a hole. Toxicon 
2001;39:1637-1645. 

14 Tweten RK, Sellman BR: Clostridium septicum pore-forming and lethal a-toxin; in Alouf JE, 
Freer JE (eds): The Comprehensive Sourcebook of Bacterial Protein Toxins. London, Academic 
Press, 1999, pp435^M2. 

15 Tweten RK: Clostridium perfhngens beta toxin and Clostridium septicum alpha toxin: Their 
mechanisms and possible role in pathogenesis. Vet Microbiol 2001 ;82: 1-9. 

16 McClane BA, Rood Jl: Clostridial toxins involved in human enteric and histotoxic infections; in 
Bahl H, Diirre P (eds): Clostridia. Weinheim, Willey-VCH, 2001, pp 169-209. 

17 Murphy JR, vanderSpek J, Lemichez E, Boquet P: Diphtheria toxin and related fusion proteins: 
Autonomous systems for the delivery of proteins and peptides to the cytosol of eukaryotic cells; 
in Moss J, Iglewski B, Vaughan M, Tu AT (eds): Bacterial Toxins and Vimlence Factors in Disease. 
New York, Marcel Dekker, 1 995, vol 8, pp 23^5. 

18 Ren J, Kachel K, Kim H, Malenbaum SE, Collier JR, London E: Interaction of diphtheria toxin 
T domain with molten globule-like proteins and it5 implications for translocation. Science 1999; 
284:955-957. 

19 Hammond K, Caputo GA, London E: Interaction of the membrane-inserted diphtheria toxin 
T domain with peptides and its possible implications for chaperone-like T domain behavior. 
Biochemistry 2002;4 1 :3243-3253. 



Popoff 



50 



20 Lemichez E, Bomsel M, Devilliers G, vanderSpek J, Murphy JR, Lukianov Ey Olsnes S, Boquet P: 
Membrane translocation of diphtheria toxm fragment A exploits early to late endosome trafficking 
machinery. Mol Microbiol I997;23:445^57. 

21 Ratts R, Zeng H, Berg EA, Blue C, McCom ME, Coslello CE, vanderSpek JC, Murphy JR: The 
cytosohc entry of diphtheria toxin catalytic domain requires a host cell cytosolic translocation 
factor complex, J Cell Biol 2003; 160: 11 39- 1 150. 

22 Pizza M, Masignani V, Rappuoli R: Molecular, functional and evolutionary aspects of ADP- 
ribosylating toxins; in Alouf JE, Freer JH (eds): The Comprehensive Sourcebook of Bacterial 
Protein Toxins. London, Academic Press, 1999, pp 45-72. 

23 West SEH: Pseudomonas aeruginosa Exotoxin A: Structure/function, production, and intoxica- 
tion of eukaryotic cells; in Aktories K, Just 1 (eds): Bacterial Protein Toxins. Berlin, Springer, 2000, 
pp 67-89. 

24 Mallard F, Antony C, Tenza D, Salamero J, Goud B, Johannes L: Direct pathway from early/recy- 
cling endosomes to the Golgi apparatus revealed through the study of Shiga toxin B-fragment 
transport. J Cell Biol 1998;143:973-990. 

25 Johannes L, Lamaze C: Clathrin-dependent ornot: Is it still the question? Traffic 2002;3:443^5 1 . 

26 Fuji! J, Malsui T, Heatherly DP, Schlegel KH, Lobo PI, Yutsudo T, Ciraolo GM, Morris RE, ObrigT: 
Rapid apoptosis induced by Shiga toxin in Hela cells. Infect Immun 2003;71:2724— 2735. 

27 Waldor MK, Mekalanos JJ: Lysogenic conversion by a filamentous phage encoding cholera toxin. 
Science I996;272:I91C^19I4. 

28 Holmes RK, Jobling MG, Conell TD: Cholera toxin and related enlerotoxins of gram-negative 
bacteria; in Moss J, Iglewski B, Vaughan M, Tu AT (eds); Bacterial Toxins and Virulence Factors 
in Disease. New York, Marcel Dekker, 1995, pp 225-255. 

29 HLrstTR: Cholera toxin and Escherichia cob' heat-labile enterotoxin; in Alouf JE, Freer JH (eds): 
The Comprehensive Sourcebook of Bacterial Protein Toxins. London, Academic Press, 1999, 
pp 10^129. 

30 Orlandj PA, Fishman PH: Filipin-dependent inhibition of cholera toxin: Evidence for toxin internali- 
zation and activation through caveolae-like domains. J Cell Biol 1998; 141 :905-915. 

31 Majoul I, Sohn K, Wieland FT, Pepperkok R, Pizza M, Hillemann J: KDEL receptor (Erd2p)- 
mediated retrograde transport of the cholera toxin A subunil from the Golgi involves COPI, p23, 
and the COOH terminus of Erd2p. J Cell Biol 1998;143:601-612. 

32 Schmitz A, Herrgen H, Winkeler A, Herzog V: Cholera toxin is exported from microsomes by the 
Sec61p complex, J Cell Biol 2000;148:1203-1212, 

33 Kaper JB, Morris JG, Levine MM: Cholera. Clin Microbiol Rev 1995;8:48-86. 

34 Locht C, Antoine R, Veilhen A, Raze D: Pertussis toxin: Structure-function relationship; in 
Aktories K, Just 1 (eds): Bacterial Protein Toxins. Berlin, Springer, 2000, pp 167-185. 

35 Guermonprez P, Khelef N, Blouin E, Rieu P, Ricciardi-Castagnoli P, Guiso N, Ladant D, Leclerc C: 
The adenylate cyclase toxin of Bordelella pertussis binds to target cells via the aMp2 integrin 
(CDllb/CDI8). J Exp Med 2001;193:1035-1044. 

36 Gueirard P, Druiihe A, Preto)ani M, Guiso N: Role of adenylate cyclase-hemolysin in alveolar 
macrophage apoptosis during Bordetetla pertussis infection in vivo. Infect Immun 1998;66: 
1718-1725. 

37 Leppla SH: Anthrax toxin; in Aktories K, Just I (eds): Bacterial Protein Toxins, Beriin, Springer, 
2000, pp 445^72. 

38 Petosa C, Collier JR, KJimpel KR, Leppla SH, Liddington RC: Crystal structure of the anthrax 
toxin protective antigen. Nature 1997;385:833-838. 

39 BradJey KA, Mogridge J, Mourez M, Collier RJ, Young JAT: Identification of the cellular receptor 
for anthrax toxin. Nature 2001;414:225-229. 

40 Cunningham K, Lacy DB, Mogridge J, Collier RJ: Mapping the lethal factor and edema factor bind- 
ing sites on oligomeric anthrax protective antigen. Proc Natl Acad Sci USA 2002;99:7049-7053. 

41 Mogridge J, Cuningham K, Lacy DB, Mourez M, Collier RJ: The lethal and edema factors of 
anthrax toxin bind only to oligomeric forms of the protective antigen. Proc Natl Acad Sci USA 
2002;99:7045-7048. 

42 Abrami L, Liu S, Cosson P, Leppla SH, van der Goot FG: Anthrax toxJn triggers endocytosis of 
its receptor via a lipid raft-mediated clathrin-dependent process. J Cell Biol 2003;160:321-328. 



Bacterial Exotoxins 



51 



43 Guidi-Rontani C, Weber-Lev^ M, Mock M, Cabiaux V: Translocation of Bacillus anthracis lethal 
and edema factors across endosome membranes. Cell Microbiol 2000;2:259-264, 

44 Cories-Bratti X, Frisan T, Thelestam M: The cytolethal distending toxins induce DNA damage and 
cell cycle arrest. Toxicon 2001;39:1729-1736. 

45 Cortes-Bratti X, Karlsson C, Lagergard T, Thelestam M: The Haemophilus ducreyi cytolethal 
distending toxin induces cell cycle arrest and apoptosis via the DNBA damage check point 
pathways. J Biol Chem 2001;276:5296-5302. 

46 Ricci V, Galmiche A, Doye A, Necchi Y Solcia E, Roquet P: High cell sensitivity to Helicobacter 
pylori VacA toxin depends on a GPl-anchored protein and is not blocked by inhibition of the 
clathrin-mediated pathway of endocytosis. Mol Biol Cell 2000;11:3897-3909. 

47 Montecucco C, Papini E, de Bernard M, Telford JL, Rappuoli R: Helicobacter pylori vacuolating 
cytotoxin and associated pathogenic factors; in Alouf JE, Freer JH (eds): The Comprehensive 
Sourcebook of Bacterial Protein Toxins. London, Academic Press, 1999, pp 264—283. 

48 Galmiche A, Rassow J, Doye A, Cagnol S, Chambard JC, Contamin S, de Thillot y Just 1, Ricci V, 
Solcia E, van Obberghen E, Boquet P: The N-terminal 34kDa fragment of Helicobacter pylori 
cytotoxin targets mitochondria and induces cytochromec release. EM BO J 2000;19:6361-6370. 

49 Hanna P: Anthrax pathogenesis and host response. Curr Top Microbiol Immunol 1998;225: 
13-35. 

50 Pannifer AD. WongTY, Scharzenbacher R, Renatus M, Petosa C, Bienkowska J, Lacy DB, Collier RJ, 
Park S, Leppla SH, Hanna P, Uddington RC: Crystal structure of the anthrax lethal factor. Nature 
2001;414:229-232. 

51 Duesbery NS, Webb CP, Leppla SH, Gordon VM, Klimpel ICR, Copeland TD, Ahn NG, 
Oskarsson MK, Fukasawa K, Paul KJD, Vande Woude GF; Proteolytic inactivation of MAP- 
kinase-kjnase by anthrax lethal factor. Science 1998;280:734-737, 

52 Vitale G, Bernardi L, Napolitani G, Mock M, Montecucco C: Susceptibility of milogen-activated 
protein kinase kinase family members to proteolysis by anthi'ax lethal factor. Biochem J 2000;352: 
739-745. 

53 Park JM, Greten FR, Li ZW, KarLm M: Macrophage apoptosis by lethal factor through p38 MAP 
kinase inhibition. Science 2002;297:2048-2051. 

54 Meunier FA, Schiavo G, Molgo J: Botulinum neurotoxins: From paralysis to recovery of func- 
tional neuromuscular transmission, J Physiol 2002;96:105-1 13. 

55 Schiavo G, Matteoli M, Montecucco C: Neurotoxins affecting neuroexocytosis. Physiol Rev 2000;80: 
717-766, 

56 Humeau Y, Doussau F, Grant NJ, Poulain B: How botulinum and tetanus neurotoxins block 
neurotransmitter release. Biochimie 2000;82:427^M6. 

57 Meunier FA, Herreros J, Schiavo G, Poulain B, Molgo J: Molecular mechanism of action of 
botulinal neurotoxins and the synaptic remodeling they induce in vivo at the skeletal neuro- 
muscular junction; in Massaro J (ed): Handbook of Neurotoxicology. Totowa, Humana Press, 2002, 
vol l,pp 305-347. 

58 Bigalke H, Shoer LF: Clostridial neurotoxins; in Aktories K, Just 1 (eds): Bacterial Protein Toxins. 
Berlin, Springer, 2000, pp 407-443. 

59 Nishiki T, Tokuyama Y, Kamata Y, Nemoto Y, Yoshida A, Sato K, Sekigichi M, Taakahashi M, 
Kozaki S: The high-affinity of Clostridium boiitlinum type B neurotoxin to synaptotagmin 11 asso- 
ciated with gangliosidesGT IB/GDI a. FEBS Lett 1996;378:253-257, 

60 Niemann H: Molecular biology of Clostridial neurotoxins; in Alouf JE, Freer J (eds): Sourcebook 
of Bacterial Protein Toxins. New York, Academic Press, 1991, pp 299-344. 

61 Popoff MR, Marvaud JC: Structural and genomic features of clostridial neurotoxins; in Alouf JE, 
Freer JH (eds): The Comprehensive Sourcebook of Bacterial Portein Toxins. London, Academic 
Press, 1999, pp 1 74-20 L 

62 Warren G, Koziel M, Mullins MA, Nye G, Carr B, Desai N, Kostichka K, Duck N, Estruch JJ: 
Novel pesticidal proteins and strains. World Intellectual Properiy Organization Patent Application, 
WO 96/10083, 1996. 

63 Barth H, Blocker D, Behlke J, Bergsma-Schutier W, Brisson A, Benz R^ Aktories K: Cellular 
uptake of Clostridium botulinum C2 toxin requires oligomerization and acidification. J Biol Chem 
2000;275:18704-18711. 



Popoff 



52 



64 Knapp 0, Benz R, Gibert M, Marvaud JC, Popoff MR: Interaction of Clostridium perfringens 
iota-toxin with lipid bilayer membranes, J Biol Chem 2002;277:6143-6152. 

65 Bachmeyer C, Benz R, Barlh H, Aktories K, Gibert M, Popoff MR: Interaction of Closihdiam 
botutimim C2 toxin with lipid bilayer membranes and Vero cells: Inhibition of channel function in 
chloroquine and related compounds in viti'o and toxjn action in vivo. FASEB J 2001;15:1658-1660, 

66 Stiles BG, Hale ML, Mar\'aud JC, Popoff MR: Clostriditun perfringens iota toxin: Characterization 
of the cell-associated iot^ b complex. Biochem J 2002;367:801-808. 

67 AktoTies KL, Koch G: Modification of actin and Rho proteins by clostridial ADP-ribosylating 
toxins; in Moss J, Iglewski B, Vaughan M, Tu AT (eds): Bacterial Toxins and Virulence Factors in 
Disease. New York, Marcel Dekker, 1995, vol 8, pp 491-520. 

68 Aktories K: Bacterial protein toxins as tools in cell biology and pharmacology; in Cossart P, Boquet 
P, Normark S, Rappuoli R (eds): Cellulaj* Microbiology. Washington, ASM Press, 2000, pp 221-237. 

69 Ohishi I: Stiucture and function of actin-adenosine-diphosphate-ribosylating toxins; in Aktories K, 
Just I (eds): Bacterial Protein Toxins, Berlin, Springer, 2000, vol 145, pp 253-273, 

70 Richard JF, Mainguy G, Gibert M, Marvaud JC, Stiles B, Popoff MR: Transcytosis of iota toxin 
across polarized CaCo-2 ceJI monolayers. MoJ Microbiol 2002;43:907-9J7. 

71 Ermert L, Duncker HR, Bruckner H, Grimminger F, Hansen T, Rossig R, Aktories K, Seeger W; 
Ultrastructural changes of lung capillary endothelium in response to botulinum C2 toxin, J Appl 
Physiol 1997;82:382-388. 

72 Lesnick ML, Reiner NE, Fierer J, Guiney DG: The Salmonella spvB virulence gene encodes an 
enzyme that ADP-ribosylates actin and destabilizes the cytoskeleton of eukaryotic cells. Mol 
Microbiol 2001 ;39: 1464^1470, 

73 Burr SE, Stuber K, Wahli T, Frey J: Evidence for a type ill secretion system in Aeromonas 
salmonicida subsp, salmonicida, J Bacteriol 2002;184:5966-5970. 

74 Boquet P: The cytotoxic necrotizing factor I (CNF I) from Escherichia coli. Toxicon 200l;39: 
1673-1680. 

75 Bluetow L, Flatau G, Chiu K, Boquet P, Ghosh P: Structure of the Rho-activating domain of 
Escherichia coll cytotoxic necrotizing factor J. Nat Struct Biol 2001 ;8:584— 588. 

76 Flatau G, Lemichez E, Gauthier M, Chardin P, Paris S, Fiorentini C, Boquet P: Toxin-induced acti- 
vation of the G protein p2l Rho by deamidation of glutamine. Nature 1997;387:729-733. 

77 Schmidt G, Sehr P, Wilm M, Selzer J, Mann M, Aktories K: Gln63 of rho is deaminated by 
Escherichia coli cytotoxic necrotizing factor- 1. Nature 1997;387:725-729, 

78 Boquet P, Fiorentini C: The cytotoxic necrotizing factor 1 from Escherichia coli; in Aktories K, 
Just 1 (eds): Bacterial Protein Toxins. Beriin, Springer, 2000, pp 361-384. 

79 Doye A, Mettouchi A, Bossis G, Clement R, Buisson-Tonati C, Flatau G, Gagnoux L, Piechaczyk M, 
Boquet P, Lemichez E: CNFl exploits theubiquitin-proteasome machinery to restrict RhoGTPase 
activation for bacterial host cell invasion. Cell 2002; 1 1 1:553-564. 

80 Hardt WD, Chen LM, Schuebel KE, Bustelo XR, Galan JE: Salmonella typhimurium encodes an 
activator of Rho GTPases that induces membrane ruffling and nuclear response in host cells. Cell 
1998;93:815-826. 

81 Friebel A, llchmann H, Aepfelbancher M, Ehrbar K, Machleidt W, Hardt WD: SopE and SopE2 
from Salmonella typhimurium activate different sets of RhoGTPases of the host cell. J Biol Chem 
2001;276:34035-34040. 

82 Buchwald G, Friebel A, Galan JE, Hardt WD, Wittinghofer A, Scheffzek K: Structural basis for 
the reversible activation of a Rho protein by the bacterial toxin SopE. EMBO J 2002;2I: 
3286-3295. 

83 Fujihara H, Walker LA, Gong MC, Lemichez E, Boquet P, Somlyo AV, Somlyo AP: Inhibition of 
RhoA n^nslocation and calcium sensitization by in vivo ADP-ribosylation with the chimeric toxin 
DC3B. Mol Biol Cell l997;8;2437-2447. 

84 Czech A, Yamaguchi T, Bader L, Linder S, Kamisnki KL, Sugai M, Aepfeibacher M: Prevalence of 
Rho-inactivating epidermal cell differentiation inhibitor toxins in clinical Staphylococcus aureus 
isolates. J Infect Dis 200 1 ; 1 84:785-788. 

85 Wei Y, Zhang Y, Derewenda U Liu X, Minor W, Nakamoto RIC, Somlyo AY Somlyo AP, 
Derewenda ZS: Crystal structure of RhoA-GDP and its functional implications. Nat Struct Biol 
1997;4:699-703. 



Bacterial Exotoxins 



53 



86 Ren XD, Bokoch GM, Traynor- Kaplan A, Jenkins GH, Anderson RA, Schwartz MA: Physical 
association of the small GTPase Rho with a 68-kDaphosphatidylinositol 4-phosphate 5-kinase in 
Swiss 3T3 cells. Mol Biol Cell I996;7:435^42. 

87 Sehr P, Gili J, Genth H, Just I, Pick E, Aktories K: Glucosylation and A DP ribosylation of Rho 
proteins: Effects on nucleotide binding, GTPase activity, and effector coupling. Biochemistry 
I998;37:529^5304. 

88 Just I, Hofinann F, Aktories K: Molecular mechanism of action of the large clostridial cytotoxins; 
in Aktories K, Just 1 (eds): Bacterial Protein Toxins. Berlin, Springer, 2000, pp 307-331. 

89 Barth H, Pfeifer G, Hofmann F, Maier E, Benz R, Aktories K: Low pH-induced formation of ion 
channels by Clostridium difficile toxin B in target cells. J Biol Chem 2001;276:10670-10676. 

90 Qa'dan M, Spyres LM, Ballard JD: pH-induced cytopathic effects of Clostridium sordellii lethal 
toxin. Infect Immun 2001;69:5487-5493. 

91 Just I, Wilm M, Selzer J, Rex G, von Eichel-Streiber C, Mann M, Aktories K: The enterotoxin 
from Clostridium difficile (ToxA) monoglucosylates the Rho proteins. J Biol Chem 1995;270: 
13932-13936. 

92 Popoflf MR, Chaves-OJarte E, Lemichez E, Von Eichel-Streiber C, TheJestam M, Chardin P, 
Cussac D, Antonny B, Chavrier P, Flatau G, Giry M, de Gunzburg J, Boquet P: Ras, Rap, and Rac 
small GTP-binding proteins are targets for Clostridium sordellii lethal toxin glucosylation. J Biol 
Chem 1996;271:10217-10224. 

93 Genth H, Aktories K, Just I: Monoglucosylation of RhoA at threonine 37 blocks cytosol mem- 
brane recycling. J Biol Chem 1999;274:29050-29056. 

94 Vetter IR, Hofmann F, Wohlgemuth S, Hermann C, Just I: Structural consequences of mono- 
glucosylation of Ha-Ras by Clostfidium sordellii lethal toxin. J Mol Biol 2000;301:1091-1095. 

95 Hermann C, Ahmadian MR, Hofmann F, Just 1: Functional consequences of monoglucosylation 
of Ha-Ras at effector domain amino acid threonine 35. J Biol Chem 1998;273:16134— 16139. 

96 Shao F, Merritt PM, Bao Z, Innes RW, Dixon JE: A Yersinia effector and a Pseudomonas avirul- 
ence protein define a family of cysteine proteases functioning in bacterial pathogenesis. Cell 
2002;109:575-588. 

97 Fu Y, Gallan JE: A Salmonella protein antagonizes Rac-1 and Cdc42 to mediate host-cell recov- 
ery after bacterial invasion. Nature 1999;401:293-297. 

98 Stebbins CE, Galan JE; Modulation of host signaling by a bacterial mimic: Structure of the 
Salmonella effector SptP bound to Rac 1. Mol Cell 2000;6:1449-1460. 

99 Stebbins CE, Galan JE: Structural mimicry in bacterial virulence. Nature 2001;412:701-705. 

100 Galan JE: Salmonella interactions with host cells: Type HI secretion at work. Annu Rev Cell Dev 
Biol 2001;17:53-86. 

101 Kubori T, Galan E: Temporal regulation of Salmonella virulence effector function by proteasome- 
dependent protein degradation. Cell 2003; 1 15:333-342. 

102 Evdokimov A, Tropea JE, Routzahn KM, Waugh DS: Crystal structure of the Yersinia peslis 
GTPase activator YopE. Protein Sci 2002; 1 1:401^08. 

103 WCirtele M, Wolf E, Pederson KJ, Bucwald G, Ahmadian MR, Barbieri JT, Wittinghofer A: How 
the Pseudomonas aeruginosa ExoS toxin downregulates Rac. Nat Struct Biol 2001;8:23-26. 

104 Kazmierczak BI, Engel JN: Pseudomonas aeruginosa ExoT acts in vivo as a GTPase-activating 
protein for RhoA, Racl and Cdc42. Infect Lnmun 2002;70:2198-2205, 

105 Ganesan AK, Frank DW, Misra RP, Schmidt G, Barbieri JT: Pseudomonas aeruginosa exoenzyme 
S ADP-ribosylates Ras at multiple sites. J Biol Chem 1998;273:7332-7337. 



Michel R. Popoff 

Unite des Bactdries ana6robies et Toxines, Institut Pasteur 

28 rue du Dr Roux, FR-75724 Paris Cedex 15 (France) 

Tel. +33 1 456838307, Fax +33 I 40613123, E-Mail mpopoff@pasteur.fr 



Popoff 



54 



Toxins 

Russell W, Herwald H (eds): Concepts in Bacterial Virulence! 
Contrib Microbiol. Basel, Karger, 2005, vol 12, pp 55-66 



Capsular Polysaccharides and 
Their Role in Virulence 



Clare M. Taylor, Ian S. Roberts 

School of Biological Sciences, University of Manchester, Manchester, UK 



Bacterial pathogens exhibit a number of virulence factors that enable them 
to invade and colonize the tissues of host organisms. A number of these viru- 
lence factors are displayed on the cell surface and include adhesins that medi- 
ate attachment to host cells, toxins that may be secreted resulting in host tissue 
damage, and the possession of molecules that render them resistant to host 
antimicrobial defences. Capsular polysaccharide (CPS) has long been recognized 
as an important virulence determinant in isolates capable of causing infection 
in humans and animals [1]. CPS is found on the outermost surface of a wide 
range of the bacteria [2] and may be linked to the cell surface via covalent 
attachments to phospholipid or lipid A molecules [3]. In contrast, extracellular 
polysaccharide (EPS) molecules appear to be released onto the cell surface with 
no visible means of attachment. Such EPS can be loosely associated with the 
cell surface and easily sloughed off as slime. 

CPS molecules are highly hydrated and typically constitute more than 95% 
water [4]. They are composed of repeating single monosaccharide units that are 
joined by glycosidic linkages. CPS may be homo- or heteropolymers and can be 
substituted with both organic molecules such as acetyl groups, and inorganic mole- 
cules such as phosphate. In addition, two monosaccharides may be joined m a 
number of configurations due to the presence of multiple hydroxyl groups within 
each monosaccharide that may be involved in the glycosidic linkage. Thus, CPS 
are a diverse range of molecules that can differ not only in their constituent mono- 
saccharides but also in the manner in which they are joined. This diversity is illus- 
trated in bacterial species such as Escherichia coli where over 80 distinct capsular 
serotypes have been described while in Streptococcus pneumoniae , there are over 
90 capsular serotypes. The introduction of branches and substitution with organic 
or inorganic molecules to polysaccharide chains adds a further layer of structural 
complexity. However, chemically identical CPS may also be synthesised by 



different bacterial species. The group B capsule of Neisseria meningitidis, a homo- 
polymer of ct2, 8-1 inked N-acety I neuraminic acid (NeuNAc), is identical to the Kl 
antigen oiE. coli [5], while the CPS of Pas teu re I la muitocida type D is identical 
to the E. coli K5 capsule which comprises repeating disaccharides of glucuronic 
acid linked to N-acetylglucosamine [6]. The apparent conservation of particular 
CPS structures between taxonomically diverse genera of bacterial species raises 
intriguing questions regarding the evolution of capsule diversity and the acquisi- 
tion of capsule biosynthesis genes. 



Functions of Bacterial Capsules 

As the polysaccharide capsule represents the outermost layer of the bacte- 
rial cell, it is not surprising that the capsule mediates interactions between the 
bacterium and its immediate environment. Accordingly, a number of functions 
has been ascribed to bacterial capsules. Each of these functions (resistance to 
desiccation, adherence, resistance to nonspecific host immunity, resistance to 
specific host immunity) is directly relevant to pathogenicity and as such con- 
tributes to the role of CPS as a virulence factor. 

Resistance to Desiccation 

As CPS are highly hydrated molecules that surround the cell surface, they 
may protect bacteria from the harmful effects of desiccation [7]. This property is 
probably most relevant in the transmission and survival of encapsulated bacteria 
in the environment demonstrated in the cases of isolates off. coli, Acinetobacter 
calcoaceticus and Erwlnia stewartii, which have been shown to be more resis- 
tant to desiccation than their isogenic acapsular mutants [8]. Furthermore, the 
capsule probably provides protection during transmission from host to host. In 
the case of E. coli, genes encoding enzymes for the biosynthesis of capsular 
colanic acid have been shown to be upregulated m response to desiccation [8]. 
While the mechanism of regulation is unclear, it is thought that external osmo- 
larity is altered during desiccation, and it has been shown tliat expression of 
alginate EPS of Pseudomonas aeruginosa as well as expression of the Vi CPS 
of Salmonella typhi, which is essential for virulence, are increased in response 
to high osmoJarity [9, 10]. 

Adherence 

CPS may mediate adhesion of bacteria to surfaces (both biotic and abiotic) 
and to each other. Adhesion to abiotic surfaces may result in the establishment of 
biofihns and EPS-mediated interspecies co-aggregation within biofilms can 
enhance colonization of various ecological niches [11]. In addition, growth of 



Taylor/Roberts 



56 



bacteria as a biofilm may offer some protection from phagocytic protozoa and pre- 
sent nutritional advantages, while it is thought that the presence of EPS acts as a 
permeabiMty barrier against antimicrobial agents [12]. While adhesion to host tis- 
sues is undoubtedly a multifactorial process involving an array of bacterial surface 
components, CPS has been implicated in the adhesion of a number of human 
pathogens to host tissues. Streptococcus pyogenes or group A Streptococcus 
(GAS) is responsible for a range of clinical infections including skin infections, 
acute rheumatic fever, streptococcal pharyngitis, streptococcal toxic shock syn- 
drome and necrotizing fasciitis [13, 14]. In the development of pharyngitis, colo- 
nization of the pharynx by streptococci not only represents a vital stage in the life 
cycle of GAS, but it is also likely that that the pharynx serves as a reservoir for 
infection from which GAS may be disseminated to other hosts as well as causing 
invasive infections such as necrotizing fasciitis. It has been demonstrated that 
the hyaluronic acid capsule of GAS binds to CD44 molecules on the surface of 
human keratinocytes, the predominant cell type in skin and the pharyngeal 
epithelium [15]. Once bound, bacterial contact with the epithelial surface 
induces lamellipodia formation on the surface of keratinocytes, which is not 
observed in an isogenic acapsular mutant [16]. Gram-negative pathogens such 
as Salmonella and Shigella spp. also induce lamellipodia formation following 
binding to host epithelial cells; however subsequent fusion of the lamellipodia 
entraps the bacteria, resulting in their internalization. GAS are inefficiently 
internalized as a consequence of the possession of their hyaluronic acid cap- 
sule. Furthermore, the binding of GAS to CD44 induces marked cytoskeletal 
rearrangements and cell signalling events leading to the opening of intercellular 
junctions, which is thought to promote tissue penetration by GAS [16]. Clearly 
this is not the case for all encapsulated pathogens, as the case of GAS involves 
molecular mimicry, with the CPS being identical to host hyaluronic acid. In 
other pathogens, initial attachment to host cells has been shown to be inhibited 
by encapsulation, as is the case for binding oi Klebsiella pneumoniae to epithelial 
cell lines in vitro [17]. Paradoxically, encapsulated isolates of the same straui 
adhered better to a mucus-producing cell line than an acapsular mutant. These data 
suggest tliat in some cases the CPS may promote initial colonization of the mucus 
layer, while subsequent interaction with the underiying epithelial layer is reduced 
by the presence of a capsule, presumably due to the masking of bacterial 
components required for specific interaction with the epithelial surface. These 
observations support the notion that there is some form of co-ordinate regulation 
of capsule expression during the early stages of infection. 

Resistance to Non-Specific Host Immunity 

During invasive infections of humans and animals by encapsulated 
pathogens, interactions between the bacterial CPS and immune system of the 



Capsules and Virulence 57 



host play a critical role in determining the fate of the infection [18]. During 
an innate host response, the bacterial capsule may confer some resistance to 
complement-mediated killing. The main function of the complement system is 
the binding of host peptides to foreign organisms. Once bound, these are rec- 
ognized by specific complement receptors on host phagocytes that facilitate 
opsonization and subsequent destruction. Thus, activation of the complement 
cascade involves an array of serum and cell surface proteins and three pathways 
of activation are recognized. In the classical pathway, an antibody response is 
generated, while the alternative pathway can be activated in the absence of spe- 
cific antigen-antibody recognition. The mannan-binding lectin pathway recog- 
nizes surface polysaccharides and then activates the complement cascade [19]. 
These pathways generate C3 convertases that cleave C3 (the major complement 
component) to C3b, which can then bind to the cell surface. Factor C3b and its 
degradation product iC3b are the primary complement opsonins [20]. In the 
absence of specific antibody, CPS is thought to activate the alternative pathway 
in which C3b binds non-specifically to the bacterial surface. Bound C3b is then 
activated by interaction with factor B and forms the C3 convertase C3bBb, 
which binds to the bacterial surface along with further C3b. This complex 
termed C3b2Bb acts as the C5 convertase and promotes formation of the mem- 
brane attack complex (MAC), which can form pores in certain bacteria, causing 
their destruction. 

CPS that contain NeuNAc are known to be poor activators of the alternative 
pathway [21 , 22] and it is thought that this is because NeuNAc binds directly to 
factor H [21]. Bound factor H promotes the binding of factor 1 to C3b, forming 
iC3b, which breaks the amplification loop of the cascade, which in turn prevents 
formation of the MAC [23]. In such cases, the bacterial capsule usually acts in 
concert with other surface structures such as the 0-antigen of lipopoly saccharide 
to confer resistance to complement-mediated killing [24]. Thus, a particular 
combination of surface structures can confer a high degree of resistance to the 
innate immune response. In the case of other encapsulated pathogens, it is thought 
that the presence of a CPS may actually provide a barrier to complement compo- 
nents by pJiysically maskhig underlying surface stiiictures that would normally be 
potent activators of the alternative pathway [24]. 

Finally, CPS may confer resistance to complement-mediated opsonophago- 
cytosis. In the case of Staphylococcus aureus^ the presence of a thick capsule has 
been shown to be antiphagocytic, as it interfered with recognition of cell-bound 
C3b and iC3b by phagocytic receptors [25]. Shnilar observations have been 
made in the case of yS*. pneumoniae where CPS also appears to block cell-bound 
C3b [26]. Furthermore, many CPS are highly negatively charged molecules and 
may also confer resistance to phagocytosis [1, 27, 28]. In addition to these direct 
interactions between CPS and components of the complement system, certain 



Taylor/Roberts 



58 



CPS may modulate the host's immune system by stimulating the release of 
certain cytokines resulting in the disruption of the cell-mediated immune response 
[29]. One such example is the CPS of K. pneumoniae, which was shown to 
induce high levels of interleukin-lO (IL-10) in experimentally infected mice, in 
contrast to an acapsular mutant [30]. High levels of lL-10 inhibit gamma 
interferon-induced activation of macrophages, and therefore cell-mediated 
reactions such as delayed-type hypersensitivity, which are normally visible 
24—48 h after infection. 

Resistance to Specific Host Immunity 

Although many CPS elicit a specific (antibody-mediated) immune response 
in the host, a certain small set of CPS are able to confer some resistance. 
Capsules such as those that contain NeuNAc, e.g. E. coli K 1 and N. meningitidis 
serogroup B [31] in addition to the E. coli K5 polysaccharide which is identical 
to N-acetyl heparosan (precursor in heparin/heparan sulfate biosynthesis) [32], 
are poorly immunogenic. Infected individuals only mount a poor immune 
response to these antigens as a consequence of the structural similarities of these 
capsules to host polysaccharides encountered abundantly in the extracellular 
matrices [18, 33]. As a result, the expression of these capsules that mimic host 
structures provides protection against the specific arm of the host's immune 
response. 



Polysaccharide Capsules of Pathogenic E. coll 

A large number of capsule gene clusters, representing various capsular 
serotypes, have been identified and cloned from a number of gram-negative 
pathogens. In all cases, the capsule genes are clustered at a single locus allow- 
ing for the co-ordinate regulation of capsule gene expression. Each of the cap- 
sule serotypes appears to be represented within E. coli, and to date, E. coli 
capsules are amongst those most intensively studied. Thus capsule gene clus- 
ters of E. coli are regarded as a paradigm for capsule gene clusters in gram- 
negative bacteria. 

As previously mentioned, over 80 different serologically and chemically 
distinct types of polysaccharide capsule have been described in E. coli [34]. 
Termed K antigens, these have been classified into four functional groups 
(table 1) based on a number of biochemical and genetic criteria [35]. Most 
pathogenic extra-intestinal E. coli express group 2 K antigens [2]. Group 2 
CPS represent a heterogeneous group concerning composition, while in 
terms of structure and cell surface assembly they resemble the capsules of 
other gram-negative pathogens, N. meningitidis and Haemophilus influenzae. 



Capsules and Virulence 59 



Table L Classification of £. coli capsules [adapted from 34] 



Characteristics 


Group 










1 


2 


3 


4 


Foriner K antigen 


lA 


11 


1/n or 111 


IB (0-antigen 


group 








capsules) 


Co-expressed with 


Limited range 


Many 


Many 


Often 08, 09 but 


serogroups 


(08,09,020,0101) 






sometimes none 


Co-expressed with 


No 


Yes 


Yes 


Yes 


colanic acid 










J hermostabiJity 


Yes 


No 


No 


Yes 


lerminal lipid moiety 


Lipid A core 


OL- 


ot- 


_.ipid A core in 




in Klps; unhiown 


G ycerophosphate 


Glycerophosphate 


Klps; unknown 




for K antigen 






for K antigen 


Direction of chain 


Reducin:^ terminus 


Non-reducing 


Non-reducing 


Reducing 


growth 




terminus 


terminus? 


terminus 


Polymerization system 


Wzy-dependent 


Processive 


Processive? 


Wzy-dependent 


Transplasma 


Wzx (PST) 


ABC-2 exporter 


ABC-2 exporter? 


Wzx (PST) 


membrane export 










E evated levels o:^ 


No 


Yes 


No 


No 


CMP-Kdo synthetase 










Genetic locus 


cps near his and rfb 


kps near serA 


kps near serA 


rfb near his 


Thermoreeulated (not 


No 


Yes 


No 


No 



expressed below 20°C) 



Positively regulated 


Yes 


No 


No 


No 


by Res system 










Model system 


K30 


K1,1C5 


KIO, K54 


K40, 0111 


Sijnilar to 


Klebsiella^ Erwinia 


Neisseria, 
Haemophilus 


Neisseria, 
Haemophilus 


Many genera 



A model for assembly and attachment of group 2 capsules to the cell surface 
is shown in figure I . 



Genetic Organization and Regulation of £. co/i Group 2 
Capsule Gene Clusters 

A number of group 2 capsule gene clusters have been cloned, and analysis 
has revealed that they have a conserved modular genetic organization, consisting 



Taylor/Roberts 



60 



Biosynthesis of 
Phosphatidyl-Kdo (PA-Kdo) 



Ligation of PA-Kdo 





^ 



Polymerization 



initiation on an 
acceptor 



OM 




o 




Phosphatidic acid 

2-Keto-deoxymanno-octonic 
acid (Kdo) 

Repeat monosaccharide 



Fig. L A model for the assembly of group 2 capsules. Polysaccharide is poJymerized at 
the non-reducing terminus and subsequently ligated to phosphatidyl-Kdo (PA-Kdo) at the reduc- 
ing end prior to export across the cytoplasmic membrane. The presence of PA-Kdo nnay act as 
a motiF for the export proteins as structurally diverse group 2 polysaccharides are all exported 
via the same conserved export proteins. LM = Inner membrane; OM = outer membratie. 



of three functiona] regions (fig. 2). Furthermore it appears that this modular 
organization is applicable to capsule gene clusters of other bacteria [2]. Gene 
expression is achieved following transcription from two convergent promoters 
PI and P3 which flank regions I and 3, respectively. Regions I and 3 are con- 
served amongst group 2 gene clusters and encode proteins necessary for the 
transport of the polysaccharide from its site of synthesis to the cell surface. 
Region 2 is serotype specific and encodes the enzymes responsible for biosyn- 
thesis (where necessary) and polymerization of the individual monosaccharides 
that comprise the particular polysaccharide. The size of this region is variable; 
however, size is thought to reflect the complexity of the polysaccharide, as 
region 2 in isolates that produce CPS with complex structures often encodes a 
larger number of open reading frames [36]. 

Region 1 comprises 6 genes kpsFEDUCS organized in a single transcrip- 
tional unit (fig. 2) that encode proteins involved in transport of the polysaccharide. 
A single E. coli of^^ promoter (PI) has been mapped 225 bp upstream of /g^^Fand 
transcription from PI generates an 8.0-kb polycistronic transcript that is subse- 
quently processed to generate a stable 13-kb A??55'-specific transcript [37]. This 
may facilitate the differential expression of KpsS, which may mfluence the 
attachment of phosphatidyl-Kdo (2-keto-3-deoxymanno-octonic acid) to nascent 



Capsules and Virulence 



61 





H-NS/BipA 




-4- 



> "♦■ 






kps F E D U C 



T M 



kfi D C 



B 



A 



Fig. 2. Genetic organization and regiilarion of £". coli group 2 capsule gene clusters. In 
this example, the gene cluster of £'. coli K5 is shown. The numbers at the top refer to the three 
functional regions; the serotype-specific region, region 2, is shaded. PI and P3 represent the 
region 1 and 3 promoters, respecrively, and the straight arrows denote the major transcripts. 



polysaccharide and regulate its entry into the export machinery. An intragenic 
Rho-dependent transcriptional terminator has also been identified within fqysF, 
This may play a role in regulating transcription by preventing synthesis of 
untranslated region 1 transcripts under conditions of physiological stress [38]. 

Region 3 of the gene cluster contains two genes kps M and /^^ 7" organized 
in a single transcriptional unit [2, 39]. The promoter (P3), which has a typical 
E. coli a70 -10 consensus sequence but no -35 motif, has been mapped 741 bp 
upstream of the initiation codon of I<psM, No consensus binding sites for other 
alternative a-factors or other DNA-binding proteins have been identified [40]. 
However, region 3 is subject to control by an antitermination process, conferred 
by RfaH and ops elements. A cis-act'mg regulatory sequence termed ops, which 
is essential for the function of RfaH has been identified 33 bp upstream of the 
initiation codon of kpsM [40]. The ops element^ with the sequence GGCGGTAC, 
is contained within a larger regulalory element of 39 bp termed JHLTMPsLart (just 
upstream from many polysaccharide-associated gene starts) [41]. RfaH is known 
to regulate a number of gene operons in E. coli including the hemolysin operon 
and the gene clusters for LPS core and 0-antigen biosynthesis [42, 43]. In addi- 
tion, RfaH is a homolog of NusG, an essential transcription elongation factor 
that is necessary for Rho-dependent transcription termination and bacteriophage 
\-N-mediated antitermination. RfaH is thought to act as a transcriptional elon- 
gation factor that allows transcription to proceed over long distances. As such, 
mutations in rfaH gwQ rise to increased transcription polarity throughout RfaH- 
regulated operons without disrupting initiation from operon promoters [42]. 



Taylor/Roberts 



62 



To act, ops elements must be located on the nascent mRNA transcript, where they 
recruit RfaH, and perhaps other proteins, promoting transcription elongation. It 
is thought that the JUMPstart sequence on the mRNA molecule may permit the 
formation of stem-loop structures at the 5' end, which mediate the interaction 
with RfaH [41]. A mutation in rfaH or deletion of the JL'MPstart sequence has 
been shown to abolish capsule production in E, coli Kl and K5 [40] and serves 
to confirm the role of RfaH in the regulation of group 2 capsule gene clusters. 

The genetic organization of region 2 is serotype specific and differs among 
group 2 K antigens. In the case of K5, region 2 comprises 4 genes kfiABCD 
[44] while Kl comprises 6 genes neiiDBACES [39], In each case, transcription 
of region 2 proceeds in the same direction as that of region 3, which is impor- 
tant in the RfaH-mediated regulation of region 2 expression [40]. 

A feature of group 2 capsule gene expression pertinent to pathogenicity is 
that transcription from both PI and P3 is temperature regulated, enabling capsule 
expression at 37°C but not at 18°C [37, 45]. Temperature regulation is in part 
achieved via the action of the global regulatory protein H-NS (histone-like 
nucleoid-associated protein), since hns mutants show comparable levels of tran- 
scription from PI at both 18 and 37°C^ albeit lower than those usually seen in a 
wild-type strain at 37''C, indicating that H-NS is required for maximal transcrip- 
tion at 37°C as well as repression at 1 8°C [46]. This situation is analogous to the 
H-NS-mediated thermoregulation of the virB promoter in Shigella flexneri. In 
this system however, activation of the virB promoter has an absolute requirement 
for the AraC-like protein VirF [47]. It is not yet clear whether an AraC-like tran- 
scriptional activator is involved in modulating transcription from PI and P3. 

Mutations in bipA also result in increased transcription at 18°C and reduced 
transcription at 37°C [44]. Although this phenomenon mirrors the effect of muta- 
tions in hns^ the phenotype of a bipA mutant cannot be explained by a loss of 
H-NS function as this is unaffected in a bipA mutant. BipA was first described as 
a tyrosine-phosphorylating protein in enteropathogenic E, coli (EPEC) [48]. 
EPEC bipA mutants are unable to trigger cytoskeletal rearrangements in host cells, 
are hypersensitive to BPI (bactericidal/permeability-increasing protein) protein 
and demonstiate increased flagella expression and motility [48], Fuithermore, 
BipA is a GTPase with similarity to the TetO resistance protein and elongation 
factor G (EF-G), both of which interact with ribosomes. These data have led to 
the suggestion that BipA may represent a novel class of regulators that interact 
directly with the ribosome by regulating translation elongation [48]. It is therefore 
likely that BipA does not regulate PI and P3 directly, but that regulation is 
achieved via interaction with other proteins that do modulate transcription of PI 
and P3. This hypothesis is currently under investigation in our laboratory. 

At 37°C, the mechanism of temperature regulation is further complicated 
by the interaction of integration host factor (IHF) with PI . IHF is required for 



Capsules and Virulence 



63 



optimal capsule gene expression and IHF binding sites have been identified that 
flank P I [40], IHF usually acts to facilitate the activity of other regulatory proteins 
[49] and as such it is likely that LHP also acts in concert with an as yet unidenti- 
fied regulatory protein or proteins that act to control transcription from regions 
1 and 3 at 37°C. However, the lack of IHF binding sites in the region 3 promoter 
[40] demonstrates that the requirement for IHF is not absolute, 

Ln summary, the regulation ofE. coli group 2 capsules is complex, hivolving 
a number of overlapping regulatory circuits. However, of relevance to patho- 
genicity and virulence, there are still many unanswered questions. How are 
changes in temperature, such as those concomitant with entry into a susceptible 
host, sensed and transduced to induce changes in gene expression? How is cap- 
sule gene expression regulated in response to attachment and interaction with 
host cells? While it is known that encapsulation is an important virulence fac- 
tor, understanding the regulation of capsule expression during the stages of 
infection still represents an interesting challenge. One fiirther important area 
for fijture research is the understanding of the export of CPS onto the bacterial cell 
surface. Such understanding will lend itself to the design of chemotherapeutic 
agents targeted to selectively disrupt capsule export and therefore combat infec- 
tions caused by encapsulated bacteria. 



References 

1 Moxon ER, Kroll JS: The role of bacterial polysaccharide capsules as virulence factors. Curr 
Microbiol Immunol 1990;21:221-231. 

2 Roberts IS: The biochemistry and genetics of capsular polysaccharide production in bacteria, 
Annu Rev Microbiol 1996;50:285-315. 

3 Whitfield C, Valvano M: Biosynthesis and expression of c^ 11 -surface polysaccharides in gram- 
negative bacteria. Adv Microbio] Phys 1993;35:135-146. 

4 Costerton JW, Irvin RT, Cheng K-J: The bacterial glycocalyx in nature and disease. Annu Rev 
Microbiol 1981;35:299-324. 

5 Grados O, Ewing VM: Antigenic relationships between Escherichia co// and Neissena memngitidis 
group B. J Infect Dis 1970;122:100-103, 

6 DeAngelis PL, White CL: Identification and molecular cloning of a heparosan synthase from 
Pasieurella muttocida type D. J Biol Chem 2002;277:7209-7213. 

7 Roberson E, Firestone M: Relationship between desiccation and exopolysaccharide production in 
soil Pseudomonas sp. Appl Environ Microbiol 1992;58: 1284— 1291. 

8 Ophir T, Gutnick D: A role for exopolysaccharides in the protection of micro-organisms from 
desiccation. Appl Environ Microbiol 1994;60:740-745. 

9 Berry A, De Vault J, Chakrabaj'ty A: High osmolarity is a signal for enhanced algD transcription 
in mucoid and nonmucoid Pseudomonas aetvginosa strains. J Bacteriol 1989;171:2312-2317. 

10 Pickard D, Roberts M, Maskell D, Hone D, Levine M: Characterization of defined ompR mutants 
of Salmonella typhi: OmpR is involved in the regulation of Vi polysaccharide. Infect Immun 
1994;62:3984-3993. 

1 1 Costerton JW, Cheng K-J, Geesey GG, Ladd TI, Nickel JC, Dasgupta M, Marrie T: Bacterial 
biofilms in nature and disease. Annu Rev Microbiol 1987;41:435-464. 



Taylor/Roberts 



64 



12 Costerton JW, Stewart PS^ Greenberg EP: Bacterial biofilms: A common cause of persistent 
infections. Science 1999;284:1318-1322. 

13 Hoge CW, Scwartz B, Talkington DF, Breiman RF, MacNeill EM, Englender SJ: The changing 
epidemiology of invasive group A streptococcal infections and the emergence of streptococcal 
toxic shock-like syndrome. A retrospective population-based study. JAMA 1993;269:384-389. 

14 KauJ R, McGeer A, Low DE, Green K, Scwartz B: Population based surveillance for necrotizing 
fasciitis: Clinical features, prognostic indicators, and microbiologic analysis of seventy-seven cases. 
Ontario Group A Streptococcal Study. Am J Med 1997;103:18-24. 

15 Cywes C, Stamenkovic T, WesseIsM: CD44 as a receptor for colonization of the pharynx by group 
A Streptococcus. J Clin Invest 2000;106:995-1002. 

16 Cywes C, Wessels M: Group A Streptococcm tissue invasion by CD44-mediated cell signalling. 
Nature 2001;414:648-652. 

17 Favre-Bonte S, Joly B, Forestier C: Consequences of reduction oi Klebsiella pneumoniae capsule 
expression on interactions of this bacterium with epithelial cells. Infect Immun 1999;67:554-561. 

18 Roberts IS, Saunders FK, Boulnois GJ: Bacterial capsules and interactions with complement and 
phagocytes. Biochem Soc Trans 1 989; 1 7:462^64. 

1 9 Frank MM, Fries LF: The role of complement in the defense against bacterial disease; in Bailliere's 
Clinical Immunology aiid Allergy. Philadelphia, BailliereTindall, 1998, vol 2, pp 335-361. 

20 Morgan BP: Physiology and pathophysiology of complement: Progress and trends. Crit Rev Clin 
LabSci 1995;32:265-298. 

21 Michaiek M, Mold C, Bremer E: Inhibition of the alternative pathway of human complement by 
structural analogues of sialic acid. J Immunol 1988;140:1588-1594. 

22 Stevens P, Huang SNH, Welch WD, Young LS: Restricted complement activation by Escherichia 
coll with the K 1 capsular serotype: A possible role in pathogenicity. J Immunol 1 978; 1 2 1 :2 1 7 1 -2 1 80. 

23 Frank M, Joiner K, Hammer C: The function of antibody and complement in the lysis of bacteria. 
Rev Infect Dis 1987;9:S527-S545. 

24 Howard CJ, Glynn AA: The virulence for mice of strains of Escherichia coli related to the effects 
of IC antigens on their resistance to phagocytosis and killing by complement. Immunology 
1971;20:767-777. 

25 Cunnion M, Lee JC, Frank MM: Capsule production and growth phase influence binding of com- 
plement to Staphylococcus aureus. Infect Immun 2001;69:6796-6803, 

26 AbeytaM, Hardy GG, YotherJ: Genetic alteration ofcapsule type but not PspA type affects acces- 
sibility of surface-bound complement and surface antigens of Streptococcus pneumoniae. Infect 
Immun 2003;71:218-225. 

27 Brown EJ, Joiner KA, Garther TA, Hammer CH, Frank MM: The interaction of C3b bound to 
pneumococci with factor H (beta IH globulin), factor I (C3b/C4b inactivator), and properdin 
factor B of the human complement system. J Immunol 1 983; 1 3 1 :409^ 1 5. 

28 Hor\vilz MA, Silvei'stein SC: Influence of the Escherichia coli capsule on complement fixation 
and on phagocytosis. J Clin Invest 1990;65:82-94. 

29 Cross A: The biological significance of bacterial encapsulation. Curr Top Microbiol Immunol 
1990;150:87-95. 

30 Yoshida K, MatsumotoT,TatedaIC, Uchida K,Tsujimoto S,Yamaguchi K: Induction of interleukin- 
10 and down-regulation of cytokine production by Klebsiella pneumoniae capsule in mice with 
pulmonary infection. J Med Microbiol 200l;50:456— ^61. 

31 Bhanacharjee A, Jennings H, Kenny C, Martin A, Smith I: Structural determination of the sialic 
acid polysaccharide antigens of Neisseria meningitidis serogroups B and C with carbon 1 3 nuclear 
magnetic resonance. J Biol Chem 1975;250:1926-1932. 

32 Vann WF, Schmidt M, Jann B, Jann K: The struchjre of the capsular polysaccharide (K5 antigen) 
of urinary tract infective Escherichia coli O10:K5:H4. A polymer similar to desulfo-heparin. 
Eur J Biochem 1981;116:359-364. 

33 Wyle F, Artensteiji M, Brandt BL, Tramont EC, Kasper DL: [mmunological response of man to 
group B meningococcal polysaccharide vaccines. J Infect Dis 1972;126:514-521. 

34 Jann K, Jann B: Capsules of Escherichia coli, expression and biological significance. Can J Microbiol 
1992;38:705-710. 



Capsules and Virulence 65 



35 Whitfield C, Roberts IS: Structure, assembly and regulation of expression of capsules in Escherichia 
coli. Mol Microbiol 1999;31:1307-1319. 

36 Boulnois G, Drake R, Pearce R^ Roberts 1: Genon^e diversity at the serA-linked capsule locus in 
Escherichia coli. FEMS Microbiol Lett 1992;100:121-124. 

37 Simpson DA, HammaJtonTC, Roberts IS: Transcriptional organization and regulation of expression 
of region 1 of the Escherichia coli K5 capsule gene cluster. J Bacteriol 1996;178:6466-6474. 

38 Richardson JP: Preventing the synthesis of unused transcripts by Rho factor. Cell I99l;64: 
1047-1049. 

39 Bliss JM, Silver RP: Coating the surface: A model for expression of capsular polysialic acid in 
Escherichia coli Kl. Mol Microbiol I996;2I :22I-23L 

40 Stevens MP, Clarke BR, Roberts IS: Regulation of the Escherichia coli K5 capsule geae cluster 
by transcription antiterminalion. Mol Microbiol 1997;24:1001-1012. 

41 Hobbs M, Reeves PR: The JUMPstart sequence: A 39 bp element common to several poly- 
saccharide gene clusters, Mol Microbiol 1994;12:855-856, 

42 Bailey MJ, Hughes C, Koronakis V: RfaH and the ops element, components of a novel system 
controlling bacterial transcription elongation. Mol Microbiol 1997;26:845-851. 

43 Marolda CL, VaJvano MA: Promoter region of the Escherichia coli 07-specific I ipopoly saccharide 
gene cluster: Structural and functional characterization of an upstream untranslated mRNA 
sequence. J Bacteriol 1998;180:3070-3079. 

44 Petit C, Rigg GP, Pazzani C, Smith A, Sieberth V, Boulnois G, Jann K, Roberts IS: Region 2 of 
the Escherichia coli K5 capsule gene cluster encoding proteins for the biosynthesis of the K5 
polysacchai'ide. Mol Microbiol 1995;17:611-620, 

45 Cieslewicz M, Vimr E: Thermoregulation of kpsF, the first region 1 gene in the kps locus for 
polysialic acid biosynthesis in Escherichia coli KL J Bacteriol 1996;178:3212-3220, 

46 Rowe S, Hodson N, Griffiths G, Roberts IS: Regulation of the Escherichia coli K5 capsule gene 
cluster; evidence for the role of H-NS, BipA and IHF in the regulation of group II capsule gene 
clusters in pathogenic E. coli, J Bacteriol 2000;182:2741-2745. 

47 Dorman CJ, Porter ME: The Shigella virulence gene regulatory cascade: A paradigm of bacterial 
gene control mechanisms. Mol Microbiol 1998;29:677-684. 

48 Farris M, Grant A, Richardson TB, 0*Connor CD: BipA: A tyrosine-phosphorylaled GTPase that 
mediates interactions between enteropathogenic Escherichia coli (EPEC) and epithelial cells, Mol 
Microbiol 1998;28:265-279. 

49 Freundlich M, Ramani N, Malhew E, Sikiro A, Tsui P: The role of integration host factor in gene 
expression in Escherichia coli, Mol Microbiol 1992;6:2557-2563. 



Clare M. Taylor 

School of Biological Sciences, 1.800 Stopford Building, University of Manchester 

Oxford Road, Manchester, M 13 9PT (UK) 

Tel. +44 161 2755601, Fax +44 161 2755656, E-Mail clare.taylor@man.ac.uk 



Taylor/Roberts 



66 



Adhesins 

Russell W, Herwald H (eds): Concepts in Bacterial Virulence.! 
Contrib Microbiol. Basel, Karger, 2005, vol 12, pp 67 89 



Fimbriae, Pili, Flagella and 
Bacterial Virulence 



Ann-Beth Jonson, Stajfan Normark, Mikael Rhen 

Microbiology and Tumor Biology Center, Karolinska Institute, 
Stockholm, Sweden 



Filamentous surface structures have been detailed on gram-negative entero- 
bacteria since the introduction of electron microscopy [1]. The bacteria 
appeared to express two types of extruding appendages: wavy flagella exceed- 
ing the length of the bacterium itself, and more rigid but somewhat thinner 
'fimbriae' [2] or 'pili' [3]. Soon after the description of fimbriae and pili, it was 
realized that their expression correlated with the ability of the bacteria to bind 
to cells from potential host organisms. Fimbriated and piliated bacteria agglu- 
tinated erythrocytes in a fashion resembling classical hemagglutination and 
adhered to host epithelial cells [2, 4-6]. Moreover, for some strains bacteria- 
induced hemagglutination was inhibited by the addition of the monosaccharide 
mannose. This suggested that mannose is used as a receptor for adherence and 
that the free mannose functions as a hapten. For other bacteria-erythrocyte 
reactions hemagglutination was not inhibited by mannose implying another 
receptor selectivity in the binding reaction [7-9]. 

Since the initial notion that fimbriae or pili function as specific adhesive 
organelles that aid bacterial colonization of mucosal surfaces, a myriad of 
bacterial adhesive factors have been described, many of which have turned out 
to act as virulence factors and to have a fimbriaJ morphology. Thematically, 
therefore, the unique but often separate binding specificity expressed by the vari- 
ous fimbriae participates in determining host and mucosal tropism (fig. 1) [10, 
1 1]. While such notions remain rather unchallenged, many recent observations 
imply additional functions for fimbriae. Distinct fimbriae are known to bind 
plasma proteins and to initiate intrinsic proteolytic cascades [12], whereas others 
are capable of activating Ca^+ influx and signal transduction cascades in host 
target cells [13]. In addition, fimbriae have been shown to act as invasion and 
motility factors, whereas bacterial flagella that typically mediate bacterial 



p 



+ 




p 




a 




Fig. h a Transmission electron nnicroscopy of type IV piliated (P^) and nonpiliated 
(P") N. gonorrhoeae, b The adherence of fluoreseently labelled P^ and P" bacteria to 
human cervical tissue sections is shown. The presence of type IV pili enables the bacteria 
to attach. 



motility have also been ascribed functions in terms of bacterial adherence [14] 
and in the initiation of proinflammalory responses [15]. One purpose of this 
review is to highlight the more recently defmed virulence-associated functions of 
fimbriae, pili (see The Role of Fimbriae in Pathogenesis of Mammalian Hosts as 
Illustrated through a Few Examples) and flagella (see Flagella as Virulence 
Factors) using a few illustrating examples, and to argue that these organelles have 
a role in the infection pathogenesis beyond the first step of surface colonization. 



Classification and Biosynthesis of Fimbria! 
Adhesive Factors 

The early notion of variation among fimbrial adhesive virulence factors 
brought the need for classification schemes [16]. However, the multitude by 



Jonson/Normark/RJien 



68 



which various bacterial organelles with different binding specificities started to 
emerge soon implied that typing approaches could become difficult based on a 
single characteristic, such as antigenicity or receptor specificity. Even for a 
single fimbrial type, the antigenic variation could be significant [17], and for 
many fimbriae no defined receptor structure was identified. Furthermore, not 
all adhesive factors appeared typically 'fimbria!' in morphology although they 
showed receptor-specific binding abilities [18, 19], Finally, in selected cases 
even flagellaare known to function as adhesive organelles [14]. 

While fimbriae and flagella can be defined as distinct structures, they 
share the need to create a polymer architecturally outside the ordinary bacterial 
anabolic machinery. This is also reflected in the complex organization of genes 
that are needed for either fimbrial or flagellar biosynthesis. The elucidation of 
the precise events involved in fimbrial biosynthesis by several laboratories has 
clearly formulated distinct fnnbrial 'families' and assembly pathways that 
actually can define groups of fimbrial types. Thus, a given assembly pathway 
can be used as a gross classification criterion [20], within which fimbriae can 
be defined based on receptor specificity or antigenic variation. 

Fimbriae Produced through the 'Chaperone/Usher' Pathway 

The classical 'common' type-1 fimbriae that mediate mannose-sensitive 
hemagglutination, and the P-blood-group-antigen-binding P-fimbriae, or Pap piU, 
are produced through the so-called 'chaperone/usher' pathway [21]. The biogene- 
sis and the basis for receptor recognition have been extensively studied for these 
two types of fimbriae. Therefore, the defined biogenesis and function of type I 
and Pap pili have functioned as guidelines when dissecting the molecular biology 
of fimbriae not only belonging to this class, and as a 'reference system' when 
dissecting other types of bacterial adhesins. 

Yet, fimbriae that belong to this 'chaperone-usher' family come in several 
different variants, and are not onJy defined to Escherichia coli. Gene clusters 
that provide the fimbrial subunits, protein chaperones and outer membrane 
anchors for the fimbrial shaft, as well as specific fimbrial regulatory genes 
code for these fimbriae. Altogether nine 'biosynthelic' genes and two iiitrijisic 
funbrial regulatory genes are included in the E. coli pap gene cluster responsi- 
ble for the expression of P-fimbriae [21, 22]. The fimbrial constituents are 
translocated to the periplasm through the housekeeping Sec system, and are met 
by the fimbrial chaperones once translocated. Principally, the chaperones 
translocate fimbrial subunits to the usher which then initiates translocation and 
polymerization of the funbrial subunits across the outer membrane. Thus, the 
f unbria elongates from the proxunal end of its shaft. 

Initially, these fimbriae were considered genuine homopolymers of the 
fimbrial protein subunit, the fimbrillin [23]. Furthermore, isolated fimbriae that 



Fimbriae, Pili, Flagella and Bacterial Virulence 69 



appeared as one major protein species in Coomassie-blue-stained SDS 
polyacrylamide gels couJd agglutinate cells, implying that receptor recognition 
was closely associated with fimbrial subunits [24]. However, during the dissection 
of P-fimbrial biosynthesis it became evident that the formation of fimbria and 
the ability of fimbriae to mediate adhesion or hemagglutination (receptor 
recognition) could be separated [25, 26]. This showed that the P-fimbriae actually 
were composite fibers. TTie fimbriaJ fibers include at least two distinct func- 
tions: the constitution of a filament and the recognition of the receptor; these 
functions were separable [27]. indeed, in addition to the major fimbrial subunit 
PapA, P-fimbrial filaments were found to contain minor subunits, including the 
PapE, PapF, PapK and PapG proteins located at the distal end of the fiber [21, 
27]. The ability to bind the receptor resided in the PapG subunit, whereas other 
tip-located Pap proteins functioned as initiators of fimbrial polymerization and 
for adapting PapG to the fimbrial shaft. However, PapA alone forms the 
micrometer long shaft and hence substantially dominates preparations of 
isolated fimbriae [26]. This may explain why isolated fimbriae perform receptor 
recognition but require sensitive staining techniques to reveal minor components 
in gel analyses. 

Not surprisingly, type 1 fimbriae are also composite fibers [22, 28-30], 
and may even include minor components scattered throughout the filament 
[31]. This could be due to a need to enhance fimbrial polymerization, and/or 
due to a need to include receptor-binding entities along the fimbrial shaft [20, 
31]. That is to say, the addition of minor nucleator or lectin components along 
the shaft could increase the efficiency of polymerization, or the avidity of the 
receptor-recognizing potential of the fimbrial filament. Other fimbriae belonging 
to this class include the E. coli S-fimbriae recognizing sialyl galactosides and 
type IC fimbriae [21, 22, 32]. 

Crystallographic studies have demonstrated that the periplasmic chap- 
erone not only fulfils a transporting function for the respective pilus subunit 
proteins in the periplasmic space as initially thought [33]. The pilus subunit 
proteins have an incomplete immunoglobulin fold, due to the lack of the 
seventh p-strand creating a large hydrophobic groove in the pilus subunit 
protein. In pilus biogenesis this groove is transiently occupied by the Gl strand 
of the chaperone [34]. At the site of the usher, the chaperone Gl strand is 
replaced by the amino-terminal extension of the next subunit protein to become 
incorporated via a donor strand exchange mechanism [35]. During donor strand 
exchange, the subunit undergoes a topological transition that triggers the 
closure of the groove and seals the amino-terminal extension of the incoming 
subunit in place [36]. These findings help explain the ordered assembly of pili 
heteropolymers. A contributing factor to the ordered assembly is the different 
affinities that chaperone-subunit complexes have for the outer membrane usher 



Jonson/Normark/RJien 70 



protein [37]. Outer-membrane PapC molecular ushers discriminately recognize 
peripJasmic chaperone-pilus subunit complexes. That the initiating step in pilus 
assembly is an interaction between the adhesin, in complex with the chaperone 
and the outer membrane usher, explains why the adhesin ends up at the pilus tip 
[38]. Evidently, comparable complex strategies of assembly are aJso applied by 
other fimbria and fimbrial types, and reflected in the multitude of participating 
gene functions [20, 39]. 

The atomic structures of three minor fimbrial lectin subunits or lectin 
domains associated with the cognate receptor have been determined [34, 40, 41], 
While these three lectin proteins do not share obvious sequence identity, they share 
a remarkably similar structural outline. The three protems in question that, respec- 
tively, recognize mannosides (FimH of the type 1 fimbriae), Galal -^ 4Galp 
(PapG of the P-fimbriae) and terminal N-acetyl-Z)-glucosamine (GafD or F17- 
G of the F17 fimbriae) share an immunoglobulin-like folding pattern forming an 
ellipsoid structure [41]. The receptor-binding pockets, however, seem to be 
somewhat differently positioned in relation to the superimposed ijnaginary core 
structure [41]. Thus, although all these fimbrial lectin proteins share the ability 
to bind a small carbohydrate epitope and to become integrated into the fimbrial 
filament, the lectin proteins apparently have not evolved just through modifi- 
cations in one existing carbohydrate-binding pocket. 

Fimbrial lectins are interesting candidate antigens for vaccine development. 
Due to the incomplete structural nature of the adhesin, vaccine trials have been 
conducted with adhesin-chaperone dimeric complexes. The FimH/FimC complex 
provided protection against uropathogenic E, coli in both a murine and a primate 
cystitis model [42^ 43]. 

The CSl Fimbrial Family 

Fimbriae belonging to the class of the CSl fimbrial family are assembled 
in a manner that phenotypically resembles the 'chaperone-usher' pathway [20]. 
The CSl fimbria forms the prototype of this class that includes several antigenic 
variants, including the classical CFA/1 fimbriae of enterotoxigenic E, coli 
(ETEC), and the type II pili o^ Burkholderia cepacia [44, 45]. Tlie CSl fimbrial 
subunit CooA is translocated to the periplasm through a Sec-dependent path- 
way, and then assisted by a protein CooB with chaperone-like function [20, 46, 
47]. CooA is then fed to a larger transmembrane protein CooC concomitant 
with fimbrial polymerization. However, polymerization needs the presence of a 
minor fimbrial subunit protein CooD, which functions both as an initiator and 
the lectin subunit [45, 48]. 

The constituents of the transport and assembly machinery do not show 
apparent amino acid sequence homology to the P-fimbrial chaperone or usher 
components. In addition, the number of CSl -specific genes that participate in 



Fimbriae, Pili, Flagella and Bacterial Virulence 71 



funbrial biogenesis as well as the number of specific fimbrial components 
tend to be more restricted within the CSl family. Still, the number of 
genes involved may not be a definitive characteristic of a fimbrial class; 
N-acetyj-D-glucosamine-binding F17 fimbriae also need only four genes for 
their expression in E. coll K12 and yet show many characteristics of the 
P-fimbrial family [48, 49]. 

T^pe IV Pili 

Type IV pili are multifunctional adhesive structures expressed by a num- 
ber of diverse microorganisms, including Neisseria meningitidis. Neisseria 
gonorrhoeae, Pseudomonas aeruginosa, Dichelobacter nodosus and Moraxella 
bovis [50]. Related structures have also been identified in Vibrio cholerae 
(toxin-coregulated pili, Tcp) and enteropathogenic E. coli (bundle-forming pili, 
Bfp) [51, 52]. Type IV pili are typically 5-7 nm in diameter and can extend 
several micrometers in length (fig. 1). They share an unusual (amino-termmal) 
N-methyl phenylalanine, a high conservation of the amino-termLnal 32 amino 
acids, and a proposed immunogenic carboxy-terminal disulfide-bound region. 
As with other types of fimbriae, type IV pili are composed primarily of a smgle 
protein subunit, termed pilin, which are arranged in a helical conformation with 
5 subunits per turn. In addition, and somewhat unorthodoxal for prokaryotic 
structural proteins, type IV pili can be glycosylated and/or phosphorylated 
depending on the bacterial species [53-57]. Type IV pilus assembly is hypothe- 
sized to occur within the cytoplasmic membrane or periplasm. The assembly of 
pili requires a nucleotide-binding protein, a polytopic inner membrane protein, 
the prepilin peptidase, and a multimeric outer membrane protem that forms a 
pore in the outer membrane for pilus protrusion [58]. 

One most astonishing aspect of type IV pili is their ability to intimate their 
initial contact through pilus retraction. A core set of mechanisms, fiber assem- 
bly and extension, fiber adhesion, fiber disassembly and retraction, account for 
these functions. Genetic analysis has revealed multiple clusters of genes, scat- 
tered through the microbial genome coding for type IV pilus biogenesis genes, 
as well as for major pilin and minor pilin-like proteins. The fact that ahnost 40 
genes have been identified in P. aeruginosa as essential for biogenesis and func- 
tionality of type IV pili evidently reflects the complexity both in pilus assem- 
bly and function [59, 60]. While bacterial fimbriae belonging to chaperone/usher 
or CSl family may have evolved through a divergent evolutionary need to pro- 
duce sticky, surface-located adhesive organelles [20], type IV pili may share 
evolutionary origins with filamentous bacteriophages [61], and with genes 
required for bacterial type II protein export and DNA uptake systems [62]. 

Type IV pili bind to a variety of surfaces, including both 'inert' nonbiological 
surfaces, to other bacteria, as well as to eukaryotic cells. In the case of type IV 



Jonson/Normark/RJien 72 



pili, the tip of the pilus binds to specific receptors on mammalian epithelial 
cells as an initial engaging event. Pili attached to cells are always observed 
anchored to surfaces at their distal end, and broken pili also only attach via an 
end [63]. In P. aeruginosa, the above-mentioned carboxy-terminal disulfide- 
bonded region is exposed at the tip of the pilus and binds the carbohydrate 
moiety of the asialo-GMl and asialo-GM2 glycosphingolipids on epithelial 
cells [64, 65]. 

Consequently, type TV pili of Neisseria are composed of a major pilus 
subunit PilE and several other pilus-associated proteins, which have different 
functions in pilus assembly and adhesion [66, 67]. One of these proteins is PilC, 
which is associated with the tip and the shaft of the pili [68] and the basal part 
in the outer membrane [69]. Adhesion o^ Neisseria to cells requires PilC, which 
appears to function as a tip adhesin, although it is also found in the cell membrane. 
The pili of Neisseria recognize and interact with the cell surface receptor 
complement regulator CD46 [70]. 

Fimbriae Produced through the Extracellular 

Nucha tor Pathway: Curli Organelles 

Many enterobacteria are capable of expressing elongated surface 
organelles, called AgfA fimbriae, with an 'aggregative' and chemically robust 
character [71, 72]. AgfA fibers appear not as straight but rather as twisted, 
curly structures and hence are referred to as 'curli' fimbriae [73]. Curli fibers 
of £". coli and Salmonella enterica sv Tyhpimurium are coded for by the cfg 
gene cluster. The cluster consists of two divergently transcribed units that 
include the csgABC diud csgDEFG genes, respectively. Although curli fibers are 
coded for genetic elements comparable in size to the P-fimbrial pap operon [74, 
75], the curli fiber polymerization process is apparently different. Interestingly, 
curli fibers show all the typical characteristics of amyloid fibers, such as the 
binding to the dye Congo red. However, unlike amyloid formation in human 
neurodegenerative disorders such as Alzheimer's disease, curli amyloids require 
a specific assembly machinery [76]. Thus, the CsgA and CsgB fimbrial subunits 
appear to be secreted out from the bacteria [72, 74], where after an interactioji 
between the subunits in the extracellular compartment then leads to polymer- 
ization. The CsgA subunit occurs in excess in the isolated filament, whereas 
in vitro both the CsgB subunit [72] and the isolated CsgA subunit [76, 77] are 
capable of self-polymerization. Thus, as in analogy with type 1, P- and CSI 
funbriae the assembly of curli organelles also involves a nucleator component 
(CsgB), proteins with apparent chaperone functions (CsgE) [76], or a nucleator 
center (CsgG) [78]. As with type IV pili, curli fibers have a rather diverse 
spectrum of receptor targets. Curli fibers are reported to mediate binding to 
mouse small intestinal epithelial cells [73], in addition to various plasma and 



Fimbriae, Pili, Flagella and Bacterial Virulence 73 



extracellular matrix proteins [12, 71, 79, 80]. One reason for this promiscuity 
might reside in the participation of curii in the formation of biofilms [81, 82]. 
A more flexible binding specificity might be more efficient in collecting 
various organic molecules into the biofilm as compared to an organelle with a 
highly specific, but concomitantly more narrow receptor repertoire. Since the 
CsgD transcriptional regulator also affects bacterial production of cellulose an 
important role of curli might be to interact with cellulose fibrils in an extra- 
cellular matrix [83]. 



The Role of Fimbriae in Pathogenesis of Mannnnalian 
Hosts as Illustrated through a Few Examples 

Chape roneAJsher Fimbriae and Urinary Tract Infection 

Adhesion 

E. coli is by far the most common causative agent of urinary tract infections 
(UTI) [84]. Consequently, the role of £". coli fimbriae in the infection patho- 
genesis of UTi has been given much attention, and has been used as a template 
for the analysis of other fimbria! structures [21, 85]. 

The ability to express certain types and sets of fimbriae seems overrepre- 
sented among urinary tract isolates of £. coli. The expression of type 1 fimbriae 
appears to be both an miportant colonization factor and a factor contributing to 
the persistence in the bladder epithelium [85]. However, the pattern of mannose 
binding by the protein FimH is somewhat different among commensal and UTI 
E. coli; UTI isolates seem capable of binding Z)-mannose whereas commensals 
seem to prefer trimannoside structures [86]. This difference in specificity 
resides in minute differences m the FimH fimbrial lectin molecule as coded for 
by separate alleles of JimH. Uroplakins, or rather mannosides contained on 
uroplakin, are believed to be the actual epithelial receptor in the urinary tract 
[84, 87]. Thus, it appears that the type 1 fimbria can be equipped with differ- 
ent variants of FimH, and that the receptor preferences expressed by FimH 
in tuni steer the mucosal tropism of the bacterial even within a single host 
organism. 

The P-fimbriae is another group of bacterial adhesins often expressed by UTI 
isolates of £". coli, in particular among strains causing upper UTI and urosepsis 
[84, 88]. P-fimbriae recognize the core Gala I -^ 4Gal|3 entity contained in 
blood group antigen-carrying globoseries glycolipids [78]. Thereby, as the 
receptor is present on cells linmg the human urinary tract, it provides an adhe- 
sion target for P-fimbriated bacteria ascending from the bladder to the ureter 
and further up into the kidney [84]. As with the FimH protein of type 1 pili, 
PapG possesses allelic polymorphism: the class 1, 11 and 111 adhesins. Of these, 



Jonson/Normark/RJien 74 



the class II G adhesin recognizes most members of Galal — > 4Gal|3-containing 
globoseries glycolipids and has been considered important for kidney infection 
in persons with a nonobstiucted urinary tract [84, 89-91]. 

Beyond Adherence 

Besides mediating adherence to the urinary tract epithehum, type 1 and 
P-fimbriae have been implicated in the later phases of infection, and in the 
generation of innate proinflammatory responses in the infected urinary tract 
epithelium. First, although cystitis-associated E. coLi have generally been 
regarded as noninvasive bacteria, type I fimbriated E. coli have been observed 
to enter human bladder epithelial cell lines in vitro in a FimH-dependent manner 
[87]. Invasion could be mimicked by applying FimH-coated beads, and invasion 
was associated with host protein tyrosine phoshorylation and host actin 
cytoskeleton rearrangement [92]. This suggests that FimH alone, in analogy 
with Yersinia invasion factor Inv [93], can activate host signal transduction 
events that subsequently trigger actin cytoskeletal rearrangements in the host 
leading to bacterial uptake. Later it was observed in a mouse cystitis model that 
the bacteria were internalized into bladder epithelial cells and subsequently 
formed a biofilm-like mass [94]. Apart from type 1 fimbriated bacteria, uroplakin 
was also found in the biofihn. Thus, type 1 fimbriae appear multifunctional in 
the pathogenesis of UTI; they mediate initial adherence, invasion and seem to 
participate in the formation of an intracellular biofihn. 

Many types of fimbriae, including type 1, type IC and P-fimbriae have all 
been associated with the induction of proinflammatory responses in epithelial 
cells [95-97]. Type 1 fimbriated E. coli induce cytokine expression from both 
A498 kidney epithelial cells as well as in bladder cell lines [96, 98]. However, 
in bladder epithelial cells the majority of the IL-6 response seems to derive 
from lipopolysaccharide (LPS) signalling through the CD14-TLR4 pathway 
[98]. Still, type \lfimH^ fimbriae appear to be somewhat more potent inducers 
of IL-6 as compared to type \lfimH~ bacteria in LPS-hyporesponsive A498 
cells. Likewise^ type IC fimbriae, also associated with cystitis, augment bacte- 
rial lL-8 release from A498 cells [95]. It is thus possible that bacterial attach- 
ment, the prerequisite for the infection in the first place, is also one cause foq 
the symptoms of cystitis. 

The mechanism by which P-fimbriae induces signal transduction 
casacades in kidney A498 cells appears complex, and differs from those mecha- 
nisms used by type 1 fimbriae [96, 99]. Binding of P-fimbriated bacteria causes 
a release of ceramide in the target cells concomitant with an activation of 
cytokine release [96]. Cytokines, such as TNF-a, also cause the release 
of ceramide from sphingomyelin, which eventually results in the activation of 
transcription factor NF-kB [100]. It has thus been suggested that ceramide 



Fimbriae, Pili, Flagella and Bacterial Virulence 75 



Initial adherence 



CD46 



Tight adherence 




Cellular responses 



Pilus retraction 




CD44 



ICAM-1 



.2 + 



Ca signalling 

Trigger lysosome exocytosis/LAMP-1 to cell surface 



Fig. 2. Initial adherence of type FV piliated Neisseria involves initial contact with ceJJ 
surface receptors followed by sophisticated cell signalling leading to tight adherence and 
invasion of host cells. Failure in the pilus retraction events and/or host cell signalling leads 
to lost or changed adherence patterns, and a loss of ability to enter and invade host cells. 



release caused through attachment of P-fimbriated E. colt could induce nuclear 
responses as a result of ceramide release. Furthermore, the LPS-recognizing 
Toll-like receptor TLR4 has been implicated in P-fimbria-induced host responses 
[99]. Possibly^ P-fhnbriae can adapt both cermJde- and TLR4-mediated signals 
to induce NF-kB nuclear translocation. Binding of P-fimbriated bacteria to A498 
cells also caused an upregulation in the expression of TLR4mRNA suggesting 
that one function of P-fimbria-mediated host cell responses might be to modify 
the surface of the host cell to better accommodate or promote the infection. 

Type IV Pili in Sequential Attachment and Invasion 

of Pathogenic Neisseria 

Adhesion 

The important initial interaction between pili of Neisseria and its host cell 
occurs through the receptor molecule CD46;, a human cell surface protein 
involved in the regulation of complement activation. In cultured epithelial cells, 
binding of pili to CD46 is followed by release of Ca^"^ from intracellular stores 
[13, 101], This Ca^"^ transient is sufficient to mediate exocytosis of a pool of 
the lysosomal/late endosomal vacuoles resultmg in the increase of surface 
lysosomal components such as h-Lamp-1, and possibly other factors that could 
contribute to a tighter adherence of bacteria. During initial contact between bacte- 
ria and cells, pilus retraction exerts tensile forces upon the plasma membrane 
(fig. 2) [102]. The mechanical forces applied to the plasma membrane trigger actm 
polymerization accompanied by accumulation of phosphotyrosine-containing 



Jonson/Normark/RJien 



76 



proteins, which leads to the formation of compact microcolonies and so-called 
pilus-associated cortical plaques on the host cell [103, 1 04]. The cortical plaque 
structures are characterized by the accumulation of actin and actin-associated 
proteins, and trigger recruitment of transmembrane proteins such as CD44, 
ICAM-1, EGFR, and components of the cortical cytoskeleton, i.e. ezrin and 
cortactin, and contain tyrosine-phosphorylated host cell proteins beneath the 
microcolony [104, 105]. 

At later times after infection, bacteria disperse from the microcolonies, pili 
disappear, and individual diplococci become intimately associated with the host 
plasma membrane. Pilus loss, bacterial dispersal, and intimate adhesion are all 
blocked in ap/'/r mutant [106, 107]. The full set of rearrangements requires the 
expression of both type IV pili and PilT. Obviously, pilus retraction could account 
for elongation of microvilli towards the bacterial microcolony and bring the 
host cell and bacterial membrane into close contact [108]. For example pilT 
mutants of N. gonorrhoeae are unable to make intimate contact with or form 
attaching effacing lesions on epithelial cells [95]. P. aeruginosa pilT mutants 
are not infective in corneal tissue and exhibit reduced cytotoxicity to epithelial 
cells in culture [109 1 1 !]. 

To summarize, type IV pili do not only simply anchor the bacteria at the cell 
surface, they initiate a multistep adhesion cascade, which starts with a loose 
adherence and ends with the intimate attachment of bacteria [1 12]. Establishment 
of intimate attachment appears to require an intensive host-pathogen cross talk, 
and a complex sequence of bacteria-host cell interactions. Type IV pili also assist 
in the formation of biofilms [1 12] that may support further tissue colonization 
and protect the bacteria against antibodies and antibiotics. 



Beyond Adhesion 

In an experimental model system oi Neisseria infection, using transgenic 
mice expressing human CD46, the crossing of the blood-brain barrier by 
bacteria occurred in CD46 mice but not in nontransgenic mice, indicating an 
important role for CD46 in meningococcal meningitis [113]. Intranasal 
infection of CD46 mice required piliated bacteria for the development of disease, 
supportmg that CD46 facilitates pilus-dependent mteractions at the epithelial 
mucosa. 



Binding ofFimbrial Structures to Extracellular Components 
Although a primary role of fimbriae indeed might be to mediate adhesion 
and subsequent events through binding to specific structures on host (epithe- 
lial) cells, it has recently become evident that fimbriae can also bind various 
connective tissue proteins, as well as plasma and serum proteins. Moreover, 



Fimbriae, Pili, Flagella and Bacterial Virulence 77 



binding to selected plasma components can induce subsequent intrinsic cascades 
leading to the activation of zymogen proteases and the release of biologically 
active host peptides [12, 80], Such observations illustrate that fimbriae may 
contribute to the infection pathogenesis even after they have assisted adhesion 
and invasion. 

The F17 fimbriae occur characteristically in E. coli isolates causing 
diarrhea and septicemia in newborn calves. F17 fimbriae mediate binding to the 
calf intestinal epithelium, which suggests a role for F17 fimbria in the intesti- 
nal colonization. In addition, the F17 fimbria is capable of binding plasmino- 
gen [114] and the extracellular matrix protein laminin [1 15]. Binding to laminin 
is inhibited by the receptor analogue N-acetyl-Z)-glucosamine, indicating that 
carbohydrate receptors on the extracellular matrix protein are recognized by the 
minor fimbrial lectin protein GafD [115]. The binding to plasminogen is not 
inhibited by the receptor analogue, but instead the binding leads to conversion 
of plasminogen to proteolytically active plasmin. Binding and activation of 
plasmin is not unique to F17 fimbriae. For instance, it has been shown that 
meningitis-associated S-fLmbriae and S. enterica sv Typhimurium type 1 fim- 
briae as well as curii fibers are both plasminogen binders and activators [79, 
1 14, 1 16, 1 17]. Such observations suggest that fimbriae may assist bacteria dur- 
ing tissue dissemination by directing them to extracellular matrix proteins, and 
by coating them with proteolytically active proteins that enable the bacteria to 
penetrate through the tissue. Indeed, enterobacteria capable of binding and acti- 
vating plasminogen have been shown to degrade extracellular matrix proteins, 
and to penetrate reconstituted basement membranes in vitro [117]. 

Yet another aspect of binding to plasma proteins is illustrated by the ability 
of bacterial curIi fimbriae to activate the contact phase pathway of the coagu- 
lation system, and thereby to induce proinflammatory reactions [12, 80]. Factor 
XI, factor XII, prokallikrein and H-kJninogen are absorbed to curilated E. coli 
and S. enterica sv Typhimurium, but not to isogenic noncurliated mutants. 
Binding of the contact phase proteins by the curiiated bacteria lead to a rapid 
release of vasodilatory bradykinin from kininogens and to prolonged clotting 
times of the infected plasma. While it is difficult to ascertain the biological 
fijnction of such reactions, possibly they reflect an aspect of the innate line of 
defenses, such observations imply that a more massive encounter with curiiated 
bacteria may contribute to the symptoms of septic shock [80]. 

Pili and Motility 

The term twitching motility was used by Lautrop [118] in 1961 to describe 
flagella-independent spasmodic movements of bacteria. Twitching motility 
occurs in a wide range of bacteria, and has been well studied in N. gonorrhoeae 
and P. aeruginosa. It occurs on solid, wet surfaces and is mediated by type IV 



Jonson/Normark/RJien 78 



pili. Twitching motility occurs by extension, tethering, and then retraction of type 
TV pili, which operate in a manner similar to a grapping hook, which has been 
shown by elegant studies in A', gononhoeae [1 02], Myxococcus xanthiis [119], and 
P. aeruginosa [1 11], 

Type IV pili serve as an initial bridge between bacteria and cells, and twitch- 
ing motility allows bacteria to spread in the infected tissue. P. aeruginosa is 
an important pathogen, being the major cause of lung damage in patients 
suffering from cystic fibrosis as well as of opportunistic infections in immuno- 
compromised individuals, such as burn victims or patients undergoing chemo- 
therapy. Twitching motility has been shown to be important for infection by 
P. aeruginosa as well as for biofilm formation, which appears to be involved in 
chronic infection [110, 112]. 

Type JV pili generate considerable force by retraction [102, 120]. For 
some pilus-dependent functions, the amount offeree is critical, e.g. in host-cell 
responses and movement of bacteria through viscous mucous layers. Although 
pilT mutants adhere and colonize surfaces, pilT mutants are aviruient in many 
experimental model systems. PilT mutants are unable to retract their pili, lead- 
ing to hyperpiliation and loss of twitching motility. It could be speculated that 
a signal could pass from the tip of the fiber to its base, by the propagation of a 
helix dislocation or a mechanical force such as tension, compression or flexion. 
The dislocation signal that reaches the base of the pilus could induce a beneficial 
movement response to the cell. 

PilT, an ATPase associated with various cellular activities (AAA), seems 
to act as a molecular motor [121, 122]. Pilus retraction is thought to occur by 
filament disassembly mediated by PilT, a process that has been estimated to 
occur at around 1 ,000 pilin subunits per second. Genetic studies and structural 
data support the following molecular model. The cytoplasmic membrane has a 
reservoir of the prepilin subunits that are cleaved by PilD, the prepilin pepti- 
dase, and then polymerized into pili. In the model PilT is actively involved in 
the dissociation of a pilus. PilT is a member of the GspE family of hexameric 
AAAs, and one PilT unit could hydrolyze several (up to six) ATP molecules in 
the process of dissociating one pilin subunit. It is possible that epithelial cells 
sense the amount of force generated by pilus retraction and respond in a similar 
manner. 

Phase Variation of Pilus Structures 

As mentioned above, fimbriae and pili of the same type can be expressed as 
antigenic variants. For example, separate strains of UTI E. coli can express sepa- 
rate antigenic variants of the major fimbrial subunit protein [123], and a single 
strain can contain more than one P-fimbrial gene cluster [124]. Furthermore, as 
different P-fimbrial gene clusters may contain separate /?£j!/?G alleles [89], and 



Fimbriae, Pili, Flagella and Bacterial Virulence 79 



as P-fimbriae are subject to phase variation [125], the set-up provides E. coli 
with flexibihty in terms of varying antigenicity and ftinction of P-fimbriae. 
Still, for a given strain the repertoire is restricted to the number of fimbria! gene 
clusters contained, and hence usually narrow. 

One extraordinary characteristic of the pathogenic Neisseria species is 
their enormous capability to vary their surface pili [17]. In this context, the 
changing in the antigenic structures of surface proteins is certainly an impor- 
tant immune escape mechanism [ 1 26]. Furthermore, the variation also modifies 
the function of these adhesions [127-130]. Small alterations on the primary 
structures of neisserial pilins cause changes in immunoreactivity, post- 
translational modification, adhesive function, and ability to form bundles of pili. 
As a consequence, the pathogens can selectively interact with certain cell types 
and thus occupy special niches in their host. Many pilin variants that promote 
strong adhesion to host cells also aggregate into laminar bundles, whereas vari- 
ants that promote weaker adhesion tend to exist as single filaments [131, 132]. It 
is unclear how bundling promotes adhesion. Bundles could promote bacterial 
aggregation, increase receptor avidity by oligomerizing binding sites, or increase 
pilus stiffness. Bundles might also promote twitching motility by promoting 
coordinated fiber extensions and retraction processes that would be unfeasible 
with less-ordered structures. 



Flagella as Virulence Factors 

Like fimbriae, flagella are protein polymers, each flagellum consisting of 
thousands of flagell in monomers [1 4]. These filaments are connected to the cell 
surface through the 'hook' structure, and the basal structure that forms the rota- 
tion device and that traverses the bacterial cell wall. Consequently, flagella are 
complex structures and coded for by a large set of genes. While the primordial 
role of flagella is to ensure motility, either as swimming movement in liquid 
medium or as swarming on solid surfaces, these traits are also applied in bacte- 
rial virulence [14]. For example, flagel la-mediated moLilily acts as a virulence 
function for V. cholerae [133], Helicobacter pylori [134] and for Proteus 
mirabilis [135]. The former two pathogens are noninvasive colonizers of the 
digestive tract. Evidently, these bacteria apply motility to gain contact with the 
intestinal or gastric mucosal cells, respectively, and thus to establish the infection. 
P. mirabilis, on the other hand, is believed to apply motility for ascending from 
the ureter to the bladder, and further up to kidney structures. For V. cholerae and 
//. pylori, the role of flagella as virulence factors is also supported through 
transcriptome analyses, which show an upregulation of motility genes in de facto 
infecting bacteria [136-138]. For P. mirabilis, the swarming state involves a 



Jonson/Normark/RJien 



80 



transition to a hyperflagellated state and an upreguiation, the expression of 
selected virulence functions [135]. 

Besides mediating motility, flagella are in many instances known to adapt 
functions typically ascribed to fimbriae. The flagellar FliC and FliD proteins of the 
gram-positive anaerobe Clostridium difficile, a causative agent of pseudo- 
membranous colitis, have been shown to bind both to mouse catcall mucous 
and cultured cells [139]. Similarly, a nonflagellated P. aeruginosa mutant was 
shown to be attenuated in a mouse pneumonia infection model. In parallel 
flagellin was been shown to bind GMl, asialoGMl and GDI glycolipids 
in vitro [140]. For S. enterica sv Enteritidis, nonflagellated mutants are abro- 
gated for their ability to adhere to gut epithelium and epithelial cells, and for 
their ability to invade host cells [141]. 

Whereas some bacteria, like S. enterica sv Typhimurium, can phase-variate 
between the expression of two alternative flagellar subunit proteins, others, 
like Vibrio parahaemolyticus and Aeromonas spp., apply two separate sets of 
flagella: polar and lateral sets [14]. The different flagellar sets expressed by 
Aeromonas primarily associate with a shift in motility, the lateral set being used 
for swarming. However, there is also evidence for different adhesive characters 
disposed by polar and lateral Aeromonas flagella [142]. Therefore, as for R 
mirabilis, the switch to a swarming phenotype reflects a more fiindamental 
alteration in the expression of the bacterial virulence potential. 

As with fimbriae, flagella also activate host cell signal transduction cas- 
cades and inflammatory responses. At least in part, this response originates 
from the fact that flagella, like bacterial LPS, act as pattern molecules that 
are recognized by the host innate responses. While LPS is recognized by 
TLR4, flagellin from both gram-positive and gram-negative bacteria is rec- 
ognized by TLR5 [15]. The interaction between flagellin and TLR5 signals 
via Myd88 to cause activation of inflammatory responses [143]. Both LPS 
and flagellin can cause tolerance in host cells, the cells becoming non- or 
hyporesponsive after prior exposure to the ligand. What is interesting in this 
context is that LPS and flagellin can cause cross-tolerance, at least in cell 
lines [144]. 

The flagellar assembly pathway is related to the contact-dependent, so-called 
type 111 protein secretion pathway that is applied by many pathogens, like 
S. enterica sv Typhimurium and Yersiniae^ for the translocation of bacterial viru- 
lence protein into host cells [145]. In selected cases it has been observed that 
the flagellar basal body and hook structures in >S'. enterica sv Typhimurium can 
substitute for the transport function of virulence proteins [146, 147]. While this 
was observed against a background with the ordinary secretion machinery inac- 
tivated, it suggests that the flagellar protein secretion potential, normally reserved 
for flagellar components, also could be applied for more sinister purposes. 



Fimbriae, Pill, Flagella and Bacterial Viruience 81 



Concluding Rennarks 

The ability to express surface structures related to adhesiveness and motility 
appears to be a widespread ability among prokaryotes, reflecting the necessity of 
corresponding traits for microorganisms. In many cases these organelles medi- 
ate colonization and adhesion of the bacteria to their growth niche: the plant root 
or a vertebrate epithelium. However, the further investigations proceed from 
describing adhesion to resolving the biogenesis and host responses, the more 
complex the functions of the ad priori adhesive and motility organelles appear. 
Indeed, type IV pili are known not merely to function as passive adhesive 
fibers, but in addition as dynamic machines that participate in a surprising num- 
ber of functions: adhesion to host cell surfaces, modulation target cell specificity, 
twitching motility, DNA transformation, and bacterial autoagglutination. 
Furthermore, fimbrial receptor recognition can actually represent the prelude to 
a much more elaborated host-parasite cross talk. This is illustrated by type 1 
fimbria-mediated activation of host signal transduction cascades that result in 
concomitant bacterial internalization, or by P-fimbria that activates TLR4- 
mediated proinflammatory and causes increase in TLR4 expression to further 
amplify the process. Considering the impact of adhesion and motility in viru- 
lence, it is interesting to note that, at least in selected cases, the same protein- 
aceous extensions are being applied both for adhesion and motility. Perhaps it 
is the extendedness of the structure that makes it suitable for such purposes. It 
is important to mention that the host has evolved systems that recognize bacte- 
rial flagellar It remains to be evaluated whether there are specific innate 
specific recognition systems for fimbriae or whether the ability of fimbriae to 
initiate proinflammatory responses in fact reflects attempts to eradicate bacte- 
ria] colonization. 



References 

1 Houwink AL, Iterson W: Electron microscopical observations on bacterial cytology. 11. A study on 
fiagellation. Biochim Biophys Acta 1950;5:1(M4. 

2 Duguid JP, Smith IW, Dempster G.Edmunds PNrNon-flagellar filamentous appendages (fimbriae) 
and haemagglutination activity in Bacterium coli. J Pathol Bacteriol 1955;70:335-348. 

3 Brinton CC Jr: The structure, function, synthesis and genetic control of bacterial pili and a molecular 
model for DNA and RNA transport in gram negative bacteria. Trans NY Acad Sci 1965;27: 
1003-1054. 

4 Jojies GW, Rulter JM: The association ofKSS aiitigeji with haejnagglutinatioji activity in porcine 
strains of Escherichia coli. J Gen Microbiol 1974;84:135-144, 

5 Burrows MR, Sellwood R, Gibbons RA: Haemagglutination and adhesive properties associated 
with K99 antigen of bovine strains of Escherichia coli. J Gen Microbiol 1976;96:269-275. 

6 Isaacson RE, Fusco PC, Brinton CC, Moon H^^^: In vitro adhesion of Escherichia coli to porcine 
small intestinal epithelial cells: Pili as adhesive factors. Infect Immun 1978;21:392-397. 



Jonson/Normark/Riien 



82 



7 Evans DJ, Evans DG, DuPonL HL: Hemagglutination patterns of enterotoxigenic and entero- 
pathogenic Escherichia coli determined within human, bovine, chicken, and guinea pig erythrocytes 
in the presence and absence of mannose. Infect Immun 1979;23:336-346. 

8 Kallenius G, Mollby R, Svenson SB, Winberg J, Lundblad A, Svensson S, Cedergren B: The P^ 
antigen as a receptor for the haemagglulinin of pyelonephritogenic Escherichia coli. FEMS 
Microbiol Lett 1980;7:297-302. 

9 Leffler H, Svanborg-Ed6n C: Chemical identification of a glycosphingolipid receptor for Escherichia 
co// attaching to human urinary tract epithelial cells and agglutinating human erythrocytes, FEMS 
Microbiol Lett 1980;8:127-134. 

10 Nowicki B, Holthofer H, SaranevaT, Rhen M, Vaisanen-Rhen V, KorhonenTK: Location of adhesion 
sites for P-fimbriated and for 075X-positive Escherichia coli in the human kidney. Microb Pathog 
1986;1:169-180. 

1 1 KorhonenTK, Parkkinen J, Haker J, Finne J, Pere A, fUien M, Holthofer H: Binding oi Escherichia 
coli S fimbriae to human kidney epithelium. Infect Immun 1986;54:322-327, 

12 Herwald H, Morgelin M, Ols6n A, Rhen M, Dahlback B, Miiller-Esterl W, Bjorck L: Activation of 
contact-phase system on bacterial surfaces -A clue to serious complications in infectious diseases. 
Nat Med 1998;4:298-302. 

13 Kallstrom H, Islam MS, Berggren P-0, Jonsson A-B: Cell signalling by the type IV pilus of patho- 
gen ic A/m^en^. J Biol Chem 1998;273:21777-21782. 

14 Kirov SM: Bacteria that express lateral flagella enable dissection of the multifunctional roles of 
flagella in pathogenesis. FEMS Microbiol Lett 2003;224:151-159. 

1 5 Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, Eng JK, Akira S, Underhil I DM, 
Aderem A: The innate immune responses to bacterial flagellin is mediated by Toll-like receptor 5. 
Nature 2001;410:1099-1 103. 

16 Nimmich W, Zingler G, 0rskov 1: Fimbrial antigens o{ Escherichia coli 0I:KI:H7 and 0I:K1: 
H-strains isolated from patients with urinary tract infections. Zentralbl Bakteriol Mikrobiol Hyg 
1984;258:104-111. 

17 Hagblom P, Segal E, Billyard E, So M: Inrragenic recombination leads to pilus antigenic variation 
in Neisseria gonorrhoeae. Nature 1985;315:156-158. 

18 Labigne-Roussel AJF, Lark D, Schoolnik G, Falkow S: Cloning and expression of an afimbrial 
adhesin (AEA-1) responsible for P-blood-group-independent mannose-resistant hemagglutination 
from a pyelonephritogenic Escherichia coli strain. Infect Immun 1984;46:251-259. 

19 Rhen M, Klemm P, Korhonen TK: Identification of two new hemagglutinins oi Escherichia coli: 
N-acetyl-D-glucosajnine-specific fimbriae and a blood group M-specific agglutinin, by cloning 
the corresponding genes in Escherichia coli K-\2. J Bacteriol 1986;168:1234-1242. 

20 Smyth CJ, Marron MB, Twohig JMGJ, Smith GJS: Fimbrial adhesins: Similarities and variations 
in structure and biogenesis. FEMS Immunol Med Microbiol 1996;16:127-139. 

21 Hung DL, Hultgren S: Pilus biogenesis via the chaperon/usher pathway: An integration of struc- 
ture and function. J Struct Biol 1998;124:201-220. 

22 Krogfelt KA: Bacterial adliesion: Genetics, biogenesis, and role in pathogenesis of fimbrial 
adhesins in Escherichia coli. Rev Infect Dis 1991;13:721-735. 

23 Brinton CC Jr: The structure, function, synthesis and genetic control of bacterial pili and a molecular 
mechanism for DNA and RNA transport in gram negative bacteria. Trans NY Acad Sci 1965;27: 
1003-1054. 

24 Korhonen TK: Yeast cell agglutination by purified enterobacterial fimbriae. FEMS Microbiol Lett 
I979;6:421^25. 

25 Norgren M, Normark S, Lark D, O'Hanley P, Schoolnik G, Falkow S, Svanborg-Ed6n C, BSgaM, 
LHilin BE: Mutations in E. coli cisti'ons affecting adhesion to human cells do not abolish Pap fiber 
formation. EMBO J 1984;3:1 159-1 165. 

26 Lindberg FP, Lund B, Normark S: Genes of pyelonephritogenic E. coli required for digalactoside- 
specific agglutijiation of human cells. EMBO J 1984;3: 1 167-1 173. 

27 Kuehn MJ, Heuser J, Normark S, Hultgren S: P pili in uropathogenic E. coli are composite fibers 
with distinct fibrillar adhesive tips. Nature 1992;356:252-255. 

28 Klemm P, Christiansen G; Threey?w genes required for the regulation of length and mediation of 
adhesion of Escherichia coli type 1 fimbriae. Mol Gen Genet 1987;208:439^M5. 



Fimbriae, Pili, Flagella and Bacterial Virulence 83 



29 Jones CH, Pinkner JS, Nicholes Ay Slonim LN, Abraham SN, Hultgren SJ: FimC is a periplasmic 
PapD-like chaperone that directs assembly of type 1 pili in bacteria. Proc Natl Acad Sci USA 
1993;92:2081-2085. 

30 Jones CH, Pinkner JS, Roth R, Heuser J, Nicholes AV, Abraham S, Hultgren SJ: FimH adhesin of 
type I pili is assembled into a fibrillar tip structure in Enterobacieriaceae. Proc Natl Acad Sci USA 
1995;90:8397-8401. 

3 1 Klemm P, Kxofelt KA: Type I fimbria of Escherichia coii\ in Klemm P (ed): Fimbriae: Aspects of 
Adhesion, Genetics, Biogenesis and Vaccines. Boca Raton, CRC Press, J 994, pp 9-26. 

32 SchmollT, Morschhauser J, Ott M, Ludwig B, van Die I, Hacker J: Complete genetic organization 
and functional aspects of the Eschehchia coli S fimbrial adhesion determinant: Nucleotide sequence 
of the genes sfa B, Q A E, F. Microb Pathog I990;9:33 1-343. 

33 Lindberg F, Tennel JM, Hultgren SJ, Lund B, Normai'k S: PapD, aperiplasmic transport protein in 
P pilus biogenesis, J Bacteriol 1989;171:6052-6058. 

34 Chodhury D, Thompson A, Stojanoff V, Langermann S, Pinkner J, Hultgren SJ, Knight SD: X-ray 
structure of the FimC-FimH chaperon-adhesin complex fi-om uropathogenic Escherichia cod. 
Science 1999;285:1061-1066. 

35 Sauer FG, Futter K, Pinkner JS, Dodson KW, Hultgren SJ, Waksman G: Structural basis of chap- 
erone function and pilus biogenesis. Science 1999;285:1058-1061. 

36 Sauer FG, Pinkner JS, Waksman G, Hultgren SJ: Chaperone priming of pilus subunits facilitates 
a topological transition that drives fiber formation. Cell 2002; I 1 1:543-551 . 

37 Dodson KW, Jacod-Dubuisson F, Striker RT, Hultgren SJ: Outer-membrane PapC molecular usher 
protein. Proc Natl Acad Sci USA 1993;90:3670-3674. 

38 Barnart MM, Sauer FG, Pinkner JS, Hultgren SJ: Chaperon-subunit-usher interactions required 
for donor strand exchange during bacterial pilus assembly. J Bacteriol 2003;185:2723-2730. 

39 Mooi FR, Wouters C, Wijfjes A, de Graaf FK: Construction and characterization of mutants 
impaired in the biosynthesis of the K88ab antigen. J Bacteriol 1982;150:512-521. 

40 Dodson KW, Pinkner JS, RoseT, Magnusson G, Hultgren SJ, Waksman G; Structural basis of the 
interaction of the pyelonephritogenic Escherichia coli adhesin to human kidney receptor Cell 
2001;105:733-743. 

41 Buts L, Bouckaerl J, De Genst E, Loris R, Oscarson S, Lahman M, Messens J, Brosens E, Wyns L, 
De Greve H: The fimbrial adhesin F17-G of enterotoxigenic Escherichia coli has an immunoglobul in- 
like lectin domain that binds N-acetylglucosamine. Mol Microbiol 2003;49:705-715, 

42 Langermann S, Palaszynski S, Barnhart M, Auguste G, Pinkner JS, Burlein J, Barren P, Koenig S, 

Leath S, Jones CH, Hultgren SJ: Prevention of mucosal Escherichia coli infection by FimH- 
adhesin-based systemic vaccination. Science 1997;276:607-61 1. 

43 Langermann S, Mollby R, Burlein JE, Palaszynski SR, Auguste CG, DeFusco A, Strouse R, 
Schenerman MA, Hultgren SJ, Pinkner JS, Winberg J, Guldevall L, Soderhall M, Ishikawa K, 
Normark S, Koenig S: Vaccination with FimH adhesin protects cynomolgus monkeys from colo- 
nization and infection by uropathogenic Escherichia coli. J Infect Dis 2000; 181 :774-778. 

44 Sajjan US, Sun L, Goldstein R, Forstner JF: Cable (Cbl) type II pili of cystic fibrosis-associated 
Burholderia (Pseudomonas) cepacia: Nucleotide sequence of the cblA major pilin subunit pilin 
gene and novel morphology of the assembled appendage fibers, J Bacteriol 1995;30:1030-1038, 

45 Sakellaris H, Scott JR: Novel tools in an old trade: CSl pilus morphogenesis. Mol Microbiol 
1998;30:681-688, 

46 Voegele K, Sakellaris H, Scott JR: CooB plays a chaperone-like role for the proteins involved in 
formation of CSl pili of enterotoxigenic Escherichia coli. Proc Natl Acad Sci USA 1997;94: 
13257-13261. 

47 Sakellaris H, Munson GP, Scott JR: A conserved residue in the lip proteins of CSl and CFA/I pili 
of enterotoxigenic Escherichia coli that is essential for adherence, Proc Natl Acad Sci USA 1 997; 
96:12828-12832. 

48 Saarela S, Taira S, Nurmiaho-Lassila E-L, Makkonen A, Rhen M: The Escherichia coli G-fimbrial 
lectin protein participates both in fimbrial biogenesis and in recognition of the receptor 
N-acetyl-D-glucosamine. J Bacteriol 1995;177:1477-1484. 

49 Lintermanns P: KaJ'akierizatie van de F17 en Fl 1 1 fimbriae van Escherichia coli en genetische 
analyse van de F17 genkluster; Academi Dissertation Rijksuniversitet Gent, 1990. 



Jonson/Normark/Riien 



84 



50 Strom MS^ Lory S: Structure-function and biogenesis of the type IV pili, Armu Rev Microbiol 
1993;47:565-596, 

51 Faast R, Ogierman MA, Stroeher UH, Manning PA: Nucleotide sequence of the strucmral gene, 
tcpA, for a major pilin subunit of Vibrio cholerae. Gene 1989;85:227-231. 

52 Donnenberg MS, Giron JA, Nalaro JP, Kaper JB: A plasmid-encoded type IV fimbrial gene of 
enteropathogenic Escherichia colt associated with localized adherence, Mol Microbiol 1992;22: 
3427-3437. 

53 Gastric P, Gassels FJ, Carlson RW: Structural characterization of the Pseudomonas aeruginosa 
1244 pilin glycaii. J Biol Chem 2001;276:26479-26485. 

54 Forest KT, Dunham SA, Koomey M, Tainer JA: Crystal lographic structure reveals phosphorylated 
pihn from Neisseria: Phosphoserine sites modify type IV pilus surface chemistry and fibre morpho- 
logy. MoJ Microbiol l999;3]:743-752. 

55 Marceau M, Forest K, Beretti JL, Tainer J, Nassif X; Consequences of the loss of 0-linked 
glycosylation of meningococcal type IV pilin on piliation and pilus-mediated adhesion. Moi 
Microbiol 1998;27:705-715. 

56 Stimson E, Virji M, Makepeace K, Dell A, Morris HR, Payne G, Saunders JR, Jennings MP, 
Baj'ker S, Panico M, et al: Meningococcal pilin: A glycoprotein substituted with digalactosyl 2,4- 
djacetamido-2,4,6-trideoxyhexose. Mol Microbiol 1995;17:1201-1214. 

57 Stimson E, VLrji M, Barker S, Panico M, Blench I, Saunders J, Payne G, Moxon ER^ Dell A, 
Morris HR: Discovery of a novel protein modification: Alpha-glycerophosphate is a substituent of 
meningococcal pilin. Biochem J 1996;316:29-33. 

58 Tonjum T, Koomey M: The pilus colonization factor of pathogenic neisserial species: Organelle 
biogenesis and structure/function relationships- A review. Gene 1997;192/1:155-163. 

59 Ahn RA, Mattick JS: Genes involved in the biogenesis and function of type-4 fimbriae in 
Pseudomonas aeruginosa. Gene 1997;192:89-98. 

60 Beatson SA, Wliitchurch CB, Sargent JL, Levesque RC, Mattick JS: Differential regulation of 
twitching motility and elastase production by Vfr in Pseudomonas aeruginosa, J Bacteriol 2002; 
184:3605-3613. 

61 Karaolis DK, Somara S, Maneval DR Jr, Johnson JA, Kaper JB: A bacteriophage encoding a patho- 
genicity island, a type-lV pilus and a phage receptor in cholera bacteria. Nature 1999;399:375-379. 

62 Fussenegger M, Rudel T, Barten R, Ryll R, Meyer TF: Transformation competence and type-4 
pilus biogenesis in Neisseria gonorrhoeae - A review. Gene 1997;192:125-134. 

63 Skerker JM, Berg HC: Direct observation of extension and retraction of type IV pili. Proc Natl 
Acad Sci USA 2001;98:6901-6904. 

64 Lee KK, Shelh HB, Wong WY, Sherburne R, Paranchych W, Hodges RS, Lingwood CA, Krivan H, 
Irvin RT: The binding of Pseudomonas aeruginosa pili to glycosphingolipids is a tip-associated event 
involvmg the C-terminal region of the structural pilin subunit. Mol Microbiol 1994;1 1:705-713. 

65 Hazes B, Sastry PA, Hayakawa K, Read RJ, Irvin RT: Crystal structure of Pseudomonas aeruginosa 
PAK pilin suggests a main-chain-dominated mode of receptor binding. J Mol Biol 2000;299: 
1005-1017. 

66 Wolfgang M, van Putten JP, Hayes SF, Dorward D, Koomey M: Components and dynamics of fiber 
formation define a ubiquitous biogenesis pathway for bacterial pili, EMBO J 2000;19:6408-6418. 

67 Scheuerpflug I, Rudel T, Ryll R, Pandit J, Meyer TF: Roles of PilC and PilE proteins in pilus- 
mediated adherence of Neisseria gonorrhoeae and Neisseria meningitidis to human erythrocytes 
and endothelial and epithelial cells. Infect Immun 1999;67:834— 843. 

68 Rudel T, Scheurerpflug 1, Meyer TF: Neisseria PilC protein identified as type-4 pilus tip-located 
adhesin. Nature 1995;373:357-359. 

69 Rahman M, KaJlsti'om H, Normark S, Jonsson AB: PilC of pathogenic Neisseria is associated with 
the bacterial cell surface. Mol Microbiol 1997;25:1 1-25. 

70 KailstrSm H, Liszewski MK, Atkinson JP, Jonsson AB: Membrane cofactor protein (MCP or 
CD46) is a cellular pilus receptor for pathogenic Neisseria. Mol Microbiol 1997;25:639-647. 

71 01s6n A, Jonsson A, Normark S: Fibronectin binding mediated by a novel class of surface 
organelles on Escherichia coli. Nature 1989;338:652-655. 

72 Bian Z, Normark S: Nucleator function of CsgB for the assembly of adhesive surface organelles 
in Escherichia coli. EMBO J 1997;16:5827-5836. 



Fimbriae, Pili, Flagella and Bacterial Virulence 85 



73 Sukupolvi S, Lorenz RG, Gordon Jl, Bian Z, Pfeifer JD, Normark SJ, Rhen M: Expression of thin 
aggregative fimbriae promotes interaction of Salmonella typhimiirium SR-1 1 with mouse intestinal 
epithelial cells. Infect Immun 1997;65:5320-5325. 

74 Hammar M, Amqvist A, Bian Z, Ols6n A, Normark S: Expression of two csg operon is required 
for production of fibronectin- and Congo red-binding curii polymers in Escherichia coli K-12. 
Mol Microbiol 1995;18:661-670. 

75 Romling U, Bian Z, Hamniar M, Sierralta WD, Normark S: CurIi fibers are highly conserved 
between Salmonella typhimurium and Escherichia coli with respect to operon structure and regu- 
lation. J Bactriol 1998;180:722-731. 

76 Chapman MR, Robinson LS, Pinkner JS, Roth R, Heuser J, Hammar M, Normark S, Hultgren SJ: 
Role of Escherichia coli curIi operons in directing amyloid fiber formation. Science 2002;295: 
851-855. 

77 Hammar M, Bian Z, Normark S: Nucleator-dependent intercellular assembly of adhesive curIi 
organelles in Escherichia coli. Proc Natl Acad Sci USA 1996;93:6562-6566. 

78 Loferer H, Hammar M, Normark S: Availability of the fibre subunit CsgA and the nucleator protein 

CsgB during assembly of fibronectin-binding curii is limited by the intracellular concentration of 
the novel lipoprotein CsgG. Mol Microbiol 1997;26:l 1-23. 

79 Sjobring U, Pohl G, Ols^n A: Plasminogen, absorbed by Escherichia coli expressing curli or by 
Salmonella enteritidis expressing thin aggregative fimbriae, can be activated by simultaneously 
captured tissue-type plasminogen activator (t-PA). Mol Microbiol 1994; 14:443^52. 

80 Ben Nasr A, Olsen A, Sjobring U, MuUer-Esterl W, Bjorck L: Assembly of human contact phase 
proteins and release of bradykinin at the surface of curli-expressing Escherichia coli, Mol 
Microbiol 1996;20:927-935. 

81 Prigent-CombaretC, PresnierG, LeThiTT, Vidal O, Lejeune P, Dorel C: Developmental pathway for 
biofilm formation in curli-producing Escherichia coli strains: Role of flagella, curli and colanic 
acid. Environ Microbiol 2000;2:450-464. 

82 Zogaj X, Bokranz W, Nimz MM, Romling U: Production of cellulose and curli fimbriae by mem- 
bers of the family Enterobacteriaceae isolated from the human gastrointestinal tract. Infect Immun 
2003;71:4I5I^158. 

83 Zogaj X, Nimz M, Rohde M, Boki'anz W, Romling U: The multicellular morpholypes of 
Salmonella typhimurium and Escherichia coli produce cellulose as a second component of the 
exn^cellular man-ix. Mol Microbiol 2001;39:1452-1463. 

84 Wullt B, Bergsten G, Samuelsson M, Svanborg C: The role of P fimbriae for Escherichia coli 
establishment and mucosal inflammation in the human urinary tract. Int J Antimicrob Agents 
2002;19:522-538. 

85 Schilling JD, Mulvey MA, Hultgren SJ: Structure and function of Escherichia coli type 1 pili: 
New insight into the pathogenesis of urinary tract infection. J Infect Dis 2001;I83:S36-S40. 

86 Hung C-S, Bouckaert J, Hung D, Pinkner J, Widbei^ C, DeFusco A, Augusle CG, Strouse R, 
Langermann S, Waksman G, Hultgren SJ: Structural basis of trophism of Escherichia coli to the 
bladder during urinary tract infection. Mol Microbiol 2002;44:903-915. 

87 Mulvey MA, Lopez-Boado YS, Wilson CL, Roth R, Parks WC, Heuser J, Hultgren SJ: Induction 
and evasion of host defenses by type 1-piliated uropathogenic Escherichia coli. Science 1998; 
282:1494-1497, 

88 Soderhall M, Bergerheim US, Jacobson SH, Lundahl J, Mollby R, Normark S, Winberg J: Molecular 
evidence for /?a^»G-s peel fie adhesion of Escherichia coli to human renal cells. J Urol 1997; 157: 
346-350. 

89 Haslam DB, Bor6n T, Falk P, liver D, Chou A, Xu Z, Normark S: The amino-terminal domain of 
the P-pilus adhesin determines receptor specificity. Mol Microbiol 1 994; 14:399^09. 

90 Jantunen ME, Siitonen A, Koskimies O, Wikstrom S, Karkkainen S, Salo E, Sax6n H: Predominance 
of class ]l papG allele of Escherichia coli in pyelonephritis in infants with normal urinary tract 
anatomy J Infect Dis 2000;181:1822-1824. 

91 Tseng CC, Huang JJ, Ko WC, Yan JJ, Wu JJ: Decreased predominance of papG class II allele in 
Escherichia coli strains isolated from adults with acute pyelonephritis and urinary tract abnor- 
malities. J Urol 2001;166:1643-1646. 



Jonson/Normark/Riien 



86 



92 Martinez JJ, Mulvey MA, Schilling JD, Pinkner JS, Hultgren SJ: Type 1 pilus-mediated bacterial 
invasion of bladder epithelial cells, EMBO J 2000;19:2803-2812. 

93 Marra A, Isberg RR: Invasin-dependent and invasin-independent pathways for translocation of 
Yersinia pseudotuberculosis across the Peyer's patch intestinal epithelium. Infect Immun 1997;65: 
3412-3421. 

94 Anderson GG, Palermo JJ, Schilling JD, Roth R, Heuser J, Hultgren SJ: Intracellular bacterial 
biofilm-like pods in urinary tract infection. Science 2003;30l : 105-107, 

95 Backhead F, Als^n B, Roche N, Angstrom J, von Euler A, Breimer ME, Westerlund-Wickstrom B, 
Teneberg S, Richter-Dahlfors A: Identification of target tissue glycosphingolipids for uropatho- 
genic, FlC-fimbriated Escherichia coli and its role in mucosal inflammation. J Biol Chem 2002; 
277:18198-18205. 

96 Hedlund M, Svensson M, Nilsspn A, Duan R-D, Svanborg C: Role of ceramide-binding in 
cytokine responses to P-fimbriated Escherichia coli, J Exp Med 1996; 183:1037-1044. 

97 Khigatian M, Nassar H, Chou HH, Gibson FC 3rd, Genco CA: Fimbria-dependent activation of 
cell adhesion molecule expression in Porphyromonas gingivalis-\r\?tcitd endothelial cells. Infect 
Jmmun 2002;70:257-267. 

98 Schilling JD, Martin SM, Hunstad DA, Patel ICP, Mulvey MA, Justice SS, Lorenz RG, Hultgren SJ: 
CD14- and Toll-like receptor dependent activation of bladder epithelial cells by lipopoly- 
saccharide and type 1 piliated Escherichia coli. hifect Immun 2003;71:1470-1480. 

99 Frend6us B, Wachtler C, Hedlund M, Fischer K, Samuelsson P, Svensson M, Svanborg C: Escherichia 
coli P fimbriae utilize the Toll-like receptor 4 pathway for cell activation. Mo) Microbiol 2001; 
40:37-51. 

100 Schiitze S. Potthoff K, Machleidt T, Berkovic C, Weigman K, Kronke M: TNF activated NF-kB by 
phosphatidylcholine-specific phospholipase C-induced 'acidic' sphingomyelin breakdown. Cell 
1992;71:765-776. 

101 Ayala BP, Vasquez B, Clary S, Tainer JA, Rodland K, So M: The pilus-induced Ca^"^ flux triggers 
lysosome exocytosis and increases the amouiit of Lampl accessible to Neisseria IgAl protease. 
Cell Microbiol 2001;3:265-275. 

102 Merz AJ, So M, Sheetz MP: Pilus retraction powers bacterial twitching motility. Nature 2000; 
407:98-102. 

103 Merz AJ, So M: Attachment of piliated, Opa- and Ope- gonococci and meningococci to epithelial cells 
elicits cortical actin rearrangements and clustering of tyrosine-phosphorylated proteins. Infect 
Immun 1997;65:4341^349, 

104 Merj AJ, Enns CA, So M: Type IV pili of pathogenic Neissehae elicit cortical plaque formation 
in epithelial cells. Mol Microbiol 1999;32:1316-1332. 

105 Popp A, Billker O, Rudel T: Signal transduction pathways induced by virulence factors of 
Neisseria gonorrhoeae. Int J Med Microbiol 2001;291:307-314. 

1 06 Pujol C, Eugene E, de Saint Martin L, Nassif X: Interaction of Neisseria meningitidis with a polar- 
ized mojiolayer of epithelial cells. Infect Inunun 1997;65:4S30-^842. 

107 Pujol C, Eugene E, Marceau M, Nassif X: The meningococcal PilT protein is required for mduc- 
tion of intimate attachment to epithelial cells following pilus-mediated adhesion. Proc Natl Acad 
SciUSA 1 999;96:40 17^022. 

108 Merz AJ, So M: Interactions of pathogenic neisseriae with epithelial cell membranes. Annu Rev 
Cell Dev Biol 2000;16:423^57. 

109 Hazlett LD, Moon MM, Singh A, Berk RS, Rudner XL: Analysis of adhesion, piliation, prote- 
ase production and ocular infectivity of several R aeruginosa strains. Curr Eye Res I991;10: 
351-362. 

1 10 Comolli JC, Hauser AR, Waite L, Whitchurch CB, Mattick JS, Engel JN: Pseudomonas aeruginosa 
gene products PilT and PilU are required for cytotoxicity in vitro and virulence in a mouse model 
of acute pneumonia. Infect bnmun 1999;67:3625-3630. 

1 1 1 Zolfaghar 1, Evans DJ, Fleiszig SM: Twitching motility contributes to the role of pili in corneal 
infection caused by Pseudomonas aeruginosa. Infect Immun 2003 ;71 :5389-5393. 

1 12 O'Toole GA, Kolter R: Flagellar and twitching motility are necessary fox Pseudomonas aeruginosa 
biofilm development. Mol Microbiol 1998;30:295-304, 



Fimbriae, Pili, Flagella and Bacterial Virulence 87 



1 13 Johansson L, Rytkonen A, Bergman P, Albiger B, Kallstrom H, HokfeltT, Agerberth B, Cattaneo R, 
Jonsson AB: CD46 in meningococcal disease. Science 2003;301:373-375, 

114 Kukkonen M, Saarela S, Lahteenmaki K, Hyvonen U, Westerlund-Wickstrom B, Rhen M, 
Korhonen TK: Identification of Kvo laminin-binding fimbriae, the type I fimbria of Salmonella 
enfehca serovar lyphimuxium and the G fimbria of Escherichia coli, as plasminogen receptors. 
Infect Immun 1998;66:4965^97Q. 

I 15 Saarela S, Westerlund-Wickstrom B, RJien M, Korhonen TK: The GatD protein of the G (FI7) 

fimbrial complex confers adhesiveness of Escherichia coli to laminin. Infect Immun I996;64: 

2857-2860. 
I 16 Parkkinen J, Hacker J, Korhonen TK: Enhancement of tissue plasminogen activator-catalyzed 

plasminogen activation by Escherichia coli S fimbriae associated with neonatal septicaemia and 

meningitis. Thromb Haemost 1991;65:483-486. 
117 Lahteenmaki K, Westerlund B, Kuusela P, Korhonen TK: Immobilization of plasminogen on 

Escherichia coli f^Sig^Wa. FEMS Microbiol Utt 1993;106:309-314. 
I 18 Lautrop H: Bacterium anitratum transferred to the genus cytophaga. Int Bull Bacteriol Nomencl 

1961;1:107-108. 

1 19 Sun H, Zusman DR, Shi W:Type IV pilus of Myxococcus xanthns is a motility apparatus controlled 
by the frz chemosensoi'y system. Curr Biol 2000; 10: 1 143-1 146. 

120 Maier B, Potter L, So M, Long CD, Seifen HS, Sheetz MP: Single pilus motor forces exceed 
1 00 pR Proc Natl Acad Sci USA 2002;99: 1 60 1 2- 1 60 1 7. 

121 Kaiser D: Bacterial motility: How do pili pull? Curr Biol 2000;I0:R777-R780. 

122 Mattick JS:Type IV pili and twitching motility Annu Rev Microbiol 2002;56:289-314. 

123 Rhen M, Wahlstrom E, Korhonen TK: P-fimbriae of Escherichia coli: Fractionation by immune 
precipitation. FEMS Microbiol Lett 1983;18:227-232. 

124 Rhen M, Knowles J, Penttila ME, Sarvas M, Korhonen TX: P-fimbriae of Escherichia coli: Molecular 
cloning of DNA fi'agments containing the structural genes. FEMS Microbiol Lett 1 983; 19:1 19-123. 

1 25 Rhen M, Makela PH, Korhonen TK: P-fimbriae of Escherichia coli are subject to phase variation. 
FEMS Microbiol Lett 1983;19:267-271. 

126 Seifert HS: Questions about gonococcal pilus phase- and antigenic variation. Mol Microbiol 
1 996;2 1:433^40. 

127 Rudel T, van Putten JP, Gibbs CP, Haas R, Meyer TF: Interaction of two variable proteins (PilE 
and PilC) required for pilus-mediated adherence of Neisseria gonorrhoeae to human epithelial 
cells. Mol Microbiol 1992;6:3439-3450. 

128 Virji M, Saunders JR, SimsG, Makepeace K, Maskell D, Ferguson DJ: Pilus-facilitated adherence 
of Neisseria meningiiidis to human epithelial and endothelial cells: Modulation of adherence 
phenotype occurs concurrently with changes in primary amino acid sequence and the glycosylation 
status of pilin. Mol Microbiol 1993;10:1013-1028. 

129 Nassif X, Lowy J, Stenberg P, O'Gaora P, Ganji A, So M: Antigenic variation of pilin regulates 
adhesion of Neisseria meningitidis to human epithelial cells. Mol Microbiol 1993;8:719-725. 

130 Jonsson AB, liver D, Falk P, Pepose J, Normark S: Sequence changes in the pilus subunit lead to 
tropism variation of Neisseria gonorrhoeae to human tissue. Mol Microbiol 1994;13:403-416. 

131 Marceau M, Beretti JL, Nassif X: High adhesiveness of encapsulated Neisseria meningitidis to 
epithelial cells is associated with the formation of bundles of pili. Mol Microbiol 1995;17:855-863. 

132 Park HS, Wolfgang M, van Putten JP, Dotward D, Hayes SF, Koomey M: Structural alterations in 
a type IV pilus subunit protein result in concurrent defects in multicellular behaviour and adher- 
ence to host tissue. Mol Microbiol 2001;42:293-307. 

133 Richardson K: Roles of motility and flagellar structure in pathogenicity of Vibrio cholerae: Analysis 
of motility mutants in three animal models. Infect Immun 1991;59:2727-2736. 

134 Ottemann KM, Lowenthal AC: Helicobacter pylori uses motility for initial colonization and to 
attain robust infection, infect immun 2002;70:1984-1990. 

135 Liaw SJ, Lai HC, Ho SW, Luh KT, Wang WB: Role of RsmA in the regulation of swarming motility 
and virulence factor expression in Proteus mirabilis. J Med Microbiol 2003;52:19-28. 

136 Xu Q, Dziejman M, Mekalanos JJ: Determination of the transcriptome of Vibrio cholerae during 
intra intestinal growth and midexponential phase in vitro. Proc Natl Acad Sci USA 2003; 100: 
1286-1291. 



Jonson/Normark/Riien 



88 



137 Merrell DS, Butler SM, Qadri F, Dolganw NA, Alam A, Cohen MB, CaLderwood SB, Schoolnik GK, 
Camilli A; Host-induced epidemic spread of the cholera bacterium. Nature 2002;417:642-645. 

1 38 Kavermann H, Bums BP, AngermullerK, Odenbreit S, Fischer W, Melchers K, Haas R: Identification 
and characterization of Helicobacter pylori genes essential for gastric colonization. J Exp Med 
2003;197:813-822. 

139 TasteyreA, BarcMC, Collignon A, Boureau H, KarjalainenT: RoleofFliCand Fli.D flagellar pro- 
teins of C/o5/r/£//i^mrf/^c/7e in adherence and gut colonization. Infect bnmun 2001;69:7937-7940. 

140 Feldman M, Bryan R, Scheffler L, Brunnert S, Tang H, Prince A: Role of tlagella in pathogenesis 
of Pseudomonas aemginosa 'p\\\ovc\2ixy infection. Infect Tmmun 1998;66:43-51. 

141 Allen-Vercoe E, Woodward MJ: The role of flagella, but not fimbriae, in the adherence of 
Salmonella enterica serotype enteritidis lo chick gut explant. J Med Microbiol 1999;48:771-780. 

142 Gavin R, Merino S, AltarribaM, Canals R, Shaw JG,Tomas JM: Lateral flagella for increased cell 
adherence, invasion and biofilm formation by Aeromonas spp. FEMS Microbiol Lett 2003; 
224:77-83. 

143 Gewirtz AT, NavasTA, Lyons S,Godowski PJ,Madara J L: Cutting edge: Bacterial flagel I in activates 
basolaterally expressed TLR5 to induce epithelial proinflammatory gene expression. J Immunol 
2001;167:1882-1885. 

144 MJzel SB, Snipes JA: Gram-negative flagellin-induced self-tolerance is associated with a block in 
interleukin-1 receptor-associated kinase release from toll-like receptor 5. J Biol Chem 2002;277: 
22414-22420. 

145 Hueck CJ: Type Fl! secretion systems in bacterial pathogens of animals and plants. Microbiol Mol 
Biol Rev l998;62;379-433. 

146 Young GM, Schmiel DH, Miller VL: A new pathway for the secretion of virulence factors by 
bacteria: The flagellar export apparatus flinctions as a protein-secretion system, Proc Natl Acad Sci 
USA 1999;96:6456-6461. 

147 Murray RA, Dilva M, McCormick BA, Lee CA: Salmonella typhimiirium induced enteritis: 
Identification of alternative pathways for SipA secretion; in Microbial Pathogenesis and Host 
Response. Cold Spring Harbor, Cold Spring Harbor Laboratory, 2001, p 140. 



Mikael Rhen 

Microbiology and Tumor Biology Center, Karolinska Institute 

Nobels vag 16, SE-171 77 Stockholm (Sweden) 

Fax +46 8 301797, E-Mail mikael.rhen@mtc.ki.se 



Fimbriae, Pili, Flagella and Bacterial Virulence 89 



Adhesins 



Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kai'ger, 2005, vol 12, pp 90^1 13 



Gram-Positive Adhesins 



Susanne R. Talay 

GBF-Gcrman Research Centre for Biotechnology, Braunschweig, Germany 



In the process of bacterial infection, adhesion to host tissues represents an 
initial and essential step. Adhesion allows the pathogen to attach to and colonize 
specific sites of the body, thereby withstanding eradication through cleansing 
mechanisms such as excretion and peristalsis. Once attached to the target tissue, 
bacteria may either remain extracellular, multiply, and eventually spread into 
deeper tissue, or trigger their own uptake by host cells, resulting in an intra- 
cellular location that may allow the pathogen to persist or further spread within 
the cellular or subcellular compartment. 

Bacterial surface components that mediate adherence are called adhesins. 
Among gram-positive pathogens, surface proteins represent the largest group of 
adhesins, although other factors such as polysaccharides and lipids may also 
display adhesive functions. Targets for these microbial adhesins are host mole- 
cules found on mucosal surfaces, skin, and wounds. Depending on the strength 
of this interaction, adhesins allow the pathogen to loosely associate with or 
intimately bind to specific cells or tissues. Most gram-positive pathogens express 
multiple adhesins that may bind to either the same or distinct target molecules. 
Multiple adhesins of one pathogen are likely to be involved in different stages 
of an infection, expressed under different environmentally determined condi- 
tions, and may display a redundant function. Li the present article, adhesins 
of pathogenic gram-positive bacteria belonging to the genus Streptococcus, 
Staphylococcus and Listeria, as well as the most important host molecules targeted 
by these adhesins are reviewed. 



The Extracellular Matrix -A Major Target for Pathogens 

Many adhesins function by specifically recognizing and binding to various 
components found in the extracellular matrix (ECM) of the host. The ECM 



forms the major structural support for cells and tissues and is responsible for 
maintaining the strength and elasticity of the body. Thus, it is ubiquitously present 
and frequently exposed in cases such as trauma and injury, a situation that renders 
its constituents ideal targets for many adhesins. The following section gives a 
short overview on the major ECM components, their structure and their basic 
function. 

Collagens 

Collagens are the most abundant protehis in the mammalian body and it is 
well recognized that collagens fulfill an important structural role in the ECM in 
a number of tissues. More than 25 distinct collagen types have been identified, 
in which identical or distinct a chains form a triple helix. Collagens can be 
divided into fibril-forming interstitial collagens (e.g. types I, II, III, V, and XI) 
and non-fibril-forming collagens such as type IV, VI, and X [1]. Type I collagen 
is found bi tendons and muscle, while type II collagen is the major constituent of 
cartilage. The nonfibrillar type IV collagen is the major constituent of basement 
membranes, forming a network with laminins, nidogen, and sulfated 
proteoglycans. Collagen IV is composed of six chains (al-a6) that form three 
basic sets of triple helical molecules. Collagens may interact with a variety of 
factors, Lncludmg other matrix components such as fibronectin and laminin, as 
well as matrix metal loproteinases. The binding of collagen to cells is mediated 
by integrins, which constitute another group of receptors for collagens. Currently 
four collagen-binding integrins are known, a|(3|, cd2Ph otioPi ^^id ctnPi, that 
mediate cellular binding and signalling. Bacterial binding to collagens, such as 
cartilage collagen and basement membrane collagen, represent important adhesion 
mechanisms among pathogens. 

Fibronectin 

Fibronectin, which exists both as a soluble protein in plasma and as a 
fibrillar polymer in the ECM, is a large glycoprotein involved in cell adhesion, 
migration, and differentiation. Fibronectin exists as a dimer composed of two 
250-kD subunits which are carboxy-terminally linked via a pan- of disulfide 
bonds [2]. Each subunit contains three distinct types of modules, the type I, II 
and III modules (fig. 1). Fibronectin efficiently binds to cell surfaces via 
numerous integrins, including the classic fibronectin-binding integrin, agp, 
integrin. Integrin bindmg is mediated by an RGD sequence and also involves 
secondary sites on the fibronectin molecule. In addition to the interaction with 
integrms, fibronectm associates with heparin, collagen/gelatin, and fibrin. 
Heparin binding is governed by three domains that interact with heparan sulfate 
proteoglycans. Bmding to collagen is mediated by type I repeats 6—9 and the 
two type II repeats. The two fibrin-binding sites are located at the carboxy- and 



Gram-Positive Adhesins 



91 



a 



COOH 




HOOC 



5>3-NH; 




50 Residues 



NHj- 




T ^ 



PRR 



1 I 2 1 _3 I 4 1 5"! [W| M H- COOH 



Spacer 



FnBR 




LPATG 



Fig, L a Modular structure of fibronectin (one subunit). Module types 1, 2 and 3 are 
symbolized by pentagons, hexagons, and circles, respectively. The amino-terminal domain 
('"^Fl), the collagen-binding domain (CBD), alternatively spliced sites (curved labels) and 
the major integrin-binding site (RGD) are labelled. bl\\t extended tandem |3-zipper model 
of Sfbl binding to '"^Fl. A model of the amino-terrmnal domain is shown on top. Short con- 
secutive segments of a fibronectin-binding repeat form antiparallel p-strands on triple- 
stranded (3-sheets ofall five homologous Fl modules, c Molecular organization of Sfbl from 
S, pyogenes. S = Signal peptide; A = nonhomologous region; PRR = proline-rich repeats; 
spacer — upstream fibronectin-binding region; 1-5 — Fn-binding repeats; W — cell wall 
spanning sequence; LPATG = cell wall anchor; M = membrane spanning region. (The 
tandem zipper model was kindly provided by Dr. Ulrich Schwarz-Linek, University of 
Oxford, UK,) 



ammo-teniiinal part of the molecule, the major site being formed by type I 
modules 4 and 5 located at the amino-terminal domain. Fibrin-binding and 
cross-linking to fibronectin via factor XJlla is important in the generation of 
fibrin clots that form a provisional ECM network in the wound healing process. 
Fibronectin is an ideal target for many pathogens due to its wide presence in 
exudates, blood, wounds, as well as on the surface of cells. 

Laminin 

Laminin is a 900-kD glycoprotein and is a major component of the basement 
membrane. Its macromolecular structure is formed by assembly of tbree distinct 



Talay 



92 



polypeptide chains, a, p, and 7 [3]. Laminin functionally interacts with other 
components of basement membranes such as collagen IV and a variety of proteo- 
glycans and ECM molecules. More than 10 different isoforms of laminin are 
known to be involved in cell proliferation and attachment, as well as in chemotaxis 
and angiogenesis. In the case of epithelial and endothelial injury, basement 
membrane components such as lammin are likely to be exposed and may serve 
as target structures for bacterial colonization of damaged tissue, 

Elaslin 

Elastin is the major ECM protein of lung, skin and large arteries such as 
the aorta, imparting characteristics of extensibility and elastic recoil [4]. Elastin 
is formed by polymerization and cross-linking of its precursor tropoelastin. 
This process of ordered self-aggregation is called coacervation. Once deposited 
in tissues, polymeric elastin is not subject to turnover, but is able to sustain its 
mechanical resilience through millions of cycles of extension and recoil. 
Elastin consists of approximately 36 domains with alternating hydrophobic and 
cross-linking characteristics. The rubber-like mechanical properties result from 
the repetitive hydrophobic domains of tropoelastin that display an unstructured 
organization with higher entropy in the relaxed state, and a structured organi- 
zation with lower entropy in the extended state. The major binding partners for 
tropoelastm are fibrillins, the main components of microfibrils which themselves 
may be attached to cells, Elastin serves as a target for pathogenic staphylococci, 
which use this molecule for adhesion to host tissue. 

Viironectin 

Vitronectin is a multiftinctional 75-kD glycoprotein present in blood and 
the ECM [5]. It binds collagen, plasminogen and the urokinase receptor, and 
stabilizes the inhibitory conformation of plasminogen activation inhibitor-1, 
thereby regulating the proteolytic degradation of the ECM. It further interacts 
with glycosaminoglycans via its carboxy-terminal part, and integrins of the ay 
family via an ROD sequence located at the most ammo-terminal part Binding 
of the RGD sequence to integrins induces signalling cascades, and mediates 
attachment and spreading of cells on the matrix. Through its localization in the 
ECM and its binding to plasminogen activation inhibitor-1, vitronectin can 
potentially regulate the proteolytic degradation of this matrix. In addition, 
vitronectin binds to complement factors, heparin and thrombin-antithrombin III 
complexes, and therefore participates in the regulation of clot formation. The 
biological functions of vitronectin can be modulated by proteolytic enzymes, 
and exo- and ecto-protein kinases which are present in blood. Like fibronectin, 
vitronectin is an ideal target for adhesins of pathogens due to its presence in the 
ECM, in blood, and at sites of tissue injury. 



Gram-Positive Adhesins 



93 



Fibrinogen 

Fibrinogen is a 340-kD plasma glycoprotein composed of six polypeptide 
chains, two Aa, two B[3, and two -y chains that form a dimer. In the vascular 
system, fibrinogen mediates platelet adherence and aggregation at sites of 
trauma and injury, thereby acting as an important clotting factor [6]. Upon 
interaction with thrombin, subsequent stabilization of the fibrin clot is achieved 
by transglutaminase/factor XJIla-mediated cross-linkage of the 7 and a chains 
of fibrinogen. Binding to platelets is mediated through the interaction of 
fibrinogen with integrin a]jb(33 on the platelet surface. In addition to its function 
in the coagulation system, fibrinogen also participates in inflammatory 
responses. Fibrinogen mediates leukocyte attachment to the vessel wall and 
transmigration through the endothelium. Fibrinogen binds to aMp2 integrin on 
leukocytes and to ci^\^2 ifitegrin on macrophages, thereby regulating phagocytic 
clearance of fibrin clots during wound healing. Interaction with integrins is 
governed by two RGD sequences and other defined epitopes on the fibrinogen 
molecule. In addition, fibrinogen has the ability to bind a variety of factors such 
as fibronectin, collagen, and components of the fibrinolytic system, implicat- 
ing this protein as a key factor in matrix organization, remodelling and wound 
repair. Many gram-positive pathogens have evolved distinct factors that speci- 
fically bind fibrinogen, evoking bacterial adhesion, aggregation, and evasion of 
phagocytosis. 

G lye OS am in oglycan s 

Glycosaminoglycans are polysaccharide chains covalently linked to a protein 
core to form proteoglycans. Being composed of distinct repeating disaccharide 
units, these molecules can be divided into different classes such as heparan 
sulfate, dermatan sulfate, and chondroitin sulfate. Glycosaminoglycans are pre- 
sent in the ECM of connective tissue but are also expressed on the surface of 
eukaryotic cells. Heparan sulfate [7] and dermatan sulfate [8] are ubiquitously 
found on the surface of cells and in the ECM and skin. Glycosaminoglycans 
fimction as stabilizers, cofactors and coreceptors of cytokines and chemokines, 
regulators of enzymatic activity, and signalling molecules in response lo injury 
or infection. Glycosaminoglycans may mediate adherence and entry of pathogens 
including bacteria, viruses and parasites [8]. 



Streptococcal Adhesins 

Streptococcus pyogenes 

S. pyogenes, the group A Streptococcus, is an important human pathogen 
that causes localized infections of the respiratory tract and the skin, but also in 



Talay 



94 



severe invasive diseases, such as sepsis and toxic shock-like syndrome. Severe 
nonsuppurative sequelae such as acute rheumatic fever and glomerulonephritis 
may follow primary group A streptococcal infection. S. pyogenes initiates infec- 
tion by interacting specifically with host molecules present on mucosal surfaces 
or skin. A variety of different adhesins that either bind to identical or distinct tar- 
get molecules are expressed by lS". pyogenes (table 1). Among the large number 
of bacterial factors that bind to host molecules, onJy those for which adhesive 
properties were clearly demonstrated are herein termed adhesins. 

S. pyogenes possesses at least nine distinct fibronectin-binding adhesins. 
Some of these occur in a large number of serotypes, such as Sfbl protein or 
FBP54, whereas others such as Ml or M3 protein are exclusively expressed by 
Ml or M3 serotypes, respectively. Among all fibronectin-binding adhesins of 
S. pyogenes, Sfbl protein and its allelic variant Fl are the most extensively stud- 
ied. Identified iii 1992, Sfbl/Fl was shown to act as an adhesin on epithelial cells 
[9^ 10]. Sfbl protein has a modular architecture [11], and binds to fibronectin via 
two distinct domains [12, 13]. The carboxy-terminal repeat region and the adja- 
cent nonrepetitive domain termed spacer 2 or UR synergistically bind to two dis- 
tinct regions on the fibronectin molecule: the amino-terminal fibrin-binding 
fragment (harboring fibronectin Fl modules 1-5) and the gelatine/collagen- 
binding fragment (harboring Fl modules 6-9 and the two F2 modules) [14]. The 
carboxy-terminal repeat region of Sfbl was demonstrated to be sufficient to 
mediate adherence to epithelial cells [14]. However, besides this activity, Sfbl 
acts as a potent invasin that triggers internalization into eukaryotic cells [14-18]. 
Sfbl mediates attachment to epithelial cells of the oral mucosa and the lung, but 
also to endothelial cells [1 8]. Binding to human cells was shown to be dependent 
on the presence of fibronectin-binding integrins [17], leading to the concept that 
fibronectin acts as a bridging molecule between bacteria and host cell mtegrins. 
Besides its potential to bind to cell surfaces, Sfbl has the ability to recruit collagen 
via prebound fibronectin, a mechanism that enables the bacteria to form 
aggregates and renders the organism capable of colonizing collagen matrix [19], 
The overall pathogenic potential of this protein is underlined by vaccination 
studies using recombinant Sfbl that protected mice fi'om lethal 5*. pyogenes 
infection [20]. Recently, the first three-dimensional stmcture for a bacterial 
fibronectin-binding peptide, the B3T peptide derived from the Streptococcus 
dysgalactiae FnBP, in complex with the ^FHFl module of fibronectin was 
obtained. Based on this structural information, a compelling model for the inter- 
action of the fibronectin-binding repeats of Sfbl with the amino-terminal 
domain of fibronectin was developed, termed the tandem (3-zipper model [21]. 
Short motifs within each of the carboxy-terminal repeats of Sfbl w^ere predicted 
to form antiparallel (5 strands along the five Fl modules in the amino-terminal 
domain of fibronectin (fig. 1)^ leading to binding affinities in the nanomolar 



Gram-Posilive Adhesins 



95 



Table L Streptococcal adhesins 



Adhesin 


Ligand niolecu e 


Target cells, tissue 


Reference no. 


S. pyogenes 








Sfbl/Fl 


fibronectin, collagen 


pharynx and lung 
epithelial cells, 
endothelial cells, 
CO agen matrix 


9 19,21 


F2/PFBP 


fibronectin 


n.c. 


22,23 


FBP54 


fibronectin 


buccal epithelial cells 


24 


hba 


fibronectin 


epithelial cells 


26 


hbaB 


fibronectin 


epithe ial cells 


27 


Protein H 


fibronectin, M proteins 


epithelial cells 


28,29 


Ml protein 


fibronectin 


epithelial cells 


30 32 


ITA 


fibronectin, macrophage 


epithe ial cells. 


33,34 




scavenger receptor 


macrophages 




M3 protein 


type 1 and IV collagen, 
fibronectin 


collagen matrix 


35,36 


Cpa 


type ] CO lagen 


n.c. 


37 


HA capsule 


type ] and IV collagen, 


collagen matrix. 


35,38 




Cl)44 


keratinocytes 




M6 protein 


CD46 


keratinocytes 


39,40 


M proteins 


g ucosaminoglycans, 


epithelial cells, 


29,41,42 




M proteins 


Tbroblast ce Is 




Lbp 


aniinin 


epithe ial ce Is 


43 


SpeB 


aJiLinin, glycoproteins 


n.d. 


44 


R?8 


n.d. 


cervical epithe ial cells 


45 


Scl A/Scl 1 


n.d. 


pharyngeal cells 


46,47 


Sc B/ScJ2 


n.d. 


fibrob ast ce s 


48,49 


S, agalactiae 










ScpB 


fibronectin 


pharynx and ung 


50,51 








epithelial cells 




^mb 


aminin 


n.c. 


52 


A pha C protein 


n.d. 


cervica epithelial ce s 


53 


S. pneumoniae 








SpsA/CbpA/PspC 


SC, slgA, factor H 


plgR-expressing cells 


56-61 


Phosphoiy choline 


PAF receptor 


endothelia and 
epithelial eel s 


62 


PavA 


fibronectin 


n.d. 


63,64 


n.d. ->]ot determined. 



Talay 



96 



range. This is extremely important since high affinity binding is a prerequisite 
for bacterial attachment, a mechanism that has to withstand shear forces occur- 
ring on the mucosal surfaces or during the mternalization process. 

Protein F2 or PFBP are homologous but distinct fibronectin-binding 
proteins, found in most isolates of S. pyogenes lacking the sfbllprtFl gene 
[22, 23]. Like Sfbl, protein F2 possesses two binding domains that interact with 
fibronectin. 

Among the genes encoding fibronectin-binding proteins, the gene for 
FBP54 is the most abundant and found in all S. pyogenes isolates [24]. Although 
lacking the classical membrane anchor motif of gram-positive surface proteins, 
it appears to be localized on the streptococcal surface by a distmct mechanism 
[25], thereby acting as an adhesin for buccal epithelial cells but not for HEp2 
cells [24]. These data also indicate that distinct fibronectin-binding factors may 
target different cell types and have a substantial effect on cell tropism. 

Two other recently discovered fibronectin-binding proteins are Fba and 
FbaB [26, 27]. Thtfba gene was found m 5 serotypes of S. pyogenes including 
M types 1 and 49. An Fba mutant showed diminished adhesion to HEp2 cells, 
suggesting that this protein has adhesive properties [26]. However, it should 
be noted that the FbaB protein was only found in serotype M3/M 1 8 S. pyogenes 
isolates and appears to be genetically most closely related to protein F2 [27]. 

Protein H, a member of the M protein family, binds to fibronectin in a unique 
manner [28]. Unlike the proteins described so far that mainly interact with the 
type 1 or type 11 module containing domains of fibronectin, protein H binds to the 
type III modules. In addition, protein H was shown to mediate streptococcal 
aggregation through a so-called AHP sequence that also promoted adhesion to 
epithelial cells [29]. 

Ml protein, another member of the M protein family, was demonstrated to 
bind fibronectin [30], and Ml -specific antibodies efficiently blocked adherence 
to HeLa cells. Moreover, an Ml -deficient mutant showed reduced adherence 
and invasion, indicating that Ml protein acts as an adhesin and invasin m serotype 
Ml S. pyogenes strains [31]. Importantly, as in the case of Sfbl protein, agPi 
integruis are the termmal receptor proteins on the cellular surface [32]. 

Lipoteichoic acid (LTA) was suggested to interact with fibronectin or 
hydrophobic residues on the cellular surface. It was defined as a first step 
adhesin, mediating low affinity and reversible binding to the ligand, whereas 
protein adhesins with high affinity binding to the ligand were termed second 
step adhesins [reviewed in 33]. At least one other cellular receptor exists for 
LTA: the type I macrophage scavenger receptor which exhibits a broad range of 
binding specificity [34]. 

Recent findings identified M3 protein as an important adhesin that binds 
to soluble type 1 and type IV collagen as well as to the native collagen matrix 



Gram-Positive Adhesins 



97 




Fig. Z Serotype M3 S. pyogenes adhering to collagen type I fibers via the M3 protein, 
(The scanning electron-microscopic image was kindly provided by Dr. Manfred Rohde, GBF, 
Braunschweig, Germany.) 



[35] (fig- 2). The amino- terminal variable but M3-specific region of M3 protein 
is essential for collagen binding, explaining why other M proteins lack this 
function. Besides attaching bacteria directly to collagen matrix, aggregation 
of soluble collagen on the bacterial surface leads to formation of large bacte- 
rial aggregates that facilitate the colonization process [35]. The only other 
collagen-binding protein of 5". pyogenes described so far is Cpa, which was 
identified in the M49 serotype and was suggested to mediate attachment to 
immobilized type 1 collagen [37], 

In highly encapsulated MI8 streptococci, collagen-binding activity and 
adhesive properties are mediated by the hyaluronic acid (HA) capsule. The 
assumption that M3 protein and streptococcal HA could indeed mediate adherence 
to the collagenous matrix was demonstrated ex vivo on native collagen fibers 
and in vivo by using a skin infection mouse model [35]. Apart from binding to 
collagen, HA interacts with human CD44 on the surface of keratinocytes, acting 
as an adliesin for the major cell type of the human pharyngeal epithelium and 
external skin [38]. This finding was of particular importance since former studies 
suggested an inhibitory role of HA in streptococcal cell attachment. The current 
concept, however, is that HA may act as an adhesin itself but may also mask 
binding interactions of other streptococcal surface molecules, depending on the 
type of the M serotype or tissue [38]. 

Another target receptor present on the surface of keratinocytes is 
CD46, the membrane cofactor protem which is bound by M6 protein [39]. The 
carboxy-terminal region of M6 protein as well as the short consensus domains 
3 and 4 of CD46 were shown to be crucial for M6/CD46-mediated keratinocyte 



Talay 



98 



attachment [40]. Although structurally closely related, M proteins represent a 
heterogeneous group of adhesins with respect to their ligands or target cells 
[41]. In contrast to the binding properties displayed by individual M proteins, 
such as the fibronectin-binding activity of Ml protein or the collagen-binding 
of M3 protein, homophilic interactions of M protein [29] and interactions with 
glycosaminoglycans [42] represent common adherence mechanisms among 
several types of M proteins. This is underlined by the finding that interactions 
with several types of glycosaminoglycans such as dermatan sulfate and heparan 
sulfate are predominantly, although not exclusively, mediated via the conserved 
carboxy-terminal part of the M proteins [42]. 

Laminin, another constituent of the ECM, also represents a target for 
S. pyogenes. Two lam in in- binding proteins are known, Lbp that has adhesive 
properties for epithelial cells [43], and SpeB, the secreted cysteine protease 
which also displays glycoprotein-binding activity [44]. Whether SpeB indeed 
mediates adherence to host cells remains to be determined. 

Within S. pyogenes, three adhesins have been identified of which the ligand 
molecules are still unknown. R28, a highly repetitive surface protein related to 
the Streptococcus agalactiae surface proteins Rib and a, binds to cervical 
epithelial cells [45], and two distinct collagen-like proteins termed SclA/Scll 
and SclB/Scl2 were shown to bind to pharyngeal and fibroblast cells, respec- 
tively [46-^9]. Since the set genes appear to be prevalent in all S. pyogenes 
serotypes, are differentially regulated, and display adhesive function, precise 
functional analysis of these potentially important factors will be helpful to 
understand their role in the infection process. 

During recent years a large number of S. pyogenes adhesins have been 
identified and considerable progress has been made by analyzing the molecular 
mechanisms underlying the process of bacterial attachment to host cells and tissue. 
Future challenges will be to elucidate the three-dimensional stnjcture of receptor/ 
ligand complexes that will lead to a better understanding of the molecular 
nature of these interactions, and the development and use of appropriate in vivo 
and ex vivo models for studying the role of the adhesins in the infection process. 
The emerging number of available biockout cell lines and mice will serve as 
helpful tools, defining a promising interdisciplinary cutting edge between mouse 
genomics and infection biology. 

S. agalactiae 

S. agalactiae, the group B streptococcus, is a gram-positive commensal of 
the human vagina, but also the major cause of neonatal sepsis and meningitis. 
vS". agalactiae may also cause serious infections in immunocompromised adults. 
Compared to S. pyogenes, the number of adhesins identified so far is relatively 
small (table I). The host molecules known to be targeted by S. agalactiae are 



Gram-Positive Adhesins 



99 



fibronectin [50, 51], laminin [52, 53], and cytokeratin 8 [54]. The only known 
fibronectin-binding factor of group B streptococci is C5a peptidase (ScpB), a 
large serine protease that is secreted but also attached to the streptococcal sur- 
face. Purified recombinant ScpB was demonstrated to bind to immobilized 
fibronectin [51], as well as to HEp2 and A549 cells [50], 

Lmb, a surface-associated lipoprotein belonging to the Lral family of pro- 
teins, was shown to mediate attachment of group B streptococci to laminin [52]. 
Whether Lmb indeed acts as an adhesin remains to be determined. Other data 
suggest a direct role for the alpha C protein in adherence to cervical epithelial 
cell [53]. The alpha C protein is the prototype for a family of long tandem repeat- 
containing surface proteins that also include R28 of S. pyogenes and Esp of 
Enterococcus faecalis . The cellular receptor for alpha C protein is, as in the case 
of R28, still unknown. The molecular nature of another streptococcal adhesin 
that binds to cytokeratin 8 [54], a molecule potentially important for coloniza- 
tion of keratinized epithelium or damaged cells, also remains to be identified. 

Streptococcus pneumoniae 

*S. pneumoniae, the pneumococcus, is a natural colonizer of the nasopharyn- 
geal epithelium and has the ability to penetrate the epithelial barrier, to translocate 
into deeper tissue, where it can cause severe infections such as pneumonia, 
meningitis and sepsis. Although binding Xo laminin, type IV collagen, and 
vitronectin was described over a decade ago [55], onJy three adhesins that bind 
to other target molecules have been identified in this streptococcal species. 

To date, the best-studied adhesin of S. pneumoniae is SpsA, also named 
CbpA or PspC [56-58]. SpsA binds to human secretory IgA [56, 59], mediates 
adherence to activated human cells [57], and uses the human polymeric 
immunogJobulin receptor as a terminal receptor on the surface of host cells for 
adherence and translocation [60]. In addirion to these properties, SpsA is a pro- 
tective antigen that also binds to factor H [58, 61], suggesting a multifunctional 
role for this adhesin. 

Attachment of pneumococci to activated cells was also shown to be mediated 
Lhrough phosphory] choline on the bacterial surface, employing the platelet- 
activating factor (PAF) receptor as a target molecule on the cellular surface 
[62]. PAF receptor-mediated adherence was found to be coupled to invasion of 
epithelial and endothelial cells, suggesting a direct role for this interaction in 
subcellular spreading of the pathogen [62]. 

Among the various ECM molecules, fibronectin is one of the target mole- 
cules used by pneumococci for attachment [63]. The binding site of pneumo- 
cocci was suggested to be located within the carboxy-terminal portion of 
fibronectin. Immobilized rather than soluble fibronectin was shown to be bound 
by this bacterial species, discriminating this binding factor from most of the 



Talay 



100 



fibronectm-bindiiig proteins found in S, pyogenes or Staphylococcus aureus, 
which efficiently bind to soluble fibronectin as well. PavA, a surface-associated 
pneumococcal protein, was identified as receptor for immobilized fibronectin 
[64], It displays high similarity to FBP54, its orthologue found in S, pyogenes. 
Evident data demonstrate that PavA is essential for virulence [64]; however, its 
precise role in mediating cell or tissue adherence remains to be defined. 



Staphylococcal Adhesins 

S, aureus is an important opportunistic pathogen of humans and animals. The 
spectrum of diseases ranges from superficial skin infection to serious infections 
such as endocarditis, septic arthritis, and community-acquired and nosocomial 
sepsis. Besides this, S. aureus is a major cause of infections originating from 
catheters and implanted synthetic medical devices. 

Many S. aureus isolates have the ability to bind fibronectin. Most strains 
express FnbpA and FnbpB (table 2), two related fibronectin-binding proteins 
encoded by closely linked genes [65-67]. These two proteins were shown to bind 
soluble and immobilized fibronectin via their carboxy-terminal repeat region^ 
whereas FnbpA was also shown to bind fibrinogen via its amino-terminal A 
domain [68], In vitro infection experiments employing distinct cell types as well 
as isogenic S, aureus strains either expressing or lacking one or both Fnbps 
revealed that fibronectin-coated devices, human epithelial cells, endothelial 
cells, and T lymphocytes are targets for Fnbp-mediated adhesion [69-73]. As in 
the case for the S, pyogenes fibronectin-binding proteins Sfbl/Fl and Ml, the 
underlying mechanism for this interaction was shown to be the use of fibronectin 
as a bridging molecule between the bacteria and host cell integrins such as 
a5pi integrin [71, 74, 75]. Consequently, S. aureus Fnbps may act as invasins 
governing the uptake of staphylococci by human epithelial and endothelial cells 
[74—78]. Analogous to the fibronectin-binding repeat region of Sfbl from S. pyo- 
genes^ the domain in fibronectin which is recognized by the Fnbp repeat region 
is located at the amino-tenninus of the molecuJe, being composed of five Fl 
modules [79-82]. The interacting Fnbp repeat region was suggested to be 
unfolded, undergoing a conformational shift upon interaction with the Fl mod- 
ules of fibronectin [83, 84]. Based on recent NMR-based structural data, FnbpA 
contains 1 1 fibronectin-binding repeat segments, each of which can potentially 
bind sequential Fl modules, most likely through the tandem p-zipper mecha- 
nism that has also been suggested for Sfbl protein [21] (fig. 1). Altogether, these 
findings provide substantial insight into the molecular mechanisms of fibronectin- 
mediated adherence of pathogenic cocci. Whether Fnbps of S. aureus are also 
able to recruit collagen via prebound fibronectin remains to be determined. 



Gram-Positive Adhesins 1 



Table Z S. aureus adhesins 



Adhesm 


^igand mo ecule 


Target ce Is, tissue 


Reference no. 


FnbpA 


fibronectin, fibrinogen 


epithelial cells, endothelial 
cells, mammary g ands, 
'1' lymphocytes 


65 75 


FnbpB 


fibronectin 


epithelial cells, endothelia 
cells, mammary glands 


67, 69 75 


Ebh 


fibronectin 


9 


85 


Cna 


CO agen 


cartilage 


86 94 


ClfA 


fibrinogen 


thrombi, implanted 
biomateria 


95 100 


ClfB 


fibrinogen, cytokeratin 


thrombi, implanted 
biomaterial, keratinocytes, 
nasal epithe ia ce s 


101 102 


SasG 


9 


nasa epithe ia cells 


103, 105 


Pis 


9 


nasal epithelial cells 


104, 105 


Bbp 


bone sia oprotein 


bone tissue 


106 


Spa 


vWF 


damaged endothe ium 


108 


vWbp 


vWF 


7 


109 


Map/F.ap 


fibronectin, fibrinogen, 


epithelial cells, fibroblast 


110-116 




vitronectin, bone sialo- 


ce Is 






protein, thrombospondin, 








collagen, osteopontin 








lCAM-1 






E!mp 


fibronectin, fibrinogen, 
vitronectin, collagen 


9 

t 


117 


EbpS 


elastin 


? 


118 120 


PI A 


9 


bio film formation, 
ce l-cell adhesion 


121 


Capsule 


9 

■ 


epithe ial cells, 
endothelial cells 


122 



Another fibronectin-binding protein of 5. aureus is Ebh, a large Kl- 
megadalton surface-associated protein that has been shown to bind soluble and 
irrmnobilized fibronectin [85]. The role of Ebh in cell adherence is, however, 
still undefined. 

Cna, the collagen-bindmg factor of S. aureus is an important adhesin which 
mediates attachment to collagen substrates and collagenous tissues [86, 87]. 
In addition to this, Cna is abJe to mediate adherence to cartilage, a poten- 
tially important mechanism during septic arthritis [88, 89] and/or osteomyelitis 
[90]. The ligand-binding domain of Cna was identified to be located on a 
1 68-amino-acid-long segment within the amino-terminal A domain of the protein 



Talay 



102 



[91]. A synthetic peptide mimicking a subdomain of this segment inhibited 
collagen binding to the bacteria and identified the critical residues for collagen 
binding [92]. Structural resolution of the binding domain revealed a trench- 
shaped organization of the binding module that was predicted to accommodate 
the collagen triple helix [93]. interestingly, collagen binding to S. aureus cells 
is inhibited by capsule expression, suggesting a masking role for the surface 
polysaccharide [94]. This is in contrast to the collagen-binding characteristics 
observed in S. pyogenes where HA capsule expression does not inhibit but 
enhances collagen binding of 6". pyogenes by directly binding to collagen [35]. 

S. aureus expresses two adhesins that mediate binding to fibrinogen, ClfA 
and ClfB. ClfA enables S. aureus to adhere to fibrinogen-containing substrates 
such as plasma clots and to clump in the presence of fibrinogen, giving this 
protein its name: clumping factor [95]. ClfA is a potentially important viru- 
lence factor since ClfA negative mutant staphylococci showed reduced viru- 
lence in a rat endocarditis model [96]. The ligand binding domain of ClfA was 
mapped to a 329-amino acid segment within the amino-terminal A domain [97]. 
ClfA recognizes the carboxy-terminus of the y chain of fibrinogen, a region 
also recognized by the oiu]^^^ integrin on platelets, and thus inhibits platelet 
aggregation [98]. Analogous to the integrin/fibrinogen interaction, ClfA-mediated 
fibrinogen binding is affected by Ca^"^ [99], The structural basis for this inter- 
action was found by analyzing the crystal structure of the fibrinogen-binding 
domain. A variant of the immunoglobulin (IgG) fold, a structure found in IgG, 
was defined to mediate adhesion, placing ClfA into the IgG fold group of 
adhesins [100]. 

ClfB, the second fibrinogen-binding clumping factor and adhesin of 
S. aureus, has an overall organization similar to ClfA [101]. However, in contrast 
to ClfA, ClfB binds to the a and p chains of fibrinogen. Another characteristic 
of ClfB is its abiUty to bind cytokeratin 10 via the amino-terminal A domain [102]. 
It was shown to promote adherence to human keratinocytes and desquamated 
nasal epithelial cells, suggesting that this adhesin plays an important role in 
nasal colonization [102]. 

SasG, a recently identified surface protein of S. aureus [103], and Pis, a 
surface protein of methicillin-resistant 5". aureus [104], also promote adherence 
to desquamated nasal epithelial cells [105]; their receptor on the cellular surface 
is, however, still unknown. 

Bone sialoprotein (BSP) is bound by Bbp, a surface protein of S. aureus 
[106]. BSP is present in high concentrations in newly formed bone tissue, the 
osteoid, and thus suspected to be of relevance in osteomyelitis, an infection 
mostly affecting the osteoid. Bbp, like ClfA and ClfB, belongs to the Sdr family 
of surface proteins, characterized by the presence of carboxy-terminal serine- 
apartic acid dipeptide repeats [107]. 



Gram-Positive Adhesins 



03 



S. aureus has the ability to adhere to von Willebrand factor (vWF), a 
multimeric glycoprotein present at damaged endothelial sites. Two proteins 
have been identified that mediate binding ofS. aureus to human vWF: staphylo- 
coccal protein A (Spa) and vWbp [108, 109]. Binding to soluble or immobilized 
vWF may not onJy be responsible for 5'. aureus endovascular adherence but also 
increase the risk of disturbed hemostasis and vascular thrombosis, both symptoms 
observed during severe ^S". aureus infection. 

A surface-associated protein with broad matrix protein binding specificity 
was identified in 1993 [110], and subsequently characterized as Map or Eap 
protein [111, II 2]. Map/Eap was shown to bind fibrinogen, fibronectin, thrombo- 
spondin, vitronectin, bone sialoprotein, osteopontin and collagen, and occurs as 
a secreted but also surface-associated protein [110-112]. Map/Eap was demon- 
strated to mediate adherence to cultured epithelial and endothelial cells [113, 
1 14], and appears to enhance staphylococcal internalization into eukaryotic cells 
[115]. Furthermore, due to its binding ability towards ICAM-1 and the result- 
ing impairment of leukocyte recruitment, Map/Eap plays a role as anti-inflam- 
matory immune modulator [116]. 

Emp, another surface-associated protein of 5. aureus, binds to fibronectin, 
fibrinogen, vitronectin and collagen [1 17]. Like Map/Eap, Emp lacks the carboxy- 
terminally located LPXTG membrane anchor motif present in several gram- 
positive adhesins, but is found on the surface of ^. aureus cells where it may 
display adhesive function. 

The ECM component elastin is a target for EbpS, an elastin-binding protein 
[118]. The elastin-binding domain was localized within the amino-terminal por- 
tion of the transmembrane molecule, encompassing 2 1 amino acid residues shown 
to be exposed on the surface of intact S. aureus cells [119, 1 20]. As for Ebh, vWbp, 
and Emp, its role in mediating cell adherence remains to be investigated. 

In addition to the various protein adhesins, S. aureus expresses poly- 
saccharides that display an adhesive function: PIA, the polysaccharide inter- 
cellular adhesin, is required for biofihn formation and cell-to-cell adhesion 
[121]. Capsular polysaccharide of serotype 5 or 8, most frequently found to be 
expressed by S. aureus isolated from human infections, bmds to monocytes as 
well as to epithelial and endothelial cells, demonstrating adhesive properties for 
the S. aureus capsule [122]. 



Other Gram-Positive Adhesins 

Listeria monocytogenes is a gram-positive food-borne human pathogen 
that causes listeriosis, a severe invasive infection during which bacteria are 
disseminated to the fetoplacental unit and the central nervous system. Although 



Talay 



104 



the overall number of cases of listeriosis is low, the severity of infection is high 
and the factors responsible for host cell interaction and spreading are well 
studied. L. monocytogenes expresses two important invasins, internalin A and 
B (InlA, InlB), that also mediate adhesion to host cells. The cellular receptor for 
InJA was shown to be human E-cadherin [123], a cell surface adhesion 
molecule contributing to cell cohesion via homophilic dimerization and formation 
of adherens junctions. Interestingly, the species specificity of listeriosis arises 
from a single amino acid variation in E-cadherins of distinct species: the presence 
of a proline residue at position 16 in human E-cadherin was demonstrated to be 
crucial for cell interaction, explaining the finding that mouse and rat E-cadherin 
harboring a glutamic acid residue at that position was not susceptible for 
listeriosis [124, 125]. Different mammalian cell lines have varying susceptibilities 
to InJA and InlB. The human intestinal epithelial cell line Caco-2 and the 
hepatocyte HepG2 cells are targets for InlA and InlB. Interaction with 
monkey kidney Vero cells, mouse hepatocytes, and human endothelial cells is 
mediated via InlB [125]. Three receptor molecules have been identified for 
InlB [reviewed in 126]. InlB binds to HGF-R or Met, a receptor tyrosine 
kinase that acts as a receptor for hepatocyte growth factor [127], to gClq-R 
or p32, a receptor of the complement component Clq [128], and to 
proteoglycans [129]. 

Recent work has demonstrated that autolysLns of gram-positive pathogens 
may also display adhesive properties. The first autolysin shown to act as an 
adhesin was AtlE o^ Staphylococcus epidermidis, a commensal of the skin and 
an opportunistic pathogen [1 30]. AtlE was suggested to play a role in the attach- 
ment to polystyrene surfaces and to vitronectin, thereby contributing to biofilm 
formation of S. epidermidis on implanted polymers. Aas, an orthologous 
autolysin of Staphylococcus saprophyticus, mediates adhesion and binds to 
fibronectin [131]. The third autolysin found to mediate bacterial attachment 
was Ami of L monocytogenes [132]. Adhesive properties were localized within 
the noncatalytic carboxy-terminal cell wall-anchoring domain, composed of 
so-called GW modules, short dipeptide repeats containing the amino acid 
residues glycine and tryptophane [133]. Linkage of GW modules to LTA, as 
well as to glycosaminoglycans, anchor GW module-containing proteins to the 
surface of gram-positive bacteria [134], GW modules are found within all 
adhesive autolysins described herein, but also in eight other listerial proteins 
including InlB [134]. Thus, to define the adhesive properties of the yet unchar- 
acterized GW module-containing proteins will be a future goal. Interestingly, 
Cwp66 of Clostridium difficile, the first identified adhesin of this gram-positive 
spore-forming pathogen belonging to the genus Clostridia, exhibits homology to 
the catalytic domain of CwlB, the autolysin of Bacillus subtilis [135]. In contrast 
to the above-described adhesive autolysins, Cwp66 lacks repetitive GW modules 



Gram-Positive Adhesins 105 



but may be linked to the gram-positive cell wall via an alternative mechanism, 
explaining its surface localization. 

It is important to mention that a variety of adhesins, colonization and 
cross-linking factors have been identified and characterized in commensal 
gram-positive bacteria such as oral streptococci, enterococi, and staphylococci. 
Since these adhesins were not the subject of this chapter, the reader should be 
referred to these reviews [136-140] summarizing the adhesive mechanisms of 
commensal organisms that may also play an important role as opportunistic 
human pathogens in the susceptible host. 



Concluding Remarks 

Among bacterial virulence factors adhesins represent an important group. 
Many gram-positive pathogens express adhesins with a broad specificity, as 
well as adhesins that recognize particular target molecules such as collagen or 
fibronectin. These proteins have evolved in distinct gram-positive and gram- 
negative pathogens via convergent mechanisms. Adhesins very often function 
synergistically and are highly specific factors that are a prerequisite for infec- 
tion which subsequently governs the interplay between the microbe and the 
host. In particular cases, they may even have a direct impact on the phenotype 
of a disease such as septic arthritis in case of the S. aureus collagen-binding 
adhesin Cna [88, 89] or in autoimmune reaction based on the M3 protein of 
S. pyogenes [35]. 

Further characterization of the concerted fianction of multiple adhesins is 
a hallmark in understanding the initiation and progress of infection caused by a 
particular pathogen. Defining the target molecules in the adhesion process will 
help to understand individual host susceptibilities, and will link recent data on 
molecular interactions with epidemiological data collected over a whole century. 
The growing knowledge in the field of molecular mechanisms of pathogen 
adhesion will open up new perspectives in prevention and treatment strategies. 
Rational drug design based on the availability of structural data on receptor/1 igand 
complexes, fine-tuned vaccination approaches based on minimal functional 
domains, and identification of new vaccine candidates are the challenging 
perspectives of future research in this field. 



References 

1 Cremer MA, Rosloniec EF, Kang AH: The cartilage collagens: A review of their structure, 
organization, and role in the pathogenesis of experimental arthritis in animals and in human 
rheumatic disease. J Mol Med 1998;76;275-288. 

2 Pankov R, Yamada KM: Fibronectin at a glance. J Cell Sci 2002;1 15:3861-3863. 



Talay 



106 



3 Colognato H, Yurchenco PD: Form and function: The laniinin family of heterotrimers, Dev Dyn 
2000;218:213-234. 

4 Debelle L, Tamburro AM: Elasrin: Molecular description and function. Int J Biochem Cell Bio) 
1999;31:261-272. 

5 Schvartz I, Seger D, Shaltiel S: Vitronectin, Int J Biochem Cell Biol 1 999;3 1:539-544, 

6 Dempfle CE, Mosesson NfW : Theme issue: Fibrinogen and fibrin - Structure, function, interactions 
and clinical applications. Thromb Haemost 2003;89:599-600- 

7 Capila 1, Linhardt RJ: Heparin-protein interactions. Angew Chem Int Ed Engl 2002;4 1:391^ 12. 

8 Trowbridge JM, Gallo RL: Dermatan sulfate: New functions from an old glycosaminoglycan. 
Glycobiology 2002; 1 2: 1 1 7R-1 25R. 

9 Talay SR, Valentin-Weigand P, Jerlstrom PG, Timmls KN, Chhatwal GS: Fibronectin-binding 
protein o^ Streptococcus pyogenes: Sequence of the binding domain involved in adherence of 
streptococci to epithelial cells. Infect Immun l992;60;3837-3844, 

10 Hanski E, Caparon M: Protein F, a fibronectin-binding protein, isanadhesinof thegroupA strepto- 
coccus. Proc Natl Acad Sci USA 1992;89:6172-6176. 

I I Talay SR^ Valentin-Weigand P,Timmis iCN, Chhatwal GS: Domain snucture and conserved epitopes 
of Sfb protein, the fibronectin-binding adhesin o^ Streptococcus pyogenes . Mol Microbiol 1994; 
13:531-539, 

12 Sela S, Aviv A, Tovi A, Burslein 1, Caparon MG, Hanski E: Protein F: An adhesin of 
Streptococcus pyogenes binds fibronectin via two distinct domains. Mol Microbiol I993;10: 
1049-1055. 

13 Ozeri V, Tovi A, Bujstein 1, Natanson-Yaron S, CapaJon MG, Yamada KM, Akiyama SK, 
Vlodavsky 1, Hanski E: A two-domain mechanism for group A streptococcal adherence through 
protein F to the extracellular matrix. EMBO J 1996;15:989-998. 

14 Talay SR, Zock A, Rohde M, Molinari G, Oggioni M, Pozzi G, Guzman CA, Chhatwal GS: 
Co-operative binding of human fibronectin to Sfbl protein triggers streptococcal invasion into 
respiratory epithelial cells. Cell Microbiol 2000;2:521-535. 

15 Molinari G, Talay SR-, Valentin-Weigand P, Rohde M, Chhatwal GS: The fibronectin-binding protein 
0^ Streptococcus pyogenes, Sfbl, is involved in the internalization of group A streptococci by 
epithelial cells. Infect Immun 1997;65:1357-1363. 

16 Jadoun J, Ozeri V, Burstein E, Skutelsky E, Hanski E, Sela S: Protein Fl is required for efficient 
entry o^ Streptococcus pyogenes into epithelial cells. J hifect Dis 1998;178:147-158. 

17 Ozeri V, Rosenshine 1, Mosher DF, Fassler R, Hanski E: Roles of integrins and fibronectin in the 
entry of Streptococcus pyogenes into cells via protein Fl. Mol Microbiol 1998;30:625-637. 

1 8 Rohde M, Mijller E, Chhatwal GS, Talay SR: Host cell caveolae act as an entiy-port for group A 
streptococci. Cell Microbiol 2003;5:323-342. 

19 Dinkia K, Rohde M, Jansen WTM, Carapetis JR, Chhatwal GS, Talay SR: Streptococcus pyogenes 
recruits collagen via surface-bound fibronectin: A novel colonization and immune evasion mech- 
anism. Mol Microbiol 2003;47:861-809. 

20 Guzman CA, Talay SR, Molinari G, Medina E, Chhatwal GS: Protective immune response against 
Streptococcus pyogenes in mice after inn^anasal vaccination with the fibronectin-binding protein 
Sfbl. J Infect Dis 1999;179:901-906. 

21 Schwarz-Linek U, Werner JM, Pickford AR, Gurusiddappa S, Kim JH, Pilka ES, Briggs JA, 
Gough TS, Hook M, Campbell ID, Potts JR: Pathogenic bacteria attach to human fibronectin 
through a tandem beta-zipper. Nature 2003;423:177-181. 

22 Jaffe J, Natanson-Yaron S, Caparon MG, Hanski E: Protein F2, a novel fibronectin-binding 
protein from Streptococcus pyogenes, possesses two binding domains. Mol Microbiol 1996;2I: 
373-384. 

23 Rocha CL, Fischetti VA: Identification and characterization of a novel fibronectin-binding protein 
on the surface of group A streptococci. Lifect immun 1999;67:2720-2728. 

24 Courmey HS, Dale JB, Hasty Dt: Differential effects of the streptococcal fibronectin-binding 
protein, FBP54, on adhesion of group A streptococci to human buccal cells and HEp-2 tissue culture 
cells. Infect Immun 1996;64:2415-2419. 

25 Chhatwal GS: Anchorless adhesins and invasins of gram-positive bacteria: A new class of virulence 
factors. Trends Microbiol 2002;10:205-208. 



Gram-Positive Adhesins 107 



26 Terao Y, Kawabata S, Kunitomo E, Murakami J, Nakagawa I, Hamada S: Fba, a novel fibronectin- 
bindmg protein from Streptococcus pyogenes, promotes bacterial entry into epithelial cells, and 
iht Jba gene is positively transcribed under the Mga regulator. Mo) Microbiol 2001;42:75-86. 

27 Terao Y, Kawabata S, Nakata M, Nakagav^a I, Hamada S: Molecular characterization of a novel 
fibronectin-binding protein of Sfreplococcus pyogenes strains isolated from toxic shock-like 
syndrome patients. J Biol Chem 2002;277:47428^7435. 

28 Frick IM, Crossin KL, Edelman GM, Bjorck L: Protein H - A bacterial surface protein with 
aft'inity for both immunoglobulin and fibronectin type Ul domains. EM BO J 1995;14:1674-1679. 

29 Frick IM, Morgelin M, Bj6tx;k L: Virulent aggregates o^ Streptococcus pyogenes are generated by 
homophilic protein-protein interactions. Mol Microbiol 2000;37:1232-1247. 

30 Cue D, Dombek PE, Lam H, Cleary PP: Streptococcus pyogenes serotype Ml encodes multiple 
pathways for entry into human epithelial cells. Infect Immun 1998;66:4593-4601. 

31 Dombek PE, Cue D, Sedgewick J, Lam H, Ruschkowski S, Finlay BB, Cleary PP: High-frequency 
intracellular invasion of epithelial cells by serotype MJ group A streptococci: Ml protein-mediated 
invasion and cytoskeletal rearrangements. Mol Microbiol 1999;31:859-870, 

32 Cue D, Southern SO, Southern PJ, Prabhakar J, Lorelli W, Smallheer JM, Mousa SA, CJeary PP: 
A nonpeptide integrin antagonist can inhibit epithelial cell ingestion oi^ Streptococcvs pyogenes by 
blocking formation of integrin alpha 5beta 1-fibronectin-Ml protein complexes, Proc Natl Acad 
Sci USA 2000;97:2858-2863. 

33 Courtney HS, von Kunolstein C, Dale JB, Bronze MS, Beachey EH, Hasty DL: Lipoteichoic acid 
and M protein: Dual adhesins of group A streptococci. Microb Pathog 1992; 12: 199-208. 

34 Dunne DW, Resnick D, Greenberg J, Kj'ieger M, Joiner KA: The type ] macrophage scavenger 
receptor binds to gram-positive bacteria and recognizes lipoteichoic acid. Proc Natl Acad Sci USA 
1994;91:1863-1867. 

35 Dinkla K, Rohde M, Jansen WT, Kaplan EL, Chhatwal GS, Talay SR: Rheumatic fever-associated 
Streptococcus pyogenes isolates aggregate collagen. J Clin Invest 2003;! 1 1:1905-1912. 

36 Schmidt KH, Mann K, Cooney J, Kohler W: Multiple binding of type 3 streptococcal M protein 
to human fibrinogen^ albumin and fibronectin. FEMS Immunol Med Microbiol 1993;7: 
135-143. 

37 Podbielski A, Woischnik M, Leonard BA, Schmidt KJK: Characterization of nra, a global negative 
regulator gene in group A streptococci, Mol Microbiol 1999;31:1051-1064, 

38 Schrager HM, AJberti S, Cywes C, Dougherty GJ, Wessels MR: Hyaluronic acid capsule modu- 
lates M protein-mediated adherence and acts as a ligand for attachment of group A Streptococcus 
toCD44on human keratinocytes. J Clin Invest 1998;101:1708-1716. 

39 Okada N, Liszewski MK, Atkinson JP, Caparon M: Membrane cofactor protein (CD46) is a keratino- 
cyte receptor for the M protein of the group A streptococcus. Proc Natl Acad Sci USA 1995;92: 
2489-2493. 

40 Giannakis E, JokirantaTS, Ormsby RJ, Duthy TG, Male DA, Christiansen D, Fischetti VA, Bagley C, 
Loveland BE, Gordon DL: Identification of the streptococcal M protein binding site on membrane 
cofactor protein (CD46). J Immunol 2002; 168:4585^592. 

41 Berkower C, Ravins M, Moses AE, Hanski E: Expression of different group A streptococcal M 
proteins in an isogenic backgroujid demonstrates diversity in adherence to and invasion of eukaryotic 
cells. Mol Microbiol 1999;31:1463-1475. 

42 Frick IM, Schmidtchen A, Sjobring U: hiteractions between M proteins of Streptococcus 
pyogenes and glycosaminoglycans promote bacterial adhesion to host cells. Eur J Biochem 
2003;270:2303-231 L 

43 Terao Y, Kawabata S, Kunitomo E, Nakagawa 1, Hamada S: Novel laminin-binding protein of 
Streptococcus pyogenes, Lbp, is involved in adhesion to epithelial cells. Infect Immun 2002;70: 
993-997, 

44 Hytonen J, Haataja S, Geriach D, Podbielski A, Finne J: The SpeB virulence factor of Streptococcus 
pyogenes, a multifunctional secreted and cell surface molecule with strepadhesin, laminin-binding 
and cysteine protease activity. Mol Microbiol 2001;39:512-519. 

45 Stalhammar-Carlemalm M, Areschoug T, Larsson C, Lindahl G: The R28 protein oS^ Streptococcus 
pyogenes is related to several group B streptococcal surface proteins, confers protective immunity 
and promotes binding to human epithelial cells. Mol Microbiol 1999;33:208-219. 



Talay 



108 



46 Rasmussen M, Eden A, Bjorck L: SciA, a novel collagen-like surface protein oi Streptococcus 
pyogenes. Infecllmmun 2000;68:6370-6377. 

47 Lukomski S, Nakashima K, Abdi I, Cipriano VJ, Ireland RM, Reid SD, Adams GG, Musser JM: 
Identification and characterization of the scl gene encoding a group A Streptococcus 
extraceliuJar protein virulence factor with similarity to human collagen. Infect Immun 2000;68: 
6542-6553. 

48 Lukomski S, Nakashima K, Abdi 1, Cipriano VJ, Shelvin BJ, Graviss EA, Musser JM: 
Identification and characterization of a second extracellular collagen-like protein made by 
group A Streptococcus: Control of production at the level of translation. Infect Immun 200l;69: 
1729-1738. 

49 Rasmussen M, Bjorck L: Unique regulation of SclB - A novel collagen-like surface protein of 
Streptococcvs pyogenes. Mol Microbiol 2001;40:1427-1438. 

50 Cheng Q, Stafslien D, Purushothamaii SS, Cleary P: The group B streptococcal C5a peptidase is 
both a specific protease and an invasin. Infect Immun 2002;70:2408-2413. 

51 Beckmann C, Waggoner JT), Harris TO, Tamura GS, Rubens CE: Identification of novel adhesins 
from group B streptococci by use of phage display reveals that C5a peptidase mediates fibronectin 
binding. Infect Immun 2002;70:2869-2876. 

52 Spellerberg B, Rozdzinski E, Martin S, Weber-Heynemaiin J, Schnilzler N, Liitticken R, 
Podbielski A: Lmb, a protein with sunilarities to the Lral adhesin family, mediates attachment of 
Strepiococcifs agalactiae to human laminin. Infect Immun 1999;67:871-878. 

53 Bolduc GR, Baron MJ, Gravekamp C, Lachenauer CS, Madoff LC: The alpha C protein mediates 
internalization of group B Streptococcus within human cervical epithelial cells. Cell Microbiol 
2002;4:751-758. 

54 Tamura GS, Nittayajarn A: Group B streptococci and other gram-positive cocci bind to cyto- 
keratin 8. Infect Immun 2000;68:2129-2134. 

55 Kostrzynska M, WadstromT: Binding of laminin, type TV collagen, and vitronectin by Streptococcus 
pneumoniae. Zentralbl Bakteriol 1992;277:80-83, 

56 Harmnerschmidt S, Talay SR, Brandtzaeg P, Chhatwal GS: SpsA, a novel pneumococcal surface 
protein with specific binding to secretory immunoglobulin A and secretory component. Mol 
Microbiol 1997;25:1113-1124. 

57 Rosenow C, Ryan P, Weiser JN, Johnson S, Fontan P, Ortqvist A, Masure HR: Contribution of 
novel choline-binding proteins to adherence, colonization and immunogenicity of Streptococcus 
pneumoniae. Mol Microbiol 1997;25:819-829. 

58 Brooks-Walter A, Briles DE, Hollingshead SK: The pspC gene of So'eptococcus pneumoniae 
encodes a polymorphic protein, PspC, which elicits cross-reactive antibodies to PspA and pro- 
vides immunity to pneumococcal bacteremia. Infect Immun 1999;67:6533-6542. 

59 Hammerschmidt S, Tillig MP, Wolff S, Vaerman JP, Chhatwal GS: Species-specific binding of 
human secretory component to SpsA protein of Streptococcus pneumoniae via a hexapeptide 
motif Mol Microbiol 2000,36:72(^736. 

60 Zhang JR, Mostov KE, Lamm ME, Nanno M, Shimida S, Ohwaki M, Tuomanen E: The polymeric 
immunoglobulin receptor translocates pneumococci across human nasopharyngeal epithelial cells. 
Cell 2000;102:827-837. 

61 Dave S, Brooks-Walter A, Pangbum MK, McDaniel LS: PspC, a pneumococcal surface protein, 
binds human factor H. Infect Immun 2001;69:3435-3437. 

62 Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, Tuomanen EI: Streptococcus pneumoniae 
anchor to activated human cells by the receptor for platelet-activating factor Nature I995;377: 
435^38. 

63 van der Flier M, Chhun N, Wizemann TM, Min J, McCarthy JB, Tuomanen El: Adherence of 
Streptococcus pneumoniae to immobilized fibronectin. Infect linmun 1995;63:43 17^322. 

64 Holmes AR, McNab R, Millsap KW, Rohde M, Hammerschmidt S, Mawdsley JL, Jenkinson HF: 
JhtpavA gene of Streptococcus pneumoniae encodes a fibronectin-binding protein that is essential 
for virulence. Mol Microbiol 2001;41:1395-1408. 

65 Flock Jl, Froman G, Jonsson K, Guss B, Signas C, Nilsson B, Raucci G, Hook M, Wadslrom T, 
Lindberg M; Cloning and expression of the gene for a fibronectin-binding protein from 
Staphylococcus aureus, EMBO J 1987;6:2351-2357. 



Gram-Positive Adhesins 109 



66 Signas C, Raucci G, Jonsson K, Lindgren PE, Anantharamaiah GM, Hook M, Lindberg M 
Nucleotide sequence of the gene for a fibronectin-binding protein from Staphylococcus aureus 
Use of this peptide sequence in the synthesis of biologically active peptides. Proc Natl Acad Sci 
USA 1989;86:699-703. 

67 Jonsson K, Signas C, Miiller HP, Lindberg M:Two different genes encode fibronectin binding pro- 
teins in Staphylococcus aureus. The complete nucleotide sequence and characterization of the second 
gene. Eur J Biochem J99I;202:J0^ 1-1048. 

68 Warm ER, Gurusiddappa S, Hook M: The fibronectin-binding M SCRAM M FnbpA of 
Staphylococcus aureus is a bifunctional protein that also binds to fibrinogen. J Biol Chem 2000; 
275:13863-13871. 

69 Greene C, McDevitt D, Francois P, Vaudaux PE, Lew DP, FosterTJ: Adhesion properties of mutants 
of Staphylococcus aureus defective in fibronectin-binding proteins and studies on the expression 
ofy^i genes, Mol Microbiol 1995;17:1143-1 152, 

70 Peacock SJ, Foster TJ, Cameron BJ, Berendt AR: Bacterial fibronectin-binding proteins and 
endothelial cell surface fibronectin mediate adherence of Staphylococcus aureus to resting human 
endothelial cells. Microbiology 1999;145:3477-3486. 

71 Miyamoto YJ, Wann ER, Fowler T, Duffield E, Hook M, Mclntyre BW: Fibronectin binding 
protein A of Staphylococcus aureus can mediate human T lymphocyte adhesion and coactivation. 
J Immunol 200 1 ; 1 66:5 1 29-5 1 38. 

72 Mongodin E, Bajolet O, Cutrona J, Bonnet N, Dupuit F, Puchelle E, de Bentzmann S: Fibronectin- 
binding proteins of Staphylococcus aureus are involved in adherence to human airway epithelium. 
Infect Immun 2002;70;620-630. 

73 Brouillette E, Talbot BG, Malouin F: The fibronectin-binding proteins of Staphylococcus aureus 
may promote mammary gland colonization in a lactating mouse model of mastitis. Infect Immun 
2003;71:2292-2295. 

74 Sinha B, Francois PP, Nusse O, Foti M, Hartford OM, Vaudaux P, FosterTJ, Lew DP, Herrmann M, 
Kiause KH: Fibronectin-binding protein acts as Staphylococcus aureus invasin via fibronectin 
bridging to integriji alpha5beta I . Cell Microbiol 1999;J : 1 Ol-l 1 7. 

75 Fowler T, Wann ER, Joh D, Johansson S, FosterTJ, Hook M: Cellular invasion by Staphylococcus 
aureus involves a fibronectin bridge between the bacterial fibronectin-binding MSCRAMMs and 
host cell betal integrins, Eur J Cell Biol 2000;79:672-679. 

76 Sinha B, Francois P, Que YA, Hussain M, Heilmann C, Moreillon P, Lew D, Kiause KH, Peters G, 
Herrmann M: Heterologously expressed Staphylococcus aureus fibronectin-binding proteins are 
sufficient for invasion of host cells. Infect Immun 2000;68:6871-6878. 

77 Massey RC, Kanlzanou MN, Fowler T, Day ^fP, Schofield K, Wann ER, Berendt AR, Hook M, 
Peacock SJ: Fibronectin-binding protein A of Staphylococcus aureus has multiple, substituting, 
binding regions that mediate adherence to fibronectin and invasion of endothelial cells. Cell Microbiol 
2000;3:839-851. 

78 Dziewanowska K, Palti JM, Deobald CF, Bayles KW, Trumble WR, Bohach GA: Fibronectin binding 
protein and host cell tyrosine kinase are required for internalization of Staphylococcus aureus by 
epithelial cells. Infect Immun 1999;67:4673^678. 

79 McGavin MJ, Gurusiddappa S, Lindgren PE, Lindberg M, Raucci G, Hook M: Fibronectin receptors 
from Streptococcus dysgalactiae and Staphylococcus aureus. Involvement of conserved residues 
in ligand binding, J Biol Chem 1993;268:23946-23953. 

80 Huff S, Matusuka YV, McGavin MJ, Ingham KC: Interaction of N-terminal fragments of 
fibronectin with synthetic and recombinant D motifs from the binding protein of Staphylococcus 
aureus studied using fluorescence anisotropy. J Bio] Chem 1994;269:15563-15570. 

81 Joh D, Speziale P, Gurusiddappa S, Manor J, Hook M: Multiple specificities of the 
staphylococcal and streptococcal fibronectin-binding MSCRAMMs. Eur J Biochem 1998;258: 
897-905. 

82 Penkett CJ, Dobson CM, Smith LJ, Bright J R, Pickford AR, Campbell ID, Potts JR: Identification 
of residues involved in the interaction of Staphylococcus aureus fibronectin-binding protein with 
the (4)F1(5)F1 module pair of human fibronectin using heteronuclear NMR specti"oscopy. 
Biochemistry 2000;39:2887-2893. 



Talay 



no 



83 House-Pompeo K, Xu J, Joh D, Speziale P, Hook M: Conformational changes in the fibronectin- 
binding MSCRAMMs are induced by ligand binding, J Biol Chem 1996;271:1379-1384, 

84 Penkett CJ, Redfield C, Jones JA, Dodd 1, Hubbard J, Smith RAG, Smit LJ, Dobson CM: 
Structural and dynamical characterization of a biologically active unfolded fibroneclin-binding 
protein from Siaphylococcus aureus. Biochemistry 1 998;37; 17054- 17067, 

85 Clarke SR, Harris LG, Richards RG, Foster SJ: Analysis of Ebh, a 1.1-megadalton cell wall- 
associated fibronectin-binding protein of Staphylococcus aureus. Infect Immun 2002;70: 
6680-6687. 

86 Switalski LM, Speziale P, Hook M: Isolation and characterization of a putative collagen receptor 
from Staphylococcus aureus strain Cowan I. J Biol Chem 1989;264:21080-21086. 

87 Palti JM, Jonsson H, Guss B, Switalski LM, Wiberg K, Lindberg M, Hook M: Molecular 
characterization and expression of a gene encoding a Siaphylococcus aureus collagen adhesin. 
J Biol Chem l992;267:4766-^772, 

88 Switalski LM, Patti JM, Butcher W, Gristina AG, Speziale P, H55k M: A collagen receptor on 
Staphylococcus aureus strains isolated from patients with septic arthritis mediates adhesion to 
cartilage. MoJ Microbiol 1993;7:99-107. 

89 Patli IM, Bremell T, Krajewska-Pielrasik D, Abdelnour A, Tarkowski A, Ryden C, Hook M: The 
Staphylococcus aureus collagen adhesin is a virulence determinant in experimental septic arthritis. 
Infect Immun 1994;62:152-161. 

90 Elasri MO, Thomas JR, Skinner RA, Blevins JS, Beenken KE, Nelson CL, Smelter MS: 
Staphylococcus aureus collagen adhesin contributes to the pathogenesis of osteomyelitis. Bone 
2002;30:275-280. 

91 Patti JM, Boles JO, Hook M: Identification and biochemical characterization of the ligand 
binding domain of the collagen adhesin from Siaphylococcus aureus. Biochemistry 1993;32: 
I 1428-1 1435. 

92 Patti JM, House-Pompeo K, Boles JO, Gai"za N, Gurusiddappa S, Hook M: Critical residues in the 
ligand-binding site of the Siaphylococcus aureus collagen-binding adhesin (MSCRAMM). J Biol 
Chem 1995;270:12005-12011. 

93 Symersky J, Patti JM, Carson M, House-Pompeo K, Teale M, Moore D, Jin L, Schneider A, 
DeLucas LJ, Hook M, Narayana SV: Structure of the collagen-binding domain from diSlapliylococcus 
aureus adhesin, Nat Struct Biol 1997;4:833-838. 

94 Gillaspy AF, Lee CY, Sau S, Cheung AL, Smeltzer MS: Factors affecting the collagen binding 
capacity of Staphylococcus aureus. Infect Immunol 1998;66:3170^3I78, 

95 McDevitt D, Francois P, Vaudaux P, Foster TJ: Molecular characterization of the clumping factor 
(fibrinogen receptor) of Staphylococcus aureus. Mol Microbiol 1994; 1 1:237-248. 

96 Moreillon P, Entenza JM, Francioli P, McDevitt D, Foster TJ, Francois P, Vaudaux P: Role of 
Staphylococcus aureus coagulase and clumping factor in pathogenesis of experimental endocarditis. 
Infect Immun 1995;63:4738^743. 

97 McDevitt D, Francois P, Vaudaux P, Foster TJ: Identification of the ligand-binding domain of the 
surface-located fibrinogen receptor (clumping factor) of Staphylococcus aureus. Mol Microbiol 
1995;16:895-907. 

98 McDevitt D, Nanavaty T, House-Pompeo K, Bell E, Turner N, Mclntire L, Foster T, Hook M: 
Characterization of the interaction between the Staphylococcus aureus clumping factor (ClfA) and 
fibrinogen. Eur J Biochem 1997;247:416-424. 

99 O'Connell DP, Nanavaty T, McDevitt D, Gurusiddappa S, Hook M, Foster TJ: The fibrinogen- 
binding MSCRAMM (clumping factor) of Staphylococcus aureus has a Ca^"^-dependent 
inhibitory site. J Biol Chem 1998;273:6821-6829. 

100 Deivanayagam CC, Wann ER, Chen W, Carson M, Rajashankar KR, Hook M, Narayana SV: A 
novel variant of the immunoglobulin fold in surface adhesins of Staphylococcus aureus: 
Crystal structure of the fibrinogen-binding MSCRAJVIM, clumping factor A. EMBO J 2002;2I: 
6660-6672. 

101 Ni Eidhin D, Perkins S, Francois P, Vaudaux P, Hook M, Foster TJ: Clumping factor B (ClfB), a new 
surface-located fibrinogen-binding adhesin of Siaphylococcus aureus. Mol Microbiol I998;30: 
245-257. 



Gram-Positive Adhesins 



II 



102 O'Brien LM, Walsh EJ, Massey RC, Peacock SJ, Foster TJ: Staphylococcus aureus clumping 
factor B (ClfB) promotes adherence to human type I cytoloeratin 10: Implications for nasal coloni- 
zation. Cell Microbiol 2002;4:759-770. 

103 Roche FM^ Massey R, Peacock SJ, Day NPJ, Visai L, Speziale P, Lam A, Fallen M, Foster TJ: 
Characterization of novel LPXTQ-containing proteins oC Staphylococcus aureus identified from 
genome sequences- Microbiology 2003;149:643-654. 

104 SavolainenK^ Paulin L, Westerlund-Wikstrom B, Foster TJ, KorhonenTK^ Kuusela P: Expression 
ofp/.s, agene closely associated with the mecA gene of methicillin-resistant Staphylococcus aureus, 
prevents bacterial adhesion in vitro. Infect Immun 2001;69:3013-3020. 

105 Roche FM, Meehan M, Foster TJ: The Staphylococcus aureus surface protein SasG and its homo- 
logues promote bacterial adherence to human desquamated nasal epithelial cells. Microbiology 
2003;149:2759-2767. 

106 Tung H, Guss B, Hellmaii U Persson L, Rubin K, Ryden C: A bone sialoprotein-binding protein from 
Staphylococcvs aureus'. A member of the staphylococcal Sdr family, Biochem J 2000;345:6I 1-619, 

107 Josefsson E, McCrea KW, Ni Eidhin D, O'Connell D, Cox J, Hook M, FosterTJ: Three new members 
of the serine-aspartate repeat protein multigene family of Staphylococcus aureus. Microbiology 
1998;144:3387-3395. 

108 Hartleib J, Kohler N, Dickinson RB, Chhatwal GS, Sixma JJ, Hartford OM, Foster TJ, Peters G, 
Kehrel BE, Herrmann M: Protein A is the von Willebrand factor binding protein on Staphylococcus 
aureus. Blood 2000;96:2149-2156. 

109 Bjerketorp J, Nilsson M, Ljungh A, Flock Jl, Jacobsson K, Frykberg L: A novel von Willebrand 
factor binding protein expressed by Staphylococcus aureus. Microbiology 2002;148:2037-2044. 

1 10 McGavin MH, Krajewska-Pietrasik D, Ryden C, Hook M: Identification of a Staphylococats aureus 
extracellular matrix-binding protein with broad specificity. Infect Immun 1993;61:2479-2485, 

I 1 1 Jonsson K, McDevitt D, McGavin MH, Patti JM, Hook M: Staphylococcus aureus expresses a 

major histocompatibility complex class II analog. J Biol Chem 1995;270:21457-21460. 
1 12 Palma M, Haggar A, Flock JI: Adherence of Staphylococcus aureus is enhanced by an endogenous 

secreted protein with broad binding activity. J Bactexiol 1999;18l:2840— 2845. 
I 13 Hussain M, Haggar A, Heilmann C, Peters G, Flock JI, Herrmann M: Insertional inactivation of 

Eap in Staphylococcus aureus strain Newman confers reduced staphylococcal binding to fibroblasts. 

hifect Immun 2002;70:2933-2940, 

1 14 Kreikemeyer B, McDevitt D, Podbielski A: The role of the Map protein in Staphylococcus aureus 
matrix protein and eukaryotic cell adherence, Int J Med Microbiol 2002;292:283-295. 

1 15 Haggar A, Hussain M,JLonniesH, Herrmann M, Norrby-Teglund A, Flock Jl: Extracellular adher- 
ence protein from Staphylococcus aureus enhances internalization into eukaryotic cells. Infect 
Immun 2003;71:2310-2317. 

1 16 Chavakis T, Hussain M, Kanse SM, Peters G, Bretzel RG, Flock Jl, Herrmann M, Preissner KT: 
Staphylococcus aureus extracellular adherence protein serves as anti-inflammatory factor by 
inhibiting the recruitment of host leukocytes. Nat Med 2002;8:687-693. 

117 Hussain M, Becker K^ von Eiff C, Schienzel J, Peters G, Herrmann M: Identification and 
characterization of a novel 38.5-kilodalton cell surface protein of Staphylococcus aureus with 
extended-spectrum binding activity for extracellular matrix and plasma proteins J Bacteriol 2001; 
183:6778-6786, 

I 18 Park PW, Rosenbloom J, Abrams WR, Rosenbloom J, Mecham RP: Molecular cloning and expres- 
sion of the gene for elastin-binding protein (ebpS) in Staphylococcus aureus. J Biol Chem 1996; 
271:15803-15809, 

I 19 Park PW, Broekelmann TJ, Mecham BR, Mecham RP: Characterization of the elastin binding 
domain in the cell-surface 25-kDa elastin-binding protein of Staphylococcus aureus (EbpS). J Biol 
Chem 1999;274:2845-2850. 

120 Downer R, Roche F, Park PW, Mecham RP, Foster TJ: The elastin-binding protein of 
Staphylococcus aureus (EbpS) is expressed at the cell surface as an integral membrane protein and 
not as a cell wall-associated protein. J Biol Chem 2002;277:243-250 

121 Cramton SE, Gerke C, Schnell NF, Nichols WAV, Gotz F: The intercellular adhesion (ica) locus is 
present in Staphylococcus aureus and is required for biofilm formation. Infect Immun 1999; 
67:5427-5433. 



Talay 



112 



122 Soell M, Diab M, Haan-Archipoff G, Beretz A, Herbelin C, Poutrel B, KJein JP: Capsular poly- 
saccharide types 5 and 8 of Staphylococcus aweus bind specifically to human epithelial (KB) cells, 
endothelial cells, and monocytes and induce release of cytokines. Infect Immun 1995;63:1380-1386. 

123 Mengaud J, Ohayon H, Gounon P, Mege R-M, Cossart P: E-cadherin is the receptor for intemalin, a 
sujface protein required for entry of L, monocytogenes into epithelial cells. Cell 1996;84;923-932. 

124 Lecuit M, Dramsi S, Gottardi C, Fedor-Chaiken M^ Gumbiner B, Cossart P: A single amino acid 
in E-cadherin responsible for host specificity towards the human pathogen Listeria monocytogenes. 
EMBOJ 1999;18:3956-3963. 

125 Lecuit M, Vandormael-Pournin S, Lefort J, Huerre M, Gounon P, Dupuy C, Babinet C, Cossart P: 
A transgenic model for listeriosis: Role of intemalin in crossing the intestinal barrier Science 
2001;292:1722-1725. 

126 Bierne H, Cossart P: IniB, a surface protein of Listeria monocytogenes that behaves as an invasin 
and a growth factor. J Cell Sci 2002;! 15:3357-3367, 

127 Shen Y, Naujokas M, Park M^ Ireton K: InJB-dependent internalization of Listeria is mediated by 
the Met receptor tyrosine kinase. Cell 2000;103:501-510. 

128 Braun L, Ghebrehiwet B, Cossart P: gClq-R/p32, a C J q-binding protein, is a receptor for the JniB 
invasion protein of Listeria monocytogenes. EMBO J 2000; 19: 1458-1466. 

129 Jonquieres R, Pizarro-Cerda J, Cossart P: Synergy between (he N-and C-terminal domains of InIB 
for efficient invasion ofnon-phagocytic cells by Listeria monocytogenes. Mol Microbiol 2001;42: 
955-965. 

130 Heilmann C, Hussain M, Peters G, Gotz F: Evidence for autolysin-mediated primary altachmenl 
of Staphylococcus epidcrmidis to a polystyrene surface. Mol Microbiol 1997;24:1013-1024. 

1 3 1 Hell W, Meyer HG, Gatermann SG: Cloning of aas, a gene encoding a Staphylococcus saprophyticus 
surface protein with adhesive and autolytic properties. Mol Microbiol 1998;29:871-881. 

1 32 Milohanic E, Pron B, Berche P, Gaillard JL: Identification of new loci involved in adhesion of Listeria 
monocytogenes to eukaryotic cells. European Listeria Genome Consortium. Microbiology 2000; 
146:731-739. 

133 Milohanic E, Jonquieres R, Cossart P, Berche P, Gaillard JL: The autolysin Ami contributes to the 
adhesion of Listeria monocytogenes to eukaryotic cells via its cell wall anchor. Mol Microbiol 
2001;39:1212-1224. 

134 Cabanes D, Dehoux P, Dussurget 0, Frangeul L, Cossart P: Surface proteins aiid the pathogenic 
potential of Listeria monocytogenes. Trends Microbiol 2002;10:238-245. 

1 35 Waligora AJ, Hennequin C, Mullany P, Bourlioux P, Collignon A, Karjalainen T: Characterization 

of a cell surface protein of Clostridium difficile with adhesive properties. Infect Immun 2001 ;69: 
2144-2153. 

136 Banas JA, Vickerman MM: Glucan-binding proteins of the oral streptococci. Crit Rev Oral Biol 
Med 2003;14:89-99. 

1 37 Love RM, Jenkinson HP: Invasion of dentinal tubules by oral bacteria. Crit Rev Oral Biol Med 2002; 
13:171-183. 

138 Jenkinson HP, Lamont RJ: Streptococcal adhesion and colonization. Crit Rev Oral Biol Med 1997; 
8:175-200. 

139 0*Gara JP, Humphreys H: Staphylococcus epidcrmidis biofilms: hnportance and implications. 
J Med Microbiol 2001;50:582-587. 

140 Mack D: Molecular mechanisms of Staphylococcus epidcrmidis biofilm formation. J Hosp Infect 
1999;43:SI13-S125. 



Susaniie R. Talay 

Department of Microbial Pathogenesis and Vaccine Research 

GBF-German Research Centre for Biotechnology 

Mascheroder Weg U DE-38124 Braunschweig (Germany) 

Tel. +49 5346 912110, Fax +49 531 6181 708, E-Mail sta@gbf.de 



Gram-Positive Adhesins 1 1 3 



Adhesins 



Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kai'ger, 2005, vol 12, pp 1 14-13! 



Microbial Pathogenesis and 
Biofilm Development 



Andreas Reisner^, Niels H0iby^, Tim Tolker-Nielsen^, 
S0ren Molirv" 

^BioCentrum-DTU, Technical University of Denmark, Lyngby, and 
''DepartmenL of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark 



Microbial infections constitute a major cause of premature death in large 
parts of the world, and for several years we have seen an alarming tendency 
towards increasing problems of controlling such infections by antibiotic treat- 
ments. It is hoped that an improved understanding of the infectious cycles of 
different microorganisms will eventually lead to improved treatments. Several 
bacteria have evolved specific strategies for virulent colonization of humans in 
addition to their otherwise harmJess establishment as environmental inhabitants, 
Jn many such cases biofilm development seems to play a highly significant role 
in connection with chronic infections [1]. 

Bacterial growth on surfaces depends on several factors [2], In nature, surfaces 
are probably often conditioned with a thin film of organic molecules, which may 
serve as attractants for bacterial chemotactic systems and which subsequently 
permit bacterial growth to occur. In laboratory model systems the growth of the 
surface-associated bacteria is supported by the nutrient supply in the moving or 
standing liquid, A benchnriark of biofilm formation by several organisms in vitro 
is the development of three-dimensional structures that have been termed 'mat- 
uration', which is thought to be mediated by a differentiation process. Maturation 
into late stages of biofilm development resulting in stable and robust structures 
may require the formation of a matrix of extracellular polymeric substances (EPS), 
which are most often assumed to consist of polysaccharides. A recent striking 
finding is that DNA released fi'om biofilm cells may be important as an initial 
matrix former [3]. At later times other EPS molecules may add to the shape and 
quality of the mature biofilm structure. Figure 1 summarizes the principle steps 
involved in the development of microbial biofilms. 



1) Reversible attachment 2) Irreversible attachment 3) Cell proliferation 



"v. 



c:) 






■=^> 



<? 



^ 



^=> 



d> 



^^M 




.-^^^■.^qaF^^v.^^^^ 



4) Biofilm maturation 



5) Dissolution 




Fig, L The biofilm development cycle. Biofilm development is depicted as a geiieraJ 
scheme involving attachment to the surface, formation of a tight association between bacteriaT 
cells and a surface, growth and intercellular adhesion allowing microcolony formation, mat- 
uration including EPS matrix development, and local dissolution leading to release of bacteria, 
which may eventually restart the cycle. 



How do bacteria know that they are located in a biofilm? There is no doubt 
that cell density is an important factor that distinguishes the usually dilute 
suspensions of planktonic cells in water from the very cell-dense surface com- 
munities found where organic matter is abundant. One answer to the question 
therefore is: very high cell density. Another characteristic of biofilms and other 
types of surface-associated communities is the prevalence of Lnternally hetero- 
geneous environments and microenvironments, often generated and maintained 
by the presence of EPS, For the biofilm-associated bacteria this scenario is 
recognized as gradients of nutrients and stress factors. For planktonic cells such 
gradients rarely play a role. It is often argued that attachment to surfaces is the 
most important feature, and that surface-induced gene expression is therefore 
one of the key determmants of biofilm development. It should be remembered, 
however, that cellular contact with the substratum in a biofilm is a transient 
phenomenon (but niosL likely important for early gene activation), which is 
quickly converted to a state where essentially all bacterial cells are located far 
above the surface in microcolonies or in EPS-embedded 'mushrooms'. In these 
entities it is difficult to imagine any bacterial sensing of the surface association 
as a physical signal. 

Thus, it seems that biofilm-associated bacteria must respond to the (1) very 
high cell density and (2) to the various positive and negative gradients. If it is 
assumed that bacterial evolution is mainly connected to the dominant life form 
of these organisms, and that bacteria in natural environments almost exclusively 
live an active proliferating life associated with surfaces (in biofilms), it is to be 



Biofilms and Pathogenesis 



115 



expected that evolution has provided bacteria with properties that allow adaptation 
to life under high cell density conditions in environments with nutrient and 
antagonist gradients. 

This leaves the following issues as the major comumon themes for biofilm 
investigations related to the microbial capacity to develop mature, heterogeneously 
structured surface-associated communities: How are the specific structural fea- 
tures in a biofihn created and maintained? Which functions are involved in the 
adaptation to high cell densiries and nutrient gradients? How do biofilm bacteria 
evolve, and what are the major selective forces? 

In the following we will present an overview of the current understanding 
of microbial biofilm development and its clinical relevance in relation to 
two examples of gram-negative pathogens, Escherichia coli and Pseudomonas 
aeruginosa, for which the biofilm lifestyle seems to be relevant during the 
course of infection. 



E co/j 

As the dominant facultative anaerobe of the normal human intestinal flora, 
E, coli remains harmlessly confined to the intestinaJ lumen. However, highly 
adapted clones have evolved the ability to cause a broad spectrum of diseases 
ranging from urinary tract infection (UTI) and diarrhea to sepsis and meningitis 
[4]. Many of these infections are initiated by bacterial colonization of mucosal 
surfaces of the genitourinary, gastrointestinal or respiratory tracts. Successful 
establishment in the host depends on the ability to overcome host defenses and 
shear forces present at most of these surfaces. Since biofilm formation has also 
been suggested to be an ancient bacterial survival strategy [5], it seems possible 
that at least a fraction of pathogenic E, coli clones have conserved or evolved 
the ability to enter a sessile lifestyle in multicellular biofilm communities in the 
host environment. Through investigations in recent years we now begin to realize 
that bacterial cell-cell interactions among £'. coli cells on biotic and abiotic sur- 
faces play a more significant role in pathogenicity than previously anticipated. 
It has therefore been of significant interest to clarify the mechanism(s) by 
which this organism colonizes surfaces and develops into substantial and robust 
biofilms. 

In vitro Biofilm. Development 

Since E, coli K-12 has been the workhorse bacterium for molecular bio- 
logists for neariy 50 years, standard laboratory strains became model organisms 
used in an approach to assign a developmental program to E. coli biofilms 
formed in vitro. A simple genetic screen was implemented utilizing 96-well 



Reisner/H0iby/Tolker-Nielsen/Molin 1 16 



microliter dishes as abiotic substrates for biofilm development in vitro, allowing 
large-scale isolation of mutants attenuated in biofilm formation under static 
conditions. 

Underlined by microscopic observations, the results of these initial studies 
were integrated in a developmental model for E, coli biofilm formation [6], 
According to this model, E. coli K-12 utilizes flagel la-mediated motility and 
type 1 pili to initiate early attachment processes. The major phase-variable outer 
membrane protein Ag43 was implicated in further development of microcolonies, 
and in agreement with the classical role ascribed to exopolysaccharides in 
stabilization of mature biofilms, the production of colanic acid was found to be 
required for the development of normal biofilm architecture in vitro. 

In subsequent similar approaches, additional factors have been found to 
affect biofilm formation of £. coli on abiotic surfaces in conventional growth 
media; however, only the effects of a few of them have been studied in detail 
[7]. The intracellular localization of most of the proposed effector proteins such 
as the disulfide bond formation catalyzing DsbA or the acetate kinase AckA 
suggests an indirect influence, possibly by altering expression, assembly or 
function of already implicated surface appendages and outer membrane pro- 
teins. The importance of others such as the stress-response sigma factor RpoS 
or the stringent response proteins RelA and SpoT might simply indicate the 
requirement for metabolic pathways and stress responses within the hetero- 
geneous biofilms that are less important during exponential growth in suspension. 

Interestingly, the growth of E. coli K-12 biofilms in continuous hydro- 
dynamic culture leads to the identification of biofilm-promoting factors, 
reflecting the reduced biofilm-forming capability of K-12 lab strains under these 
conditions. An E. coli ompR234 mutant was isolated from the glass surface of 
a long-term continuous culture that was found to constitutively overexpress 
curii fimbriae [8]. The significantly improved biofilm formation phenotype was 
independent of flagella [9]. In 2001, Ghigo [10] discovered that conjugative 
plasmids enhance biofibn formation on submerged Pyrex slides under continu- 
ous flow when the expression of conjugative pili is derepressed. Mutant analysis 
demonstrated that at least for plasmid F, fujictionai conjugative pili are indeed 
necessary to obtain the observed induction. In a subsequent study, evidence was 
provided that the promotion of biofilm formation in the presence of the con- 
jugative transfer genes of plasmid F is mdependent of flagella, type I pili or 
Ag43 synthesis [11]. 

As the biofilm lifestyle is thought to be fundamentally different from 
bacterial life m mixed suspension, major differences in gene expression were 
expected to be encountered upon switching from planktonic to biofilm growth. 
This view was confirmed by an experimental approach that used random chromo- 
somal insertions of a promoterless lacZ reporter gene [12], A large fraction 



Biofilms and Pathogenesis 1 17 



(38%) of 885 fusions was differentially expressed in a curli-promoted static 
E. coli K-12 biofilm when compared to planktonic cells. However, a recent 
microarray analysis of a biofilm formed by a wild-type K-12 strain under 
continuous flow indicated a more modest impact on global gene expression 
[13], The transcript level of only 5.4 and 13.6% of the 4,290 protein-encoding 
genes was found to be significantly different as compared to expression in 
either exponential or stationary planktonic culture, respectively. It is unclear 
whether these drastically different results in terms of changes in global gene 
expression can be ascribed to the different strain background and/or the exper- 
imental setup. 

Due to the exclusive focus on K-12 strains in the vast majority of genetic 
studies, the relevance of the implicated factors for biofilm formation of non- 
domesticated E. coli isolates remains uncertain. For example, whereas the role of 
type I and curli fimbriae in the adherence of Shiga toxin-producing E. coli has 
been confirmed [14], a recent study suggests that the expression of colanic acid 
blocks adhesion of uropathogenic E. coli (UPEC) to inert abiotic surfaces [15]. 
Given the significantly elevated genome size of pathogenic E. coli as compared 
to K-12, determination of the diversity of molecular mechanisms used by the 
species E. coli in bacterial cell-cell interactions will necessitate the application 
of the already established molecular approaches at least to prototypic clinical 
E. coli isolates. 

Gastrointestinal Biofilms 

As a minority member of the normal flora of the large intestine in verte- 
brates, E. coli has to compete for nutrients with approximately 500 other 
indigenous species. In principle, successful coexistence can only be achieved 
by a growth rate that is at least equivalent to the washout rate from the intestine 
or by adherence to the intestinal epithelial cells [16]. Indeed, E. coli is capable 
of growing rapidly in intestinal mucus both in vivo and in vitro, whereas growth 
in luminal contents seems to be poor [17]. In addition, in situ hybridization 
experiments detected only separated single cells of commensal E. coli strains 
within the niucus layer but no bacterial cells associated with the epithelium [17, 
18]. Thus, benign E. coli cells do not seem to be able to overcome the innate 
barriers that impede colonization in a healthy host and the natural lifestyle of 
these strains appears to be to reside and grow within the mucus layer almost 
exclusively as single cells. 

In contrast, each highly adapted E. coli clone causing diarrheal disease 
has evolved efficient ways to penetrate the mucus layer and stably adhere to 
the underlying epithelial cells even at intestinal sites normally not colonized by 
E. coli, such as the small bowel mucosa [19]. As for other mucosal pathogens, 
surface colonization by diarrheagenic E. coli is a prerequisite to initiate disease. 



Reisner/Hsiby/Tolker-Nielsen/Molin 1 18 



Not surprisingly therefore, the most useful phenotypic assay for the diagnosis 
and differentiation of diarrheagenic E. coli pathotypes is an adherence assay 
using monolayers of epithelial HEp-2 cells. Strikingly, the adherence pattern of 
members of two major pathotypes of diarrheagenic E. coli, enteropathogenic 
(EPEC) and enteroaggregative (EAEC) E. coli involves - in addition to binding 
to eukaryotic cells - apparent strong interactions between bacterial cells leading 
to three-dimensional structures typically obsei"ved in bacterial biofilms. EPEC 
develop a characteristic localized adherence pattern appearing as microcolonies 
on the surface, whereas EAEC appear to aggregate both on the surface as well 
as more distantly from the epithelium in a characteristic stacked-brick config- 
uration [19]. Most importantly, similar biofilm-like adherence patterns have 
also been observed for both EPEC and EAEC in vivo. 

While the adherence to epithelial cells has been extensively studied, little 
information is currently available about the factors that trigger bacterial cell-cell 
adherence or the relevance of the size of these cell aggregates for pathogenicity 
[20]. Although the plasmid-encoded bundle-forming pili (BFP) of EPEC have 
been suggested to mediate interbacterial interactions allowing formation of 
three-dimensional microcolonies on the surface of epithelia [21 ], BFP-expressing 
EPECs were found to bind to epithelial cells rather than to already formed 
microcolonies. Interestingly, BFP are subject to morphological changes from 
thin to thick pili as infection proceeds, resulting in loosening and dispersal of 
the aggregates [20]. A bfpF mutant that was found unable to undergo this morpho- 
logical change was significantly attenuated in virulence, indicating that formation 
and dispersal of microcolonies are both important for virulence. 

Likewise, plasmid-encoded thin aggregative adherence fimbriae were found 
to mediate the adherence and aggregation pattern of EAEC strains in vivo and 
in vitro [19]. Interestingly, the aggregative adherence pattern also requires expres- 
sion of a secreted coat protein designated Aap (antiaggregation protein), which 
appears to promote dispersal of EAEC on the intestinal mucosa by forming a protein 
capsule on the bacterial surface. Mutations in aap lead to increased aggregation 
and significantly reduced mucus penetration in vitro, indicating that bacterial 
cell-ceil adherence has to be tightly controlled in order to be advantageous iji tlie 
intestinal environment [22]. 

Nevertheless, a large fraction of EPECs and EAECs lack BFP and aggrega- 
tive adherence fimbriae, respectively [19, 23]. Thus, E. coli clones seem to have 
evolved various divergent pathways to solve the same problem. 

Inti'acellular Biofilm-Like Pods in UTI 

The human urinary tract is usually a sterile system protected from the 
intestinal microflora by nonspecific resistance mechanisms that include phago- 
cytosis, endotoxin-induced shedding of bladder epithelial cells, and the flushing 



Biofilms and Pathogenesis 1 19 



effect of urine flow. However, UTIs are considered to be the most common 
bacterial infections [24], with UPEC remaining the predominantly isolated 
species [25]. Generally, UPECs are thought to migrate from the gastrointestinal 
tract to the periurethral area where they eventually enter the bladder via the 
urethra [26]. Further transport into the kidneys may even enable an invasion 
into the bloodstream. 

Since intestuial E. coli clones are not equally able to survive within and col- 
onize the urinary tract, UPECs are thought to be equipped with a variety of vir- 
ulence factors including various adheshis of fimbrial nature such as curli, type 1 
pili, P, S, and FlC fimbriae [27]. These surface appendages bind to specific 
host cell receptor molecules and facilitate attachment of bacteria to specific 
epithelial cells they encounter during their transit [28]. However, despite the 
clear importance of cell-surface interactions dui'ing the course of infection^ bac- 
terial cell aggregates typical for biofilm formation have not been demonstrated 
on epithelial cells in vivo. 

Recent evidence suggests a novel role for biofilm-like cell-cell interactions 
during recurrent UTI. After artificial UTI infection of mice, Anderson et al. 
[29] observed large pod-like bacterial cell aggregates within superficial cells of 
dissected bladders whereas uninfected bladders appeared smooth. Bacteria 
within the pods had a uniform coccoid morphology, were interconnected by 
fibers and encased in a polysaccharide matrix. Although the presence of per- 
sistent E. coli in the bladder following acute UTI has been shown before, these 
large biofilm-like pods are observed after only 24 h of infection and represent 
a previously unrecognized intracellular microbial community and might play a 
role in the frequent recurrence of uncomplicated UTI (cystitis). However, the 
occurrence of these bacterial cell communities in human UTI has not yet been 
demonstrated. 

Colonization of Indwelling Devices 

For every artificial appliance placed in humans there is a corresponding 
microbial infection [30]. Tlie crucial importance of biofilms associated with 
conLaminaLion of medical ijnplanL devices has been well established. Although 
E. coli has been found to adhere to implanted endotracheal tubes and contact 
lenses [6, 3 1], it is predominantly isolated from the surface of urinary catheters. 
Catheter-associated UTIs are indeed the most common among nosocomial 
infections. For example, 1 0-50% of patients experiencing short-term (<7 days) 
urinary catheterization [32], and virtually all patients undergoing long-term 
(> I month) catheterization became infected [33]. 

During early stages of infection, E. coli is assumed to be present as a single 
species, whereas longer catheterization periods commonly lead to the forma- 
tion of mixed communities of mainly gram-negative opportunistic pathogens, 



Reisner/Hsiby/Tolker-Nielsen/Molin 120 



including /? aei^ginosa, Proteus mirabitis, and Klebsiella pneumoniae [34]. 
Such E. C(? //-dominated biofilms formed on the luminal surfaces can reach 
more than 400 (Jim in height, are usually embedded in a polysaccharide matrix 
[35], and can contain minerals such as hydroxyapatite and struvite that crystallize 
at the biofilm-urine interface as a result of the elevated pH achieved by bacterial 
urease activity. Although symptoms are seldom associated with the infection 
initially, ultimate blockage of the inner lumen of the catheter and/or ascent of 
bacteria to the bladder and kidney manifest severe consequences for the patient 
if left untreated. 

Further support for a biofilm mode of growth after catheter colonization is 
derived from studies indicating that bacteria in these biofilms survive the urinary 
concentrations of antibiotics generated by standard treatment [36]. As a conse- 
quence, removal of the colonized device is the only efficient way to clear the 
infection. Given these complications generated by biofihns, several attempts 
have been made to prevent infection and bacterial colonization of catheters by 
incorporating conventional antibiotics or biocides such as silver oxide into the 
catheter material [34, 36]. Unfortunately, although the onset of bacteriuria 
could be delayed for several days with some catheter materials and treatments^ 
most of these strategies were ineffective in preventing colonization [31]. 

A better insight into biofilm formation and ecology on catheters therefore 
appears to be required in order to identify more suitable and specific drug targets 
or to design more resistant catheters. It needs to be addressed whether initial 
colonization by E. colt supports a later establishment of other pathogens. 
Subsequent colonizers could attach to initial E. coli biofilms or benefit from 
provision of more suitable conditions in the local microenvironment such as 
changes of pH and nutrient supply. Interactions between different species during 
biofihn formation such as coaggregation might play an important role, as such 
phenomena have already been observed between Jactobacilli and UPEC [37]. 

However, since standardized in vitro and in vivo models are crucial for 
obtaining any relevant information about virulence mechanisms, the lack of a 
nondestructive, longitudinal monitoring system is a major problem faced in 
indwelling-device-related biofilm research. A recently described mouse model 
of chronic biofihn infection that relies on biophotonic imaging of biolumines- 
cent reporter bacteria constitutes an appealing approach to overcome this 
bottleneck [38]. 



P. aeruginosa 

R aeruginosa is an environmental microorganism found especially in fresh- 
water and soil. In humans, R aeruginosa may cause a wide range of infections. 



Biofilms and Pathogenesis 121 



The most prevalent and severe chronic lung infection in cystic fibrosis (CF) 
patients is caused by mucoid, biofilm-forming F aeruginosa, which has become 
endemic in CF patients [1]. CF is the most common congenital, inherited disease 
among Caucasian populations with an incidence rate of 1:2,500-1:4,500. The 
pathology of the lung infection, however, is similar in severe chronic obstructive 
pulmonary disease, where the number of patients is much higher. 

fn vitiv Biofilm Development 

In contrast to the biofilm development for E, coli, which appears to be a case 
of relatively simple self-assembly processes in concert with surface association, 
R aeruginosa is considered an example of a more elaborate biofilm develop- 
mental pathway involving several distinct steps of early and late maturation. 
Most of the work clarifying this developmental cycle has been performed with 
reference strains - PAOl, R aeruginosa 14 and PAK - and so far it appears that 
at least these strains share the major features of the biofihn developmental 
cycle. In particular, the highly structured R aeruginosa biofilms (comprising 
'mushrooms', 'towers', voids and water channels) observed under some condi- 
tions have been a challenge to molecular geneticists, and below we will briefly 
summarize the current understanding of how the development progresses and is 
controlled. 

It is first of all important to stress that structural biofilm development by 
R aeruginosa appears to be conditional. The immediate environment is a key 
determinant of the eventual biofilm structure, illustrated by the finding that in 
flow chambers supplied with a citrate minimal medium R aeruginosa forms a 
flat biofilm, while in flow chambers supplied with glucose minhnal medium it 
forms a heterogeneous biofilm with mushroom-shaped multicellular structures 
[39]. In a series of investigations, it was shown that the formation of the flat 
R aeruginosa biofilm occurs via initial growth of sessile bacteria forming 
microcolonies at the substratum, followed by expansive migration of the bacteria 
on the substratum, resulting in the formation of a flat biofilm [39]. Since biofilm 
formation by a /? aeruginosa pilA mutant (which is deficient in biogenesis of 
type IV pili) occurred without the expansive phase that resulLs in discrete 
protruding microcolonies, it was suggested that the expansive migration of the 
bacteria on the substratum is type IV pili-driven, and that the shift may be 
induced by some sort of limitation arising in the initial microcolonies. 

TTie formation of the mushroom-shaped structures in the heterogeneous 
glucose-grown R aeruginosa biofihn was shown to occur in a sequential process 
involving a nonmotile bacterial subpopulation^ which formed the initial micro- 
colonies by growth in certain foci of the biofilm, and a migrating bacterial subpopu- 
lation, which initially formed a monolayer on the substratum, and subsequently 
formed the mushroom caps by climbing the microcolonies [40]. 



Reisner/H0iby/Tolker-Nielsen/Molin 122 



TTie nature of bacterial cell agglutinating factor(s) in very dynamic 
P. aeruginosa biofilms is not known at present. A role of alginate as acell-to-cell 
interconnectLng substance has been proposed previously [41], but recently it 
was concluded that alginate is not expressed at any significant level in such 
in vitro biofilms and therefore cannot be a key structural determinant under the 
defined conditions [42]. As we will see later, this situation is completely reversed 
in biofilms developing in some clinical cases, where alginate production appears 
to be essential for robust biofilm development. Some bacterial cell populations 
are apparently kept in the biofilm by substances that allow type IV pili-driven 
migration. Since twitching motility is powered by a mechanism involving 
extension, grip, and retraction of type IV pili [43], it is possible that type IV pili 
can play a role as cell-to-cell and cell-to-substratum interconnecting compounds. 
It has been reported that extracellular DNA may play a role as a cell-to-cell 
interconnecting substance in P. aeruginosa biofilms [3, 44], and interest- 
ingly there is evidence that type IV pili bind to DNA [45]. Yet, other bacterial 
cell -to- sub stratum and cell-to-cell connections keep the pilA mutant bacteria 
substratum-associated and agglutinated in the biofihns. Evidence is emerging 
that a novel type of fimbriae may function as adhesin in P. aeruginosa biofilms 
[46], and that certain exopolysaccharides may function as cell-to-cell intercon- 
necting substances [Friedmann and Kolter, pers. commun.]. Such compounds 
could likely interconnect nonmigrating P. aeruginosa populations. 

The apparent complexity of the biofilm developmental cycle of/? aeruginosa 
has stimulated the search for genetic regulatory activities, and the findings of 
Davies et al. [47] that quorum-sensing control seems to be essential for normal 
biofilm formation was in accord with the characteristics of the process. In light 
of the current knowledge about the above-described steps of biofilm development 
for this organism it is, however, important to emphasize that so far no specific 
target for quorum-sensing control has been identified as relevant for these 
particular processes. It therefore remains to be seen whether quorum sensing is 
regulating any of the described process features such as bacterial cell-cell 
adherence, colony climbing or population differentiation. 

Chronic Lung Infections in CF 

CF patients are intermittently colonized with nonmucoid P. aeruginosa 
strains for an average of 12 months before the infections become chronic, and the 
presence of mucoid strains and an antibody response is a sign of chronicity [48^ 
49]. The chronic P. aeruginosa lung infections in CF patients is responsible for 
most of the morbidity and mortality of these patients [50], and this state of the 
infection constitutes a lung-associated biofilm [51, 52]. The biofilm is charac- 
terized by the mucoid phenotype of/? aeruginosa producing an abundance of 
alginate [53]. In the conductive zone of the lungs the majority of the bacteria 



Biofilms and Pathogenesis 123 



stay inside the mucus and grow under anaerobic conditions using nitrate as 
electron acceptor [54]. Most of the bacteria are not located on the epithelial 
cells, but they induce an endobronchitis and endobronchiolitis without spreading 
to the blood or to other organs [54, 55], In the respiratory zone of the airways, 
however, the environment is aerobic [56]. Foci of pneumonia in the aJveolar 
tissue with extensive infiltration of polymorphonuclear leukocytes (PMNs) 
surround localized biofilms of/? aeruginosa which are situated within the alve- 
oles and alveolar ducts [55, 57]. The location and organization of the bacteria 
in these biofilms are similar to those observed in mucoid colonies and Ln spu- 
tum fi^om CF patients with microcolonies of mucoid P aeruginosa [58]. High 
levels of antibodies are produced against alginate and other P. aeruginosa anti- 
gens, but elimination of the infections is not accomplished [59], and the result- 
mg persistent bnmune-compl ex-mediated inflammation is the major cause of the 
lung tissue damage [59]. The biofilm mode of growth is resistant to the patients' 
defense mechanisms and to antibiotic treatment [59] and is the major reason for 
the persistence of the infection lasting for more than 30 years in some patients. 

Adaptation of P. aerugmosa to CF Lungs 

The CF lung is a stressfijl environment for P. aeruginosa, and, therefore, 
they have developed a range of survival strategies. When particles of >5 [xm 
containing bacteria are inhaled, they are deposited in connection with the gel 
phase of the mucus on the airway surfaces in the relatively small conducting 
zone of the central airways, which are covered by ciliated epithelial cells and 
coordinated movements of these cilia beating in the sol phase (=epithelial lin- 
ing fluid) remove the gel phase of the mucus towards the trachea [56]. The gel 
phase of the mucus is produced by submucosal glands and goblet cells. In normal 
persons the effect of the cilia's beating (also named the mucociliary escalator) 
removes the mucus towards the trachea in this way rapidly (60 )xm/s) clearing 
the bacteria within 6h [54, 60]. This clearance mechanism is the most impor- 
tant part of the noninflammatory defense mechanism of the respiratory tract. In 
CF patients, however, the basic defect of the CFTR protein leads to a reduced 
volume of the epithelial lining fluid [60], and the mucociliary clearance of the 
bacteria is therefore greatly reduced, leading to robust bacterial growth [54] and 
recruitment of the inflammatory defense mechanisms (PMNs) [59]. When par- 
ticles of 2-5 |JLm containing bacteria are inhaled, they are deposited in the much 
larger peripheral respiratory zone of the lungs without mucus or cilia, and the 
major defense mechanism are the alveolar macrophages, which belong to the 
inflammatory defense mechanisms [56]. In accordance, bronchoalveolar lavage 
studies on CF infants have shown that recruitment of the inflammatory defense 
mechanisms (dominated by the phagocytic cells, PMNs and macrophages) 
takes place when aspirated microorganisms are colonizing the lower respiratory 



Reisner/Hsiby/Tolker-Nielsen/Molin 124 



tract [61]. When PMNs and macrophages engulf bacteria there is a metabohc 
burst in the phagosomes leading to a release of reactive oxygen species^ some 
of which are leaked to the environment [62]. These oxygen radicals induce 
killing, DNA damage and mutations in the bacteria [62, 63]. 

Oxygen radicals produced by the inflammatory response (PMNs) induce 
mutations in e.g. the mucA gene leading to the alginate production, which is 
characteristic for F aeruginosa biofilm infections in CF [64], Alginate, on the 
other hand, is an oxygen radical scavenger [65] and provides mucoid R aeruginosa 
with protection against further DNA damage compared to nonmucoid strains 
[66]. Alginate can also make the bacteria resistant to phagocytosis by PMNs 
and macrophages [67], Alginate production of P. aeruginosa biofilms in CF lungs, 
therefore, seems to be the major mechanism of adaptation permitting mucoid 
strains to persist in the hostile environment of oxygen radicals originating from 
the phagocytic cells of the inflammatory defense mechanisms. 

The lungs consist of the central conducting zone and the peripheral respi- 
ratory zone. When /? aeruginosa grow in the peripheral respiratory zone (niche)^ 
the growth condition is comparable to growth in an aerobic or microaerophilic 
incubation chamber (5-20% oxygen). The respiratory zone is the area of the 
lungs where the venous blood becomes oxygenated in the dense capillary net- 
work of the alveoles, thus providing continuous culture conditions with nutrient 
and oxygen from the blood [56]. The central conductive zone of the respiratory 
tract (the bronchi), on the other hand, where P aeruginosa is located in sputum, 
is a completely different niche^ since no oxygen is present in sputum [54]. 
Sputum consists mainly of dead PMNs and an abundance of released DNA [68] 
and leukocyte proteases [69] originating from PMNs in addition to mucus. In 
sputum the environment is anaerobic and the growth condition for P aerugi- 
nosa is comparable to a batch culture in the stationary phase. There is not so 
much blood supply of the conducting zone compared with the respiratory zone 
[56] and the bacteria are located inside sputum and not at the epithelial surface 
[54]. Under these conditions P aeruginosa may rely on anaerobic growth with 
N03~ as the electron acceptor [54]. 

In cases of infection with mucoid P aeruginosa cells, wliich dommates 
chronic infections, a pronounced antibody response against the bacteria is 
observed in connection with deteriorating lung function and poor prognosis. In 
contrast, the few CF patients colonized only with nonmucoid P aemginosa 
have a low antibody response, and they maintain their lung fiinction at the same 
nearly normal level similar to that of CF patients without chronic infection [70], 
The persistent PVJN inflammation around P aeruginosa infection areas in the 
respiratory zone destroys the lung tissue of the infected foci of the lungs of 
the CF patients [71], The alveolar macrophages in this zone [61], which migrate 
to the lymph nodes [56], are antigen-presenting cells, which are important for 



Biofilms and Pathogenesis 125 



initiating the antibody production of the B lymphocytes. Colonization of the 
conducting zone of the lungs, on the other hand, primarily leads to obstruction 
due to the abundance of mucus, and antibody production and lung tissue damage 
of the respiratory zone are normally not severe [54]. These observations sug- 
gest that severe respiratory failure in CF patients is caused by infection of the 
respiratory zone with mucoid P. aeruginosa located in biofilms [55, 57], Pieces 
of these biofilms are visible in gram-stained smears of sputum from CF patients 
[58]. Although the mucoid phenotype of P. aeruginosa is characteristic for 
colonization of the respiratory zones in CF patients, nonmucoid variants of the 
same genotype are regularly present simultaneously in sputum [66]. The reason 
for this diversity has so far been obscure [58], but indications from in vitro 
investigations of stratified bacterial populations may be relevant for a better 
understanding of the phenotypical diversity of infectious P. aeruginosa popula- 
tions in CF lungs [57, 72-74]. In a population of lung-associated mucoid 
P. aeruginosa, isogenic nonmucoid variants could represent a subpopulation of 
the original infecting cells (most likely not mucoid) occupying a niche in which 
mucoidy is not selectively favorable. Alternatively, the nonmucoid variants may 
be phenotypic revertants arising either as 'cheaters', benefiting from the algi- 
nate production of other bacteria within the biofihn, or as niche specialists in 
the anaerobic conductance zone. The fact that these variants seem to appear as 
individual bacteria outside the mucoid biofilm areas in sputum may indicate 
that they predominantly derive from the anaerobic zone. 

Antibiotic Therapy 

Bacteria growing in biofilms are often much more resistant to antibiotics 
than planktonic cells of the same isolate. Minimal inhibitory concentration and 
minLmal bactericidal concentration may be increased 100- to 1,000-fold in old 
biofilms, whereas young biofilms are less resistant [75]. In contrast, planktonic 
bacteria released from such resistant biofilms are most often found to be as 
sensitive to antibiotics as the original planktonic cells [75]. Biofilm-induced 
resistance to antibiotics can be caused by several factors, such as slow growth, 
reduced oxygen concentrations at the base of the biofilm, penetration barriers 
e.g. binding of positive charges on the antibiotic molecules to the negatively 
charged alginate polymers, the presence of 13-lactamase from the bacteria which 
cleaves and/or traps (3-lactam antibiotics and overexpression of efflux pumps 
[53, 76]. The increased resistance of biofilm bacteria usually results in the 
failure of antibacterial therapy with respect to eradication of the bacteria, but 
the antibiotic treatment regularly leads to temporary clinical improvement of 
the patient [53]. 

The development of traditional mechanisms of resistance to antibiotics 
occurs frequently in CF due to the intensive selective pressure provided by the 



Reisner/Hsiby/Tolker-Nielsen/Molin 126 



large amount of antibiotics used in these patients [53]. Mucoid and nonmucoid 
variants of the same strain are frequently simultaneously present in sputum but 
the nonmucoid variants are more resistant to antibiotics, possibly reflecting a 
higher antibiotic selection pressure outside the alginate biofilm [66]. The num- 
ber of/? aeruginosa in sputum may be as high as lO^-lO'^CFU/ml. The high 
number of bacteria implies that mutations do occur in sputum. In addition, high 
frequencies (>30%) of hypermutable F aemginosa variants have been found in 
CF lung infection [77, 78], and the mutator strains (hypermutable strains) show- 
ing >20-fold higher mutation frequency than control strains [78] were also mul- 
tiply resistant. The observations fr"om R aeruginosa strains from CF patients 
showed the occurrence of a high frequency of hypermutable P. aeruginosa, a 
high level of resistance to many antibiotics and, in the case of ciprofloxacin, 
several different mutations which increased over time [79]. Jn addition, muta- 
tions can be induced by means of oxygen radicals from PMNs, which in vitro 
leads to alginate production due to mutations in the mitcA gene [64]. Furthermore, 
there is an antioxidant imbalance in the CF lung, which leads to oxygen radical 
damage [80]. Taken together, all these observations have led us to suggest that 
it is the chronic inflammation dominated by PMNs which induces a high level 
of mutations in R aeruginosa in the CF lungs and that the resistant mutants are 
then selected by the heavy use of antibiotics. These conventional resistance 
mechanisms are then added to the physiological resistance caused by the biofilm 
mode of growth in the CF lung. 



Perspectives 

There is an increasing documentation concernmgthe importance of biofibns 
in connection with microbial infections - in particular in relation to persistent 
infections of opportunistic pathogens. The detailed investigation of several micro- 
bial biofilms has produced interesting information indicating that the multi- 
cellular life of bacteria may have its own genetic background that is controlled 
by bacterial interactions, which in some cases may resemble complex eukaryotic 
tissue development. One important question in relation to pathogenic bacteria 
is whether it is possible to extrapolate from these detailed in vitro observations 
and mechanisms to the conditions in the infected host. A word of caution is 
probably warranted: it is important to keep in mind that there is no indication 
of a consensus developmental program, and we therefore must resolve the indi- 
vidual biofilm pathways case by case. We also have strong ijidications that the 
in vitro biofilm conditions applied in the laboratory cannot be compared to those 
prevailing in the host, and it is therefore important to develop better model 
systems, if not performing the investigations in vivo. The genomic diversity of 



Biofilms and Pathogenesis 127 



bacteria is an additional complication; different isolates of the same species 
often behave quite differently from each other or when compared with reference 
strains or laboratory strains. We also have to keep in mind that simple molecu- 
lar identification and characterization of various bacterial cell-cell interaction 
mechanisms only constitute the first step in an approach to interfere with cell- 
cell interactions necessary for virulence. Since the overall physical strength and 
resistance of biofilms to shear force presumably play a critical role in vivo, a 
better understanding of the binding forces exhibited by the individual implied 
molecular factors is required to identify realistic drug targets. 

We now have some fundamental knowledge about the principles of bacterial 
life forms which seem to be important for a range of pathogens causing severe 
therapeutic problems in the clinic, and the technological and conceptual advances 
that have been made during the last 10 years of biofilm research should be applied 
with increased intensity in the investigations of infectious diseases. In particular, 
it will be important to establish the boundaries for our extrapolations from 
in vitro biofilm studies to the conditions prevaihng in clinical cases, just as we must 
expand our investigation scenarios to encompass conditions which much better 
reflect what goes on in cases of suspected biofilm infections. 



References 

1 Costerton JW, Stewart PS, Greenberg EP: Bacterial biofilms: A common cause of persistent infec- 
tions. Science 1999;284:1318-1322. 

2 Costerton JW, Lewandowski Z, Caldwell DE, Korber DR, Lappin-Scott HM: Microbial biofilms. 
Annu Rev Microbiol 1995;49:711-745. 

3 Whitchurch CB, Tolker-Nielsen T, Ragas PC, Mattick JS: Extracellular DNA is required for bac- 
terial biofilm formation. Science 2002;295:1487-1489. 

4 Dormenber^ MS: Escherichia coli: Virulence Mechanisms of a Versatile Pathogen. London, 
Academic Press, 2002. 

5 Stoodley P, Sauer K, Davies DG, Costerton JW: Biofilms as complex diflFerentiated communities. 
Annu Rev Microbiol 2002;56: 187-209. 

6 Davey ME, O'Toole GA: Microbial biofilms: From ecology to moleculai' genetics, Microbiol Mol 
Biol Rev 2000;64:847-867. 

7 Ghigo JM: Are there biofilm-specific physiological pathways beyond a reasonable doubt? Res 
Microbiol 2003;154:1-8. 

8 Vidal O, Longin R, Prigent-Combaret C, Dorel C, Hooreman M, Lejeune P: Isolation of an 
Escherichia cofi K-12 mutant strain able to form biofilms on inert surfaces: Involvement of a new 
ompR allele that increases curli expression. J Bacteriol 1998;180:2442-2449. 

9 Prigent-Combaret C, Prensier G, JLe Thi TT, Vidal O, Lejeune P, Dorel C: Developmental pathway 
for biofilm formation in curli-producing Escherichia coli strains: Role of flagella, curli and colanic 
acid. Environ Microbiol 2000;2:450-464. 

10 Ghigo JM: Natural conjugative plasmids induce bacterial biofilm development. Nature 2001; 
412:442^45. 

1 1 Reisner A, Haagensen JAJ, Schembri MA, Zechner EL, Molin S: Development and maturation of 
Escherichia coli K-12 biofilms. Mol Microbiol 2003;48:933-946. 

12 Prigent-Combaret C, Vidal O, Dorel C, Lejeune P: Abiotic surface sensing and biofilm-dependent 
regulation of gene expression in Escherichia coli. i Bacteriol 1999;181:5993-6002. 



Reisner/H0iby/Tolker-Nielsen/Molin 128 



13 Schembri MA, Kjaergaard IC, Klemm P: Global gene expression in Escherichia coli biofilms. Mol 
Microbiol 2003;48:253-267, 

14 Cookson AL, Cooley WA, Woodward MJ: The role of type I and curii fimbriae of Shiga toxin- 
producing Escherichia coli in adherence to abiotic surfaces. Int J Med Microbiol 2002;292: 195-205. 

15 Hanna A, Berg M, Stout V, Razatos A: Role of capsular colanic acid in adhesion of uropathogenic 
Escherichia coli. AppI Environ Microbiol 2003;69:4474^M8K 

1 6 Freter R, Brickner H, Fekete J, Vickerman MM, Carey KJE: Survival and implantation o? Escherichia 
coli in the intestinal tract Infect Immun 1983;39:686-703. 

17 Moller AK, Lealham MP, Conway T, Nuijten PJ, de Haan LA, Krogfelt KA, Cohen PS: An 
Escherichia coli MG 1655 lipopolysaccharide deep-rough core mutant grows and survives in mouse 
cecal mucus but fails to colonize the mouse large intestine. Infect Immun 2003;7 1 :2142-2I 52. 

18 Poulsen LK, Lan F, Kiislensen CS, Hobollh P, Molin S, Krogfelt KA: Spatial distribution of 
Escherichia coli in the mouse lai^e intestine inferred from rRNA in situ hybridization. Infect Immun 
1994;62:5191-5194. 

1 9 Mataro JP: Diarrhoeagenic Escherichia coli; in Sussman M (ed): Molecular Medical Microbiology, 
London, Academic Press, 2001, vol 2, pp 1463-1504. 

20 Nougayrede JP, Fernandes PJ, Donnenberg MS: Adhesion of enteropathogenic Escherichia coli to 
host cells. Cell Microbiol 2003;5:359-372, 

2 1 Giron JA, Ho AS, Schoolnik GK: An inducible bundle-forming pilus of enteropathogenic Escherichia 
coli. Science 1991;254:710-713. 

22 Nishi J, Sheikh J, Mizuguchi K, Luisi B, Burland Y Boutin A, Rose DJ, Blattner FR^ Nataro JP: 
The export of coat protein from enteroaggregative Escherichia coli by a specific ATP-binding cas- 
sette transporter system. J Biol Chem 2003;278:45680^5689. 

23 Trabulsi LR, Keller R, Tardelli-Gomes TA: Typical and atypical enteropathogenic Escherichia 
coli. Emerg Infect Dis 2002;8:508-513. 

24 Foxjnan B: Epidemiology of urinary tiact infections: Incidence, morbidity, and economic costs. 
Am JMed2002;113:5S-13S. 

25 Ronald A: The etiology of urinary tract infection: Traditional and emerging pathogens. Am J Med 
2002;113:14S-19S. 

26 Sussman M: Urinary tract infections: A clinical overview; in Sussman M (ed): Molecular Medical 
Microbiology. London, Academic Press, 2001, vol 2, pp 1507-1514, 

27 OelschlaegerTA, Dobrindt U, Hacker J: Virulence factors of uropathogens. Curr Opin Urol 2002; 
12:33-38. 

28 Muhldorfer 1, Ziebuhr W, Hacker J: Escherichia coli in urinary tract infections; in Sussman M 
(ed): Molecular Medical Microbiology. London, Academic Press, 2001, vol 2, pp 1515-1540. 

29 Anderson GG, Palermo JJ, Schilling JD, Roth R, Heuser J, Hultgren SJ: Intracellular bacterial 
biofilm-like pods in urinary tract infections. Science 2003;301:105-107. 

30 Bisno AL, Waldvogel FA: Infections Associated with Indwelling Medical Devices. Washington, 
ASM Press, 1989. 

3 1 Donlan RM, Coslerton JW: Biofilms: Survival mechanisms of clinically relevant microorganisms. 
Clin Microbiol Rev 2002;15:167-193. 

32 Mulhall AB, Chapman RG, Crow RA: Bacteriuria duting indwelling urethral catheterization. 
J Hosp Infect 1988;11:253-262. 

33 Kunin CM: Care of the urinary catheter; in Detection, Prevention and Management of Urinary 
Tract Infections, ed 4. Philadelphia, Lea & Febiger, 1987, pp 245-298. 

34 Donlan RM: Biofilms and device-associated infections, Emerg Infect Dis 2001;7:277-281. 

35 Ganderton L, Chawla J, Winters C, Wimpenny J, Stickler D: Scanning electron microscopy of bac- 
terial biofilms on indwelling bladder catheters. Eur J Clin Microbiol Infect Dis 1 992; 1 1 :789-796. 

36 Stickler DJ: Susceptibility of antibiotic-resistant gram-negative bacteria to biocides: A perspective 
from the study of catheter biofilms. J Appl Microbiol 2002;92:163S-170S. 

37 Rickard AH, Gilbert P, High NJ, Kolenbrander PE, Handley PS: Bacterial coaggregation: An inte- 
gral process in the development of multi-species biofilms. Trends Microbiol 2003; 1 1:94-100. 

38 Kadurugamuwa JL, Sin LV, Albert E, Yu J, Francis ICP, DeBoer M, Rubin M, Bellinger-Kawahara C, 
Parr TR Jr, Contag PR: Direct continuous method for monitoring biofilm infection in a mouse 
model. Infect Immun 2003;71:882-890. 



Biofilms and Pathogenesis 129 



39 Klausen M, Heydorn A, Ragas P, Lambettsen L, Aaes-J0rgensen A, Molin S, Tolker-Nielsen T: 
Biofilm formaiion by Pseudomonas aeruginosa wild type, flagella, and type IV pili mutants. Mol 
Microbiol 2003;48:1511-1524. 

40 Klausen M, Aaes-Jorgensen A, Molin S, Tolker-Nielsen T: Involvement of bacterial migration in 
the development of complex multicellular structures in Pseudomonas aeruginosa biofilms. Mol 
Microbiol 2003;50:61^8. 

41 Boyd A, Chakrabarty AM: Role of alginate lyase in cell detachment oi Pseudomonas aentginosa. 
AppI Environ Microbiol 1994;60:2355-2359. 

42 Wozniac DJ, WyckofTTJO, Starkey M, Keyser R, Azadi P, OToole GA, Parsek MR: Alginate is not 
a significant component of the extracellular polysaccharide matrix of PA 1 4 and PAOI Pseudomonas 
aeruginosa biofilms. ProcMatl Acad Sci USA 2003;100:7907-7912. 

43 Skerker JM, Berg HC: Direct observation of extension and retraction of type IV pili. Proc Natl 
Acad Sci USA 2001;98:6901-6904. 

44 Nemoto K, Hirota K, Murakami FCTaniguti IC, Murata H, Viducic D, MiyakeY: Effect of Varidase 
(streptodomase) on biofilm formed by Pseudomonas aeruginosa. Chemotherapy 2003;49: 
121-125. 

45 Schaik EJ, Irvin RT; DNA uptake in Pseudomonas aeruginosa: An addirional type IV pilus function 
(poster). ASM Conference, Victoria, 2003. 

46 VaUetl, Olson JW, Lory S, Lazdunski A, Filloux A:Thechaperone/usherpathvi'aysofft^(^t?m(9rta5 
aeruginosa: Identification of fimbrial gene clusters (cup) and their involvement in biofilm for- 
mation. Proc Natl Acad Sci USA 2001;98:69l 1-6916. 

47 Davies DG, Parsek MR, Pearson JP, Iglewski BH, Costerton JW, Greenberg EP: The involvement 
of cell-to-cell signals in the development of a bacterial biofilm. Science 1998;280:295-298. 

48 Heiby N: Pseudomonas aeruginosa infection in cystic fibrosis. Relationship between mucoid 
strains of Pseudomonas aeruginosa and the humoral immune response. Acta Pathol Microbiol 
Scand Microbiol Immunol 1974;82:551-558. 

49 Johansen HK, Hoiby N: Seasonal onset of initial colonisation and chronic infection with 
Psei/domonas aeruginosa in patients with cystic fibrosis in DeJimark. Thorax J992;47y2:J09-J J 1. 

50 H0iby N, Frederiksen: Microbiology of cystic fibrosis; in Hodson ME, Geddes D (eds): Cystic 
Fibrosis, ed 2. London, Arnold, 2000, pp 83-107. 

51 Singh PK, Schaefer AL, Parsek MR, Moninger TO, Welsh MJ, Greenberg EP: Quorum-sensing 
signals indicate that cystic fibrosis lungs are infected with bacterial biofilms. Nature 2000;407: 
762-764. 

52 Middleton B, Rodgers HC, Camara M, Knox AJ, Williams P, Hardman A: Direct detection of 
N-acylhomoserine lactones in cystic fibrosis sputum. FEMS Microbiol Lett 2002;207/l:l-7. 

53 Hoiby N: Understanding bacterial biofilms in patients with cystic fibrosis: Current and innovative 
approaches to potential therapies. J Cystic Fibrosis 2002;1:249-254. 

54 Worlitzsch D,Tarran R, Ulrich M, Schwab U, Cekici A, Meyer KC: Effects of reduced mucus oxygen 
concentration in airway Pseudomonas aeruginosa infections of cystic fibrosis patients. J Clin 
Invest 2002;109:317-325. 

55 Baltimore RS, Christie CDC, Smith GJ\\^: Immunohistopathologic localization of Pseudomonas 
aeruginosa in lungs from patients with cystic fibrosis - Implications for the pathogenesis of pro- 
gressive lung deterioration. Am Rev Respir Dis 1989; 140:1650-1661. 

56 Westh JB: Pulmonary Physiology and Pathophysiology. Philadelphia, Lippincott Williams & 
Wilkins, 2001. 

57 Hoffmann N, Rasmussen TB, Jensen P0, Stub C, Hentzer M, Ciofu O, Givskov M, Molin S, 
Johansen HK, Hoiby N: A novel mouse model of chronic Pseudomonas aeruginosa lung infection 
mimicking cystic fibrosis. Infect Immun, submitted. 

58 Hoiby N: Pseudomonas aeruginosa infection in cystic fibrosis. Diagnostic and prognostic signif- 
icance of Pseudomonas aeruginosa precipitins determined by means of crossed immunoelectro- 
phoresis. A survey. Acta Pathol Microbiol Scand l977;262(suppl):3-96. 

59 Hoiby N, Johansen HK, Moser C, Song ZJ, Ciofu O, Kharazmi A: Pseudomonas aeruginosa and 
the biofilm mode of growth. Microbes Infect 2001;3:1-13, 

60 Boucher RC: An overview of the pathogenesis of cystic fibrosis lung disease. Adv Drug Deliv Rev 
2002;54:1359-1371. 



Reisner/H0iby/Tolker-Nielsen/Molin 130 



61 Armstrong DS, Grimwood K, Carzino R, Carlin JB, Olinsky A, Phelan PD: Lower respiratory 
mfection and inflammation in infants with newly diagnosed cystic fibrosis, Br Med J 1995;310: 
1571-1572. 

62 Miller RA, Britigan BE: Role of oxidants in microbial pathophysiology. Clin Microbiol Rev 
1997;10:1-18. 

63 Hull J, Vervaart P, Grimwood K, Phelan P: Pulmonary oxidative stress response in young children 
with cystic fibrosis. Thorax 1997;52:557-560. 

64 Mathee fC, Ciofli 0, Sternberg C, Lindum PW, Campbell JIA, Jensen P, Johnsen AH, Givskov M, 
Ohman DE, Molin S, H0iby N, Kharazmi A: Mucoid conversion of Pseudomonas aeruginosa by 
hydrogen peroxide; A mechanism for virulence activation in the cystic fibrosis lung. Microbiology 
1999;145:1349-1357. 

65 Simpson J, Smith SE, Dean RT: Scavenging by alginate of free radicals released by macrophages. 
Free Radic Biol Med 1989;6:347-353. 

66 Ciofij 0, Fussing V, Bagge N, Koch C, Hoiby N: Characterization of paired mucoid/non-mucoid 
Pseudomonas aeruginosa isolates from Danish cystic fibrosis patients: Antibiotic resistance, beta- 
lactamase activity and RiboPrinting. J Antimicrob Chemother 2001;48:391-396. 

67 Simpson JA, Smith SE, Dean RT: Alginate inhibition of the uptake of Pseudomonas aeruginosa 
by macrophages. J Gen Microbiol 1988;134:29-36, 

68 Shah PL, Scott SF, Knight RA, Marriott C, Ranasinha C, Hodson ME: In vivo effects of recom- 
binant human DNase I on sputum in patients with cystic fibrosis. Thorax 1996;5I/2:I 19-125. 

69 Goldstein W, Doring G: Lysosomal enzymes from polymorphonuclear leukocytes and proteinase 
inhibitors in patients with cystic fibrosis. Am Rev Respir Dis 1986; 134:49-56. 

70 Pedersen SS, Hoiby N, Espersen F, Koch C: Role of alginate in infection with mucoid 
Pseudomonas aeruginosa in cystic fibrosis. Thorax 1992;47:6-13. 

71 Tiddens HAWM: Detecting early structural lung damage in cystic fibrosis. Pediatr Pulmonol 
2002;34:228-231. 

72 Spiers AJ, Buckling A, Rainey PB:The causes of Pseudomonas di'^QTsity, Microbiology 2000; 146: 
2345-2350. 

73 WyckolTTJO, Thomas B, Hassetl DJ, Wozniak DJ: Static grov^lh of mucoid Pseudomonas aeruginosa 
selects for non-mucoid variants that have acquired flagetlum-dependent motility. Microbiology 
2002; 148:3423-3430. 

74 Rainey PB, Travisano M: Adaptive radiation in a heterogenous environment. Nature 1998;394: 
69-72. 

75 Anwar H, Strap JL, Costerton JW: Establishment of aging biofilms: Possible mechanism of bac- 
terial resistance to antimicrobial therapy. Antimicrob Agents Chemother 1992;36:1347-1351. 

76 Stewart PS, Costerton JW: Antibiotic resistance of bacteria in biofilms. Lancet 2001 ;358: 135-138. 

77 Oliver A, Canton R, Campo P, Baquero F, BLazquez J: High frequency of hypermutable Pseudomonas 
aeruginosa in cystic fibrosis lung mfection. Science 2000;288:1251-1253. 

78 Oliver A, Baquero F, Blazquez J: The mismatch repair system {mutS, mulL and uvrD genes) in 
Pseudomonas aerugimjsa: Molecular characterization of naturally occuiring mutants. Mol Microbiol 
2002;43:1641-1650. 

79 Jalal S, Ciofli 0, Hoiby N, Gotoh N, Wretlind B: Molecular mechanisms of fluoroquinolone resis- 
tance in Pseudomonas aeruginosa isolates from cystic fibrosis patients. Antimicrob Agents 
Chemother 2000;44 :7 1 0-7 1 2. 

80 Wood LG, Fitzgerald DA, Gibson PG, Cooper DM, Collins CE, Garg ML: Oxidative stress in 
cystic fibrosis: Dietary and metabolic factors, J Am Coll Nutr 2001;20/2:157-165. 



Soren Molin 

Molecular Microbial Ecology Group, BioCentrum-DTU 

Building 301, DTU 

DK-2800 Lyngby (Denmark) 

Tel. +45 45252513, Fax +45 45887328, E-Mail sm@biocentrum.dtu,dk 



Biofilms and Pathogenesis 131 



Enzymes 

Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kai'ger, 2005, vol 12, pp 132-180 



Bacterial Peptidases 



Jan Potempa^, Robert N. Pike^ 

^Department of Microbiology, Faculty of Biotechnology, Jagiellonian University, 

Krakow, Poland, and Department of Biochemistry and Molecular Biology, 

University of Georgia, Athens, Ga., USA; 
''Department of Biochemistry and Molecular Biology, Victorian Center for Oral Health 

Sciences and CRC for Oral Health Sciences, Monash University, 

Clayton, Australia 



Enzymes that catalyze the hydrolysis of peptide bonds are referred 
to as proteases or peptidases. They are widely distributed in nature, where a 
variety of biological functions and processes depend on their activity. 
Regardless of the complexity of the organism, peptidases in general are 
essential at every stage m the life of every individual cell, since all protein 
molecules produced must be proteolytically processed and eventually 
degraded. Therefore, it is not surprising that throughout cellular life forms, 
genes encoding proteases occur at a relatively high frequency, ranging from 
1.15% {Pirellula sp.) to 6.06% (Buchnera aphidicola) of the total gene count, 
with the average being about 3%). Among bacterial species which are patho- 
genic for humans, the number of peptidases known and putatively functional 
ranges from 9-15 in small genomes, such as those of the Mycoplasma 
spp. (1.45-2.07%) of the total gene count) to 98 (2.64%) and 121 (2.85%) in 
genomes stich as Pseudomonas aeruginosa and Escherichia coli, respectively. 
Fortunately, only a small fraction of the expressed peptidases in any pathogen 
impose a direct or indirect deleterious effect on their human host and may 
therefore be considered a virulence factor. With respect to the number of pro- 
tease genes, the record in the microbial world goes to Bacillus cereus [179 
potentially functional peptidase genes out of a total of 5,243 genes (3.99%)]. 
In comparison, onJy three times more functional protease genes have been 
identified in Homo sapiens (489 + 143 out of 23,531, 2.7% of the total gene 
count). 



Classification of Peptidases 

Three major criteria are currently used to classify peptidases: (I) the reaction 
catalyzed, (2) the chemical nature of the catalytic site, and (3) the evolutionary 
relationship to other proteases, as revealed by the primary and/or tertiary structure 
of the protein. 

Based on the reaction they catalyze, peptidases are divided into two classes, 
comprising the exopeptidases and endopeptidases. The exopeptidases act only 
near the ends of polypeptide chains. Those acting at a free amino-terminus to 
liberate a single amino acid residue, a dipeptide or a tripeptide are referred to as 
aminopeptidases, dipeptidyl-peptidases, and tripeptidyl-peptidases, respectively. 
On the other hand, exopeptidases that cleave a single residue or dipeptide from a 
free carboxy-terminus are called carboxypeptidases and dipeptidyl-dipeptidases, 
respectively. Other exopeptidases are specific for dipeptides (dipeptidases), or 
the removal of terminal residues, either carboxy- or amino-terminal, that are 
substituted, cyclized, or linked by isopeptide bonds. Isopeptide bonds are peptide 
linkages other than those joining an ot-carboxyl to an a-amino group. This last 
group is collectively referred to as the omega peptidases and is of particular 
importance for prokaryotic organisms producing nascent proteins that start with 
N-formylmethionine at the beginning of their sequence, which needs to be 
removed. 

In contrast to the exopeptidases, endopeptidases preferentially hydrolyze 
peptide bonds in the inner regions of peptide chains, away from the termini. 
Typically, the presence of fi^ee a-amino or a-carboxyl groups has a negative 
effect on the activity of these enzymes, but it must be kept in mind that it is not 
unusual for an endopeptidase to have both exo- and endopeptidase activity. 
A subset of the endopeptidases, with activity limited to oligopeptides or fairly 
short polypeptide chains, are referred to oligopeptidases. 

According to the nature of their catalytic site, peptidases are divided 
into 6 types differing in their catalytic mechanism. The aspartic peptidases, 
sometimes incorrectly referred to as carboxypeptidases, have two aspartic acid 
residues involved in the catalytic process. The cysteine-type peptidases (incor- 
rectly called thiol peptidases) have a cysteine residue in their active center. The 
metallopeptidases use a metal ion (commonly zinc) in their catalytic mecha- 
nism. The activity of the serine-type peptidases depends on an active serine 
residue, while threonine-type peptidases utilize a catalytic threonine. The last 
group constitutes a number of peptidases that cannot yet be assigned to any par- 
ticular catalytic type. Among prokaryotic organisms, including pathogenic bac- 
teria, peptidases of all 6 catalytic types are common, although the frequency of 
their appearance is often strongly disproportionate (see following sections). 



Bacterial Peptidases 



33 



A third way to classify peptidases is based on the evolutionary and struc- 
tural relationship among enzymes, inferred from the comparison of amino acid 
sequences and/or tertiary structures. This method, introduced by Barrett et al. 
[2003], and currently implemented in the MEROPS database server (www. 
merops.ac.uk) [Rawlings et al., 2004], is a powerful tool, aJlowing the logical 
classification of all peptidases, since the structural similarities within a family 
of peptidases commonly reflect important similarities in catalytic mechanism 
and other properties. However, in some cases, the classification is not fully con- 
sistent with three-dimensional structural data, as observed for the structurally 
distinct astacins and adamolysins, englobed in the same family Ml 2, or ser- 
ralysins and matrixins, grouped into family MIO. This classification may even 
extend to assigning the biological flinction of an enzyme for which only the 
encoding DNA sequence is known. Therefore, the classification system briefly 
described below will be used here to discuss bacterial peptidases. 

The term 'family' is used to describe a group of peptidases in which each 
member shows an evolutionary relationship to at least one other, either through- 
out the whole sequence or at least in the part of the sequence responsible for cat- 
alytic activity. Each family is identified by an upper-case letter representing the 
catalytic type (A for aspartic type, C for cysteine type, M for metal lo-type, S for 
serine type, T for threonine type, and U for unknown type), followed by a unique 
number. A family that contains deeply divergent groups is sometimes divided 
into subfamilies, identified by upper-case letters. Families are further clustered 
into clans. A clan contains all the present peptidases that have evolved from a sin- 
gle origin. It represents one or more families that show evidence of their evolu- 
tionary relationship, judged by similar tertiary structures, or when structures are 
not available, by the order of catalytic-site residues in the polypeptide chain and 
often by common sequence motifs around the catalytic residues. Each clan is 
identified by two letters, the first representing the catalytic type of the families 
included in the clan (with the letter 'P' being used for a clan containing families 
of more than one of the catalytic types: serine, threonine or cysteine). 

For the purpose of this review it is worth introducing a fourth classification 
of bacterial peptidases accordhig to their role in pathogenicity. Pathogenicity, 
which is a term synonymous with virulence, is generally delineated as the abil- 
ity of a bacterium to cause infection. Virulence factors represent either bacterial 
products or a strategy that contributes to virulence, which entails the pathogen to 
colonize the host, evade host defense mechanisms, facilitate dissemination, and 
cause host damage [Jsenberg, 1988; Mekalanos, 1992]. In many respects, prote- 
olytic enzymes produced by several pathogenic bacterial species fit mto the cat- 
egory of virulence factors since they are directly involved in one or more of the 
processes listed above. Taking into account the numbers of peptidases produced 
by bacteria, relatively few can be considered sensu stricto as virulence factors. In 



Potempa/Pike 



134 



this chapter we refer to peptidases, which preferentially target host proteins as 
'primary virulence factors'. Many other peptidases are indirectly involved in 
pathogenicity, since they are indispensable for the expression of virulence factors 
per se. Such proteinases we call 'auxiliary virulence factors'. Finally, many other 
peptidases have well defined housekeeping functions. They do not harm the host 
either directly or indirectly, but are needed to withstand the stress of living in a 
hostile environment. We name them 'bystander virulence factors'. 



Aspartic Peptidases 

The MEROPS database currently (March 24, 2004) contams a total of 
19,682 peptidase-related sequences and aspartic peptidases represent 6.3% of all 
peptidases, compared with 19.8% for cysteine, 30.2% for metal lo-, 35.0% for 
serine, and 4.1% for threonine peptidases. The aspartic peptidases are subdi- 
vided into six clans. Two clans (clans AC and AF) contain enzymes present only 
in the major domain of living organisms made up by bacteria. Bacterial pepti- 
dases also constitute a separate family within clan AD. They are represented by 
three archetypal enzymes: lipoprotein signal peptidase (LspA) often referred to 
as signaJ peptidase II (SPase II), a type IV prepilin peptidase and omptin. 

SPase II participates in prolipoprotein translocation through the cytoplas- 
mic membrane of both gram-negative and gram-positive bacteria. With the 
exception of only three bacterial species, including Mycoplasma penetrans. 
Mycoplasma gallisepticum and onion yellows phytoplasma, the gene encoding 
a potentially functional protein has been found in all other species for which 
there is a completely sequenced genome (total 94). SPase II is a good example 
of a nonessential housekeeping enzyme, which, in the case of some pathogens, 
can contribute to their virulence. Apparently in Listeria monocytogenes, 
a gram-positive facultative intracellular human pathogen, temporally regulated 
expression of surface lipoproteins is critical for efficient phagosomal escape of 
L. monocytogenes. Mutants deficient in SPase II activity stayed entrapped 
inside the phagosomes of infected macrophages and have severely attenuated 
virulence [Reglier-Poupet et al., 2003]. 

The gene encoding a potentially functional homologue of the type IV 
prepilin peptidase is strongly conserved amongst bacteria (clan AD, subfamily 
24 A), although not to the same degree as SPase II. The enzyme cleaves, among 
other substrates, the leader sequence from type 4 prepilins or prepilin-like 
proteins secreted by a wide range of bacterial species. Its activity is required 
for a variety of functions, including type 4 pilus formation, secretion of tox- 
ins and other enzymes through the type II protein secretion system in gram- 
negative bacteria, gene transfer and biofilm formation. In many regards, 



Bacterial Peptidases 135 



prepilin peptidase can be considered a housekeeping enzyme, but it contributes 
to the expression of well-defined virulence factors in several pathogenic species. 
In enteropathogenic E. coii, assembly of the type IV fimbriae known as the bun- 
dle-forming pilus (BFP) is dependent on the activity of the prepilin peptidase 
encoded by the bfpP gene [Anantha et al., 2000]. Biogenesis of BFP is required 
for autoaggregation and localized adherence to host cells and enteropathogenic 
E. coli mutants deficient in these surface appendages are nonvirulent in orally 
challenged human volunteers. Similarly, a knockout of the prepilin peptidase 
gene (pilD) in Legionella pneumophila greatly impaired the ability of the bac- 
terium to grow within amoebae and human macrophage-like U937 cells [Liles 
et al., 1999]. The mutant showed strongly attenuated virulence in animal models 
due to the malfunction of the prepilin peptidase-dependent type 11 secretion 
system operating inside the phagocytes [Rossier et al., 2004]. Jn the case of 
Vibrio cholerae, functionmg of the extracellular protein secretion apparatus 
encoded by the eps gene is strongly dependent on prepilin peptidase activity. 
Deletion of the peptidase gene resulted in a dramatic decrease in cholera toxin 
secretion and abolished surface expression of the type 4 pilus responsible for 
mannose-sensitive hemagglutination [Marsh and Taylor, 1998]. 

In contrast to SPase 11 and the prepilin peptidase, which are good exam- 
ples of auxiliary virulence factors, the plasminogen activating surface pepti- 
dase, Pla, of the plague bacterium Yersinia pestis is a paradigm for the primary 
virulence factor. The Pla surface peptidase resembles mammalian plasminogen 
activators in function and converts plasminogen to plasmin by limited proteol- 
ysis. At the same time, the Pla peptidase inactivates a2-antiplasmin, a potent 
inhibitor of plasmin [Kukkonen et al., 2001], facilitating unrestrained activity 
of this broad -spectrum peptidase that in turn degrades fibrin and noncollage- 
nous proteins of the extracellular matrix and activates latent procollagenases. 
This causes local damage of the connective tissue and enables the highly 
efficient spread of Y. pestis from a subcutaneous site, where the pathogen is 
introduced by a vector bite, into the cb-culation [Sodeinde et al., 1992]. In addi- 
tion, independent of proteolytic activity, the Pla peptidase mediates Y. pestis 
adhesion to basement membrane and invasion into human endothelial cells, 
which may also contribute to dissemination of the bacterium in the host 
[Lahteenmaki et al., 2001]. 

The Pla peptidase shares significant amino acid sequence identity (about 
50%) with the E. coli integral outer membrane peptidases, OmpT and OmpR, 
referred to as omptins. Since some serine protease inhibitors weakly affect OmpT 
activity and site-directed mutagenesis studies appeared to implicate Ser99 and 
His212 as the active site residues [Kj-amer et al., 2000], the omptins have been 
classified as novel serine proteases (family S18) [Rawlings and Barrett, 1994]. 
However, the crystal structure of OmpT [Vandeputte-Rutten et al., 2001 ] followed 



Potempa/Pike 



136 



by structure-guided site-directed mutagenesis [Kramer et al., 2001] proved that 
OmpT activity depends on the Asp83-Asp85 and Asp2 1 0-His2 1 2 residues. These 
residues are strictly conserved in all OmpT homologues described to date, includ- 
ing PgtE of the Salmonella sp., peptidase SpoA of Shigella flexneri, putative 
peptidases ofRhizobium loti, a new species of legume root nodule bacteria, plant 
pathogens of the Ei-winia sp. and Agrobacterium tumefaciem, and of course 
OmpP and the Pla peptidase. It is assumed that these peptidases have a consei^ved 
fold, consisting of a lO-stranded antiparallel |3-barrel that protrudes far from the 
lipid bi layer into the extracellular space with the catalytic site located in a groove 
at the extracellular top of the vase-shaped p-barrel. Interestingly, activity of 
omptins is critically dependent on a specific interaction with lipid A of the LPS 
molecule [Kukkonen et al., 2004]. 

Omptins other than the Pla peptidase are typical housekeeping enzymes 
with their function/s not yet entirely tmderstood. Nevertheless, they also seem 
to be implicated directly or indirectly in bacterial pathogenicity [Stathopoulos, 
1998]. The presence of the ompT gene in clinical isolates of E. coli has been 
associated with complicated urinary tract disease [Webb and Lundigran, 1996], 
a notion supported by the observation that OmpT cleaves protamine, a highly 
basic antimicrobial peptide that is excreted by epithelial cells of the urinary 
tract [Stumpe et al., 1998]. Similarly, PtgE expression by Salmonella enterica 
may promote resistance to innate immunity by proteolytic inactivation of 
a-helical cationic antimicrobial peptides. On the other hand, SopA from 
S. flexneri, the causative agent of bacillary dysentery, cleaves the endogenous 
autotransporter IcsA, which has an essential role in the formation of actin tails 
in host cells, and therefore SopA might be indirectly involved in the actin-based 
motility inside infected cells [Egile et al., 1997; Shere et al., 1997]. 

Among omptins only the Pla peptidase is a potent plasminogen activator 
Interestingly, however, OmpT can be easily converted into the plasminogen 
activator by subtle mutations at surface-exposed loops. Such conversion may 
represent an interesting example of the evolution of a potent virulence factor 
from a housekeeping protein [Kukkonen et al., 2001]. In the case of PgtE 
from^S. enterica, the 0-antigen of LPS sterically prevents recognition of large- 
molecular-weight substrates, rendering plasminogen activator activity cryptic 
in this enteropathogen. The 0-antigen repeats also prevent plasminogen activa- 
tion by the Pla peptidase and, in this context, it is now clear why Y. pestis lost 
the genetic locus involved in 0-antigen synthesis [Kukkonen et al, 2004]. 

Collectively, it is apparent that the proteolytic activity of omptins con- 
tributes to virulence in a variety of ways. Their contribution ranges from bacte- 
rial defense and plasmin-mediated tissue infiltration to motility inside infected 
cells. Fortunately, they are produced by only a limited number of gram-negative 
bacteria which are pathogenic for plants and animals. 



Bacterial Peptidases 137 



Cysteine Peptidases 

The MEROPS database contains 3,897 cysteine-peptidase-related 
sequences (19.8% of the total sequences), which are divided into five phylo- 
genetically related clans of proteins (CA, CD, CE, CF, and CH) and several 
families which are provisionally without a clan assignment. Bacterial pepti- 
dases are scattered among all of the clans except clan CH. It is a paradox, how- 
ever, that although the bacterial ly derived cysteine peptidases, streptopain 
(SpeB) of Streptococcus pyogenes and clostripain from Clostridium perfrin- 
gens were among the first proteolytic enzymes ever characterized, cysteine 
peptidases are underrepresented in prokaryotic organisms and show limited 
variation. Just one family (family C40) encompasses more than one third of 
the total cysteine peptidase count in prokaryotes (about 640 sequences). These 
enzymes are exemplified by dipeptidyl-peptidase VI from Bacillus sphaericus 
and murein endopeptidases (LytE and LytF) from Bacillus subtiUs and repre- 
sent typical housekeeping peptidases. Biochemically characterized enzymes 
have N-acetylmuramoyl-L-aJanine amidase activity [Kuroda and Seikiguchi 
et aL, 1991; Moriyama et aj., 1996; Yamamoto et al., 2003] and are involved 
in a peptidoglycan turnover. They are widespread among both gram-positive 
and gram-negative bacteria and genes encoding from 1 to 6 functional homol- 
ogous are present in at least 70 bacterial species with completely sequenced 
genomes (out of 94). No association with virulence has been reported for this 
group of peptidases. 

Sortases (Family C60) 

Peptidases comprising the C60 family constitute a functionally and struc- 
turally related group of proteins expressed by all gram-positive species of 
bacteria. The prototypical enzyme, referred to as sortase A (SrtA), was first 
described in Staphylococcus aureus as an enzyme that is anchored in the plasma 
membrane and is responsible for covalent tethering of protein A to the cell wall 
[Mazmanian et al., 1999]. It is now known that SrtA attaches a range of impor- 
tant surface proteins to the peptidoglycan component of S. aureus and many 
other gram-positive bacteria, including virulence-related microbial surface com- 
ponents recognizing adhesive matrix molecules (MSCRAMs). Substrates for 
SrtA are easily recognized by a carboxy-terminally located sorting signal made 
up by an LPXTG amino acid sequential motif, where X is any amino acid, fol- 
lowed by a hydrophobic domain composed of about 20 amino acid residues and 
a tail of positively charged residues. The hydrophobic domain and charged 
residues hinder polypeptide chain translocation through the plasma membrane, 
facilitating recognition of the LPXTG motif by SrtA. In a two-step transpepti- 
dation reaction, sortase cleaves the LPXTG motif between the threonine and 



Potempa/Pike 



138 



glycine residues and covalently attaches a polypeptide chain, via the carboxy- 
terminal threonine, to the amino group of the pentaglycine crossbridge, thus 
tethering the protein to the cell wall. Although the structure of peptidoglycan 
crossbridging shows large variability in gram-positive bacteria, the mechanism 
of surface protem attachment is strictly conserved. 

A comparative genome analysis indicated that gram-positive bacteria fre- 
quently encode more than one sortase (up to 7 paralogues) and an even larger 
number of potential substrates (up to 40 per genome) with their characteristic 
LPXTG-type cell wall sorting motif or derivatives thereof [Comfort and Clubb, 
2004]. In contrast, a single gene coding for a sortase and only one potential sub- 
strate have been identified thus far in only five gram-negative bacterial species. 
The sortases can be partitioned into 6 distinct subfamilies (5 in gram-positive 
and 1 in gram-negative bacteria) based on amino acid sequence. Members of 
each subfamily are suggested to recognize a discrete variation of the sorting 
motif [Comfort and Clubb, 2004]. In the bacterial species with more than one 
sortase, usually the SrtA-like molecule is responsible for tethering of most cell 
wall proteins in an organism, while additional sortase(s) have more specialized 
functions. For example, in the case of S. aureus, sortase B (SrtB) recognizes 
and anchors a protein known as IsdD, which is involved in heme iron transport 
[Mazmanian et al., 2002, 2003]. This protein contains the NPQTN motif 
instead of the classical LPXTG sorting sequence exploited by SrtA, but other- 
wise the catalyzed reaction is identical. Also a protein, referred to as SvpA, 
which is anchored to peptidoglycan by SrtB of L. monocytogenes has the sort- 
ing motif, NAKNT, which is divergent from the one used by SrtA [Bierne et al., 
2004]. As in S. aureus, the genes encoding SrtB and its target, SvpA, are part 
of the same locus. In S. aureus, isd genes are regulated by iron and encode fac- 
tors for hemoglobin binding and the passage of iron, in the form of a heme 
group, to the cytoplasm [Mazmanian et al., 2002]. 

Some of the six sortase genes encoded in the genome of Corynebacierium 
diphtheriae are required for biogenesis of the pilus. Assembly of the fimbriae 
involves the cleavage of pilin precursors at the classical sorting signal 
(LPLTG), or at an LAFTG motif, by two different sortases, which then further 
catalyze amide bond cross-linking of adjacent subunits or tethering to peptido- 
glycan [Ton-That and Schneewind, 2003]. This covalent attachment of adjacent 
pilin subunits has probably evolved m many gram-positive bacteria, since sor- 
tase genes in close association with pilin subunit genes with sorting signals 
were found in enterococci, streptococci, Actinomyces spp., and C. perfringens. 

The NMR structure in solution of SrtA [llangovan etal., 2001] and the crys- 
tal structure of SrtB [Zong et al., 2004] from S. aureus are available, revealing an 
eight-stranded (3-barrel core structure with a helical subdomain at the amino- 
terminal end, which is unique among peptidases. The topology of the p-barrel is 



Bacterial Peptidases 139 



identical in both enzymes with the critical cysteine residue (Cysl 84 and Cys223 
in SrtB and SrtA, respectively) located at the tip of the (37 strand. Initially, it was 
predicted that Cysl 84 and His 1 20 of SrtA form a thiolate-imidazolium ion pair 
for catalysis [Ton-That et al., 2002] as in the papain cysteine peptidases. 
However, pKa measurements for SrtB Cysl 84 and His 120 residues refuted the 
involvement of the His residue in the transpeptidation reaction [Connolly et al., 
2003]. From the crystal structure of SrtB and conservation of the Arg233 
(Argl 97 in SrtA) residue it is apparent that a unique Cys-Arg catalytic dyad con- 
stitutes the foundation of the catalytic machinery of sortases. 

By exposing anchored proteins and polymeric structures such as fimbriae, 
the cell wall envelope of gram-positive bacteria can be considered to be a surface 
organelle maintaining contact between the microbe and its environment. It is now 
apparent that the assembly of these surface appendages is dependent on sortases. 
In this regard, sortases can be considered to be house-keeping enzymes. However, 
they are responsible for surface expression of acknowledged virulence factors, 
which mediate adherence to host tissues, host cell invasion, iron acquisition, and 
provide protection from assault by the formidable forces of the innate and 
acquired immune system. Therefore, sortases can be considered to be the classi- 
cal example of an auxiliary virulence factor. Indeed, it was shown that sortase 
knockouts in various pathogenic bacteria, including S. aureus, Sweptococcus 
/nutans, L. monocytogenes. Streptococcus gorcfonii, and Streptococcus pneumo- 
niae, have significantly attenuated virulence when tested in several different ani- 
mal models. In this way sortase(s) are a very good target for the development of 
therapeutic inhibitors to fight gram-positive infections. 

Family C66: IdeS Peptidase (MAC Protein) 

A streptococcal protein (Mac) has been identified as a group A Streptococcus 
(GAS)-secreted protein of 35 kD with homology to the a-subunit of Mac-1, a 
leukocyte ^2 integrin. Mac binds to CDl 6 (Fc^ROIB) on the surface of human 
polymorphonuclear leukocytes and inhibits opsonophagocytosis and production 
of reactive oxygen species, which resulted in significantly decreased pathogen 
killing [Lei et al., 2001]. Later, the MAC protein was shown to be identical to the 
IdeS peptidase (IgG-degrading enzyme of 5*. pyogenes) [von Pawel-Rammingen 
et al., 2002a, b], a previously unrecognized cysteine peptidase of 5". pyogenes. The 
IdeS peptidase is an extremely specific enzyme, which exclusively cleaves the 
heavy chain of IgG at the Gly237 residue in the hinge region. The enzyme is active 
in human plasma and its ability to interfere with Fc-mediated phagocytic killing 
has been demonstrated in a variety of bactericidal assays. These data collectively 
show that the IdeS protease contributes to evasion of the adaptive immune system 
by GAS by cleaving opsonizing IgG antibodies at the bacterial surface [von 
Pawel-Rammingen and Bjorck, 2003]. There is, however, a debate as to whether 



Potempa/Pike 



140 



the proteolytic activity of IdeS (MAC protein) is absolutely necessary for inter- 
ference with phagocytosis, which may only be dependent on molecular mimicry 
and the presence of the Arg-Gly-Asp amino acid motif in IdeS, which is involved 
in the interaction of the enzyme with the human integrins, a^^j ^nd a^^^ [Lei 
et al., 2002; von Pawel-Rammingen and Bjorck, 2003], 

The occurrence of orthologues of the IdeS peptidase is limited to a very 
small subset of the streptococci. In GAS, the enzyme occurs in two allelic 
variants among GAS serotypes, where the amino acid sequences of the variants 
differ from each other by about 15%. The only three homologues of the IdeS 
peptidase identified thus far are in the genome of Streptococcus equi (two 
genes) and m Streptococcus suis. One enzyme from S. equi was expressed and 
the recombinant protein was shown to possess the same activity as the IdeS 
peptidase [Lei et al., 2003]. A distant homologue was also identified in the 
genome of Treponema denticola. The recombinant protein was expressed in 
E. coli and shown to have a nonspecific, general peptidase activity [Potempa, 
unpubl. data]. 

The activity of IdeS depends on a thiolate-imidazolium ion pair formed by 
Cys94 and His262, which act as the active-site residues as in the papain-like 
peptidases. These residues are conserved not only in the enzymes from S. equi 
and S. suis, but also in the T. denticola homologue. However, the amino acid 
sequence is unique and the crystal structure of the IdeS peptidase needs to be 
solved to delineate the relationship of the enzyme to other cysteine peptidases. 

Based on the present cumulative knowledge, it is apparent that the IdeS 
peptidase evolved to a primary virulence factor. It is also a good example of the 
possibility that bacteria may contain more peptidases than predicted from 
sequence alignments. 

Clan CA 

All clan CA peptidases have a common fold motif, consisting of an amino- 
terminal domain that is mostly a-helical and a carboxy-terminal domain fea- 
turing an antiparallel |3-sheet, with the Cys and His catalytic residues forming 
a thiolate-imidazolium dyad. However, it is also the most divergent and popu- 
lous clan of the cysteine peptidases. The clan is divided into 12 families, of 
which bacterial peptidases are found only in 6. Two of these families encom- 
pass exclusively bacterial enzymes that have apparently evolved as important 
virulence factors. 

Family CI: The Papain Family 

It is an evolutionary paradox that this major family of cysteine peptidases, 
exemplified by papain and manmialian cysteine cathepsins and encompassing 
more than 720 sequences, has only few representatives in bacteria. All together, 



Bacterial Peptidases 



41 



only 47 homologues of papain have been identified, including 22 bacterial 
species with a completely sequenced bacterial genome. In this context, it is inter- 
esting to note that two Mycoplasma species, M. gallisepticum and M. penetrans, 
carry three and two copies of a gene encoding a potentially active papain homo- 
logue, respectively. However, among the genus Mycoplasma, these two species 
are the richest with regard to their peptidase gene count. 

Papain homologues occur predominantly in gram-positive species, the major 
representative being aminopeptidase C. This enzyme from Lactococci spp. has 
been thoroughly characterized [Vesanto et al., 1994; Fenster et al., 1997], and is 
also present in pathogens, but there are no reports that this peptidase or its homo- 
logues are involved in any aspect of bacterial pathogenicity. 

Family C2: The Calpain Family 

The protein fold of the peptidase unit for members of this family resem- 
bles that of papain. In mammals they are represented by calcium-regulated 
ubiquitous enzymes, but thus far only five highly diverged homologues have 
been identified in prokaryotes. The recombinant enzyme from Porphyromonas 
gingivalis, Tpr peptidase, was characterized as a general endopeptidase which 
also cleaves the bacterial collagenase peptide substrate. However, the enzyme 
has no collagenolytic activity [Bourgeau et al., 1992] and there is no indi- 
cation that the Tpr peptidase is associated with the virulence of this major 
periodontopathogen. 

Family CIO: The Streptopain (SpeB) Family 

The streptococcal cysteine peptidase was isolated and characterized in 
1945 and was the second proteolytic enzyme after clostripain to be isolated 
fi"om a prokaryote [Elliott, 1945]. For some time the identity of the peptidase 
was mistaken for the streptococcal pyrogenic toxin termed SpeB (streptococcus 
pyrogenic exotoxin B). The confusion ended when the entire genomes of 
several strains of GAS were sequenced, showing that SpeB and streptopain are 
the same protein. For historical reasons, however, streptopain is still very often 
referred to as SpeB. The enzyme occurs in two variants, which differ only 
in a single amino acid residue, glycine or serine, at position 164 from the amino- 
terminus of the mature enzyme. Most strains of 5. pyogenes that are associated 
with severe invasive diseases express a Gly variant and therefore present an 
integrin-binding Arg-Gly-Asp motif at the surface-exposed loop. It was suggested 
that the ability of streptopain to bind integrins may be linked to the pathogenicity 
of these strains [Stockbauer et al., 1999]. 

Despite a lack of significant sequence similarity, the crystal structure 
clearly indicates that streptopain belongs to the papain clan (superfamily) of 
cysteine peptidases. The mature peptidase portion has the two-domain fold 



Potempa/Pike 



142 



characteristic of other papain-like enzymes, with an amino- terminal domain 
composed largely of a-helices and a carboxy-terminal domain based on a four- 
stranded antiparallel (3-sheet, with the catalytic dyad in the same topological 
orientation as in actinidtn, a close relative to papain. In contrast to the peptidase 
domain, the profragment of streptopain has a unique fold. While an extended 
strand of the prosegment runs the full length of the active site cleft in a direc- 
tion opposite to that of a natural substrate, thus blocking the major specificity 
pocket in the papain-like peptidase, in prostreptopain the inactivation mecha- 
nism relies on displacement of the catalytically essential histidine residue by a 
loop inserted into the active site [Kagawa et a!., 2000]. 

For more than 50 years, streptopain was recognized as a unique cysteine 
peptidase unrelated to papain or any other known peptidase. The first homo- 
logue of streptopain was identified in P. gingivalis^ a bacterium involved in the 
pathogenesis of human periodontal disease [Madden et al., 1995], then another 
one from the same microorganism was purified and characterized [Nelson et al., 
1999]. This peptidase, referred to as periodontain, shows a strong preference 
for the degradation of unfolded polypeptide chains, with the human plasma 
proteinase inhibitor, apantitrypsin, being an important exception. This major 
inhibitor of human neutrophil elastase is very efficiently inactivated by cleavage 
in the reactive site loop [Nelson et al., 1 998]. Locally, this may lead to a loss of 
control of neutrophil peptidases and contribute to connective tissue damage. On 
the other hand, any direct role of periodontain in P. gingivalis pathogenicity is 
obscure. The enzyme, together with its homologue, is probably involved in gen- 
erating nutrients in the form of short peptides which are an indispensable source 
of carbon and energy for this asaccharolytic microorganism. 

The MEROPS database lists only three streptopain homologues, two in 
P. gingivalis and one in the genome of Baclewides thetaiotaomicron. However, 
closer analysis of partially finished bacterial genome sequences revealed that 
genes encoding potentially active streptopain-like peptidases are more widely 
spread. Three different homologues were found in the genome of Prevotella 
intermedia, two in Prevotella ruminicola, and one in each of Tannerella 
forsythensis and Bacteroides fragilis. These genes encode either secreted or 
intracellular proteins. Significantly, the potentially secreted enzymes carry 
profragments with significant similarity to the proregion of streptopain. in the 
context of streptopain, which is very likely to be a virulence factor, it would be 
very interesting to elucidate the role of these streptopain homologues from other 
bacterial species. 

Streptopain is an outstanding example of a primary virulence factor with a 
very broad spectrum of activity. The list of pathogenetically relevant, biologi- 
cally important proteins processed, activated, or otherwise altered by the enzyme 
is impressive. In vitro, streptopain cleaves the human interleukin-l|3 (IL-I|3) 



Bacterial Peptidases 



43 



precursor to form bioactive IL-1 (3 [Kapur et al., 1 993a], processes the monocytic 
cell urokinase receptor [Wolf et al., 1994] and degrades human fibronectin and 
vitronectin, two abundant extracellular matrix proteins engaged in maintaining 
host tissue integrity [Kapui" et al., 1993b]. In addition, streptopain activates latent 
human matrix metal) opeptidases (MMPs), a process hypothesized to participate 
in the extensive soft tissue destruction observed in some patients with invasive 
streptococcal disease [Bums et al., 1996]. 

Streptopain is able to cleave IgG molecules at the hinge region of the 
7-chain, generating two Fab fragments and one Fc fragment [Collin and Olsen, 
2000]. Interestingly, although streptopain can also cleave antigen-bound IgG, it 
does not affect antibodies bound to the bacterial surface through the Fc region 
[Eriksson and Norgren, 2003]. In this way, streptopain's ability to cleave off the 
Fc part of antigen-bound IgG contributes to the ability of GAS strains to escape 
opsonophagocytosis, while not interfering with the formation of a host-like coat 
of IgG immobilized on the bacterial surface through the Fc portion. This mech- 
anism may significantly remforce the defenses of 5". pyogenes against attack by 
the adaptive immune response. In addition to streptopain, this deterrence system 
consists of (1) eel I- wall-anchored surface proteins of the so-called M protein 
family, which binds IgG 'upside down' through the Fc fragment [Berge et al., 
1997]; (2) a secreted, highly specific endoglycosidase (EndoS) that targets con- 
served N-linked oligosaccharides on IgG [Collin and Olsen, 2000], and (3) the 
uniquely IgG-specific endopeptidase, IdeS (see family C66). Taken together, this 
system is very effective hi protectmg S. pyogenes against opsonin-dependent 
uptake and killmg by professional phagocytes [Collin et al., 2002]. 

Streptopain also seems to play a key role in shielding S. pyogenes from the 
innate immune system. The enzyme induces release of dermatan sulfate from 
the extracellular matrix resulting in the inactivation of antibacterial peptides 
[Schmidtchen et al., 2001] or directly eliminates the bactericidal potential of 
these peptides by degrading them [Schmidtchen et al., 2002], Finally, and pos- 
sibly the most important role of streptopain in the pathogenicity ofS. pyogenes 
is the ability of streptopain to directly release the potent peptide hormone, 
bradykinin, from high-molecular- weight kininogen. This release is not under 
the control of the host system [Hei'wald et al., 1996]. Bradykinin released 
by bacterial pathogens has been shown to contribute to the dissemination of 
infection [Sakata et al., 1996] and symptoms of sepsis and septic shock 
[Herwald et al., 1 998, 2003; Tapper and Herwald, 2000]. Studies conducted with 
animal models confumed the significant pathogenic potential of streptopain. 
The purified enzyme is lethal to mice [Geriach et al., 1983] and can cause 
myocardial necrosis when injected into rabbits, apparently due to its fibrinolytic 
activity [Kellner and Robertson, 1954]. Moreover, active immunization of mice 
with the purified streptopain elicits a protective response in a model of invasive 



Potempa/Pike 



144 



disease, while mice injected with lethal doses of S. pyogenes were cured 
by a single injection of streptopain-specific inhibitor [Bjorck et al., 1989]. 
Furthermore, experiments using a rat model of lung infection show that strep- 
topain acts synergistically with either the streptococcal cell wall antigen or 
streptolysin O to augment lung injury [Shanley et al., 1996], This observation 
is especially intriguing in the context of the recent discovery that streptolysin O 
is the functional equivalent of the type III secretion system in gram-positive 
bacteria [Madden et al., 2001] and invites specuJation that in some circum- 
stances streptopain may enter the host cell and act as an intracellular viiulence 
factor. 

Taking into account the results of in vitro and ex vivo experiments, it is 
somewhat perplexing that the importance of streptopain as an indispensable vir- 
ulence factor in vivo is still questioned. In one study, the importance of strep- 
topain for the virulence of S. pyogenes has been demonstrated in a mouse 
model using isogenic strains with the streptopain gene inactivated by genetic 
manipulation [Lukomski et al., 1997]. In the follow-up in vivo investigation, it 
was shown that streptopain helps S. pyogenes to resist phagocytosis [Lukomski 
et al., 1998], contributes to soft tissue pathology, including necrosis, and is 
required for efficient systemic dissemination of the organism from the initial 
site of skin inoculation [Lukomski et al., 1999]. In stark contrast, in a well- 
designed and executed study, Ashbaugh and Wessels [2001] proved that genetic 
inactivation of the streptopain gene did not significantly attenuate murine inva- 
sive infection, either after intraperitoneal or subcutaneous challenge. Also, in a 
model of necrotizing fasciitis, a streptopain mutant organism was found to be 
as effective in causing tissue damage, as the wild-type control strain [Ashbaugh 
et al., J 998]. These results are in keeping with the clinical observation of an 
inverse correlation between disease severity and streptopain production in vitro 
by genetically related MlTl GAS isolates associated with invasive infection 
[Kansal et al., 2000]. This paradox may be explained, at least partially, by the 
ability of streptopain to proteolytically remodel S. pyogenes surface proteins. 
Although this process is considered advantageous for bacteria [Rasmussen 
and Bjorck, 2002], two studies have suggested that the overexpression of strep- 
topain results in nonspecific degradation of the antiphagocytic protein M and 
solubilizingofthe C5a peptidase [Bergeand Bjorck, 1995; Raederetal., 1998]. 
Together with degradation of secreted key virulence factors, such as superanti- 
gens (streptococcal pyrogenic exotoxins) [Kansal et al., 2003], excessive pro- 
duction of streptopain may therefore decrease the pathogenicity ofS. pyogenes. 
This hypothesis is further corroborated by the observation that streptopain- 
negative isolates have a survival advantage in vivo [Reader et al., 2000] and the 
recent discovery that invasive MlTl GAS undergoes a stable phase shift to a 
phenotype expressing no streptopain, but instead a full repertoire of secreted 



Bacterial Peptidases 145 



proteins, which are apparently degraded by active streptopain [Aziz et al., 
2004]. This phenotypic phase shift may be related to the marked resurgence of 
severe, invasive and potentially fatal GAS infection, including the necrotizing 
fasciitis and streptococcal toxic syndrome observed during the last 20 years. 

The role of streptopain in GAS virulence confirms the ancient maxim that 
even for a bacterial pathogen too much of a 'good thing' can be bad. Indeed, 
S. pyogenes has developed its own system to regulate proteolytic activity and 
protect its surface-associated array of key virulence factors. Firstly, expression 
of streptopain is regulated at the transcriptional level [Heath et a!., 1999]; 
secondly, streptopain is produced as an inactive zymogen, which undergoes an 
autocatalytic, multistep activation process assisted by the bacterial surface [Liu 
and Elliott, 1965a, b; Collin and Olsen, 2000; Chen et al., 2003], and thirdly, 
in vivo, the pathogen can coat its surface with the broad spectrum peptidase 
inhibitor, a2"r"^croglobulin (a2M) immobilized through interaction with the 
peptidoglycan-anchored protein, G-related a,2M-binding protein (GRAB). 
Bound to GRAB, a2M protects protein M, and possibly other surface proteins, 
from being cleaved by streptopain [Rasmussen et al., 1999]. In this context, it 
is very interesting to note that S. pyogenes retains some of the streptopain 
enzyme displays associated with the bacterial cell surface, where the enzyme 
displays laminin-binding activity [Hytonen et al., 2001], Taking into account 
the mechanism of peptidase inhibition by a2M, it is tempting to speculate that 
the immobilized form of streptopain preserves proteolytic activity even in the 
presence of a high concentration of this inhibitor. Such a feature may be par- 
ticularly useful in soft tissue infections where the experimental and epidemio- 
logical evidence strongly implies that streptopain plays a critical role in 
promoting infection [Svensson et al., 2000]. 

Family C47: The Staphopain Family 

At present, this family is limited to the Staphylococcus genus. Staphopain 
occurs in two variants, apparently reflecting the duplication of an ancestral 
gene. S. aureus expresses both variants, referred to as staphopain A and 
staphopain B, which share about 47% identity at the amino acid sequence level 
of the mature enzymes. The single staphopain o^ Staphylococcus epidermidis is 
related to staphopain A (75% identity) [Dubin et al., 2001; Oleksy et al., 2004]. 
On the other hand, a gene encoding a close relative of staphopain B has been 
cloned from Staphylococcus warneri, while a cysteine peptidase similar to the 
staphopains was purified fi^om the growth medium o? Staphylococcus simulans 
biowsLT s tap hylo lytic us [Donham et al., 1988; Neumann et al., 1993]. 

Both staphopains are processed from large precursors, but so far only 
the crystal structure of the mature staphopains is available [Hofmann et al., 
1 993; Filipek et al., 2003]. Remarkably, despite the low sequence similarity to 



Potempa/Pike 



146 



papain-like peptidases, the tertiary structure of the staphopains resembles the 
overall fold of papain. 

The reciprocal relationship present between the staphopains apparent at the 
amino acid sequence level is also mirrored at the genetic level. The staphopain 
A gene (scpA) occurs in a bicistronic operon (scpA), in which it is followed by a 
gene (scpB) encoding a staphopain A-specific inhibitor. On the other hand, the 
staphopain B gene (sspB) is part of the tricistronic operon sspABC, where sspA 
and sspC encode the V8 protease and an inhibitor specific for staphopain 
B, respectively [Rzychon et al., 2003a, b]. The staphopain inhibitors, ScpB 
and SspC, termed staphostatins, have similar folds and apparently the same mech- 
anism of target peptidase inhibition although they share less than 20% sequence 
identity [Rzychon et al., 2003a, b, Dubin et al., 2003]. Nevertheless, they are 
uniquely specific; ScpB affects only staphopain A activity, while SspC exclusively 
inhibits staphopain B, without any cross-reactivity. In some cases, the reactivity of 
the inhibitor does not extend to the orthologous enzyme from other staphylococcal 
species [Dubin et al., 2004]. Apparently, evolution has hand-tailored these 
inhibitors to control the activity of the coexpressed enzyme. Interestingly, 
staphopains are secreted, while staphostatins are intracellular proteins, suggesting 
that they function as so-called threshold inhibitors protecting cytoplasmic proteins 
from any prematurely folded peptidases [Rzychon et al., 2003a, b]. The genetic 
assembly of peptidase and inhibitor genes in cotranscribed, cotranslated units 
provides the means for very efficient elimination of active staphopain fi^om the 
cytoplasm. 

The extracellular activity of S. aureus is also the subject of multilevel 
control. All secreted peptidases, including both staphopains are coordinately 
regulated at the transcriptional level by an accessory gene regulator operon 
(agr) in a cell density-dependent manner [Janzon et al., 1989]. This regulation 
is fine tuned by direct, strong repression of the transcription of the stpAB and 
sspABC operons by SarA, the product of the staphylococcal accessory regula- 
tor (sar) locus [Chan and Foster, 1998; Lindsay and Foster, 1999; Ziebandt 
et al., 2001]. Additionally, this regulatory system is indirectly affected by the 
alternative sigma factor c^ [Ziebandt et al., 2001] and probably by several 
SarA-like transcriptional factors. Collectively, this highly complex network of 
gene regulation assures the precisely coordinated synthesis of extracellular 
proteins, including staphopains and other peptidases. 

In the case of the proteinases, the regulation of their activity does not stop at 
the transcriptional level. Aureolysin, the V8 peptidase (glutamylendopeptidase I) 
and the staphopams are secreted as proenzyme forms and activated in a cascade- 
like manner. It is well documented that aureolysin activates the zymogen of the 
V8 peptidase, which in turn cleaves pro-staphopain B [Drapeau, 1978; Rice at al., 
2001]. Indeed, pro-staphopain B can be expressed in the zymogen form in E. coU 



Bacterial Peptidases 147 



and activated in vitro by the V8 peptidase (J. Potempa, unpubl. data). In contrast, 
the means by which pro-staphopain A processing/activation occurs is obscure and 
nothing is known as to whether this pro-enzyme is inactive or which proteinase 
is responsible for its processing. 

Tight regulation of staphopain expression, together with that of other 
acknowledged virulence factors, including toxins and adhesins, may be con- 
sidered as indirect evidence of their importance for the sui'vival of S. aureus 
in vivo. This association has revitalized interest in staphylococcal extracellular 
peptidases as markers of pathogenicity, a subject which has been neglected for 
many years. Unfortunately, the results of recent investigations using animal 
models of staphylococcal infection are contradictory and confusing. Firstly, it 
was shown that a mutant strain deficient in the V8 peptidase was severely atten- 
uated in virulence in mouse abscess, bacteremia and wound infection models 
[Coulter etal., 1998]. However, the reduced virulence ofthis mutant was appar- 
ently due to a polar effect on the expression of the sspB gene encoding 
staphopain B, located downstream of the V8 peptidase gene (sspA) in the same 
operon [Rice et al., 2001]. Indeed, this assumption was confirmed using a 
S. aureus strain with the staphopain B gene eliminated by means of genetic 
manipulation [Shaw et al., 2004]. In this study it was shown that only the sspB 
gene knockout strain, but not the metalloproteinase (aureolysin) and staphopain 
A-deficienr mutants were attenuated in the skin abscess model. However, these 
results were not confirmed in a model of septic arthritis in mice. The inactiva- 
tion of any of the peptidase genes did not affect the frequency or severity of 
joint disease, indicating that, at least in this model, staphopain B does not act 
as virulence factor [Calander et al., 2004]. 

Taken together, the role of staphopains in the physiology and virulence of 
staphylococci is obscure, but stringent conservation of the stpA and sspB genes 
among S. aureus strains, as well as preservation of the stpA-Wke gene among 
coagulase-negative staphylococcal species, implies that their function is impor- 
tant for staphylococcus survival in vivo. Amongst the bacterial proteinases, 
staphopains are unique with regard to their secretion as zymogens and activa- 
tion by Imiited proteolysis. In this respect they resemble sti'eptopain from 
5*. pyogenes. In addition, for an as yet not understood reason they are tightly 
regulated both at the transcriptional and posttranslational levels. At the protein 
level their activity is released in a cascade pathway unique among bacterial 
species and then is further controlled by highly specific inhibitors. 

Family C39: Bacteriocin-Processing Peptidase 

Bacteriocins are antimicrobial peptides produced by microorganisms 
belonging to different bacterial taxonomic branches and used by microorganisms 
for biological warfare and communications [Eijsink et al., 2002]. One type of 



Potempa/Pike 



148 



these peptides is posttranslationaily modified (class I lantibiotics), while a sec- 
ond type does not contain modified amino acids (class 11 nonlantibiotic bacterio- 
cins). Both classes are ribosomally synthesized in the precursor form. In most 
nonlantibiotic peptides and some lantibiotic peptides, the amino-terminal exten- 
sions are composed of a very characteristic leader sequence termed the double- 
glycine-type leader, which is cleaved after the second glycine, concomitant with 
export carried out by members of a specific family of dedicated ATP-binding 
cassette (ABC) transporters. The amino-terminal domain of these transporters, 
absent in other ABC transporters, contains conserved cysteine and histidine 
residues operating as the catalytic dyad. Also, other residues, including the glu- 
tamate and aspartate residues which participate in peptide bond hydrolysis by 
papain-like peptidases, are strictly conserved in this portion of the molecule, 
which apparently has a canonical fold characteristic of papain [Havarstein et al.^ 
1995]. The peptidase domain, together with a central hydrophobic integral mem- 
brane domain and a carboxy-terminal cytoplasmic ATP-binding domain, consti- 
tutes the dedicated transport machinery which recognizes substrates and removes 
leader peptides while translocating them across the cytoplasmic membrane. In 
addition to bacteriocins, the ABC transporters are used to translocate peptide 
pheromones [N4ichiels et al., 2001]. 

Bacteriocin-processing peptidases are widespread amongst both gram- 
positive and gram-negative bacteria and constitute the second most numerous 
family of cysteine peptidases in prokaryotes (after family C40). None has been 
implicated as a virulence factor. On the contrary, as peptidases which are indis- 
pensable for the maturation of bacteriocins, they can be utilized in expanding 
applications using bacteriocins as natural food preservatives [Riley and Wertz, 
2002]. 

Family C51: D-Alanyl-Glycyl Endopeptidase 

Representatives of this family have thus far only been found in the three bac- 
terial species, S. aureus, S. epidermidis, and S. pyogenes. The enzymes are phage- 
derived and can degrade the cell wall envelope. Autolysins LytN and LytA from 
S. aureus possess a D-alanyl-glycyl endopeptidase as well as N-acetyhnuramyl- 
L-alanyl amidase activity, which is contained within the amino-terminal portion 
of the polypeptide chain [Navarre et al., 1 999]. None of these autolysins has been 
implicated in virulence. Conversely, it has been suggested that they may be used 
to counter antibiotic-resistant staphylococcal infections [Fischetti, 2003]. 

Family C58: The YopT Peptidase Family 

Bacterial pathogens share common strategies to infect and colonize animal 
and plant host [Staskawicz et al., 2001]. One system, widespread among gram- 
negative pathogens, referred to as the type 111 secretion system [Cheng and 



Bacterial Peptidases 149 



Schneewind, 2000; Cornells and Van Gijsegem, 2000] directly delivers different 
classes of proteins to the host. These proteins, now collectively termed type IIJ 
effectors, mimic, suppress, interfere^, or modulate host defense signaling path- 
ways. Their sole function is to enhance pathogen survival, proliferation and dis- 
semination and therefore may be considered to be primary virulence factors. 
The structural scaffold to dispense type 111 effectors is conserved but 'delivered 
goods' are custom designed to serve the particular needs of a given pathogen. 
This is exemplified by the YopT peptidase [Cornelis, 2002] and its homologues 
from Yersinia spp. and plant pathogens, including Pseudomonas syringae 
[Axtell et al., 2003], which, despite sharing the same fold and catalytic mecha- 
nism, target a different set of substrates inside host cells. In addition to the YopT 
peptidase onthologues, an overlapping set of pathogens has adopted a cysteine 
peptidase with a different fold and evolutionai^y origui (clan CE) [Orth^ 2002] 
as the type III effectors. 

The YopT peptidase is one of six proteins called Yop effectors (YopH, 
YopE, YopJ/YopP, YopO/YpkA, YopM, and YopT) injected into the host cell by 
the Yersinia type III secretion system [Juris et al., 2002]. They function in con- 
cert to thwart the host immune system. YopT itself exerts a cytotoxic effect in 
mammalian cells when delivered by the type 111 secretion system [Iriarte and 
Cornelis, 1 998]. This effect is due to proteolytic cleavage of posttranslationally 
modified Rho GTPases by the YopT peptidase [Shao et al., 2002]. Apparently 
the YopT peptidase specifically recognizes prenylated Rho GTPases and exe- 
cutes a proteolytic cleavage near their carboxy-termini [Shao et al., 2003b]. 
This leads to the loss of the carboxy-terminal lipid modification on these 
GTPases, resulting in their release from the membrane and irreversible inacti- 
vation. Globally, this causes a disruption of the actin cytoskeleton, exerting a 
powerfiil antiphagocytic effect and thus protectuig the pathogen from being 
killed by phagocytes. 

AvrPphB is an avirulence (Avr) protein from the plant pathogen P. syringae 
that can trigger a disease resistance response in a number of host plants. The 
crystal structure revealed that the topology of the catalytic triad (Cys-His-Asp), 
together with other structural features, resembles that for papain-like pepti- 
dases, particularly staphopain [Zhu et al., 2004]. AvrPphB has a very stringent 
substrate specificity and apparently exerts only a single proteolytic cleavage in 
the Arabidopsis serine/threonine kinase PBSl [Shao et al., 2003a]. It is sug- 
gested that the cleavage product is recognized by RPS5, a member of the class 
of R proteins that have a predicted nucleotide-binding site and leucine-rich 
repeats. In a resistant host these molecular events induce a hypersensitive 
response. 

The avr genes of the YopT family are common amongst plant pathogens as 
well as symbiotic plant bacteria and multiple Avr proteins are found in a single 



Potempa/Pike 



150 



Pseudomonas strain. They all function as specific peptidases targeting different 
substrates in the plant host or possibly cleaving the same substrates at different 
positions, generating signals detected by distinct R proteins. It is speculated that 
the large number of YopT-like proteins found in plant pathogens may reflect 
coevolutionary pressures in which the evolution of a new R protein in the host 
that detects the cleavage products of a given peptidase selects for a pathogen 
with new protease variants [Axtell and Staskawicz, 2003; Zhu et al., 2004]. 

Clan CD 

This clan was recognized based on a conserved sequential motive His-Gly- 
spacer-Ala-Cys encompassing the catalytic His-Cys dyad present in caspases, 
peptidases involved in apoptosis and cytokine activation (family 14), gingipains 
(family 25), plant and animal legumains, processing proteinases (family 13), 
bacterial clostripain (family 1 1), and separase, a proteinase required for sister 
chromatid separation during anaphase (family 50) [Chenetal., 1998]. The addi- 
tional common feature of all these enzymes is a substrate specificity dominated 
by a specific PI residue recognition, which is asparagine (legumain), lysine 
(Kgp), arginine (Rgp, clostripain, and separase), or aspartic acid (caspases). 
Although crystal structures are only available for caspases and one gingipain, it 
is expected that representatives of other families in the clan will aJso have a 
similar fold. The hallmark of this fold is a six-stranded parallel (i-sheet in the 
middle of the molecule sandwiched by three a-helices on each side [Eichinger 
et al., 1999]. Out of the five CD clan families known so far, three are found in 
bacteria. 

Family CI I: The Clostripain Family 

Clostripain was identified and partially purified in 1937 from the culture 
filtrate of Clostridium histolyticum. The enzyme was then characterized as a 
cysteine peptidase that is strictly specific for Arg-Xaa (Xaa stands for any 
amino acid) peptidyl bonds. The mature, active clostripain is a noncovalent het- 
erodimer derived from an inactive precursor through the autocatalytic removal 
of a 9-residue linker peptide [Witle el al., 1996, 1994]. Al least 16 closLripaiii 
onthologues homologues were identified in microbial genomes, most of them 
in Clostridium spp. [Labrou and Rigden, 2004]. None of them was ever impli- 
cated as a virulence factor in clostridial infections. On the contrary, clostripain 
is a very useful enzyme in technology, both in sequence analysis and in enzy- 
matic peptide synthesis [Gunther et al., 2000]. 

Family CIS: The Legumain Family 

Mammalian asparaginyl endopeptidase (AEP) or legumain is a recently 
identified lysosomal cysteine peptidase belonging to clan CD. To date it has been 



Bacterial Peptidases 



51 



shown to be involved in antigen presentation within main-histocompatibility- 
complex (MHC) class 11-positive cells and in proprotein processing [Shirahama- 
Noda et al., 2003; Manoury et al., 1998; Sarandeses et al., 2003]. Genes 
encoding potentially active legumain homologues have thus far only been found 
in a few bacterial species, including Caulobacler crescentus, P. aeruginosa, 
Pseudomonas putida, P. syringae, Xanthomonas axonopodis, and Xanthomonas 
campestris. Their function awaits elucidation. 

Family CI 4: The Caspase Family 

Caspases are important players in the programmed cell death of multi- 
cellular organisms ranging from humans to sponges [Wiens et a!., 2003]. 
Comparative genomic studies have provided evidence which indicates that the 
eukaryotic apoptotic system emerged by acquisition of several central apop- 
totic effectors, including caspases, from a-protobacteria as a consequence of 
mitochondrial endosymbiosis [Koonin and Aravind, 2002]. Therefore, it is not 
surprising that homologues of caspases, referred to as paracaspases and meta- 
caspases [Aravind and Koonin, 2002], are abundant in diverse bacteria, par- 
ticularly those with complex development, such as Streptomyces, Anabaena, 
Mesorhizobium, Myxococcus, and a-protobacteria. The role of these ancient 
enzymes in bacterial physiology is obscure. 

Family C25: The Gingipain Family 

So far gingipains have only been found in P. gingivalis, the major pathogen 
of adult onset periodontal disease. They are represented by the products of three 
genetic loci conserved amongst clinical and laboratoi7 strains of/? gingivalis, 
one (kgp) encoding a lysine-Xaa peptide bond-specific endopeptidase (gingi- 
pain K, Kgp) and two others, rgpA and rgpB, which are arginine-Xaa-specific 
enzymes (Arg-gingipains, Rgps) [Curtis et al., 1999; Potempa et al., 1995]. The 
nascent translation products of gingipain genes undergo complex proteolytic 
processing and posttranslational modifications [Veith etal., 2002]. In the case of 
Kgp and RgpA, initial polypeptide chain fragmentation is necessary for assem- 
bly of a noncovaJenl complex composed of the catalytic, hemoglobin-binding 
and hemagglutkiation/adhesin domains [Potempa et al., 2003]. This complex is 
either anchored to the outer membrane through a glucan moiety attached to the 
carboxy-terminus of the domain derived from the carboxy-terminal portion of 
the nascent product, or released into the growth media in the nonglycated form. 
RgpB lacks the additional hemoglobin-binding and adhesin domains, but still 
undergoes complex modification consisting of the autoproteo lytic removal of 
the profragment and either truncation at the carboxy-terminus (the secreted form 
of the enzyme) [Mikolajczyk et al., 2003] or glycosylation at the carboxy- 
terminus, the latter allowing RgpB to form an association with the cell envelope 



Potempa/Pike 



152 



[Veith etal., 2002]. Collectively, gingipain activity constitutes at least 85% of the 
general proteolytic activity produced by /? gingivalis [Potempa et al., 1997]. 

In every respect, gingipains can be considered to be primary virulence 
factors for P. gmg/vfl//5-dependent initiation and/or progression of periodontal 
disease. As peptidases, tJiey target a large set of disease-relevant substrates 
which can be directly associated with the clinical hallmarks of the disease 
[Potempa et aJ., 2000]. Due to the large number of substrates it targets, gingi- 
pain activity is also thought to provide this asaccharolytic organism with nutri- 
ents. However, gingipains are certainly broad spectrum peptidases. Actually, in 
many cases they act with the precision and sophistication of the tailored host 
peptidases, mimicking their function. The best example of how P. gingivalis can 
manipulate the host is the use of the gingipains to affect the major proteolytic 
cascades of coagulation, complement activation, fibrinolysis and kinin genera- 
tion [Imamura et al., 2003]. 

The coagulation cascade is targeted at several levels by Rgps, which 
convert factor X, factor IX, protein C and prothrombin to active peptidases by 
limited proteolysis, thus mimicking the action of host enzymes [Imamura et al., 
1997, 2001a, b; Hosotaki et al., 1999]. In the case of factor X activation, this 
functional mimicry additionally involves enhancement of the Rgp-converting 
activity in the presence of phospholipids and Ca^"^, two critical cofactors of 
the normal coagulation cascade [Imamura et al., 1997]. The factor X activation 
is very efficient, with the catalytic potency in some cases matching that of 
natural activators. In this context it is worth emphasizing that gingipains are not 
controlled by host inhibitors, in stark contrast to the clotting factors. In vivo, at 
periodontal disease sites, the procoagulant activity of Rgps is apparently 
negated by the fibrinogen degradation carried out by Kgp [Scott et al., 1993; 
Imamura et al., 1995a, b], which contributes to a bleeding tendency, a hallmark 
of the disease, which correlates positively with the presence of P. gingivalis at 
discrete periodontal pockets. Collectively, the interaction of gingipains with the 
coagulation cascade leads to local, uncontrolled release of thrombin, an enzyme 
with a multitude of diverse biological activities, including the stimulation of 
prostaglandin, IL-1 and pi ate let- activating factor release by endothelial cells 
and macrophages. These mediators are considered predominant factors in the 
tissue destruction process in periodontal disease. 

Another trademark of periodontitis is the increased flow of gingival fluid 
from periodontal pockets. This symptom can be directly associated with the 
unrivalled (compared to other bacterial proteases) ability of gingipains to 
release bradykinin. Physiologically, this potent mediator is released from high- 
molecular-weight kininogen by plasma kallikrein, which in turn is generated 
from prokallikrein by activated Hageman factor (factor Xlla). Rgps shortcut 
this cascade by activation of plasma prekaMikrein, with kinetics, which are 



Bacterial Peptidases 



53 



better than those observed in prekalHkrein activation by factor Xlla [Imamura 
et al., 1994]. In addition, Rgps working in concert with Kgp, can release 
bradykinjn directly from high-molecular-weight kininogen [Imamura et al., 
1995a, b]. Bradykinin exerts powerful biological activities and is responsible 
for pain and local extravasation at the site of infection/inflammation leading to 
edema, which underlies the mechanism of generation of gingival crevicular 
fluid. 

The main targets for gingipains amongst factors of the complement 
cascade seem to be the proteins C3 and C5, but the mode of action on these 
factors is different. While C3 is destroyed, thus disabling the bactericidal and 
opsonizing ability of activated complement, the functional chemoattractant, 
C5a, is released from C5 by the action of the gingipains [Wingrove et al., 1992; 
Discipio et al., 1996]. In addition, gingipains can enhance the chemotactic 
activity of IL-8 [Mikolajczyk-Pawlinska etal., 1998]. Cumulatively, this gingi- 
pain-mediated generation of potent chemoattractants may lead to excessive 
neutrophil accumulation at periodontal sites, another clinical sign of active 
disease. 

A large set of cell surface proteins and receptors, including the LPS recep- 
tor (CD14) [Sugawara et al., 2000; Tada et al., 2002], the C5a receptor (CD58) 
[Jagels et al., 1 996], the IL-6 receptor (TL-6R) [Oleksy et al., 2002], and ICAM-1 
[Tada et al., 2003] are targeted by the gingipains. Although the cleavage of 
these proteins may significantly contribute to P. gingivalis-'mdiXxcQd. pathologi- 
cal changes in the periodontium, activation of protease-activated receptors 
(PARs) desei'ves special emphasis. PARs mediate cellular responses to a vari- 
ety of extracellular serine peptidases [Ossovskaya and Bunnett, 2004]. The four 
known PARs constitute a subgroup of the family of seven-transmembrane 
domain G protein-coupled receptors and activate intracellular signaling path- 
ways typical for this family of receptors. Activation of PARs involves prote- 
olytic cleavage of the extracellular domain, resulting in formation of a new 
amuio-terminus, which acts as a tethered ligand. PAR-1 , PAR-3, and PAR-4 are 
relatively selective for activation by thrombin whereas PAR-2 is activated by a 
variety of proteases, including trypsin and tryptase [Gabazza et al., 2004^. Rgps 
specifically activate intracellular signaling pathways through cleavage of PAR-2 
on neutrophils [Lourbakos et al., 1998], PAR-1 and PAR-4 on platelets 
[Lourbakos et al., 2001 b], and PAR-1 and PAR-2 on human oral epithelial cells 
[Lourbakos et al., 2001a] with efficiency matching that for the endogenous 
agonists. Collectively, hijacking of the PAR-dependent signaling pathways 
illustrates the ability of the gingipains to carry out functional mimicry, which 
contributes to potentiation of local inflammatory responses and can be directly 
linked to bone resorption, the most profound clinical sign of advanced peri- 
odontal disease. 



Potempa/Pike 



154 



The list of proteins cleaved by gingipains discussed above is far from com- 
plete. A more complete set includes P. gingivalis extracellular proteins, as well 
as many other host proteins, such as hemoglobin and heme/iron-binding pro- 
teins, cytokines, bactericidal peptides, host peptidase inhibitors, proteins of the 
extracellular matrix, latent matrix metalloproteinases, and epithelial junctional 
proteins. The significance of these protein cleavages for periodontal disease 
pathogenicity is often speculative, but there is no doubt that gingipains can^ 
out an extremely diverse set of interactions with the host. Consistently, strains 
with the gingipain genes disabled by genetic manipulation have severely 
decreased virulence [O'Brien-Simpson et al., 2001] and the pathogeneicity of 
P. gingivalis can be supressed in vivo by gingipain-specific inhibitors [Curtis 
et al., 2002]. Finally, immunization with the gingipains as antigens has protec- 
tive effects, as observed in animal models of P. gingivalis infection [Gibson and 
Genco, 2001; Gibson et al., 2004; Rajapakse et aJ., 2002]. 

Clan CE 

This clan contains five families recognized thus far, three are found exclu- 
sively in viruses, one is unique for bacteria (family C55) and one is widespread 
among cellular organisms, except the archae (family C48). The archetypal 
enzyme of clan CE is the cysteine peptidase from adenovirus, adenain. Although 
adenain has a unique scaffold not seen in cysteine peptidases outside clan CE, 
the active site contains a Cys-His-Glu triplet and an oxyanion hole in an 
arrangement similar to that in papain [McGrath et al., 2003; Ding et al., 1996]. 
In this respect, the CE clan peptidases represent a powerful example of conver- 
gent evolution at the molecular level. 

Family C48: The Ulpl Endopeptidase Family 

In eukaryotic cells, the modification of proteins by a small ubiquitin-like 
modifier (SUMO) plays an important role in the function, compartmentaliza- 
tion, and stability of target proteins, contributing to the regulation of diverse 
processes [Muller et al., 2004; Melchior et al., 2003]. The covalent modifica- 
tion of proteins by SUMO-l is reversible and is mediated by SUMO-specific 
proteases. These proteases are ubiquitous in eukaryota and are thought to have 
a dual function. They are responsible firstly for the initial processing of SUMO- 1 
by cleavage of the precursor peptide at the carboxyl-terminus of the protein, and 
secondly for the subsequent processing and cleavage of high molecular weight 
SUMO-1 conjugates, releasing SUMO-1 and reducing the conjugation status of 
the target proteins. Homologues of these peptidases have thus far only been 
found in a few gram-negative bacteria, including Bradyrhizobium japonicum, 
Chlamydia muridarum. Chlamydia trachomatis, Mesorhizobium loti, P. syringae 
and X. campestiis. In the genomes of these organisms, representing animal and 



Bacterial Peptidases 155 



plant pathogens and plant symbionts, up to 3 genes encoding potentially func- 
tional SUMO-specific peptidases are present, but their role in symbiosis or vir- 
ulence has not been established. However, taking into account the importance 
of SUMO conjugation for the functioning of eukaryotic cells [Yeh et al., 2000], 
it is tempting to speculate that bacterial homologues of SUMO-specific pepti- 
dases are also active inside the host cell, subverting its function to benefit the 
pathogen, as in the case of the YopJ peptidases described below. 

Family C55: The YopJ Peptidase Family 

It is fascinating to note that amongst the type 111 secretion effectors, human 
and plant pathogens, as well as plant symbionts, have evolved two conserved 
families of cysteine peptidases with completely different folds. Both families 
mimic the proteolytic activity of eukaryotic proteins that are essential for the 
normal maintenance of host signaling. Members of the YopT family discussed 
earlier have a typical papain-like fold which has been crafted by pathogen evo- 
lution to yield a new, specific role in bacterial pathogenicity. The YopJ family 
described here apparently evolved using the scaffold of SUMO-specific pepti- 
dases (see above). Regardless of their differences in structure and specificity, 
both groups of enzymes target a limited number of intracellular substrates, 
specific cleavage of which subdues the host reaction to benefit the invading 
pathogen. 

YopJ, one of the effector molecules injected into the host cell by Y. pestis 
was the first protein m this family recognized as a peptidase, based on a com- 
parison of the predicted secondary structure of YopJ to that of the known struc- 
ture of the adenovirus cysteine peptidase, which revealed significant similarity 
between these two proteins [Orth et al., 2000]. Indeed, the intact catalytic dyad 
of Cys-His is absolutely necessary for YopJ to exert biological activity in 
the host eukaryotic cell. Also, the ability of the YopJ homologue, AvrBsT (the 
effector molecule secreted via the type HI pathway by X. campestris pathovar 
campestris), to trigger the hypersensitive response in plants, was shown to be 
dependent on the proteolytic activity of AvrBsT. In the case of YopJ, the activ- 
ity was exerted by cleaving SUMO- 1 -conjugated proteins. Now, it has become 
clear that plant homologues of YopJ are also cysteine peptidases with SUMO sub- 
strate specificity, since it was shown that XopD, an X. campestris pathovar vesi- 
catoria type III effector injected into plant cells, translocated to subnuclear foci 
and hydrolyzed SUMO-conjugated proteins in vivo [Hotson et al., 2003]. This 
indicates that SUMO protein deconjugation is a common strategy utilized by 
animal and plant pathogens to alter signal transduction. The SUMO-dependant 
pathway of intracellular signaling is very ancient and evolutionarily conserved 
in eukaryotic cells. So is its sensitivity to proteolytic interference by YopJ, 
which cleaves SUMO-conjugated proteins in yeast, resulting in a blockage of 



Potempa/Pike 



156 



the mitogen-activated protein kinase (MAPK) kinase-dependent pathway of 
signaling [Yoon et al., 2003]. The cleavage of SUMO conjugates in mammahan 
cells by Yersinia YopJ peptidase also blocks MAPK kinase [Collier-Hyams 
et al., 2002] paralyzing both the innate and adaptive immune responses. There 
are, however, some differences between the function of different YopJ pepti- 
dases, which apparently reflects adaptation to the specific lifestyle of a given 
pathogen. An AvrA protein from common, mild enteropathogen of humans, 
S. enterica serovar typhimurium, although 86% similar in amino acid sequence 
to YopJ, only inhibits NF-kB signaling and augments apoptosis in human 
epithelial cells, giving rise to speculation that AvrA may limit virulence in 
vertebrates in a manner analogous to the avirulence factors in plant [Collier- 
Hyams et al., 2002]. The lack of an avrA allele in strains of Salmonella typhi 
and Salmonella paratyphi [Prager et al., 2000], which evade epithelial defenses 
and results in severe systemic diseases seems to support this hypothesis. 

In summary, in the case of animal pathogens, SUMO protein deconjugation 
interferes with the innate Lmmune response by blocking cytokine production and 
inducing apoptosis in the infected cells. The infected host cell cannot respond 
to invaders because YopJ-like peptidases disrupt an essential posttranslational 
modification that is required for activation of mammalian MAPK and NF-kB 
pathways [Orth, 2002]. 

Clan CF 

The crystal structures of two peptidases from this clan are known and they 
are clearly unique. As yet, only one family was distinguished (family CI 5). 

Family CI 5: The Pyroglutamy I- Peptidase I Family 

Pyroglutamyl-peptidases remove the amino terminal pyroglutamate (pGlu) 
residue from specific pyroglutamyl substrates [Cummins and O'Connor, 1998]. 
To date, three distinct forms of this enzyme have been identified, but only type 
1 pyroglutamyl-peptidase is a cysteme peptidase with a unique fold. The active 
enzyme is apparently a homotetramer [Odagaki et al., 1999]. Both in mammals 
aiid prokaryotes, it is located in the cytoplasm and displays a broad pyroglu- 
tamyl substrate specificity. Genes encoding pyroglutamyl-peptidase I occur in 
several, mostly gram-positive bacterial species, but there are no reports that this 
enzyme activity may be related to virulence. 



Metallopeptidases 

Metallopeptidases are hydrolases in which the nucleophilic attack on a 
peptide bond is carried out by a water molecule activated by a divalent metal 



Bacterial Peptidases 157 



cation, which is usually zinc, but examples where cobalt, manganese or nickel 
are used have been reported. The metal ion is usually immobilized by three 
amino acid ligands, His, Glu, or Asp. In addition to the metal ligands, at least 
one other residue is involved in catalytic hydrolysis of the peptide bond exercis- 
ing the functions of a general base in catalytic solvent polarization. In many 
cases this residue is a glutamate. 

At present the MEROPS database allocates metallopeptidases to 15 clans 
recognized by the type and number of metal ions required for catalysis and, 
within these broad groups, by the sequential arrangement of the metal ligands 
and the catalytic residue. Within clans, separate families are distinguished based 
on structural similarities. The most divergent and densely populated clan is MA 
featuring the zincins, in which the water nucleophile is bound by a single zinc 
ion ligated to two His residues in a sequential motif of His-Glu-Xaa-Xaa-His^ in 
which Glu is the general base and Xaa stands for any amino acid. Depending on 
the third Zn ligand, which is either a Glu or His/Asp located downstream of the 
Zn-binding motif, clan MA is divided into two subclans, MA(E) and MA(M) 
[Gomis-Ruth, 2003], respectively. These subclans putatively represent separate 
evolutionary lines of metallopeptidases after a very ancient divergence within 
clan MA. Also, peptidases grouped into clan MM utilize the His-Glu-Xaa-Xaa- 
His motif and use an Asp residue to ligate zinc, but they are structurally unre- 
lated to clan MA enzymes. The other well-defined and characteristic sequential 
motifs involved in zinc chelation include His-Xaa-Xaa-Glu and His (clan MC), 
His-Xaa-Xaa-Glu-His and Glu (clan ME), His-Xaa-Glu-Xaa-His with the third 
ligand unidentified (clan MK), His-Xaa-Xaa-Xaa-Asp and His-Xaa-His (clan 
MO) and His-Ser-His-Pro-(Xaa9)-Asp (clan MP). 

In contrast to the limited occurrence of aspartic and cysteine peptidases 
amongst bacteria, metallopeptidases are widespread and they have representa- 
tives in 50 out of the 52 distinguished families of this class of enzymes. Even 
more interestingly, three metallopeptidases, including the FtsH protease [clan 
MA(E), family M41], methionyl aminopeptidase (clan MG, family M24), and 
homologues of sialoglycoprotease from Mannheimia (Pasteurella) haemolytica 
(Clan MK, family M22) are the only peptidases of any catalytic class which are 
absolutely conserved among bacterial species. Apparently, this trio features 
essential house-keeping enzymes and, therefore, a perfect target for the devel- 
opment of inhibitors, which, by blocking the activity of these peptidases, should 
arrest or kill most bacteria. Methionyl aminopeptidase I is an especially attrac- 
tive target since the reaction it catalyzes, i.e. removal of the formylated amino- 
terminal methionine residue fi^om newly synthesized polypeptide chains, is 
unique to bacteria. TTierefore, one would expect that specific inhibitor of the 
methionyl aminopeptidase should exert no side effects on eukaryotic organisms, 
thus resembling the action of classical antibiotics. Unfortunately, however, the 



Potempa/Pike 



158 



mammalian homologues of methionyl aminopeptidase are also susceptible to 
bacterial enzyme inhibitors. Collectively, the promise of effective new drugs and 
the obstacles with regard to cross-reactivity has fuelled intense interest in the 
detailed investigation of this family of peptidases, which are of known tertiary 
structure, have a characterized mechanism of catalysis and are subject to inhi- 
bition by an array of different compounds [Bradshaw et al., 1998; Bazan et al., 
1994; Douangamath et al., 2004; Oefner et al., 2003; Hu et al., 2004; Towbin 
et al., 2003; Copik et al., 2003; Klein et al., 2003; Li et al., 2004]. 

Using the FtsH protease as a target to fight bacterial infection seems to 
be an even more challenging task than targeting the methionyl aminopeptidase 
I. FtsH is a member of the AAA superfamily (ATPases associated with diverse 
cellular activities), which includes proteins involved in a variety of cellular 
processes characterized by conserved regions which include an ATP-binding site 
and a metal lopeptidase domain. These ATP-dependent proteases mediate the 
degradation of membrane proteins in bacteria, mitochondria and chloroplasts. 
They combine proteolytic and chaperone-like activities and thus form a mem- 
brane-integrated quality control system [Langer, 2000]. In bacteria, the FtsH 
peptidase is anchored to the cytoplasmic membrane with the catalytic domains 
exposed to the cytoplasm. In addition to being involved in quality control of inte- 
gral membrane proteins, FtsH peptidase is involved in the posttranslational con- 
trol of the activity of a variety of important transcription factors [Schumann, 
1999]. In this way, FtsH peptidase is involved in the regulation of the stress 
response together with other chaperones with proteolytic activity, including 
serine peptidases such as ClpXP, ClpAP, HslUV and Lon [Hengge and Bukau, 
2003; Wong and Houry, 2004]. However, unlike the serine peptidase chaperones, 
FtsH has never been implicated as an agent contributing to pathogenic fitness of 
a pathogen until recently, when it was shown that a S. aureus ftsH mutant was 
attenuated in a murine skin lesion model of pathogenicity [Lithgow et al., 2004]. 

The biological function of the sialoglycopeptidase in M. (Pasteurella) 
haemolytica has been investigated in some detail. The 35-kD enzyme isolated 
from the culture supernatant of this bacterium is active at neutral pH and 
is remarkably specific for 0-siaJoglycoproteins. It cleaves human eiythrocyte 
glycophorin A, which is 0-glycosylated at several positions, with a major 
site of cleavage at Arg31-Asp32, but does not cleave N-glycosylated proteins 
or nonglycosylated proteins [Abdullah et al., 1992]. The importance of the 
enzyme in the pathogenicity of bovine pneumonic pasteurellosis (shipment 
fever) caused by M. (Pasteurella) haemolytica is not clear, although the enzyme 
may interfere with cell-cell adhesion or with cytokine receptor binding through 
the cleavage of the cell surface 0-sialoglycoproteins [Sutherland et al., 1992] 
during the development of the host immune response in the cattle lung. Also, 
the sialoglycopeptidase-mediated enhanced adhesion to bovine platelets may 



Bacterial Peptidases 159 



initiate platelet aggregation and fibrin formation in alveolar tissue in pneu- 
monic pasteurellosis [Nyarko et al., 1998]. 

Genes encoding potentially active homologues of the sialoglycopeptidase 
are conserved across all cellular forms of life, but their biological function is 
still a puzzle. The essentiality nature of this gene for some bacteria indicates 
that the enzyme has a very important biological function, but either we do not 
know its physiological substrate(s) or the protein carries out a function unre- 
lated to proteolytic activity. At least in the case Schizosacchawmyces pombe the 
sialoglycopeptidase homologue has been shown to be involved in pro-protein 
processing [Ladds and Davey, 2000]. 

The large number of bacterial metallopeptidases excludes the possibility of 
a systematic description of each family of these peptidases in the context of 
their involvement in pathogenicity. Jt is interesting to note that a relatively large 
number of peptidase families in clans MA(E) (7 out of 16) and MA(M) (6 out 
of 12) have no counterparts in any other cellular form of life outside the 
(archae) bacterial kingdom. In addition to peptidases, which are strongly impli- 
cated as virulence factors, only members of families specific for bacteria are 
discussed below in more detail. 

Family M4: Thermolysin Family 

Thermolysin, an extracellular metallopeptidase isolated from Bacillus 
thermoproteolyticus, constitutes an archetype, not only of this family, but 
also for bacterial metallopeptidases in general. Enzymes homologous to ther- 
molysin are expressed by several pathogens, including L. monocytogenes, 
S. epidermidis, S. aureus, Enterococcus faecalis, C. perfringens, Helicobacter 
pylori, P. aeruginosa and V cholerae. Their involvement in pathogenicity is 
generally related to the broad substrate specificity of these peptidases, which 
can attack several physiologically important host proteins. A significant 
amount of data has been generated regarding the destructive function of 
pseudolysin from P. aeruginosa, an enzyme known for its strong elastinolytic 
activity [Wretlind and Wadstrom, 1977; Galloway, 1991]. This peptidase, also 
referred to as P. aeruginosa eiastase, exerts its destructive action by direct 
degradation of several connective tissue proteins [Kessler et al., 1977; Heck 
et al., 1986; Galloway, 1991] and, indirectly, by inactivation of host proteinase 
inhibitors, including a (-antitrypsin [Morihara et al., 1979]. Through its fib- 
rinogenolytic and fibrinolytic activities, the eiastase may disturb homeostasis 
and induce changes in the structure of the vascular wall, causing leakage of the 
plasma component, including cells into the extravascular tissue. This activity 
can potentially induce a hemorrhagic tendency and damage of infected tissue 
[Komori et al., 2001]. In lungs, the enzyme may degrade surfactant proteins 
SP-A and SP-D, which have important roles in the innate immune response. 



Potempa/Pike 



160 



This mechanism significantly contributes to the virulence mechanism in the 
pathogenesis of chronic P. aeruginosa infection [Mariencheck et al., 2003]. 
This data correlate well with the observation suggesting that the P. aeruginosa 
elastase is a potent inflammatory factor in a mouse model of diffuse panbron- 
chiolitis [Yanagihara et al., 2003] and that the control of elastase release by 
P. aeruginosa may be beneficial for patients with diffuse panbronchiolitis. Also, 
pseudolysin seems to play an essential role in the initiation and/or maintenance 
of a corneal infection [Hobden, 2002]. 

The role of pseudolysin orthologues in other pathogenic bacteria is less well 
understood and requires further investigation. Nevertheless, aureolysin from 
S. aureus has been shown to contribute to connective tissue degradation by host 
peptidases through inactivation of host proteinase inhibitors [Potempa et al., 
1986, 1991]. It may also assist m S. aureus dissemination by degradation of bac- 
terial adhesins [McAleese et al., 2001]. A similar function is suggested for the 
hemagglutinin/peptidase of V cholerae, which may be responsible for the detach- 
ment of these bacteria from cells through digestion of several putative adhesion 
receptors [Finkelstein et al., 1992]. On the other hand, the L. pneumophila Msp 
protease can significantly suppress antibacterial human phagocyte responses and 
contribute to the pathogenesis of Legionnaire's disease [Sahney et al., 2001]. A 
totally different mechanism seems to be utilized by the gelatinase (GelE) secreted 
by E.faecalis. This enzyme, which is also termed coccolysin, is implicated as a 
virulence factor by both epidemiological data and animal model studies and can 
apparently contribute to the dissemination of E. faecalis by fibrin degradation 
[Waters et al., 2003]. It is also possible that some of the manifestations of inflam- 
matory conditions in the presence of E. faecalis are related to coccolysin- 
catalyzed inactivation of endothelin [Makinen and Makinen, 1994]. 

Family M6: Immune Inhibitor A Family 

The name of this family, also known as thuringilysm family and belonging 
to the metzincin clan (MA(M)) [Gomis-Ruth, 2003], refers to the ability of pro- 
teins initially isolated from Bacillus thuringiensis to inactivate the antibacterial 
activity of insect hemolymph [Ediund et al., 1976]. It is now known that this 
protein is a metallopeptidase, exerting its insecticidal activity by proteolytic 
degradation of attacins and cecropins, two classes of antibacterial proteins in 
insects, and thus kills insect larvae [Dalhammar and Sterner, 1984; Lovgren et 
al., 1990]. This unique property contributes to the use of 5. thuringiensis in bio- 
logical pest control. Fortunately, this kind of peptidase, which is very effective 
in disabling the most important weapon of the host innate defense, is limited to 
insect pathogens. Nevertheless, several bacterial peptidases of different catalytic 
classes have been described to be able to inactivate human antibacterial peptides, 
once again indicating the importance of this activity in bacterial pathogenesis. 



Bacterial Peptidases 



61 



Family M9: Microbial Collagenase 

By virtue of being able to degrade collagen, one of the major proteinaceous 
constituents of the connective tissue and extracellular matrix, bacterial peptidases 
with this activity are by default recognized as virulence factors [Harrington, 1996]. 
The members of this family are common among Clostridium spp., Bacillus spp., 
and Vibrio spp. Despite the potential ability to inflict extensive tissue damage 
and facilitate spreading of infection, the precise role of microbial collagenases m 
pathogenicity remains unclear. 

Family MI 

This family is divided into two subfamilies in MEROPS, though according 
to somewhat dubious criteria. Both belong to the metzincin clan [Gomis-Riith, 
2003], as well as those of the -equally cryptically subdivided- family 12. Subfamily 
lOA encompasses predominantly eukaryotic MMPs. Probable orthologues have 
been identified in the genomes of archaebacteria {Methanosarcina acetivorans, 
Methanosarcina mazei Gol, Methanosarcina bar/ceri), uncultured crenarchaeote, 
and bacteria (Bacillus anthracis, Listeria innocua, L. monocytogenes, Leptospira 
intenvgans, and S. pneumoniae). In the latter cases, function as putative virulence 
factors or housekeeping enzymes remains to be assessed. According to MEROPS, 
subfamily lOA would fiarther encompass a secreted 20-kD metallopeptidase toxin, 
B.Jragilis toxin (BFT). The toxin also known as fragilysin is considered an impor- 
tant factor in the pathogenicity of infections with enterotoxigenic B. fragilis 
(ETBF), a recently identified enteric pathogen of children and adults. Fragilysin 
can directly damage human colonic mucosa [Riegler et al., 1999]. This effect is 
apparently dependent on cleavage of E-cadherin, the primary protein of the zonula 
adherens, leading to modification of epithelial cell morphology in vitro and result- 
ing in increased fluid secretion into the intestine, which is clinically manifested 
as diarrhea [Wu et al., 1998; Sears, 2001]. AJso, fragilysin contributes to mtesti- 
nal mucosal inflammation by stimulation of the expression of the neutrophil 
chemoattractant cytokine, IL-8 [Sanfilippo et al., 2000]. According to another 
classification, fragilysin, together with three paralogues and an orthologue m the 
photosynthetic cyanobacterium Nostoc punctiforme, would constitute an indepen- 
dent family within the metzincins, though structurally probably related to MlVTPs 
[Gomis-Ruth, 2003]. 

Only bacterial peptidases are grouped in subfamily lOB, which are exem- 
plified by the major metalloproteinase secreted by Serratia marcescens, termed 
serralysin. The other members of the subfamily include aeruginolysm, an alka- 
line protease from P. aeruginosa, mirabilysin (ZapA protease) from Proteus 
mirabilis, and several peptidases from Erwinia spp. Aeruginolysin seems to play 
a major role in the pathogenesis of eye infections by enhancing P. aeruginosa 
attachment to corneal epithelium [Pillar et al., 2000] and is a target for vaccine 



Potempa/Pike 



162 



development, and chemotherapy for bacterial eye infections. On the other hand, 
mirabilysin is considered to be an important virulence factor because it degrades 
host immunoglobulins, contributing to immune evasion during urinary tract 
infection [Walker et al., 1999; Almogren et al., 2003]. 



Family M26: IgA 1 -Specific Peptidase 

Many of the important mucosal bacterial pathogens, including Haemophilus 
influenzae, Neisseria gonorrhoeae, Neisseria meningitides, S. pneumoniae and 
successful members of the human resident flora, such as Streptococcus mitis, 
Streptococcus oralis, and Streptococcus sanguinis, have developed peptidases 
exclusively specific for cleavage at the hinge region of IgAl. These peptidases 
apparently belong to three catalytic classes, but only enzymes belonging to 
the serine (family S6) and metal 1 ©peptidase (family M26) classes have been 
thoroughly characterized. The IgAl-metallopeptidases are produced by 
Streptococcus spp., with a significant exception being GAS (S. pyogenes), while 
Haemophilus and Neisseria spp. produce serine-type IgA peptidases. Taken 
together, these peptidases are a striking example of convergent evolution to the 
same function by bacterial vinilence factors [KJlian et al., 1996]. All these 

enzymes cleave peptide bonds at a PI proline residue within the hinge region of 
IgAl, separating the antigen-binding Fab fragment from the Fc fragment. This 
mode of cleavage, which removes the Fc effector domain of the IgAl molecule, 
not only eliminates the protective effect of the immunoglobulins, but can also 
serve to camouflage the bacteria with Fab fragments, which mask the epitopes 
recognized by intact, functional antibodies. Despite this narrow specificity, which 
is precisely aimed to not only disable the effector molecules of host immune sys- 
tem and to take advantage of them, the exact role of these enzymes in bacterial 
pathogenesis is still unclear. This is due to the lack of an appropriate animal 
model to test the contribution of these enzymes to pathogenicity, since they only 
cleave human, gorilla or chimpanzee IgAl molecules [Reinholdt and Kilian, 
1991]. 

In the context of convergent evolution it is worth mentioning the IgA spe- 
cific metal lopeptidase produced by Clostridium ramosum here (family M64) 
[Kosowska et al., 2002]. This enzyme has specificity for cleavage of both IgAl 
and IgA2 molecules, which is a clear adaptation to the commensal lifestyle in 
the human gut, where both IgA isotypes are abundant. 

Family M27: Tentoxilysin 

Neurotoxins produced by several serotypes of Clostridium botulinum 
(BoNT type A-G) and Clostridium tetanum (TeNT) are the most potent natural 
toxins known to date. The toxins exert their biological effects at subfemtomolar 



Bacterial Peptidases 



63 



concentrations and they are released into the environment upon bacterial lysis 
as a single polypeptide chain of 150kD. Proteolytic cleavage executed by host 
peptidases generates a two-chain, mature, active neurotoxin composed of a 
heavy chain ( 1 00 kD) and a light chain (50 kD) held together by a single disul- 
fide bridge. The heavy chain is responsible for the specific binding of the toxin 
to presynaptic membranes and the translocation of the light chain into the 
neuron. The light chain is a very specific metal lopeptidase with activity limited 
to a small subset of proteins, including VAIVTP/synaptobrevin, SNAP-25 and 
syntaxin, which play key roles in synaptic signal transduction [Schiavo et al., 
1992a, b; Montecucco and Schiavo, 1994]. Cleavage of these proteins directly 
leads to the clinical manifestations of tetanus and botulism. 

Cumulatively, tentoxilysins represent a very interesting example of the 
development of extremely specific and potent virulence factors. Fortunately, 
their occurrence is limited to a few Clostridium spp. 

Family M34: Anthrax Lethal Factor 

The anthrax toxin is one of the most lethal natural toxins. It is produced by 
Bacillus anthracis and spores of these bacteria are the active component of the 
most deadly bioweapon developed by mankind. The toxin is composed of three 
proteins, includmg protective antigen (PA), edema factor (EF) and lethal factor 
(LF). PA binds to specific cell surface receptors and, upon proteolytic activa- 
tion by cell membrane-associated furm-like host peptidases, forms a membrane 
channel through which EF and LF enter the cell. LF is a unique multidomain 
metal lopeptidase with a very narrow specificity to cleave the amino-terminus 
of mitogen-activated kinase kinases 1 and 2 (MMPKKl and MMPKK2). The 
cleavage inactivates the signal transduction pathway dependent on these 
kinases. This signaling pathway plays a fiindamental role in the overall intra- 
cellular signaling network, thus the overall signaling in the cell is compromised. 

Family M 56: BlaRJ Peptidase (S. aureus) 

The BlaRl peptidase from S. aureus is a metallopeptidase which cleaves a 
repressor (Blal) of the synthesis of the (3-lactamase enzyme BlaZ by this bac- 
terium [Hackbarth and Chambers, 1993]. Thus, this peptidase controls antibiotic 
resistance by controlling the production of the |3-lactamase. The BlaRl pepti- 
dase orthologue, Mec Rl, only found in methicillin resistant 6". aureus (MRSA), 
controls the formation of the penicillin-binding protein 2a (PBP 2a) and thereby 
controls the resistance of the bacterium to methicillin [Hackbarth and Chambers, 
1993; Brakstad and Maeland, 1997]. The BlaRl molecule consists of two 
domains, an extracellular penicillin-binding domain and an integral-membrane 
zinc metallopeptidase domain [Zhang et al., 2001]. Upon penicillin binding, 
the BlaRl peptidase autoactivates, then cleaves the repressor of p-lactamase 



Potempa/Pike 



164 



synthesis, providing an interesting 'signal transduction' system which mediates 
this antibiotic resistance in the highly pathogenic staphylococcus species. 

Family M66: StcE Protease 

The StcE metallopeptidase, member of the cholorerilysins within the 
metzincin clan MA(M) [Gomis-Riith, 2003] is produced by the enterohemor- 
rhagic 0157:H7 strain of £". coll, which causes diarrhea, hemorrhagic colitis, 
and the hemolytic uremic syndrome, specifically cleaves CI inhibitor (also 
known as CI esterase inhibitor). The peptidase is quite specific for CI inhibitor 
and does not appear to cleave other proteins, although it has been shown to 
cause aggregation of cultured T cells, the significance of which is not com- 
pletely understood [Lathem et al., 2002]. CI inhibitor is known to control 
potent proinflammatory and procoagulant enzymes, and thus its inactivation by 
the bacterial peptidase is likely to cause proinflammatory effects which may be 
consistent with the disease outcomes caused by this strain of E. coli. Further 
experiments will be required to elucidate how critical this enzyme is to patho- 
genesis by this strain of the bacterium. 

Family M73: Camelysin 

Camelysin (casein-cleaving metal loprotease) is found on the surface of 
B. cereijs, whose genome encodes a total of four paralogues. Possible ortho- 
logues have been identified in the genomes of Oceanohacillus iheyensis (five 
sequences) and B. anthracis (two sequences). Single sequences are further found 
in B. thuringiensis, B. subtilis, and Bacillus halodurans (Gomis-Ruth; personal 
communication). This bacterium is known to cause food poisoning and nosoco- 
mial diseases. Camelysins do not have a sequence consistent with metallopro- 
teases, but the enzyme is active against a broad range of proteinaceous substrates, 
and mass spectrometry analyses strongly indicate the association of a zinc ion 
with each enzyme molecule. Disruption of the gene for the enzyme causes a 
marked loss in the proteolytic activity of membranes from the bacterium and it| 
is possible that the enzymatic activity plays a role in the pathogenic activity of 
the organism, although this remains to be fimily established [Grass et al., 2004].' 



Serine Peptidases 

Peptidases which utilize a serine residue as the main catalytic residue are 
the biggest group of peptidases, making up 35% of the total peptidases listed in 
MEROPS. The serine peptidases are widespread across all organisms and are 
divided into 10 clans on the MEROPS database [SB, SC, SE, SF, SH, SJ, SK, 
SP, SR and S- (the last contains currently unassigned peptidases)]. Bacterial 



Bacterial Peptidases 165 



proteases are present in all of these clans, except SH, SP and SR, which will 
therefore not be considered any further here. 

By definition, this catalytic class contains a serine residue acting as the 
nucleophile during catalysis. Usually (as applies to enzymes in clans SB, SC and 
SK) the catalytic Ser residue combines with His and Asp residues to form the 
classical catalytic triad exemplified by chymotrypsin, the archetypal enzyme of 
the serine protease class. Variations on this do exist, for instance enzymes in 
clans SE and SJ use a Ser/Lys dyad to accomplish catalysis, while those in SF 
use either a Ser/Lys or a Ser/His dyad. 

There are over 60 families represented within the serine-type catalytic 
class and many of these are subdivided into subfamilies. The sheer number of 
proteases in this catalytic class which are found in bacteria defies their being 
mentioned in any representative manner here. Thus the most interesting or well- 
characterized examples with direct relevance in pathogenicity were selected for 
presentation here. 

Family SIB 

The glutamyl endopeptidase I, better known as endoproteinase GluC or the 
V8 protease from S. aureus, is a member of the SIB family. The roles of this 
enzyme are somewhat related to pathogenicity (see section Family C47: The 
Staphopain Family above), but this enzyme is better known for its widespread 
biotechnological use as a specific protease in sequencing applications. Its struc- 
ture has recently been solved [Prasad et al., 2004]. This family also contains the 
Spl peptidases, which have recently been identified as a new operon which is 
positively controlled by the Agr virulence regulator, indicating a possible role 
in pathogenesis by S. aureus [Reed et al., 2001]. 

Family SIC 

An interesting group of peptidases is formed by members of the SIC family, 
which is required for growth at high temperatures by a number of organisms, such 
as E. coli. Some of these enzymes, generically termed protease Do (also referred 
Lo as DegP or HtrA), have been characterized as being associated with the viru- 
lence of 5. enterica serovar typhimurlum. Yersinia enterocoUtica and S. pyogenes. 
DegP from E. coli has a fascinating dual function of acting as a chaperone and a 
peptidase, depending on the temperature of the environment. In the chaperone 
phase, a hydrophobic patch of amino acids plays the presumptive role of binding 
unfolded proteins and mediating their refolding. During chaperone operation, the 
active site for the peptidase is 'walled off', preventing substrate binding and catal- 
ysis. A change in the environmental conditions triggers the opening of the active 
site to substrates and allows catalysis. This fascinating mechanism allows the pep- 
tidase to process many different proteins needed for pathogenesis by the bacteria. 



Potempa/Pike 



166 



Family SID 

The family is entirely composed of the endoproteinase lysC and endopro- 
teinase Arg-C, which have applications in the sequencing of proteins due to 
their high specificity for lysine and arginine amino acids at the cleavage point, 
respectively. An endoproteinase Arg-C orthologue from P. aeruginosa is 
thought to act as a virulence factor in cornea] infections by this bacterium 
[Engel etal., 1998]. 

Family S6 

The IgA I -specific serine endopeptidases which are found in Neisseria 
spp. and some Haemophilus spp. are typical members of the S6 family. In 
A^. gonorrhoeae, the enzyme has been postulated to play a role in evading the 
host immune response by specifically cleaving IgAl [Vitovski et al., 1999]. 
It has been suggested that the enzyme plays a role in bacterial invasion of host 
cells [Lin et al., 1997]. However, whether the IgAl -specific serine endopep- 
tidase is a crucial virulence factor has yet to be determined [Johannsen et al., 
1999]. 

Family S8A 

This group of serine proteases contain enzymes generally referred to as 
subtilisin-like enzymes, named after the archetypal enzyme of the group. The 
family contains a large number of enzymes, most likely second only to family 
SIA which contains the mammalian chymotrypsin-like enzymes. The subtil- 
isins and chymotrypsin-like enzymes are examples of convergent evolution, 
arriving at the same function and catalytic groups, but grafted onto very differ- 
ent scaffolds. 

Perhaps the best-characterized virulence factor of this family is the C5a pep- 
tidase from group A and group B Streptococci, exemplified by the enzyme from 
S. pyogenes. As the name suggests, this enzyme cleaves the C5a component of 
complement, destroying its ability to act as a chemotaxin for polymorphonuclear 
leukocytes [Hill et al., 1 988]. Recent studies suggest that this enzyme is also able 
to bind to fibronectin, which may be important in the binding and invasion of 
host cells by group B streptococci [Beckmann et al., 2002; Cheng et al., 2002b]. 
Recently, much effort has been invested into the development of C5a peptidase- 
based vaccines for the treatment of group A and B streptococcal infections [Shet 
et al., 2003; Cheng et al., 2002a]. 

Family S9B 

Members of the family S9B are generally dipeptidyl peptidases, which 
cleave two amino acids at a time from the terminii of proteins. The bacterial 
peptidases in this subfamily are exemplified by the dipeptidyl aminopeptidase 



Bacterial Peptidases 167 



IV, from organisms such as R gingivalis [Banbula et al., 2000; Kumagai et al., 
2000]. The enzyme is apparently important for the virulence of P. gingivalis, 
since bacteria lacking the protease or with a mutation in the catalytic domain 
have attenuated virulence [Kumagai et al., 2003]. 

Family SI 4 and SI 6 

The S14 family is primarily composed of the endopeptidase Clp enzymes, 
originally discovered and characterized m E. coli. Endopeptidase Clp enzymes 
are rather similar to Lon proteases (SI 6 family) in that their activity as a pepti- 
dase is linked to the hydrolysis of ATP. The enzymes contain an ATP binding 
and catalysis domain and a distinct peptidase domain [Wang et al., 1997]. Some 
studies suggest that these enzymes are the functional equivalents of the protea- 
some complex found in all mammalian cells, which is crucial for the control of 
protein turnover in these cells. Interesting support for this hypothesis is pro- 
vided by a recent study which suggests that the Clp enzyme is important for 
survival of bacteria which are in the stationary phase [Weichart et al., 2003]. 
The catalytic dyad of Lon proteases consists of Ser and Lys. The enzyme 
is normally induced under stress conditions [Botos et al., 2004], and animal 
studies suggest it is highly unportant S. enterica serovar typhimurium virulence 
[Takayaetal., 2003]. 



Conclusions 

As is evidenced by the above review, which is by necessity not absolutely 
comprehensive, there is a wealth of information about bacterial peptidases. In 
many instances, however, knowledge is just starting to be accumulated about 
specific families or enzymes within families. Bacterial peptidases span a 
tremendous range of mechanisms, and frequently have surprising associations 
with additional domains which carry out separate functions. This adds a fasci- 
nating range to the potential activities of these enzymes. In many cases, the 
potential for inhibitors of the enzymes to be used as antibacterial agents will 
continue to drive the active and thriving research in this important field. 



References 

Abdullah KLM, Udoh EA, Shewen PE, Mellors A: A neutral glycoprotease of Pasieurella haemotylica Al 
specifically cleaves 0-sialoglycoproteins. Infect Immun 1992;60:56-62. 

Almogren A, Senior BW, Loomes LM, Kerr MA: Structural and functional consequences of cleavage of 
human secretory and human serum immunoglobulin A 1 by proteinases fi'om Proteus mirabilis and 
Neisseria meningitidis. Infect Immun 2003;71:3349-3356. 



Potempa/Pike 



168 



Anantha RP, Stone KD, Donnenberg MS: Effects of bfp mutations on biogenesis of functional 
enteropathogenic Escherichia coli type IV pili, J Bacteriol 2000;182:2498-2506. 

Aravind L, Koonin EV: Classification of the caspase-hemcglobinase fold: Detection of new families and 
implications for the origin of the eukaryotic separins. Proteins 2002;46:355-367. 

Ashbaugh CD, Warren HB, Carey VJ, Wessels MR: Molecular analysis of the role of the group A strep- 
tococcal cysteine protease, hyaluronic acid capsule, and M protein in a murine model of human 
invasive soft-tissue infection. J Clin hivest 1998;102:550-560. 

Ashbaugh CD, Wessels MR: Absence of a cysteine protease effect on bacterial virulence in two murine 
models of human invasive group A streptococcal infection. Infect Immun 2001;69:6683-6688. 

Axtell MJ, Chisholm ST, Dahlbeck D, Staskawicz BJ: Genetic and molecular evidence that the 
Pseitdomonas syringae type 111 effector protein AvrRpt2 is a cysteine protease. Mol Microbiol 
2003;49:1537-1546. 

Axtell MJ, Staskawicz BJ: hiitiation of RPS2-specified disease resistance in Arabidopsis is coupled to 
the AvrRpt2-directed elimination of RIN4. Cell 2003;! 12:369-377. 

Aziz [IK, Pabst MJ, Jeng A, Kansal R, 1-X>w DE, Nizet V, Kotb M: Invasive M ITl group A Streptococcus 
undergoes a phase-shift in vivo to prevent proteolytic degradation of multiple virulence factors by 
SpeB. Mol Microbiol 2004;51:123-134. 

Banbula A, Bugno M, Goldstein J, Yen J, Nelson D, Travis J, Potempa J: Emerging family of proline-speci fie 
peptidases of Porphyromonas gingivah's: Purification and characterization of serine dipeptidyl pepti- 
dase, a structural and functional homologue of mammalian prolyl dipeptidyl peptidase fV. Infect 
Immun 2000;68:1176-1 182. 

Barrett AJ, Toll e DP, Rawlings ND: Managing peptidases in the genomic era. Biol Chem 2003;384:873-882. 

Bazan JF, Weaver LH, Roderick SL, Huber R, Matthews BW: Sequence and structure comparison sug- 
gest that methionine aminopeptidase, prolidase, aminopeptidase P, and creatinase share a common 
fold. Proc Natl Acad Sci USA 1994;91:2473-2477. 

Beckmann C, Waggoner JD, Harris TO, Tamura GS, Rubens CE: Identification of novel adhesins from 
group B Sn-eptococci by use of phage display reveals that C5a peptidase mediates fibronectin 
bindmg. Infect Immun 2002;70:2869-2876. 

Berge A, Bjorck L: Streptococcal cysteine proteinase releases biologically active fragments of strepto- 
coccal surface proteins. J Biol Chem 1995;270:9862-9867. 

Berge A, Kihlberg BM, Sjoholm AG, Bjorck L: Streptococcal protein H forms soluble complement- 
activating complexes with IgG, but inhibits complement activation by IgG-coated targets. J Biol 
Chem 1997:272:2077^20781. 

Bierne H, Garandeau C, Pucciarelli MG, Sabet C, Newton S, Garcia-del Portillo F, Cossart P, Charbit A: 
Sortase B, a new class of sortase in Listeria monocytogenes. J Bacteriol 2004;186:1972-1982. 

Bjorck L, Akesson P, Bohus M, Trojnar J, Abrahamson M, Olafsson I, Grubb A: Bacterial growth 
blocked by a synthetic peptide based on the structure of a human proteinase inhibitor. Nature 
1989;337:385-386. 

Botos 1, Melnikov EE, Cherry S, Tropea JE, Khalatova AG, Rasulova E, Dauter Z, Maurizi MR, 
Rotanova TV, Wlodawer A, Gustchina A: The catalytic domain of £. coli Lon protease has a 
unique fold and a Ser-Lys dyad in the active site. J Biol Chem 2004;279:8140-8148. 

Bourgeau G, Lapointe H, Peloquin P, Mayrand D: Cloning, expression, and sequencing of a protease gene 
(//?/■) from Porphyromonas gingivatis W83 in Escherichia coli. hifect bnmun 1 992;60:3 1 86-3 1 92. 

Bradshaw RA, Brickey WW, Walker KW: N-terminal processing: The methionine aminopeptidase and 
N alpha-acetyl transferase families. Trends Biochem Sci 1998;23:263-267. 

Brakstad OG, Maeland JA; Mechanisms of methicillin resistance in staphylococci. APMIS 1997;105: 
264-276. 

Burns EH Jr, Marciel AM, Musser JM: Activation of a 66-kilodalton human endothelial cell matrix met- 
alloprotease by Streptococcus pyogenes extracellular cysteine protease. Infect Immun 1996;64: 
4744^750. 

Calander AjM, Jonsson IM, Kanth A, Arvidsson S, Shaw L, Foster SJ,Tarkowski A: Impact of staphylo- 
coccal protease expression on the outcome of infectious arthritis. Microbes Infect 2004;6: 
202-206. 

Chan PF, Foster SJ: Role of SarA in virulence determinant production and environmental signal trans- 
duction \r\ Staphylococcus aureus, J Bacteriol 1998;180:6232-6241. 



Bacterial Peptidases 



169 



Chen CY, Luo SC, Kuo CF, Lin YS, Wu JJ, Lin MT, Liu CC, Jeng WY, Chuang WJ: Maturation pro- 
cessing and chat^cterization of streptopain. J Biol Chem 2003;278:17336-17343, 

Chen JM, Rawlings ND, Stevens RA, Barrett AJ: Identification of the active siteof legumain links it to 
caspases, clostripain and gingipains in a new clan of cysteine endopeptidases. FEBS Lett 1998; 
441:361-365. 

Cheng LW, Schneewind O: Type 111 machines of Gram-negative bacteria: Delivering the goods. Trends 
Microbiol 2000;8:214-220. 

Cheng Q, Debol S, Lam H, Eby R, Edwards L, Matsuka Y, Olmsted SB, Cleary PP: Immunization with 
C5a peptidase or peptidase-type (IT polysaccharide conjugate vaccines enhances clearance of 
group B Streptococci from lungs of infected mice. Infect Immun 2002a;70;6409-64l5, 

Cheng Q, Stafslien D, Purushothaman SS, Cleary P: The group B streptococcal C5a peptidase is both 
a specific protease and an invasin. Infect Immun 2002b;70:2408-24l3. 

Collier-Hyams LS, Zeng H, Sun J, Tomlinson AD, Bao ZQ, Chen H, Madara XL, Orth K, Neish AS: 
Cutting edge: Salmonella AvrA effector inhibits the key proinflammatory, anti-apoptotic 
NF-kappa B pathway, J Immunol 2002;169:2846-2850. 

Collin M, Olsen A: Generation of a mature streptococcal cysteine proteinase is dependent on ceil wall- 
anchored Ml protein. Mol iVIicrobiol 2000;36:1306-1318. 

Collin M, Svensson MD, Sjoholm AG, Jensenius JC, Sjobring U Olsen A: EndoS and SpeB from 
Streptococcus pyogenes inhibit immunoglobulin-mediated opsonophagocytosis. hifect Immun 
2002;70:6646-6651. 

Comfort D, Clubb RT: A comparative genome analysis identifies distinct sorting pathways in gram- 
positive bacteria. Infect Immun 2004;72;27 10-2722, 

Connolly KM, Smith BT, Pilpa R, llangovan U, Jung ME, Clubb RT: Sortase from Staphylococcits 
aureus does not contain a thiolate-imidazolium ion pair m its active site. J Biol Chem 2003;278: 
34061-34065. 

Copik AJ, Swierczek SI, Lowther WT, D'souza VM, Matthews BW, Holz RC: Kinetic and spectroscopic 
characterization of the H178A methionyl aminopeptidase from Escherichia coli, Biochemisti*y 
2003;42:6283-6292. 

Cornelis GR: The Yersinia Ysc-Yop 'type IIP weaponry. Nat Rev Mol Cell Biol 2002;3:742^52. 

Cornelis GR, Van Gijsegcm F: Assembly and fijnction of type III secretory systems. Annu Rev Microbiol 
2000;54:735-774. 

Coulter SN, Schwan WR, Ng EY, Langhorne MH, Ritchie HD, Westbrock-Wadman S, Hufiiagle WO, 
Folger KR, Bayer AS, Stover CK: Staphylococcus aureus genetic loci impacting growth and 
survival in multiple infection environments. Mol Microbiol I998;30:393^04. 

Cummins PM, O'Connor B: Pyroglutamyl peptidase: An overview of the three known enzymatic forms. 
Biochim Biophys Acta 1998;1429:1-17. 

Curtis MA, Aduse Opoku J, Rangarajan M, Gallagher A, Sterne JA, Reid CR, Evans HE, Samuelsson B: 
Attenuation of the viiulence of Porphyromonas gingivalis by using a specific synthetic Kgp 
protease ijiliibitor. Infect Lnmun 2002;70:6908-6975. 

Curtis MA, Kuramitsu HK, Lantz M, Macrina FL, Nakayama K, Potempa J, Reynolds EC, Aduse-Opoku J: 
Molecular genetics and nomenclature of proteases of Porphyromonas gingivalis. J Periodontal Res 
1999;34:46^M72. 

Dalhammar G, Steiner H: Characterization of inhibitor A, a protease from Bacillus thuringiensis which 
degrades attacins and cecropins, two classes of antibacterial proteins in insects. Eur J Biochem 
1984;139:247-252. 

Ding J, McGrath WJ, Sweet RM, Mangel WF: Crystal structure of the human adenovirus proteinase with 
its II amino acid cofactor. EMBO J 1996;15:1778-1783. 

Discipio RG, Daffern PJ, Kawahara M, Pike R, Travis J, Hugli TE, Potempa J: Cleavage of human com- 
plement component C5 by cysteine proteinases from Porphyromonas (Bacteroides) gingivalis. 
Prior oxidation of C5 augments proteinase digestion of C5. hnmunology 1996;87:660-667. 

Donham MC, Heath HE, LeBlanc PA, Sloan GL: Characteristics of extracellular protein production by 
Staphylococcus simulans biovar staphyhlyticus during aerobic and anaerobic growth. J Gen 
Microbiol 1988;134:2615-2621. 

Douangamath A, Dale GE, D'Arcy A, Almstetter M, Eckl R, Frutos-Hoener A, Henkel B, lUgen K, 
Nerdinger S, Schulz H, MacSweeney A, Thormann M, Treml A, Pierau S, Wadman S, Oefner C: 



Potempa/Pike 



170 



Crystal structures of Staphylococcus aureus methionine aminopeptidase complexed with keto het- 
erocycle and aminoketone inhibitors reveal the formation of a tetrahedral intermediate. J Med 
Chem 2004;47:1325-1328. 

Drapeau GR: Role of melalloprotease in activation of the precursor of staphylococcal protease. J Bacteriol 
1978;136:607-613. 

Dubin G, Chmiel D, Mak P, Rakwalska M, Rzychon M, Dubin A: Molecular cloning and bio- 
chemical characterisation of proteases from Staphylococcus epidermidis. Biol Chem 2001 ;382: 
1575-1582, 

Dubin G, Krajewski M, Popowicz G, Slec-Niemczyk J, Bochtler M, Potempa J, Dubin A, Holak TA: 
A novel class of cysteine protease inhibitors: Solution structure of staphostatin A from 
Staphylococcus aureus. Biochemistry 2003;42:13449-13456. 

Dubin G, Slec-Niemczyk J, DylagT, Silberring J, Dubin A, Potempa J: Chai'acterizalion of a highly spe- 
cific, endogenous inhibitor of cysteine proteases from Staphylococcus epidermidis, a new mem- 
ber of the staphostatin family, Biol Chem, in press. 

Ediund T, Siden 1, Boman HG: Evidence for two immune inhibitors ft^om Bacillus thuringiensis inter- 
fering mth the humoral defense system of satumiid pupae. Infect Immun 1976;14:934-941. 

Egile C, d*Hauteville H, Parsot C, Sansonetti PJ: SopA, the outer membrane protease responsible for 
polar localization of IcsA in Shigella Jlexneri. Mol Microbiol 1997;23:1063-1073. 

Eichinger A, Beisel HG, Jacob U Huber R, Medrano FJ, Banbula A, Potempa J, Travis J, Bode W: 
Crystal strucmre of gingipain R: An Arg-specific bacterial cysteine proteinase with a caspase-like 
fold. EMBOJ 1999;18:5453-5462. 

Eijsink VG, Axelsson L, Diep DB, Havarstein LS, Holo H, Nes IF: Production of class II bacteriocins 
by lactic acid bacteria; an example of biological warfare and communication. Antonie Van 
Leeuwenhoek 2002;81:639-654. 

Elliott SD: A proteolytic enzyme produced by group A streptococci with special reference to its effect 
on the type-specific M antigen. J Exp Med 1945;81:573-592. 

Engel LS, Hill JM, Caballero AR, Green LC, O'Callaghan RJ: Protease IV, a unique extracellular pro- 
tease and vimlence factor from Pseudomonas aervgitjosa. J Biol Chem 1998;273:16792-16797. 

Eriksson A, Norgren M: Cleavage of antigen-bound immunoglobulin G by SpeB contributes to 
streptococcal persistence in opsonizing blood. Infect Immun 2003;71:211-217. 

Fenster KM, Parkin KL, Steele JL: Characterization of a thiol-dependent endopeptidase from 
Lactobacillus helveiicus CNRZ32. J Bacteriol 1997;179:2529-2533. 

Filipek R, Rzychon M, Oleksy A, Gruca M, Dubin A, Potempa J, Bochtler M: The staphostatin-staphopain 
complex: A forward binding inhibitor in complex with its target cysteine protease. J Biol Chem 
2003;278:40959-40966. 

Finkelstein RA, Boesman-Finkelstein M, Chang Y, Hase CC: Vibrio cholerae hemagglutinin/protease, 
colonial variation, virulence, and detachment. Infect Immun I992;60:472^78. 

Fischetti VA: Novel method to control pathogenic bacteria on human mucous membranes. Ann NY Acad 
Sci 2003;987:207-214. 

Gabazza EC, Taguchi O, Kamada H, Hayashi T, Adachi Y, Suzuki K: Progress in the understanding of 
protease-activated receptors, Int J Hematol 2004;79:117-122, 

Galloway DR: Pseudomonas aeruginosa elastase and elastolysis revisited: Recent developments. Mol 
Microbiol 1991;5:2315-232]. 

Gerlach D, Knoll H, Kohler W, Ozegowski JH, Hribalova V: Isolation and characterization of erythrogenic 
toxins. V Communication: Identity of erythrogenic toxin type B and streptococcal proteinase 
precursor. Zentralbl Bakteriol Mikrobiol Hyg 1983;255:221-233. 

Gibson FC III, Genco CA: Prevention of Porphyromonas gingivalis-y\\d\M:Q6 oral bone loss following 
immunization with gingipain RI. Infect Immun 2001;69:7959-7963. 

Gibson FC III, Gonzalez DA, Wong J, Genco CA: Porphyromonas gingivalis-spQCif'K immunoglobulin 
G prevents P gingivalis-c\ic]ic6 oral bone loss in a murine model, hifect Immun 2004;72: 
2408-24 1 1 . 

Gomis-Rijth FX: Structural aspects of the metzincin clan of metalloendopeptidases. Mol Biotech 2003; 
24:157-201. 

Grass G, Schierhorn A, Sorkau E, Muller H, Rucknagel P, Nies DH, Fricke B: Camelysin is a novel 
surface metalloproteinase from Bacillus cereus. Infect hnmun 2004;72:219-228, 



Bacterial Peptidases 



71 



Gunther R, Stein A, Bordusa F: bivestigations on the enzyme specificity of clostripain: A new efficient 

biocatalyst for the synthesis of peptide isosteres, J Org Chem 2000;65:1672-1679. 
Hackbarth CJ, Chambers HF: blal and blaR 1 regulate beta-lactainase and PBP 2a production in methicilHn- 

resistant Staphylococcus aureus, Antimicrob Agents Chemother 1 993; 37: 1 144-1 149, 
Harrington DJ: Bacterial collagenases and collagen-degrading enzymes and Iheir potential role in 

human disease. Infect Immun 1996;64:1885-1891. 
Havarstein LS, Diep DB, Nes IF: A family of bacteriocin ABC transporters carry out proteolytic 

processing of their substrates concomitant with export. Mol Microbiol 1995;16:229-240. 
Heath A, DiRita VJ, Barg NL, Engleberg NC: A two-component regulatory system, CsrR-CsrS, 

represses expression of three Streptococcus pyogenes virulence factors, hyaluronic acid capsule, 

streptolysin S, and pyrogenic exotoxin B. Infect Immun 1999;67:5298-5305. 
Heck LW, Morihara IC, Abrahamson DR: Degradation of soluble laminin and depletion of tissue-associated 

basement membrane laminin by Pseudomonas aeruginosa elastase and alkaline protease. Infect 

Immun 1986;54:149-153. 
Hengge R, Bukau B: Proteolysis in prokaryotes: Protein quality control and regulatory principles. Mol 

MicrobioJ 2003;49:1451-1462. 
Herwald H, Collin M, Muller-Esterl W, Bjorck L: Streptococcal cysteine proteinase releases kinins: 

A virulence mechanism. J Exp Med 1996;184:665-763. 
Herwald H, Morgelin M, Bjorck L: Contact activation by pathogenic bacteria: A virulence mechanism 

contributing to the pathophysiology of sepsis. Scand J Infect Dis 2003;35:604-607. 
Herwald H, Morgelin M, Olsen A, Rhen M, Dahlback B, Muller-Esterl W, Bjorck L: Activation of the 

contact-phase system on bacterial surfaces - A clue to serious complications in infectious diseases. 

Nat Med 1998;4:298-302. 
Hill HR, Bohnsack JF, Morris EZ, Augustine NH, Parker CJ, Cleary PP, Wu JT: Group B streptococci inhibit 

the chemotactic activity of the fifth component of complement J Immunol 1988;141:3551-3556. 
Hobden JA: Pseudomonas aeruginosa proteases and corneal virulence. DNA Cell Biol 2002;21: 

391-396. 
Hofinann B, Schomburg D, Hecht HJ: Crystal structure of a thiol proteinase from Staphylococcus aureus 

V-8 in the E-64 inhibitor complex. Acta CrystallogrA (Suppl) I993;49:I02. 
Hosotaki K, Imamura T, Potempa J, KitamuraM, Travis J : Activation of protein C by arginine-specific 

cysteine proteinases (gingipains-R) from Porphyromonas gingivalis, Biol Chem 1999;380:75-80, 
Hotson A, Chosed R, Shu H, Orth K, Mudgett MB: Xanthomonas type III effector XopD targets SUMO- 

conjugated proteins in planta. Mol Microbiol 2003;50:377-389. 
Hu X, Zhu J, Srivathsan S, Pei D: Peptidyl hydroxamic acids as methionine aminopeptidase inhibitors. 

Bioorg Med Chem Lett 2004;14:77-79. 
Hytonen J, Haataja S, Gerlach D, Podbielski A, Finne J: The SpeB virulence factor of Streptococcus 

pyogenes, a multiftinctional secreted and cell surface molecule with strepadhesin, lammin-binding 

and cysteine protease activity. Mol Microbiol 2001;39:512-519. 
Ilangovan U, Ton-That H, Iwahara J, Schneewind O, Clubb RT: Structure of sortase, the transpeptidase 

that anchors proteins to the cell wall of Staphylococcus aureus. Proc Natl Acad Sci USA 2001;98: 

605^6061. 
Imamura T, Banbula A, Pereira PJ, Travis J, Potempa J: Activation of human prothrombin by arginine- 
specific cysteine proteinases (Gingipains R) from Porphyromonas gingivalis. J Biol Chem 2001a; 

276:18984-18991. 
Imamura T, Pike RN, Potempa J, Travis J: Pathogenesis of periodontitis: A major arginine-specific cysteine 

proteinase from Porphyromonas gingivalis induces vascular permeability enhancement through 

activation of the kallDkrein/kinin pathway. J Clin Invest 1994;94:361-367. 
Imamura T, Potempa J, Pike RN, Moore JN, Barton MH, Travis J: Effect of free and vesicle-bound cysteine 

proteinases of Porphyromonas gingivalis on plasma clot formation; Implications for bleeding 

tendency at periodontitis sites. Infect hnmun 1995a;63:4877^882, 
Imamura T, Potempa J, Pike RN, Travis J: Dependence of vascular permeability enhancement on cysteine 

proteinases in vesicles of Porphyromonas gingivalis. Infect Immun 1995b;63: 1999-2003. 
ImamuraX Potempa J^Tanase S, Travis J: Activation of blood coagulation factor X by arginine-specific 

cysteine proteinases (gingipain-Rs) from Porphyromonas gingivalis. J Biol Chem 1997;272: 

16062-16067. 



Potempa/Pike 



172 



Imamura T, Tanase S, Hamamoto T, Potempa J, Travis J: Activation of blood coagulation factor IX by 

gingipains R, arginme-specific cysteine proteinases from Porphytvmonas gingivaUs. Biochem J 

2001b;353:325-331. 
Imamura T, Travis J, Potempa J: The biphasic virulence activities of gingipains: Activation and inactivation 

of host proteins. Cuit Protein Pept Sci 2003;4:443^50. 
Iriarte M, Cornehs GR: YopT, a new Yersinia Yop effector protein, affects the cyioskeleton of host cells. 

Mol Microbiol 1998;29:915-929. 
Isenberg HD: Pathogenicity and virulence: Another view. Clin Microbiol Rev 1988; I :'^0-53. 
Jagels MA, Travis J, Potempa J, Pike R, Hugli TE: Proteolytic inactivation of the leukocyte C5a receptor 

by proteinases derived from Porphyromonas gingivaUs. Infect Immun 1996;64:1984-1991. 
Janzon L, Lofdahl S, Arvidson S: Identification and nucleotide sequence of the delta-lysin gene, hid, 

adjacent to the accessory gene regulator (agr) o{ Staphylococcus aureus. Mol Gen Genet 1989; 

219:480^85. 
Johannsen DB, Johnston DM, Koymen HO, Cohen MS, Cannon JG: A Neisseria gonorrhoeae 

immunoglobulin Al protease mutant is infectious in the human challenge model of urethral infec- 
tion. Infect Immun 1999;67:3009-3013. 
Juris SJ, Shao F, Dixon JE: Yersinia effectors target mammalian signalling pathways. Cell Microbio 

2002;4:20J-2M. 
Kagawa TF, Cooney JC, Baker HM, McSweeney S, Liu M, Gubba S, Musser JM, Baker EN: Crysta 

structure of the zymogen form of the group A Streptococcus virulence factor SpeB: An integrin- 

binding cysteine protease. Proc Natl Acad Sci USA 2000;97:2235-2240. 
Kansal RG, McGeer A, Low DE, Norrby-Teglund A, Kotb M: Inverse relation between disease 

severity and expression of the streptococcal cysteine protease, SpeB, among clonal MlTl iso- 
lates recovered from invasive group A streptococcal infection cases. Infect Immun 2000;68: 

6362-6369. 
Kansal RG, Nizet Y Jeng A, Chuang WJ, Kotb M: Selective modulation of superantigen-induced 

responses by streptococcal cysteine protease. J Infect Dis 2003; 187:398^07. 
Kapur y Majesky MAV, Li LL, Black RA, Musser JM: Cleavage of Lnterleukin I beta (JL-J beta) pre- 
cursor to produce active IL-I beta by a conserved extracellular cysteine protease from 

Streptococcus pyogenes. Proc Natl Acad Sci USA 1993a;90:7676-7680. 
Kapur V, Topouzis S, Majesky MW, Li LL, Hamrick MR, Hamill RJ, Patti JM, Musser JM: A conserved 

Streptococcus pyogenes extracellular cysteine protease cleaves human fibronectin and degrades 

vitronectin. Microb Pathog 1 993b; 15:327-346. 
Kellner A, Robertson T: Myocardial necrosis produced in animals by means of crystalline streptococcal 

proteinase. J Exp Med 1954;99:495-503. 
KesslerE, Kennah HE, Brown SI: Pseudomonas protease. Purification, partial characterization, and its 

effect on collagen, proteoglycan, and rabbit corneas. Invest Ophthahnol Vis Sci 1 977; 16:488^97. 
Kilian M, Reijiholdt J, Lomholt H, Poulsen K, Frandsen EV: Biological significance of IgAl proteases 

in bacterial colonization and pathogenesis: Critical evaluation of experimental evidence. APMIS 

1996;104:321-338. 
Klein CD, Schiffmann R^ Folkers G, Piana S, Rothlisberger U: Protonation states of methionine 

aminopeptidase and their relevance for inhibitor binding and catalytic activity. J Biol Chem 2003; 

278:47862^7867. 
Komori Y, Nonogaki T, Nikai T: Hemorrhagic activity and muscle damaging effect of Pseudomonas 

aeruginosa metalloproteinase (elastase). Toxicon 2001;39:1327-1332. 
Koonin EV, Aravind L: Origin and evolution of eukaryotic apoptosis: The bacterial connection. Cell 

Death Differ 2002;9:394-^04. 
Kosowska K, Reinholdt J, Rasmussen LK, Sabat A, Potempa J, Kilian M, Poulsen K: The Clostridium 

ramosum IgA proteinase represents a novel type of metalloendopeptidase. J Biol Chem 2002;277; 

11987-11994. 
Kramer RA, Dekker N, Egmond MR: Identification of active site serine and histidine residues in 

Escherichia coli outer membrane protease OmpT FEBS Lett 2000;468:220-224. 
Kramer RA, Vandeputte-Rutten L, De Roon GJ, Gros P, Dekker N, Egmond MR: Identification of essential 

acidic residues of outer membrane protease OmpT supports a novel active site. FEBS Lett 2001; 

505:426^30. 



Bacterial Peptidases 



73 



Kukkonen M, Lahteenmaki K, Suomalainen M, Kalkkinen N, Emody L, Lang H, KorhonenTK: Protein 

regions important for plasminogen activation and inactivation of alpha2-antiplasmin in the surface 

protease Pla of Yersinia pes Us. Mol Microbiol 2001;40:1097-1 111. 
Kukkonen M, Suomalainen M, Kyllonen P, Lahteenmaki K, Lang H, Virkola R, Helander IM, Hoist O, 

Korhonen TK: Lack of O-anligen is essential for plasminogen activation by Yersinia pestis and 

Salmonella enterica. Mol Microbiol 2004;51:215-225. 
Kumagai Y, Konishi K, Gomi T, Yagishita H, Yajima A, Yoshikawa M: Enzymatic properties of dipeptidy I 

aminopeptidase IV produced by the periodontal pathogen Porphyromonas gingival is and it5 partici- 
pation in virulence. Infect Immun 2000;68:716-724. 
Kumagai Y, Yajima A, Konishi K: Peptidase activity of dipeptidyl aminopeptidase IV produced by 

Pofphyromonas gingivalis is important but not sufficient for virulence. Microbiol Immunol 2003; 

47:735-743. 
Kuroda A, Sekiguchi J: Molecular cloning and sequencing of a major Bacillus subiilis autolysin gene. 

JBacteriol 1991;173:7304-7312, 
Labrou NE, Rigden DJ: The structure-function relationship in the clostripain family of peptidases, Eur 

J Biochem 2004;271:983-992. 
Ladds G, Davey J: Identification of proteases with shared functions to the proprotein processing 

protease KrpI in the fission yeast Schizosaccharomyces pombe. Mol Microbiol 2000;38: 

839-853. 
Lahteenmaki K, Kukkonen M, Korhonen TK: The Pla surface protease/adhesin of Yersinia pestis mediates 

bacterial invasion into human endothelial cells. FEBS Lett 2001;504:69-72. 
LangerT; AAA proteases; Cellular machines for degrading membrane proteins. Trends Biochem Sci 

2000;25:247-25 L 
Lathem WW, Grys TE, Witowski SE, Torres AG, Kaper JB, Tarr PI, Welch RA; StcE, a metalloprotease 

secreted by Escherichia coli OI57:H7, specifically cleaves CI esterase inhibitor. Mol Microbiol 

2002;45:277-288. 
Lei B, DeLeo FR, Hoe NP, Graham MR, Mackie SM, Cole RL, Liu M, Hill HR, Low DE, Federle MJ, 

Scott JR, Musser JM: Evasion of human mnafe and acquired irmnunity by a bacterial homolog of 

GDI lb that inhibits opsonophagocytosis. Nat Med 2001;7:1298-1305. 
Lei B, DeLeo FR, Reid SD, Voyich JM, Magoun L, Liu M, Braughton KJl, Ricklefs S, Hoe NP, Cole RL, 

Leong JM, Musser JM: Opsonophagocytosis-inhibiting mac protein of group A Streplococciis: 

Identification and characteristics of two genetic complexes. Infect Immun 2002;70:6880-6890. 
Lei B, Liu M, Meyers EG, Manning HM, Nagiec MJ, Musser JM: Histidine and aspartic acid residues 

important for immunoglobulin G endopeptidase activity of the group A Streptococcus 

opsonophagocytosis-inhibiting Mac protein. Infect Immun 2003;71:2881-2884. 
Li JY, Cui YM,Chen LL, Gu M, Li J, Nan FJ,Ye QZ: Mutations at the SI sites of methionine aminopep- 

tidases from Escherichia co/Zand Homo sapiens reveal the residues critical for substrate specificity. 

J Biol Chem 2004;279:21 128-21 134. 
Liles MR, Edelstein PH, Cianciotto NP: The prepilin peptidase is required for protein secretion by and the 

virulence of the \n{v^zQ\\\i\m' }^i^\{Qg^n Legionella pneumophila . Mol Microbiol 1999;31:959-970. 
Lin L, Ayala P, Larson J, Mulks M, Fukuda M, Carlsson SR, Enns C, So M: The Neisseria type 2 IgA 1 

protease cleaves LAMPl and promotes survival of bacteria within epithelial cells, Mol Microbiol 
1997;24:1083-1094. 

Lindsay JA, Foster SJ: Interactive regulatory pathways control virulence determinant production and 

stability in response to environmental conditions in Staphylococcus aureus. Mol Gen Genet 1999; 

262:323-331. 
Lithgow JK, Ingham E, Foster SJ: Role of the hprT-ftsH locus in Staphylococcus aureus. Microbiology 

2004;150:373-381. 
Liu TV, Elliott SD: Activation of streptococcal proteinase and its zymogen by bacterial cell walls. Nature 

1965a;206:33-34. 
Liu TY, Elliott SD: Streptococcal proteinase: The zymogen to enzyme transfromation. J Biol Chem 

1965b;240:l 138-1 142. 
Lourbakos A, Chinni C, Thompson P, Potempa J, Travis J, Mackie EJ, Pike RN: Cleavage and activation 

of proteinase-activated receptor-2 on human neutrophils by gingipain-R from Porphyromonas 

gingivalis, FEBS Utt 1998;435:45^8, 



Potempa/Pike 



174 



Lourbakos A, Potempa J, Travis J, D'Andrea MR, Andrade-Gordon P, Santulli R, Mackie EJ, Pike RN: 
Arginine-specific protease Trom Porphyromonas gingivalis activates protease-activated receptors 
on human oral epithelial cells and induces interleukin-6 secretion. Infect Immun 2001 a;69: 
5121-5130. 

Lourbakos A, Yuan YP, Jenkins AL, Travis J, Andrade-Gordon P, Santulli R, Potempa J, Pike RN: 
Activation of protease-activated receptors by gingipains from Porphyromonas gingivalis leads to 
platelet aggregation: A new trait in microbial pathogenicity. Blood 200lb;97:3790-3797. 

Lovgren A, Zhang M, Engstrom A, Dalhammar G, Landen R: Molecular characterization of immune 
inhibitor A, a secreted virulence protease from Bacillus thuringiensis. Mol Microbiol I990;4: 
2137-2146. 

Lukomski S, Burns EH Jr, Wyde PR, Podbielski A, Rurangirwa J, Moore-Poveda DK, Musser JM: Genetic 
inactivation of an extracellular cysteine protease (SpeB) expressed by Sirepiococcus pyogenes 
decreases resistance to phagocytosis and dissemination to organs. Infect Immun 1998;66:771-776. 

Lukomski S, Montgomery CA, Rurangirwa J, Geske RS, Barrish JP, Adams GJ, Musser JM: 
Extracellular cysteine protease produced by Streptococcus pyogenes participates in the pathogen- 
esis of invasive skin infection and dissemination in mice. Jnfect Immun 1999;67:1779-1788, 

Lukomski S, Sreevatsan S, Amberg C, Reichardt W, Woischnik M, Podbielski A, Musser JM; 
Inactivation o^ Streptococcus pyogenes extracellular cysteine protease significantly decreases 
mouse lethality of serotype M3 and M49 strains. J Clin Invest 1997;99:2574-2580. 

Madden TE, Clark VL, Kuramitsu UK: Revised sequence of the Porphyromonas gingivalis prtT cysteine 
protease/hemaggluiinin gene: Homology with streptococcal pyrogenic exotoxin B/streptococcal 
proteinase. Infect Immun 1995;63:238-247, 

Madden JC, Ruiz N, Caparon M: Cytolysin-mediated translocation (CMT): A functional equivalent of 
type 111 secretion m gram-positive bacteria. Cell 2001 ;104: 143-152. 

Makinen PL, Makinen KK: The Enterococcus faecal is extracellular metalloendopeptidase (EC 3.4.24.30; 
coccolysin) inactivates human endolhelin at bonds involving hydrophobic amino acid residues. 
Biochem Biophys Res Commun 1994;200:981-985. 

Manoury B, Hewitt EW^ Morrice N, Dando PM, Barren AJ, Watis C: An asparagmyl endopeptidase 
processes a microbial antigen for class II MHC presentation. Nature 1998;396:695-699. 

Mariencheck Wl, Alcorn JF, Palmer SM, Wright JR: Pseudomonas aeruginosa elastase degrades 
surfactant proteins A and D, Am J Respir Cell Mol Biol 2003;28:528-537. 

Marsh JW, Taylor RK: Identification of the Vibrio cholerae type 4 prepilin peptidase required for cholera 
toxin secretion and pilus formation. Mol Microbiol 1998;29:1481-1492. 

Mazmanian SK, Liu G, Ton-That H, Schneewind O: Staphylococcus aureus sortase, an enzyme that 
anchors surface proteins to the cell wall. Science 1999;285:760-763. 

Mazmanian SK, Skaar EP, Caspar AH, Humayun M, Gornicki P, Jelenska J, Joachmiak A, Missiaka5 
DM, Schneewind O: Passage of heme-iron across the envelope of Staphylococcus aureus. Science 
2003;299:906-909. 

Mazmanian SK, Ton-That H, Su K, Schneewind O: An iron-regulated sortase anchors a class of 
surface protein during Staphylococcus aureus pathogenesis. Proc Natl Acad Sci USA 2002;99: 
2293-2298. 

McAleese FM, Walsh EJ, Sieprawska M, Potempa J, Foster TJ: Loss of clumping factor B fibrinogen 
binding activity by Staphylococcus aureus involves cessation of transcription, shedding and cleav- 
age by metalloprotease. J Biol Chem 2001;276:29969-29978. 

McGrath WJ, Ding J, Didwania A, Sweet RM, Mangel WF: Crystallographic structure at 1 .6-A resolu- 
tion of the human adenovirus proteinase in a covalent complex with its I l-amino-acid peptide 
cofactor: Insights on a new fold. Biochim Biophys Acta 2003;1648:1-1 I. 

Mekalanos JJ: Environmental signals controlling expression of virulence determinants in bacteria. 
J Bacteriol 1992;174:1-7, 

Melchior F, Schergaut M, Pichler A: SUMO: Ligases, isopeptidases and nuclear pores. Trends Biochem 
Sci 2003;28:612-628. 

Michiels J, Dirix G, Vanderleyden J, Xi C: Processing and export of peptide pheromones and bacteri- 
ocins in Gram-negative bacteria. Trends Microbiol 2001;9:164-168. 

Mikolajczyk J, Boatright KM, Stennicke HR, Nazif T, Potempa J, Bogyo M, Salvesen GS: Sequential 
autolytic processing activates the zymogen of Arg-gingipain. J Biol Chem 2003;278:10458-10464. 



Bacterial Peptidases 175 



Mikolajczyk-Pawlinska J, Travis J, Potempa J: Modulation of interleukin-8 activity by gingipains from 

Porphyromonas glngivalis: Implications for pathogenicity of periodontal disease. FEBS Lett 

1998;440:282-286. 
Montecucco C, Schiavo G: Mechanism of action of tetanus and botulinum neurotoxins. Mol Microbiol 

1994;13:1-8. 
Morihara K,Tsuzuki H, Oda K: Protease and elastase of Pseudowonas aeruginosa: Inactivation of human 

plasma alpha- 1 -proteinase inhibitor Infect Lmmun 1979;24:188-193. 
Moriyama R, Kudoh S, Miyata S, Nonobe S, Hattori A, Makino S: A germination-specific spore cortex- 

lytic enzyme from Bacillus cereus spores: Cloning and sequencing of the gene and molecular 

characterization of the enzyme. J Bacteriol 1996;178:5330-5332, 
MuDer S, Ledl A, Schmidt D: SUMO: A regulator of gene expression and genome integrity. Oncogene 

2004;23:1998-2008. 
Navarre WW, Ton-That H, Faull ICF, Schneewind 0; Multiple enzymatic activities of the murein hydro- 
lase from staphylococcal phage phi II. Identification of a D-alanyl-glycine endopeptidase activity, 

J Biol Chem 1999;274:15847-15856. 
Nelson D, Potempa J, Kordula T, Travis J: Purification and characterization of a novel cysteine proteinase 

(periodonLain) from Porphyromonas gingivalis. Evidence for a role in the inactivation of human 

alphal -proteinase inhibitor. J Biol Chem 1999;274:12245-12251. 
Nelson D, Potempa J, Travis J: Inactivation ofalphal -proteinase inhibitor as a broad screen for detecting 

proteolytic activities in unknowTi samples. Anal Biochem 1998;260:230-236. 
Neumann VC, Heath HE, LeBlanc PA, Sloan GL: Extracellular proteolytic activation of bacteriolytic 

peptidoglycan hydrolases o'i Staphylococcus simulans hiovwr staphylolyticus. FEMS Microbiol 

Lett 1993; 110:205-211. 
Nyarko KA, Coomber BL, Mellors A, Gentry PA: Bovine platelet adhesion is enhanced by leukotoxin and 

sialoglycoprolease isolated from PasteurellahaemolyticaA] cultures. Vet Microbiol 1998;61:81-91. 
O'Brien-Simpson NM, Paolini RA, Hoffmann B, Slakeski N, Dashper SG, Reynolds EC: Role of RgpA, 

RgpB, and Kgp proteinases in virulence of Porphyromonas gingivalis W50 in a murine lesion 

model. Infect bnmun 2001;69:7527-7534. 
Odagaki Y, Hayashi A, Okada K, Hirotsu K, KabashimaT, Ito K, YoshimotoT, Tsuru D, Sato M, Clardy J: 

The crystal structure of pyroglutamyl peptidase I from Bacillus amyloliquefaciens reveals a new 

structure for a cysteine protease. Structure Fold Des 1999;7:399^1 1. 
Oefher C, Douangamath A, D'Arcy A, Hafeli S, Mareque D, Mac Sweeney A, Padilla J, Pierau S, Schuiz H, 

Thormann M, Wadman S, Dale GE: The 1.15A crystal structure of the Staphylococcus aureus 

methionyl-aminopeptidase and complexes with triazole based inhibitors. J Mol Biol 2003;332: 

13-21. 
Oleksy A, Banbula A, Bugno M, Travis J, Potempa J: Proteolysis of interleukin-6 receptor (IL-6R) by 

Porphyromonas gingivalis cysteine proteinases (gingipains) inhibits interleukin-6-mediated cell 

activation. Microb Pathog 2002;32:173-181. 
Oleksy A, Ewa Golonka E, Banbula A, Szmyd G, Moon J, Kubica M, Bogyo M, Foster TJ, Travis J, 

Potempa J: Growth-phase dependent production of a cell wall-associated elastinolytic cysteine 

proteinase by Staphylococcus epidermidis, Biol Chem, in press. 
Orth K, Xu Z, Mudgett MB, Bao ZQ, Palmer LE, Bliska JB, Mangel WF, Staskawicz B, Dixon JE: 

Disruption of signaling by Yersinia effector YopJ, a ubiquitin-like protein protease. Science 2000; 

290:1594-1597, 
Orth K: Function of the Yersinia effector YopJ. Curr Opin Microbiol 2002;5:38^3. 
Ossovskaya VS, Bunnett NW: Protease-activated receptors: Contribution to physiology and disease. Physiol 

Rev 2004;84:579-621. 
Pillar CM, Hazlett LD, Hobden JA: Alkaline prolease-deficient mutants of Pseudomonas aeruginosa are 

virulent in the eye. Curr Eye Res 2000;21:730-739. 
Potempa J, Banbula A, Travis J: Role of bacterial proteinases in matrix destruction and modulation of 

host responses. Periodontol 2000 2000;24:153-192. 
Potempa J, Fedak D, Dubin A, Mast A, Travis J: Proteolytic inactivation of alpha- 1-anli-chymotrypsin. 

Sites of cleavage and generation of chemotactic activity. J Biol Chem 1991;266:21482-21487. 
Potempa J, Pavloff N.Travis J: Porphyromonas gingivalis: A proteinase/gene accounting audit. Trends 

Microbiol 1995;3:430-^34. 



Potempa/Pike 



176 



Potempa J, Pike R, Travis J: Titration and mapping of the active site of cysteine proteinases from 
Porphyromonas gingivalis (gingipains) using peptidyl chloromethanes, Biol Chem 1997;378: 
223-230. 

Potempa J, Sroka A, Imamura T, Travis J: Gingipains, the major cysteine proteinases and virulence fac- 
toi"s of Porphyromonas gingivalis: Structure, function and assembly of multidomain protein com- 
plexes. Curr Protein Pept Sci 2003;4:397^07. 

Potempa J, Watorek W, Travis J: The inactivation of human plasma alpha 1 -proteinase inhibitor by pro- 
teinases from Staphylococcus aureus. J Biol Chem I986;26I:M330^I4334, 

Prager R, Mirold S,Tietze E, Strutz U, Knuppel B, Rabsch W, Hardt WD, Tschape H: Prevalence and poly- 
morphism of genes encoding translocated effector proteins among clinical isolates of^ Salmonella 
enterica. Int J Med Microbiol 2000;290:605-617. 

Prasad L, LeducY, Hayakawa K, Delbaere LT: The structure of a universally employed enzyme: V8 protease 
from Staphylococcus aureus. Acta Crystal logr D Biol Crystal logr 2004;60;256-259, 

Raeder R, Harokopakis E, HoMingshead S, Boyle MD: Absence of SpeB production in virulent large 
capsular forms of group A streptococcal strain 64, Infect fmmun 2000;68:744— 751. 

Raeder R, Woischnik M, PodbieJski A, Boyle MD: A secreted streptococcal cysteine protease can cleave 
a surface-expressed Ml protein and alter the immunoglobulin binding properties. Res Microbiol 
1998;149:539-548. 

Rajapakse PS, O'Brien-Simpson NM. Slakeski N, Hoffinann B, Reynolds EC: Immunization with the 
RgpA-Kgp proteinase-adhesin complexes of Porphyromonas gingivalis protects against periodontal 
bone loss in the rat periodontitis model. Infect Immun 2002;70:2480-2486. 

Rasmussen M, Bjorck L: Proteolysis and its regulation at the surface of Streptococcus pyogenes. Mol 
Microbiol 2002;43:537-544. 

Rasmussen M, Muller HP, Bjorck L: Protein GRAB of Streptococcus pyogenes regulates proteolysis at 
the bacterial surface by binding alpha2-macroglobulin. J Biol Chem 1999;274: 15336-15344. 

Rawlings ND, Bai'rett AJ: Families of serine peptidases. Methods Enzymol 1994;244:19-61. 

Rawlings ND, Tolle DP, Barrett AJ: MEROPS: the peptidase database. Nucleic Acids Res 2004;32 
(Database issue):DI60-D164. 

Reed SB, Wesson CA, Liou LE, Trumble WR, Schlievert PM, Bohach GA, Bayles KW: Molecular char- 
acterization of a novel Staphylococcus aureus serine protease operon. Infect Immun 2001 ;69: 
1521-1527. 

Reglier-PoupetH, Frehel C, Dubail I, Beretti JL, Berche P, Charbit A, Raynaud C: Maturation of lipopro- 
teins by type II signal peptidase is required for phagosomal escape of Listeria monocytogenes, J Biol 
Chem 2003;278:49469^9477. 

Reinholdt J, Kilian M: Lack of cleavage of immunoglobulin A (IgA) from rhesus monkeys by bacterial 
IgAl proteases. Infect Immun 1991;59:2219-2221. 

Rice K, Peralta R, Bast D, de Azavedo J, McGavin MJ: Description of staphylococcus serine protease 
(ssp) operon in Staphylococcus aureus and nonpolar inactivation of sspA-encoded serine protease. 
Infect Immun 2001;69:159-169. 

Riegler M, Loiz M, Sears C, PothoulakJs C, Castagliuolo I, Wang CC, Sedivy R, SogiikogluT, Cosentini E, 
Bischof G, Feil W.Teleky B, Hamilton G, LaMont JT, Wenzl E: Bacteroides jragiUs toxin 2 damages 
human colonic mucosa in vitro. Gut 1999;44:504-510. 

Riley MA, Wertz JE: Bacteriocins: Evolution, ecology, and application. Annu Rev Microbiol 2002;56: 
117-137. 

Rossier O, Starkenburg SR, Cianciono NP: Legionella pneumophila type II protein secretion promotes 
virulence in the A/J mouse model of Legionnaires' disease pneumonia. Infect Immun 2004;72: 
310-321. 

Rzychon M, Filipek R, Sabat A, Kosowska Y^ Dubin A, Potempa J, Bochtler M: SLaphostatins resemble 
lipocalins, not cystatins in fold. Protein Sci 2003;12:2252-2256, 

Rzychon M, Sabat A, Kosowska K, Potempa J, Dubin A: Staphostatins: An expanding new group of pro- 
temase inhibitors with a unique specificity for the regulation of staphopains. Staphylococcus spp. 
cysteine proteinases. Mol Microbiol 2003;49:1051-1066. 

Sahney NN, Summersgill JT, Ramirez JA, Miller RD: Inhibition of oxidative burst and chemotaxis in 
human phagocytes by Legionella pneumophila zinc metalloprotease. J Med Microbiol 2001 ;50: 
517-525. 



Bacterial Peptidases 177 



Sakata Y, AkaikeT, Suga M, Ijiri S, Ando M, Maeda H: Bradykinin generation triggered by Pseudomonas 
proteases facilitates invasion of the systemic circulation by Pseudomonas aeruginosa. Microbiol 
[mmunol 1996;40:415^23. 

Sanfilippo L, Li CIC, Seth R, BaKvin TJ, Menozzi MG, Mahida YR: Bacteroides fragilis enterotoxin 
induces the expression of lL-8 and traJisforn:iing growth factor-beta (TGF-bet^) by human colonic 
epithehal cells. Clin Exp Immunol 2000;! 19:456^63. 

Sarandeses CS, Covelo G, Diaz-Jullien C, Freire M: Prothymosin a is processed to thymosin al and 
thymosin all by a lysosomal asparaginyl endopeptidase. J Biol Chem 2003;278: 13286-13293. 

Schiavo G, Benfenati F, Poulain B, Rossetto O, Polverino de Laureto P, DasGupta BR, Montecucco C: 
Tetanus and botulinum-B neurotoxins block neurotransmitter release by proteolytic cleavage of 
synaptobrevin. Nature )992a;359:832-835. 

Schiavo G, Poulain B, Rossetto O, Benfenati F, Tauc L, Montecucco C: Tetanus toxin is a zinc protein 
and its inhibition of neurotransmitter release and protease activity depend on zinc, EMBO J 
1992b;ll:3577-3583. 

Schmidtchen A, Frick IM, Andersson E, Tapper H, Bjorck L: Proteinases of common pathogenic bacteria 
degrade and inactivate the antibacterial peptide LL-37. Mol MicrobioJ 2002;46:157-168. 

Schmidtchen A, Frick IM, Bjorck L: Dermalan sulphate is released by proteinases of common patho- 
genic bacteria and inactivates antibacterial alpha-defensin. Mol Microbiol 2001;39:708-713. 

Schumann W: FtsH -A single-chain charonin? FEMS Microbiol Rev 1999;23:1-1 1. 

Scott CF, Whitaker EJ, Hammond BF, Colman RW: Purification and characterization of a potent 70-kDa 
thiol lysyl-proteinase (Lys-gingivain) from Porphyromonas gingivalis that cleaves kininogens and 
fibrinogen. J Biol Chem 1993;268;7935-7942. 

Sears CL: The toxins of Bacferoides fragilis, Toxicon 2001;39:1737-1746. 

Shanley TP, Schrier D, Kapur V, Kehoe M, Musser JM, Ward PA: Streptococcal cysteine protease augments 
lung injury induced by products of group A streptococci. Infect Immun 1996;64:870-877. 

Shao F, Golstein C, Ade J, Stoutemyer M, Dixon JE, Innes RW: Cleavage of Arabidopsis PBSl by a bac- 
terial type 111 effector. Science 2003a;301: 1230-1 233. 

Shao F, Merritt PM, Bao Z, Iruies RW^ DLxon JE: A Yersinia effector and a Pseudomonas avimlence protein 
define a family of cysteine proteases functioning in bacterial pathogenesis. Cell 2002;109:575-588. 

Shao F, Vacratsis PO, Bao Z, Bowers KE, Fierke CA, Dixon JE: Biochemical characterization of the 
Yersinia YopT protease: Cleavage site and recognition elements in Rho GTPases, Proc Natl Acad 
Sci USA 2003b; 100:904-909. 

Shaw L, Golonka E, Potempa J, Foster SJ: The role and regulation of the extracellular proteases of 
Staphylococcus aureus. Microbiology 2004;150:217-228. 

Shere KD, Salluslio S, Manessis A, D^Aversa TG, Goldberg MB: Disruption of IcsP, the major Shigella 
protease that cleaves IcsA, accelerates actin-based motility. Mol Microbiol 1997;25:451^62. 

Shet A, ICaplan EL, Johnson DR, Cleary PP: Immune response to group A streptococcal C5a peptidase 
m children: Implications for vaccine development. J Infect Dis 2003;188:809-817. 

Shirahama-Noda K, Yamamoto A, Sugihara K, Hashimoto N, Asano M, Nishimura M, Hara-Nishimura 
I: Biosynlhetic processing of cathepsins and lysosomal degradation are abolished in asparaginyl 
endopeptidase-deficient mice. J Biol Chem 2003;278:33194-33199. 

Sodeinde OA, Subrahmanyam YV, Stark K, Quan T, Bao Y, Goguen JD: A surface protease and the inva- 
sive character of plague. Science 1992;258:1004-1007. 

Staskawicz BJ, Mudgett MB, Dangl JL, Galan JE: Common and contrasting themes of plant and animal 
diseases. Science 2001;292:2285-2289. 

Stathopoulos C: Structural features, physiological roles and biotechnological applications of the mem- 
brane proteases of the OmpT bacterial endopeptidase family: A micro-review. Membr Cell Biol 
1998;12:1-8. 

Stockbauer ICE, Magoun L, Liu M, Bums EH Jr, Gubba S, Renish S, Pan X, Bodary SC, Baker E, 
Cobum J, Leong JM, Musser JM: A natural variant of the cysteine protease virulence factor of 
group A Streptococcus with an arginine-glycine-aspartic acid (RGD) motif preferentially binds 
human integrins a^^^ and a„b33. Proc Natl Acad Sci USA 1999;96:242-247. 

Stumpe S, Schmid R, Stephens DL, Georgiou G, Bakker EP: Identification of OmpT as the protease that 
hydrolyzes the antimicrobial peptide protamine before it enters growing cells of Escherichia coli. 
JBacteriol 1998;180:4002-4006. 



Potempa/Pike 



178 



Sugawara S.NemotoE, TadaH.Miyake K, ImamuraT, TakadaH: Proteolysisof human monocyte CD 14 

by cysteine proteinases (gingipains) from Porphyromonas gingival is leading to lipopolysaccharide 

hyporesponsiveness. J Immunol 2000; 1 65:4 1 1^18. 
Sutherland DR, Abdullah KM, Cyopick P, Mellors A: Cleavage of the cell-surface O-sialoglycoproteins 

CD34, CD43, CD44, and CD45 by a novel glycoprotease from PasteureUa haemolytica. J Immunol 

1992;148:1458-1464. 
Svensson MD, ScaramuzzinoDA, Sjobring U, Olsen A, Frank C, Bessen DE: Role for a secreted cysteine 

proteinase in the establishment of host tissue tropism by group A streptococci, Mol Microbiol 

2000;38:242-253. 
Tada H, Sugawara S, Nlemoto E, ImamuraT, Potempa J,Travis J, Shimauchi H,Takada H: Proteolysisof 

ICAM-I on human oral epithelial cells by gingipains. J Dent Res 2003;82:796-801. 
Tada H, Sugawara S, "Nemolo E, Takahashi N, Imamura T, Polempa J, Travis J, Shimauchi H, Takada H: 

Proteolysis of CD 1 4 on human gingival fibroblasts by arginine-specific cysteine proteinases from 

Porphyfvmonas gingivalis leading to down-regulation of lipopolysaccharide-induced interleukin-8 

production. Infect Immun 2002;70:3304^3307, 
Takaya A, Suzuki M, Matsui H, Tomoyasu T, Sashinami H, Nakane A, Yamamoto T: ton, a stress- 
induced ATP-dependent protease, is critically important for systemic Salmonella enlerica serovar 

typhimiinum infection of mice. Infect Immun 2003;71:690-696. 
Tapper H, Herwald H: Modulation of hemostatic mechanisms in bacterial infectious diseases. Blood 

2000;96:2329-2337. 
Ton-That H, Mazmanian SK, Alksne L, Schneewind O: Anchoring of surface proteins to the cell wall of 

Staphylococcus aureus. Cysteine 184 and hislidine 120 of sortase form a thiolate-imidazolium ion 

pair for catalysis. J Biol Chem 2002;277:7447-7452, 
Ton-That H, Schneewind O: Assembly of pili on the surface of Corynebaaeriwn diphfheriae, Mol 

Microbiol 2003;50:1429-1438. 
Towbin H, Bair KW, DeCaprio JA, Eck MJ, Kim S, Kinder FR, Morollo A, Mueller DR, Schindler P, 

Song HK, van Oosti'um J, Versace RW, Voshol H, Wood J, Zabludoff S, Phillips PE: Proteomics- 

based target identification: Bengamides as a new class of methionine aminopeptidase inhibitors. 

J Biol Chem 2003;278:52964-52971. 
Vandeputte-Rulten L, Kramer RA, Kroon J, Dekker M, Egmond MR, Gros P: Crystal structure of the 

outer membrane protease OmpT from Escherichia coli suggests a novel catalytic site. EMBO J 

2001;20:5033-5039. 
Veith PD, Talbo GH, Slakeski N, Dashper SG, Moore C, Paolini RA, Reynolds EC: Major outer membrane 

proteins and proteolytic processing of RgpA and Kgp of Porphyromonas gingivalis W50. Biochem 

J 2002;363:105-1 15. 
Vesanto E, Varmanen P, Steele JL, Palva A: Characterization and expression of the Lactobacillus 

helveticus pepC gene encoding a general aminopeptidase. Eur J Biochem 1994;224;99 1-997. 
Vitovski S, Read RC, Sayers JR: Invasive isolates of Neisseria meningitidis possess enhanced 

immunoglobulin A 1 protease activity compared to colonizing strains. FASEB J 1999;13:331-337. 
von Pawel-Rammingen U, Bjorck L: IdeS and SpeB: Immunoglobulin-degrading cysteine proteinases of 

Strepiococcvs pyogenes. Curr Opin Microbiol 2003;6:50^55, 
von Pawel-Rammingen U, Johansson BP, Bjorck L: IdeS, a novel streptococcal cysteine proteinase with 

unique specificity tor immunoglobulin G. EMBO J 2002a;2 1:1607-1 61 5. 
von Pawel-Rammingen U, Johansson BP, Tapper H, Bjorck L: Streptococcus pyogenes and phagocytic 

killing. Nat Med 2002b;8: 104^1 045. 
Walker KE, Moghaddame-Jafari S, Lockatell CV, Johnson D, Belas R: ZapA, the IgA-degrading metal- 

loprotease of Proteus mirabilis, is a virulence factor expressed specifically in swarmer cells. Mol 

Microbiol 1999;32:825-836. 
Wang JM, Hartling JA, Flanagan JM: The structure of ClpP at 2.3 A resolution suggests a model for 

ATP-dependent proteolysis. Cell 1997;91:447^56. 
Waters CM, Antiporta MH, Murray BE, Dunny GM: Role of the Enterococciis faecalis GelE protease 

in determination of cellular chain length, supernatant pheromone levels, and degradation of fib- 
rin and misfolded surface proteins. J Bacteriol 2003;185:3613-3623, 
Webb RM, Lundigran MD: OmpT in Escherichia coli correlates with severity of disease in urinary tract 

infections. Med Microbiol Lett 1996;5:8-14. 



Bacterial Peptidases 179 



Weichart D, Querflirth N, Dreger M, Hengge-Aronis R: Global role for ClpP-containing proteases in sta- 
tionary-phase adaptation of Escherichia coli, J Bacteriol 2003; 185:1 15-125. 

Wiens M, Krasko A, Perovic S, Muller WE: Caspase-mediated apoptosis in sponges: Cloning and func- 
tion of the phylogenetic oldest apoptotic proteases from Melazoa. Biochim Biophys Acta 
2003;1593:179-189. 

Wingrove JA, DiScipio RG, Chen Z, Potempa J, Travis J, Hugh TE: Activation of complement compo- 
nents C3 and C5 by a cysteine proteinase (gingipain- 1 ) from Porphyromonas (Bacieroides) gingi- 
valis. J Biol Chem 1992;267:18902-18907. 

Wine y Wolf N, Dargatz H: Clostripain hnker deletion vainants yield active enzyme in Escherichia coli: 
A possible function of the linker peptide as inn^molecular inhibitor of clostripain automaturation, 
Curr Microbiol l996;33(5):28l-286. 

Witle V, Wolf N, Diefenthal T, Reipen G, Dargatz H: Heterologous expression of (he clostripain gene 
from Closiridium hisiolyticum in Escherichia coli and Bacillus sublilis: Maturation of the clostri- 
pain precursor is coupled with self-activation. Microbiology 1994; 140:1 175-1 182, 

Wolf BB, Gibson CA, KapurV, Hussaini IM, Musser JM, Gonias SL: Proteolytically active streptococcal 
pyrogenic exotoxin B cleaves monocytic cell urokinase receptor and releases an active fragment of 
the receptor from the cell surface. J Biol Chem 1994;269:30682-30687. 

Wong P, Houry WA: Chaperone networks in bacteria: Analysis of protein homeostasis in minimal cells, 
J Struct Biol 2004;146:79-89. 

Wretlind B, Wadstrom T: Purification and properties of a protease with elastase activity from 
Pseudomonas aeruginosa. J Gen Microbiol 1977;103:319-327. 

Wu S, Lim KC, Huang J, Saidi RF, Sears CL: Bacteroides fragilis enterotoxin cleaves the zonula 
adherens protein, E-cadherin. Proc Natl Acad Sci USA 1998;95:14979-14984, 

Yamamoto H, Kurosawa S, Sekiguchi J: Localization of the vegetative cell wall hydrolases LytC, LytE, 
and LytF on the Bacillus sublilis cell surface and stability of these enzymes to cell wall-bound or 
extracellular proteases. J Bacteriol 2003; 185:6666— 6677. 

Yanagihara K, Tomono K, KanekoY, Miyazaki Y,Tsukamoto K, Hirakata Y, Mukae H, Kadota J, Murata 
1, Kohno S: R^ole of elastase iji a mouse model of chronic respiratory Pseudomonas aeruginosa 
infection that mimics diffuse panbronchiolitis. J Med Microbiol 2003;52:531-535. 

Yeh ET, Gong L, Kamitani T: Ubiquitin-like proteins: New wines in new bottles. Gene 2000;248: 1-14. 

Yoon S, Liu Z, EyoboY, Orth K: Yersinia effector YopJ inhibits yeast MAPK signaling pathways by an 
evolutionarily conserved mechanism. J Biol Chem 2003;278:2131-2135. 

Zhang HZ, Hackbarth CJ, Chansky KM, Chambers HF: A proteolytic transmembrane signaling pathway 
and resistance to beta-lactams in staphylococci. Science 2001;291:1962-1965. 

Zhu M, Shao F, Innes RW, Dixon JE, Xu Z: The crystal structure of Pseudomonas avirulence protein 
AvrPphB: A papain-like fold with a distinct substrate-binding site, Proc Natl Acad Sci USA 
2004;101:302-307. 

Ziebandt AK, Weber H, Rudolph J, Schmid R, Hoper D, Engelmarm S, Hecker M: Extracellular proteins 
of Staphylococcus aureus and the role of SarA and sigma B. Proteomics 2001;l:480-^93. 

Zong Y, Mazmanian SK, Schneewind O, Narayana SV: The structure of sortase B, a cysteine transpep- 
tidase that tethers surface protein to the Staphylococcus aureus cell wall. Structure 2004; 
12:105-112. 



Jan Potempa 

Department of Microbiology, Faculty of Biotechnology 

Jagiellonian University 

ul. Gronostajowa 7 

PL-30-387 Krak6w (Poland) 

Tel. +48 12 664 6343, Fax +48 12 664 6902, E-Mail poiempa@arches.uga.edu 



Potempa/Pike 



180 



Enzymes 

Russell W, Herwald H (eds): Concepts in Bacterial Virulence.! 
Contrib Microbiol. Basel, Karger, 2005, vol 12, pp 181 209 



Bacterial Invasins: Molecular Systems 
Dedicated to the Invasion 
of Host Tissues 



Eric D. Cambronne, Olaf Schneewind 

Committee on Microbiology, University of Chicago, Chicago, 111., USA 



Bacterial pathogens have devised several strategies for their survival in 
the tissues of vertebrate hosts. Some of these strategies are common to a wide 
distribution of bacterial species, while others are quite speciaHzed and unique 
to a particular pathogen. The term invasin has been traditionally assigned to 
virulence factors that specifically promote Lntemalization of a bacterium by a 
host cell. This designation may also be collectively assigned to general viru- 
lence strategies required for host colonization. A typical pathogen must use 
one or a combination of mechanisms to colonize the host. Factors that pro- 
mote colonization can be functionally quite diverse, from the release of a 
toxin into its surrounding environment, to the display of an individual surface 
ligand promoting receptor binding on a host cell. Many pathogens have 
evolved specialized macromolecular structures dedicated to the delivery of 
effector molecules directly into the cytosol of target cells. These injection sys- 
tems bypass the requirement for association of a toxin with a target through 
diffusion, and therefore appear to be highly efficient pathogenic strategies. 
Collectively, these meclianisms allow the pathogen Lo maniptilate molecular 
processes of host cells to promote adhesion, cytotoxicity, in some cases 
phagocytosis, and often general subversion of both the innate and adaptive 
immune systems. The end result is the establishment of an environmental 
niche in host tissues that will allow for the perpetuation of the bacterium. The 
secretion of polypeptides from the bacterial cytosol to targets in or beyond the 
cell wall envelope is a requirement common amongst most pathogenic strate- 
gies. Generalized secretion pathways are utilized or modified to accommo- 
date these virulence strategies, resulting in specialized systems dedicated to 
the invasion of host tissues. 



The Bacterial Cell Wall 

The bacterial cell wall envelope provides the molecular scaffolding for the 
display of virulence factors and also provides the framework for the assembly 
of dedicated secretion systems. With few exceptions, bacteria may be catego- 
rized based on the composition and morphology of their cell wall. All bacteria 
contain an inner cytosolic compartment that is surrounded by a phospholipid 
membrane. Morphological distinctions become apparent beyond this primary 
barrier. In gram-positive bacteria, the inner membrane is surrounded by 
an elaborate peptidoglycan cell wall consisting of polymerized subunits of 
N-acetylmuramic acid-(pl-4)-N-acety]glucosamine. These glycan polymers 
are cross-linked via transpeptidation of murein peptides that are covalently 
attached to the D-lactyl groups on N-acetylmuramic acid. In general, the 
framework of the gram-positive cell wall is further supported by the inclusion 
of techoic acid, lipotechoic acid, or lipoglycan polymers, which may be cova- 
lently linked to the wall peptidoglycan or anchored to the outer leaflet of the 
inner membrane through lipid modification [I ]. The gram-negative cell wall 
consists of a thin layer of peptidoglycan beyond the inner membrane. Outside 
of the peptidoglycan wall, a second phospholipid outer membrane is assem- 
bled. The outer leaflet of the outer membrane is composed of lipopolysaccharide 
(LPS or endotoxin), consisting of lipid A, an oligosaccharide core, and a 
distal 0-antigenic polysaccharide [2]. The 0-antigen is a key virulence deter- 
minant, often leading to the promotion of inflammation at the infection site. 
LPS can be described as the molecular signature of a particular pathogen, and 
several pathogens have devised schemes to alter their LPS structure, promot- 
ing serum resistance [3]. The outer membrane is also the destination of surface 
proteins required for adherence and for pore-forming protein complexes [4]. 
The double membrane arrangement of the gram-negative bacterium provides 
a compartment distinct from the cytoplasm called the periplasm. This com- 
partment is typically rich in enzymatic factors required for adaptation to the 
extracellular environment, and proteins that influence the proper folding of 
secretion substrates [5]. Proteins destined for display on the surface of a gram- 
negative bacterium must therefore contain information that will provide for 
navigation through the inner membrane, the periplasmic space, and for inser- 
tion into the outer membrane. This contrasts with the navigation of a surface- 
displayed protein in a gram-positive bacterium, which need only contain 
information for secretion beyond the inner membrane. Gram-positive bacteria 
have devised a strategy for the display of proteins on their surface involving 
the covalent linkage between the polypeptide and the peptidoglycan itself, 
while the mechanisms for the secretion of factors beyond the cell wall are 
poorly understood. 



Cambronne/Schneewind 182 



Generalized Secretion Strategies 

Proteins that are destined for localization in compartments outside of the 
bacterial cytosol must be translocated across the inner membrane. The Sec 
pathway is often required for this translocation process. This involves the deliv- 
ery of a nascent signal-bearing precursor polypeptide to a specialized secretion 
apparatus (Sec translocase) in the inner membrane [6]. This process may be 
accomplished using either a posttranslational or cotranslational mechanism. 
The posttranslational secretion mechanism is thought to be utilized primarily 
for soluble proteins synthesized in the cytosol [7]. In the gram-negative 
bacterium Escherichia coli, newly translated polypeptide harboring an amino- 
terminal secretion signal is bound by a cytosolic chaperone SecB. This secre- 
tion signal consists of 18-30 hydrophobic residues that are preceded by a 
charged domain and succeeded by a signal peptidase cleavage site [6]. SecB 
delivers the signal-bearing precursor to the Sec translocase complex, composed 
of SecD, SecE, SecF, SecG, SecY, and YajC [7]. SecB is predicted to maintain 
the signal-bearing polypeptide in a secretion-competent conformation [8]. 
SecB associates with another factor SecA, which is a soluble translocation 
ATPase. SecA associated with signal-bearing precursor polypeptide binds the 
SecY translocase component, and the hydrolysis of ATP to ADP by SecA 
promotes delivery of short segments of the polypeptide into the transloca- 
tion channel, consisting of SecE, SecG, and SecY [9]. The signal sequence is 
retained by the translocase until a signal peptidase cleaves the signal peptide at 
a particular site, allowing for the release of the mature polypeptide. Signal peptid- 
ases have been identified for the general secretion of soluble proteins (signal 
peptidase 1), lipoproteins (signal peptidase 11), and for virulence-associated 
prepilins (prepilin peptidase) for the assembly of type IV pili [10-12]. 

Cotranslational secretion of signal-bearing precursor polypeptides is in 
general thought to be associated with the proteins destined for insertion in 
the inner membrane after translocation [7]. The secretion mechanism involves 
the stalling of translation initiated thi'ough the binding of the bacterial signal 
recognition particle (SRP) to the signal sequence. SRP is a ribonucleoprotein 
complex consisting of the factor Ffh (P48) and a 4.5S ribonucleic acid [13]. 
Translation resumes when SRP binds to the inner membrane receptor FtsY and 
is displaced. The ribosome proceeds with translation and provides the force for 
the translocation of the nascent polypeptide through the Sec translocase [14], 
Proteins destined for insertion into the inner membrane may be retained by two 
mechanisms durmg Sec -mediated translocation. Type I membrane proteins con- 
tain a noncleavable signal/anchor sequence that mserts into the membrane, 
whereas type C membrane proteins contain a downstream stop transfer/membrane 
anchor sequence that retains the mature polypeptide after cleavage [15, 16]. 



Bacterial Invasins 



83 



Outer membrane proteins often assume (3-barrel structures prior to insertion. 
Proteins that are destined for insertion into the outer membrane or secreted 
beyond the outer membrane are often folded in the periplasm. DsbA and DsbC 
catalyze disulfide bond formation, and are both important in several of the 
gram-negative secretion systems [5]. In general, the Sec pathway is thought to be 
conserved among the gram-positive bacteria. Bacillus subtillis carries homologs 
of all of the Sec genes except SecB and SecG. It is assumed that gram-positive 
bacteria may utilize functional homologs of SecB for the chaperone-mediated 
delivery of secretion substrates to the Sec translocase [17]. 

A second general secretion system has been described recently. This sys- 
tem is called the twin-arginine translocation system or TAT pathway, named for 
the secretion signals consei"ved in the amino-termini of the substrates. The con- 
sensus amino-terminal secretion signal consists of a positively charged segment 
bordered by two consecutive arginine residues, a nonspecific amino acid, and 
two hydrophobic residues [18, 19]. This translocation system is likely devoted 
to the secretion of prefolded substrates and enzyme complexes in the cytoplasm 
that are required for general physiology and destined for localization in the 
periplasm [20]. Recent observations suggest that the TAT pathway may be asso- 
ciated with the secretion of virulence factors however, and examples of this 
requirement have been demonstrated in E. coli and Pseudomonas aeruginosa 
[21, 22]. In E. coli, the TAT translocation system is composed of TatA, TatB, 
TatC, and TatE, all integral membrane proteins. TatA, TatB, and TatC have been 
purified in complexes and are believed to be the structural components of the 
translocase [23, 24]. This relatively simple complex has been implicated in 
the secretion of Shiga toxin in enterohemorrhagic E, coli and was found to be 
required for toxin-mediated cytotoxicity of cultured cells [22]. The TAT system 
appears to be conserved among most bacteria, including gram-positive species, 
and future investigation may show this mechanism to be an important virulence 
determinant [25]. The general strategies required for protein secretion in bacte- 
ria are represented in figure I . 



Invasive Strategies of Gram-Positive Pathogens 

Typical gram-positive pathogens have only a single barrier separating then" 
cytoplasmic membrane from the extracellular environment. Secretion beyond or 
localization to the cell wall represent two general mechanisms employed to 
establish infections [1]. The secretion of toxins is a common feature, but the 
mechanistic process is poorly understood beyond the scope of Sec-mediated 
translocation. The display of virulence factors on the surface of the pathogen is 
a second common feature of the gram-positive pathogen. This mechanism 



Cambronne/Schneewind 184 




Fig. L General strategies involved in the secretion of bacterial proteins, a Gram-positive 
bacteria translocate signal-bearing precursor proteins through the inner membrane (IM) via the 
Sec pathway. Extracellular soluble proteins may passage through the peptidoglycan to diffuse 
in the extracellular space (1). Proteins destined for display on the surface of the bacterium 
(2) are often covalently linked to the cell wall peptidoglycan (CW). b Gram-negative bacteria 
transport signal-bearing soluble proteins to the periplasm via the Sec translocase. Proteins may 
be transported to the extracellular space by mechanisms requiring specialized secretion sys- 
tems (1), or fold and insert into the outer membrane (OM) (2), Proteins destmed for insertion 
m the mner membrane are bound by bacterial SRP at which time ribosomal synthesis is stalled, 
SRP binding to the membrane-boiuid receptor (R) promotes release of SRP from the precursor 
at the Sec translocase, where translation will resume, promoting insertion of the membrane 
protein (3). The Tat system is employed for the transport of prefolded substrates and enzyme 
complexes that localize in the periplasm (4). 



involves the covalent linkage of secreted polypeptides to the peptidoglycan itself 
This process is accomplished by a specialized membrane bound transpeptidase 
called sortase [26, 27], The sortase mechanism was first characterized in 
Staphlyococcus aureus, a ubiquitous pathogen that causes a variety of human 
infections. Sortases have subsequently been identified in numerous gram-positive 
pathogens, including Listeria monocytogenes. Streptococcus spp.. Bacillus 
anthracis, and others [28]. Staphylococcal protein A (Spa) is a surface protein 
that binds serum immunoglobulins to protect the bacterium from complement- 
mediated destiuction. Spa is synthesized as a signal-bearing precursor that will 
promote its Sec-mediated translocation. Spa also contains a consensus carboxy- 
terminal sorting signal that consists of an LPXTG sequence motif followed by a 
hydrophobic stretch of 15-19 amino acids and a distal positively charged tail of 
5-10 residues [29]. The hydrophobic/charged domam m the carboxy-terminus 
functions to retain Spa in the membrane after signal peptidase cleavage (P2 



Bacterial Invasins 



185 



NK 



NH. 



NH, 



NK 



NH, 



-GN-MN- 
I 
L-Ala 
I 

I 
Gtyg-L-Lys 

D-AJa 



t 



-GN-MN- 
I 
L-Ala 



GN-MN 



L-Ala 



Gly^-L-Lys 



D-Afa 




LPXTG 



+ 



Fig. 2, The sorting reaction in S. aureus. PI precursor protein substrates that harbor an 
amino-termina] signal peptide and a carboxy-terminal sorting signal are exported from the 
cytoplasm through the Sec translocase (1). The amino-terminal secretion signal is cleaved by 
signal peptidase generating a P2 precursor, which is retained in the plasma membrane by the 
carboxy-terminal sortijig signal (2), Sortase (SrtA) catalyzes a cleavage reaction between the 
threonine and glycine residues of the LPXTG motif, generating a thioester enzyme interme- 
diate (3). The acyl-enzyme intermediate is resolved through nucleophilic attack by a free 
amine group on lipid n, resuhing in amide linkage of the sortase substrate to the pentaglycine 
cross-bridge (4). The mature surface protein is incorporated into the cell wall through a trans- 
glycosylation reaction (5), IM = Inner membrane. 



precursor). Sortase catalyzes a transpeptidation reaction between the threonine 
and glycine residues of the LPXTG motif, where a proteolytic cleavage event 
links the thieonyl carboxyl group to an active site cysteine, generating an acyl- 
enzyme intermediate through thioester linkage [30], The carboxyl group of thre- 
onine is then amide linked to the amino group of the pentaglycine cross-bridge 
in the murein tetrapeptide segment of the lipid II cell wall precursor [31]. The 
reaction product is incorporated into new peptidoglycan polymers through 
transglycosylation, resulting in the mature cell wall-anchored Spa polypeptide. 
The general staphylococcal sorting reaction is represented in figure 2. 

The identification of cell wall-anchored surface protems based on carboxy- 
terminal sequence analysis has revealed the potential for numerous virulence- 
associated factors, including C5 peptidase in Sti^eptococcus pyogenes, internal in A 



Cambronne/Schneewind 



186 



in L. monocytogenes, and neuraminidase in Streptococcus pneumoniae [28]. 
Variations on the consensus sorting reaction have also emerged, which include 
multiple sortase enzymes encoded by the same organism that recognize alternate 
sorting signals, such as the recognition of the NPQTN consensus sorting signal 
by SrtB in S. aureus [32]. Further, evidence suggests that the expression of sortase 
enzymes is environmentally regulated, promoting the display of a particular set 
of surface proteins under specific conditions. 

L monocytogenes is a food- and water-borne pathogen that causes infec- 
tions ranging from gastroenteritis to septicemja. It is an intracellular pathogen 
that employs a particularly interesting mechanism for migration through host 
tissue. The bacterium requires at least two surface factors for entry into cultured 
cells. Intemalin A (InlA) and internalin B (InlB) each contain Sec-mediated 
amino-terminal signal sequences but are recruited to the bacterial surface 
by two different mechanisms [33]. L. monocytogenes harbors two sortase 
genes, sriA and srtB. SrtA is required for the cell wall anchoring of InlA, which 
contains a consensus LPXTG sorting signal. Deletion of the srtA locus results 
in a defect in invasiveness sunilar to that of an inlA mutant [34]. InJB con- 
tains carboxy-terminal repeat regions that promote a noncovalent interaction 
with lipoteichoic acids in the cell wall peptidoglycan [35]. InJA binds to the 
E-cadherin receptor on epithelial cells, while InlB interacts with the comple- 
ment receptor gClqR, glycosaminoglycans, and the tyrosine kinase receptor Met 
[36—38]. Activation of signal transduction cascades promotes phagocytosis and 
the bacterium is enveloped in a phagocytic vesicle. To combat the acidification 
of the phagocytic vacuole, the bacterium expresses the enzymes listeriolysin O 
(LLO) and phosphatidylinositol phospholipase C (PlcA), which are secreted 
and promote degradation of the vacuolar membrane [39, 40]. This event allows for 
escape from the phagocytic vacuole and promotes bacterial multiplication in the 
host cell cytoplasm. Listeria expresses another factor ActA, a membrane pro- 
tein exposed on the bacterial surface. ActA recruits the host Arp 2/3 complex 
resulting in nucleation of actin filaments at the surface of the bacterium [41, 
42]. ActA acts as a molecular mimic, functioning in a similar fashion to the 
WASP family of proteins. The WASP proteins are activated tlirough binding of 
cellular GTPases, and conformational changes promote Arp 2/3 complex 
recruitment [43]. The assembly of an actin tail propels the bacterium through 
the cytoplasm, generating pseudopod-like extensions that promote phago- 
cytosis by neighboring cells, resulting in the formation of a double membrane 
vacuole in the neighbor cell. In addition to the secretion of LLO and PlcA, 
phosphatidylcholine phospholipase C (PIcB) has been implemented in the 
escape of the bacterium from this specialized vacuole [44]. The intracellular 
growth cycle of the bacterium has been shown to result in localized tissue 
destruction with minimal exposure to components of the immune system. 



Bacterial Invasins 



187 



Invasive Strategies of Gram-Negative Pathogens 

Gram-negative pathogens have devised an array of mechanisms to pro- 
mote colonization. Most of these strategies incorporate the modification of a 
generalized secretion pathway to either promote the display of a surface molecule 
for colonization, or deliver eifector molecules beyond the bacterial envelope. 
Specialized secretion systems may be generally divided into two categories, those 
that promote the release of diffusible protein factors to the surrounding envi- 
ronment and systems that promote the delivery of effector proteins directly 
into the cytosol of target cells. There are currently five specialized secretion 
systems described for gram-negative bacteria (type 1-V) that appear dedicated 
to virulence. Type 1 secretion incorporates a Sec-independent process to 
deliver toxin to the extracellular space without a periplasmic intermediate. 
Type 11 secretion, the main terminal branch of the general secretory pathway 
(GSP), represents a two-step translocation mechanism where factors secreted 
by the Sec pathway are transported by a protein complex that contains a char- 
acteristic outer membrane secretin. The type III secretion mechanism is a Sec- 
independent translocation process that involves the direct delivery of effector 
molecules from the bacterial cytoplasm to the cytosol of a target cell through 
a specialized channel or needJe complex. Type IV secretion systems are simi- 
lar to bacterial conjugational systems and harbor the ability to transfer proteins 
and/or nucleic acids into a target cell using either a one- or two-step translo- 
cation process. Type V secretion represents an alternate terminal branch of the 
GSR Often referred to as the autotransporter mechanism, type V substrates are 
secreted by the Sec pathway and contain information in their carboxy-termini 
that promotes incorporation in the outer membrane and delivery of the amino- 
terminal domain outside of the cell. Each of these systems are discussed in detail 
below and figure 3 is a representation of the basic features associated with these 
secretion mechanisms. 

Surface Proteins 

The expression of a molecule on the suiface of the bacterium, not unlike 
the display of surface proteins in gram-positive pathogens, represents a mecha- 
nism for colonization in some gram-negative bacteria. A prototypical example 
of this mechanism is the display of the factor invasin in Yersinia pseudotuber- 
culosis and Yersinia enterocolitica. The expression of invA m a noninvasive 
strain off. coli results in the phagocytosis of the bacterium by cultured mam- 
malian cells [45]. Invasin is a modular protein that harbors an amino-terminal 
outer membrane localization domain, as well as an extracellular carboxy-terminal 
domain that consists of repeats of an IgG-like fold, and an adhesive tip [46, 47]. 
It has been determined that invasin binds to pi integrin receptors localized on 



Cambronne/Schneewind 1 88 




OM 



irr 



cwc 



IM 



1*r 




Fig. 3. Basic features of the five classes of gram-negative protein secretion systems, 
(I) The type I mechanism involves a single, Sec-independent translocation event that incorpo- 
rates an inner membrane (IM) ABC transporter for energy generation, (II) The type II mecha- 
nism represents a two-step translocation process, where signal-bearing precursor proteins are 
traiisported to the periplasm via the Sec pathway. Mature substrates are transported through an 
outer membrane (OM) secretin. (DQThe type III mechanism incorporates a single translocation 
step to transport substrates from the bacterial cytoplasm into the cytosol of eukaryotic cells. The 
substrate is transported through a basal body complex, an outer membrane secretin, and a nee- 
dle complex that penetrates the target cell membrane. (fV, left) The type IV secretion system is 
employed to transfer substrates into host cells. This process requires the assembly of a pilus 
structure at the outer membrane, a core assembly in the periplasm, and inner membrane- 
associated ATPases. (FV, right) Pathogens may also employ the type FV mechanism for secretion 
of diffusible toxins to the environment in a Sec-dependent manner. (V) Autotransporters are 
secreted by the type V pathway A typical substrate is traiislocated to the periplasm by the Sec 
translocase. Insertion into the outer membrane promotes the secretion of the amino-terminal 
passenger domain, Autoproteolysis releases the diffusible mature protein. Localization of energy 
generatmg enzymes are indicated by *. CW = Cell wall peptidoglycan. 



the apical surface of M cells, located amongst the folHcle-associated epithelia 
and lymphoid follicles of the small mtestine, commonly referred to as Peyer's 
patches [48, 49]. M cells sample contents of the intestinal lumen and transport 
particles contained in vesicles to the basolateral surface^ which is rich in immune 
cells such as macrophages and polymorphonuclear leukocytes. The bmding of 
invasin to M cells may therefore represent an early mechanism mvolved in the 
Yersinia infection process, as the bacteria have a specific tropism for lymphoid 
tissues. Numerous examples of adhesion factors have been identified, often 
associated with the protein subunits localized in the tip of pili or fimbriae. 



Bacterial Invasins 



189 



Examples mclude the PapG adhesin of type I pili in E, coli, and the major pilin 
subunit PilA on the type IV pili ofP aeruginosa [50, 51]. 

Type I Secretion 

The type I secretion mechanism involves the one-step translocation of a 
secretion substrate in a Sec- independent manner [52], This mechanism is 
employed for the secretion of diffusible toxins into the extracellular space. The 
type I mechanism has been demonstrated for the secretion of a-hemolysin (HlyA) 
in E. coli, as well as for the secretion of Bordetella pertussis adenylate cyclase 
and P aeruginosa protease [53]. In each instance, the toxin is recruited to a 
translocation complex that assembles upon association with the substrate. The 
type I secretion system is relatively simple in architecture, consisting of only three 
factors, each required for transport of the substrate, A characteristic feature of the 
system is the presence of an ATP-binding cassette (ABC) protein transporter [52, 
54], ABC transporters are inner membrane proteins that are found in a wide range 
of organisms, including gram-positive bacteria, lower eukaryotic, and mammalian 
cells, and are normally associated with the transport of small molecules. The 
secretion of HlyA has been extensively studied and the synthesis of the pro- 
hemolysin precursor protein (proA) requires a lipid modification for activation to 
mature HlyA [55], The cytosolic factor HlyC as well as an acyl-carrier protein 
(ACP) are required for the myristoylation or palmitoylation of two lysine residues 
[56]. HlyC acts as an acyl-transferase for this process. Although this lipid modi- 
fication step is requu"ed for the hemolytic activity of HlyA, this event is not 
required for the type I-dependent secretion of HlyA [57]. After modification, 
HlyA binds to the ABC transporter HlyB at the inner membrane [58], The 
sequence information required for secretion of HlyA is contained in the poly- 
peptides carboxy-terminal 48 amino acids, and unlike signal sequences in Sec- 
mediated substrates, the signal sequence of type 1 substrates is not cleaved after 
translocation [59, 60]. The H lyB transporter associates with a second factor HlyD 
independent of substrate binding. The HlyD protein spans both the inner and 
outer membrane and trimerization of HlyD in the presence of HlyB bound to 
HlyA results in the recruitment of the outer membrane protein TolC [58], Each 
subunit of the trimeric TolC contains an amino-terminal p-sheet domain that 
inserts into the outer membrane. A second carboxy-terminal a-helical domain 
extends deep into the periplasmic space and forms a barrier between the 
periplasm and the amino-terminal pore-like structure [61], The binding of ATP to 
HlyB in the presence of HlyA may result in the specific recruitment of TolC by 
HJyD, The HlyB/HlyD/TolC complex then supports the HJyB mediated trans- 
location of HlyA through successive rounds of ATP hydrolysis [62], resultmg in 
the delivery of HlyA to the extracellular space. Eleven tandem glycine-rich 
repeats (LXGGXGND) contained in the carboxy-terminus of HlyA are required 



Cambronne/Schneewind 1 90 



for calcium binding. Calcium-bound HlyA is competent for insertion in the host 
cell membrane, and results in pore-mediated leakage of the target cell [63]. 

Type II Secretion 

The GSP represents the primary route for translocation of polypeptides to 
the extracellular space among gram-negative bacteria. The type EI secretion 
mechanism represents the archetype for protein secretion in the GSP, and has 
been designated the main terminal branch. The type II pathway is associated 
with the secretion of virulence factors in several bacterial pathogens. Alternate 
GSP branches include the secretion of autotransporters (type V secretion), the 
chaperone/usher-mediated assembly of P or type I pili in E. coli, the assembly 
of type IV pili in P. aeruginosa and Neisseria gonorrhea, and the assembly of 
curli in E. coli [64]. The factors required for extrusion of filamentous bacterio- 
phage from the bacterial envelope also share conserved components with the GSP 
[65]. One common feature associated with all of these strategies is the requirement 
for the Sec-mediated translocation of secretion substrates to the periplasm. The 
type II secretion pathway therefore represents a two-step translocation process, 
incorporating distinct secretion reactions for translocation across the inner and 
outer membranes. 

The type Il-dependent secretion of pullulanase in Klebsiella oxytoca is a well- 
studied example of this secretion mechanism. I\illulanase (PuIA) is a lipoprotem 
of the a-amylase family that enzymatically degrades the complex carbohydrate 
pullulan to maltotriose subunits, a substrate that may be transported into the bac- 
terium [66]. The secretion of PulA requires the products of at least 25 genes, 14 
of which are specifically involved in the translocation of PulA beyond the outer 
membrane [67]. After secretion to the periplasm through the Sec pathway, The 
PulA precursor is subjected to diacyl glyceride modification and cleaved by 
signal peptidase [68]. The lipid-modified PulA is retained in the outer leaflet of 
the inner membrane by an aspartyl residue located at the amino-terminus of the 
mature polypeptide. Factors required for the type ll-dependent translocation 
step are localized within several compartments. A cytoplasmic ATPase (GspE) 
associates willi the inner membrane through interaction with a second factor 
(GspL), an inner membrane protem that harbors a carboxy-terminal cytoplasmic 
domain [69, 70]. This interaction, coupled with ATP hydrolysis by GspE, may 
provide the energy required for PulA transport. Four additional integral mem- 
brane proteins GspC, GspF, GspM, and GspN are thought to assemble into a basal 
body complex, since the factors harbor carboxy-terminal domains that extend 
into the periplasm [71]. A characteristic feature of the type II apparatus is the 
requirement for periplasmic pseudopilin proteins. These factors, all harboring 
prepilin signal sequences, are secreted by the Sec pathway and processed by the 
inner membrane-associated prepilin peptidase GspO, which will also N-methlyate 



Bacterial Invasins 



91 



the pseudopilin subunits [72]. Five pseudopilin factors, GspG, GspH, Gspl, GspJ, 
and GspK, are processed in this manner, and assemble into a channel-hke pilus 
structure Ihiking components of the inner and outer membranes [73], GspD is an 
outer membrane secretin required for the export of type II substrates. GspD is 
inserted into the outer membrane and assembles into a dodecameric channel- 
forming structure^ a process that requires the outer membrane chaperone GspS 
[74], Functional homologs of the GspD secretin are conserved amongst most of 
the alternate branches of the GSP and the GspD secretin is also conserved 
among the type 111 secretion systems (see below). The type ll-mediated export 
of PulA may occur through its association with the basal body complex, with 
signal recognition most likely residing in the secondary or tertiary structure of 
the secretion substrate after folding in the periplasm. PulA is transported to the 
outer membrane secretin GspD and exported to the extracellular space. Similar 
type II secretion mechanisms have been identified for the release of exotoxin A, 
elastase, and phospholipase C in /? aeruginosa [71]. 

The secretion of AB-type holotoxins is also mediated by a type Il-dependent 
process. This class of toxins includes the cholera toxin of Vibrio cholerae, E. coli 
enterotoxin, and the Shiga-like toxins of £". coli and Shigella dysenteriae [75]. 
Cholera toxin is composed of two separate polypeptides CtxA and CtxB. Sec- 
dependent secretion of the subunits to the periplasm results in proteolytic cleavage 
of signal peptides and the formation of an intramolecular disulfide bond in CtxA 
prior to cleavage. The CtxB subunits assemble into a pentameric ring structure 
and bind the carboxy-terminal domain of the CtxA subunit, generating the 
CtxA|-CtxB5 holotoxin [76]. Secretion of the holotoxin requires components of 
the eps gene cluster, which encodes several factors homologous to the type II 
secretion system in Klebsiella [77]. Export of the holotoxin will result in the 
binding of the CtxBg subunits to a G^vii ganglioside on the surface of intestinal 
epithelial cells [78], Reduction of the disulfide in CtxA by host cytosolic 
thioredoxin promotes the release of the mature CtxA toxin from the CtxBs 
pentameric ring, where it will function to activate host cell adenlyate cyclase, 
resulting in the massive cellular fluid loss associated with the diarrhea in 
cholera disease [79]. 

Type II J Secretion 

The delivery of polypeptides from the bacterial cytoplasm directly into the 
cytosol of target host cells without the generation of an extracellular interme- 
diate is the halknark feature of the type III secretion system [80]. Effector pro- 
terns that are translocated into host cells harbor enzymatic activities that 
manipulate cellular processes of the eukaryotic host, resulting in a variety of 
processes that culminate in perpetuation of the bacterium at the infection site 
[81]. The type III secretion mechanism was first characterized in pathogenic 



Cambronne/Schneewind 1 92 



Yersinia species, but has subsequently been identified and extensively studied 
in various pathogens including enteropathogenic E. colt (EPEC), P. aeruginosa, 
Salmonella enterica and Shigella flexneri [82]. Analysis of genetic infornnation 
has revealed the potential for type III systems in several other gram-negative 
bacteria, and thus may represent a highly conserved pathogenic strategy [83]. 

Even though the process of injection of virulence factors by the type III 
pathway is a recently described phenomenon, the type III secretion apparatus 
appears both structurally and fiinctionally similar to the basal body of the fla- 
gellar secretion system in gram-negative bacteria. In fact, the flagellar secretion 
system is now considered a type III pathway and recent observations suggest 
that the flagellar export system may also support the secretion of virulence factors 
[84]. Type III secretion systems most likely evolved from the flagellar machinery 
to support colonization in new nutrient-rich environments such as those found 
in higher eukaryotes. Yersinia species employ the type III pathway to maintain 
an extracellular lifestyle in the lymphoid tissues of their mammalian hosts and 
cause a variety of diseases ranging from bubonic plague in Yersinia pestis to 
acute enteritis in Y. enterocolitica. This is accomplished through the type III 
injection of effector proteins called Yops {Yersinia outer proteins) into host 
macrophages, resulting in prevention of phagocytosis and eventual apoptotic 
death of the host cell [85]. 

With the exception of the assembly of the secretion apparatus, the transloca- 
tion of type in secretion substrates represents a Sec-independent process. The type 
III secretion system consists of three principle components, an inner membrane- 
associated basal body, an outer membrane secretin, and an extracellular needle 
complex. Assembly of a jflinctional Yersinia type III apparatus requires the prod- 
ucts of at least 21 ysc {Yersinia secretion) genes [86-88]. Eleven of these genes are 
conserved amongst other type IJI systems, including nine that are conserved with 
the flagellar basal body [89]. In general, the Yersinia type III secretion apparatus 
must be assembled in a similar fashion to the flagellar secretion system, where 
assembly of the basal body complex precedes any substrate dehvery. The initiation 
of the assembly of the basal body complex in Yersinia likely begins with mem- 
brane inseition of the FliF homolog YscJ, after Sec-mediated translocation [86, 90, 
9 1 ]. This event will allow the association of inner membrane proteins YscD, YscR, 
YscU, YscV and accessory factors to form the basal body complex. YscN is 
homologous to the Flil ATPase in flagellar secretion and contains the Walker boxes 
A and B, which are characteristic conserved ATP-binding domains [92]. YscN is 
predicted to provide the energy for the transport of type 111 secretion substrates and 
is required for Yop secretion. YscC is homologous to the GspD secretin involved 
in the transport of molecules in the type II padiway, and requires the outer 
membrane lipoprotein YscW for its localization and for the formation of the char- 
acteristic dodecameric rings in the outer membrane, resulting in outer membrane 



Bacterial Invasins 



93 



channel formation [93, 94]. Accessory factor association between the basal body 
complex and the YscC secretin provides a conduit between the two components of 
type III secretion system, an assembly step that is not conserved with flagellar 
secretion. Secretion of the factors YscF,YscO,YscP, and YscX promote the assem- 
bly of the needle complex [95-97], 

Although the type III needle complex remains to be isolated from Yersinia, 
needle complexes have been purified from Salmonella, Shigella, and E. coll 
[98-100], The YscF protein has been determined to be the main component of 
the needle complex, where the protein multimerizes in a right-handed helical 
fashion. The YscF homolog MxiH of Shigella displays 5.6 subunits per turn, 
and is polymerized from the distal tip into a conduit as long as 50 nm with a 
width of Vnmi and a central tube of 2-3 nm [101, 102]. YscO and YscP are 
secreted by the basal complex. YscP has recently been suggested to participate 
in substrate recognition, as yscP mutant strains secrete an increased amount of 
the needle component YscF but fail to secrete Yop proteins in vitro. Mutations 
in the amino-terminus of YscU suppress the yscP mutant phenotype^ reducing 
the amount of secreted YscF to wild-type levels and restoring the secretion of 
Yop proteins [95]. These results suggest that YscO, YscP^ and YscU may control 
type ill secretion at the level of substrate specificity, allowing for a switch 
between the secretion of structural components and the delivery of Yop sub- 
strates, similar to the switch between hook and filament proteins in the flagel- 
lar apparatus [89], Assembly of the YscF needle complex would then represent 
the final step of assembly and provide a switch for the recognition of type 111 
secretion substrates and the delivery of effector Yop proteins. 

The hydrophobic nature of the YscF polymer has been predicted to provide 
a mechanism for the piercing of the host cell cytoplasm [96]. An alternate 
hypothesis suggests that three secretion substrates, YopB, YopD, and LcrV, each 
required for the translocation of Yop proteins^ form a translocation pore in the 
host cell membrane allowing for subsequent delivery of effector proteins [80, 
103-106], 

X enterocolitica secretes 14 polypeptides via the type III pathway. One 
cui'ious feature of each of these proteins is tliat they do not contain any amino 
acid sequences that would suggest the presence of a conserved type 111 secre- 
tion signal. Experiments performed usuig reporter proteins have revealed the 
presence of minimal secretion information contained in the amino-terminal 
8-15 residues [107-110]. The nature of the minimal secretion information 
remains controversial. Scanning mutagenesis studies employed to determine 
the residues required for secretion of YopE revealed that no specific residues 
were necessary. Further, introduction of frameshift mutations in the minunal 
signal did not affect the secretion of reporter fusion constructs [107]. These 
results prompted the hypothesis that the minimal signal information is actually 



Cambronne/Schneewind 1 94 



contained in the mRNA rather than the protein. Th^ yop mRNA might therefore 
recruit a translational complex to the type Til apparatus promoting a cotransla- 
tiojial secretion mechanism. 

The nature of the 5' mRNA/amino-terminal signal hypothesis has been 
highJy contested however, and independent studies suggested that a mutant that 
generated multiple mutations in the mRNA sequence without affecting the amino 
acid sequence of the protein was indeed secreted [109], This implied that the 
amino acid sequence, rather than the mRNA, contained the information required 
for type 111 secretion. Construction of a synthetic amphipathic amino -terminal 
signal that contained alternating serine and isoleucine residues between positions 
2 and 9 of YopE also supported secretion [109], Recent observations in the mini- 
mum secretion signals of YopE and YopQ have again resurrected the mRNA 
secretion signal argument, as it was discovered that minimum signals, such as the 
1-10 positions of YopQ, do not tolerate frameshifts unless a downstream sup- 
pressor region of mRNA is included that contains codons 1 1-13 [1 10]. Further, 
single substitutions in codons 2 and 10 caused a defect in the secretion reporter 
fusions in the context of 10 but not 15 codons. Finally, multiple mutations in the 
wobble positions of yopQ 1-10 did not support the secretion of the reporter, again 
suggesting that mRNA rather than protein sequences initiate the transport of sub- 
strates via the type III pathway [1 1 0], 

Beyond the context of the amino- terminal minimal secretion signal, experi- 
mental evidence suggests that type III substrates may require a second signal for 
their injection into the cytosol of host cells, and the presence of Syc (specific Yop 
chaperone) proteins may be required for the injection process [105, 1 11]. In gen- 
eral, Syc proteins are small acidic proteins that form dimers in the bacterial cyto- 
plasm. Each chaperone appears to specifically bind a partner effector Yop protein 
in the cytoplasm and structures have been determined for secretion substrate/ 
chaperone complexes [112]. Studies that examined the role of both the amino- 
terminal and chaperone-mediated secretion signals demonstrated that a defective 
secretion signal, when linked in context to the full length YopE protein^ v^^s 
secreted in an SycE-dependent manner, suggesting that the chaperone mediated 
secretion signal does not requii'e the presence of a functional amino-terniinal 
signal [113], This prompted the hypothesis that Yop proteins harbor two indepen- 
dent secretion signals, the first required for initiation of the substrate into the type 
III pathway^ and the second for injection into host cells. 

K enterocolitica transports a class of at least six factors into the cytosol of 
the host cell, each of which harbors an enzymatic function. All of these factors, 
which include YopE, YopH, YopM, YopO, YopP, and YopT, share sequence 
homology to proteins of eukaryotic origin, suggesting the pathogen evolved 
these strategies through intimate interaction with the host over time. Although 
all of the type III pathogens share a conserved mechanism for the delivery of these 



Bacterial Invasins 



195 



effector proteins, the effector proteins themselves may or may not be conserved 
between pathogens. YopE is only cytotoxic to HeLa cells when injected via the 
type 111 pathway. Jt is a characteristic GTPase-activating protein (GAP) that acts 
upon the Rho family ofeukaryotic GTPases. YopE inactivates RhoA, Racl, and 
CDC42 by accelerating the conversion of GTP to GDP in these factors [114, 
115]. This mechanism results in an inhibition of actin polymerization at the site 
of bacterial contact, YopH is a protein tyrosine phosphatase (PTPase) that is 
involved in the dephosphorylation of focal adhesions [1 16]. The amino-terminal 
domain of YopH appears to be a targeting domain that binds to pi 30^^ and 
focal adhesion kinase (FAK) [1 17]. The carboxy-terminal domain harbors the 
phosphatase domain and acts specifically to dephosphorylate these substrates, 
resulting in an interruption of stress fiber formation [118], YopM is required for 
virulence in mice and has been suggested to target to the nucleus and may influ- 
ence transcription in the host cell, inhibiting inflammatory cytokine production 
[119, 120]. YopO is similar in sequence to RhoA kinase. YopO functions as an 
autophosphorylating serine threonine kinase that is activated in vitro through bind- 
ing to actin [121]. The protein is believed to phosphorylate the Rho family of 
GTPases and enhance the inhibition of actin polymerization [122]. YopP acts as 
an inhibitor of IkB in the NF-kB pathway and also inhibits the MAP kinase path- 
way [123-125]. It has been reported that YopP is a cysteine protease that may 
function through a protein degradation pathway [126]. The cumulative effects 
of disrupting the NF-kB and MAP kinase pathways result in inhibition of the 
proinflammatory response, thus preventing the production of the cytokines 
TNF-a and lL-8 [123, 125]. YopP also induces apoptosis in macrophages, 
which is likely to be a cumulative result of the failure to activate the NF-kB sig- 
naling pathway and through the cleavage of Bid, a proapoptotic member of the 
Bcl-2 family [127]. YopT is also a cysteine protease that has been demonstrated 
to cleave RhoA^ Racl, and CDC42 at their carboxy-termini, sites that are prenyl- 
ated for membrane anchoring [128]. Cleavage releases the factors from the 
membrane resulting in a defect in actin polymerization at the site of bacterial 
contact. 

EPEC, a food- and water-borne pathogen that is a causative agent of 
human infantile diarrhea, uses the type III pathway to establish attaching and 
effacing lesions on intestinal epithelium [129, 130], These bacteria inject a pro- 
tein, translocated intimin receptor (Tir), which is subsequently displayed on the 
surface of the gastric epithelial cell [131]. The bacterial cell displays a ligand 
for this receptor on its outer membrane, called intimin. interaction between the 
two factors results in tight binding between the bacterium and host cell. This 
event coupled with the cumulative effects of the type III injection of other factors 
will promote actin pedestal formation at the site of contact, allowing extra- 
cellular colonization and destruction of surrounding tissue. 



Cambronne/Schneewind 1 96 



S. enterica serovar spp. cause a variety of diseases in humans and animals, 
ranging from acute food poisoning and gastrointestinal inflammation to typhoid 
fever and septicemia. In general pathogenic Salmonella species are food- 
and water-borne pathogens that have a tropism for the intestinal epithelium. 
S. enterica serovar spp. harbor the genes encoding two separate type 111 secre- 
tion systems on their chromosome. The first system, designated Salmonella 
pathogenicity island 1 (SPI-I), is employed to invade nonphagocytic epithelial 
cells [132]. Salmonella uses the SPI-1 type 111 pathway to inject several effec- 
tor molecules that leads to a massive reorganization of actin filaments promot- 
ing the formation of membrane ruffles and eventual phagocytosis. Similar to 
effector proteins in Yersinia, many Salmonella effectors target the signaling 
processes governing actin polymerization. Sip A stabilizes F-actin through 
binding to the T-plastin protein [133]. SopE and SopE2 function as guanine 
nucleotide exchange factors (GEF) to activate Rac-1 and CDC42 [134, 135]. In 
order to promote recovery of the cytoskeleton, SptP is injected. SptP is a multi- 
functional enzyme that contains an amino-terminal YopE-like GAP domain and 
a carboxy-terminal YopH-like tyrosine phosphatase domain. SptP counteracts 
the enzymatic effects of SopE and SopE2 by downregulating Rac-1 and CDC42 
[136, 137]. The second type HI system located at SPI-2 appears to manipulate 
vesicular trafficking, allowing for perpetuation of the bacterium in a special- 
ized vacuole, and is required for systemic infections [138]. 

Pathogenic Shigella species are typically water-borne pathogens that are a 
causative agent of bacillary dysentery, an infection of the colon. Shigella species 
utilize a type III secretion for the invasion of epithelial cells. Shigella are 
believed to enter epithelial cells from the basolateral surface. After engulfment 
by intestinal M cells and presentation to lymphoid macrophages, Shigella 
secretes an apoptotic factor IpaB which allows the bacterium to spread to adja- 
cent cells [139]. Shigella also employs the type III pathway to promote phago- 
cytosis by the epithelial cell through the cumulative effects of IpaA, IpaB, IpaC, 
and IpaD [140]. Unlike Salmonella^ Shigella escape from acidified vesicles and 
reside in the host cell cytoplasm. An outer membrane protein IcsA nucleates 
actin polymerization tlii'ough the bindmg to N-WASP and the fomiation of die 
Arp2/3 complex [141, 142]. Production of cytoskeletal filaments at the pole of 
the bacterium propels the organism into neighboring cells, similar to the process 
described fori, monocytogenes. 

Type IV Secretion 

The type IV secretion mechanism is employed for a wide range of func- 
tions in gram-negative bacteria. Several species utilize the type IV mechanism 
for interbacterial conjugative transfer of mobilized genetic elements. Pathogenic 
Agi'obacterium tumefaciens employs the type IV system for the transfer of 



Bacterial Invasins 



197 



tumorigenic DNA and protein into host plant cells, and several vertebrate 
pathogens such as Brucella spp,, B, pertussis, Helicobacter pylori, and Legionella 
pneumophila use a modified type IV secretion system for the secretion of 
toxins or the delivery of effector proteins into the host cell [143, 144], There is 
evidence to suggest that the secretion of substrates through the type IV appara- 
tus requires a Sec-dependent translocation step, such as in the secretion of 
pertussis toxin; however, specialized systems such as those for H, pylori and 
L pneumophila may bypass this requirement [ 1 45], All type IV systems represent 
a modification of the conjugative transfer system found in strains of £". coli. 
In general, the mechanism involves the assembly of a secretion apparatus with 
a pilus-like projection that will provide intimate contact between the donor and 
recipient cell [146]. 

The transfer of DNA from bacterium to host in the pathogen A. tumefaciens 
represents the archetype for the type IV secretion pathway. Substrate translocation 
requires the products of the VirB-encoded system: VirBl-1 1 and VirD4 [147]. 
The mechanisms of VirB-mediated type IV transport have been extensively 
studied, and the system is currently employed as the general model for the type 
IV mechanism in animal pathogens [144]. Evidence suggests that the type IV 
apparatus is assembled to extract the major pilin subunit VirB2, a cyclic 
polypeptide, through the outer membrane [148]. The secretion and processing 
of the pilin subunits is a Sec-dependent process. VirB2 forms a pilus through 
multimerization and contains a second minor pilin subunit VirB5. Pilin subunits 
interact with an outer membrane lipoprotein VirB7 and are thought to assemble 
at the outer membrane [149]. VirB6 is an inner membrane protein that may 
provide the connection between components of the inner and outer membrane, 
as well as guide assembly of the periplasmic core [150]. VirB? also interacts 
with VirB9, an outer membrane component, and VirB8, a muramidase localized 
in the periplasm that may provide for organization of the complex through 
wall peptidoglycan [149, 151]. Energy for the transport of type FV substrates 
is provided by the activities of three separate ATPases, The VirB4 dimer is 
localized in the inner membrane. A second inner membrane ATPase, hexa- 
meric VLi'Bl I assembles into a ring stmcture and may provide a route for trans- 
location of type IV substrates [152^ 153]. VirB 11 also interacts with the 
periplasmic core component VirB 10. The third ATPase VirD4, also called 
'coupling protein', is localized in the bacterial cytoplasm and is involved in 
substrate recognition [144]. 

The ptl system in B. pertussis represents an interesting link between the 
type II and type IV secretion pathways. 5. pertussis is the causative agent of 
whooping cough, where pertussis toxin, an AB-type holotoxin, is the primai^ 
virulence determinant. Pertussis toxin is exported by the Ptl type IV secretion 
apparatus. The Ptl system appears functionally distinct from other type IV 



Cambronne/Schneewind 1 98 



secretion systems. Rather than supporting injection into host cells, the Ptl system 
exports pertussis toxin to the extracellular space, where the toxin is available to 
associate with the target cell membrane [145]. The mature enzyme acts as an 
ADP-ribosylating factor of G proteins in the host cell. Unlike the T-DNA 
translocation process, the PtxA and PtxB subunits are translocated to the 
periplasm, where they are processed and assembled into the PtxA|-PtxB5 holo- 
toxin [154]. This protein complex then becomes a substrate for type IV-mediated 
export. Nine structural components of the Ptl system are homologous to the 
VirB system, where PtxA represents the major pilin subunit. The system also 
contains two membrane-associated ATPases, PtIC and PtlH, which are homo- 
logous to VirB4 and VirBll, respectively. 

Recent discoveries have provided evidence that type IV secretion systems 
are competent in the delivery of effector proteins directly into the cytosol of 
host cells. H. pylori is a causative agent of several gastrointestinal syndromes 
ranging from peptic ulcers to MALT lymphoma and adenocarcinoma. 
Pathogenic strains of H, pylori harbor the CAG pathogenicity island which 
encodes a VirB-like type IV secretion system [155]. CagA is translocated 
by the type IV pathway into the host cell cytosol where it becomes tyrosine- 
phosphorylated and proteolytically processed to a carboxy-terminal phosphoryl- 
ated fragment. The injection of CagA results in a change in the 
phosphorylation state of associated host cell factors, and is required for 
virulence [156], L. pneumophila and Brucella species require type IV secre- 
tion systems for survival in intracellular vacuoles [157]. L, pneumophila is 
the causative agent of Legionnaire's disease, a severe respiratory pneumonia. 
L. pneumophila targets alveolar macrophages where it employs the Dot/Icm 
type IV secretion system for intracellular survival. The Dot/Icm transporter is 
more distantly related to the VirB system, but is homologous to the IncI con- 
jugation system in 5. flexneri, and is competent for conjugational transfer of 
DNA [158]. L. pneumophila bypasses destruction mediated by the endocytic 
pathway by creating an endoplasmic reticulum-like vacuole, presumably 
through the injection of effector molecules into the host cytosol [157], RalF 
was the first factor identified to be an effector substrate. RalF is a guanine 
nucleotide exchange factor that functions to activate the ADP ribosylation 
factor (ARf) family of GTPases [159]. The factor has been localized on the 
surface of the Legionella-conx&mxng vacuole in a Dot/Icm-dependent manner^ 
and is required for the early recruitment of ARFl, but is not required for 
intracellular survival of the bacterium. A second factor LidA has recently 
been identified to be exported in a Dot/Icm-dependent manner and localizes to 
the phagosomal surface [160]. It has been hypothesized that this factor may 
function as a gatekeeper for the premature release of other factors. It is not yet 
clear how L. pneumophila modulates the type IV pathway to support the export 



Bacterial Invasins 



199 



of nucleic acid/protein hybrids or effector protein substrates under different 
environmental stimuli. The requirement for at least 24 genetic loci to promote 
intracellular survival suggests that the Dot/Icm transporter may be far more 
sophisticated than the VirB-like type IV systems [157]. 

Type V Secretion (Autotrunsporters) 

The autotransporter pathway represents an alternate branch of the GSP that 
serves as a simplified mechanism for the translocation of substrates out of the 
cell. Analysis of various genomes suggests that the autotransporter mechanism 
is widely conserved across gram-negative bacteria [83]. Rather than a require- 
ment for factors in the periplasm or outer membrane translocases, the auto- 
transporter secretion substrate harbors information in its carboxy-terminus for 
insertion into the outer membrane, and for translocation of the amino-terminal 
domain out of the bacterium. Pathogenic species of Neisseria secrete Igal pro- 
tease using the type V mechanism to promote sui'vival in interstitial fluids. The 
activated enzyme, once exported will function to degrade secretory antibodies 
[161]. Igal protease is synthesized as a preproenzyme that harbors an amino- 
termmal signal sequence to initiate its translocation across the inner membrane 
through the Sec pathway. The signal sequence is cleaved by signal peptidase, and 
the carboxy-terminaJ domain folds into a (5-barrel stiuctui'e promoting insertion 
of the proenzyme in the outer membrane [162]. Insertion in the outer membrane 
generates a porin-like channel for the export of the amino-terminal passenger 
domain. Transport of the amino-terrmnal domain through the channel promotes 
an autoproteolysis event, cleaving the proenzyme between the N- and C-terminal 
domains at a proline residue [163], This proteolytic event will result in the 
release of a diffusible active enzyme. 

Examples of autotransport have also been described for the Hap adhesin of 
Haemophilus influenzae, the IcsA protein ofS./lexneri, and the adhesin YadA of 
X enlerocolitica. The IcsA autotransporter represents a modification of the type 
V pathway, where an outer membrane serine protease SopA is required for the 
cleavage of the IcsA passenger domain, which promotes proper actin cytoskele- 
Lal nuclealion [164]. YadA in X enterocoliiica may represent another modifi- 
cation of the type V pathway, where the amhio-terminal passenger domain is 
delivered to the extracellular space, but is not cleaved from the carboxy-terminal 
p-barrel [165], Further, YadA assembles into trimers in the outer membrane and 
the amino-terminal heads assume a lollipop-like structure extending from the 
narrow stalk domain. YadA is involved in the resistance to complement mediated 
lysis. 

A second variation of the type V pathway involves a two-component 
secretory system, where the synthesis and secretion of an enzyme substrate 
requires a single outer membrane transporter for delivery to the extracellular 



Cambronne/Schneewind 200 



space. The ShlA hemolysin of Serratia marcescens is synthesized as a pro- 
enzyme that contains an amino-terminal signal sequence^ promoting its Sec- 
dependent translocation to the periplasm [166]. The ShlB polypeptide also 
contains an amino-terminal signal sequence and is exported through the 
Sec pathway [167]. Both ShlA and ShlB are processed by a signal peptidase 
and fold into mature species. The ShlB protein folds into a p-barrel structure 
that inserts into the outer membrane. This event is required for the trans- 
location of the enzymatic substrate ShlA. Other examples of this modified 
type V pathway include the secretion of filamentous hemagglutinin (FHA) 
by B, pertussis^ and the secretion of the HpmA hemolysin by Proteus 
mirabiiis [168], 



Concluding Remarks 

Molecular mechanisms that promote bacterial colonization are seemingly 
countless, however the accumulation of an ever-increasing body of information 
has allowed for the detection of common themes in pathogenesis. Strategies 
employed for the translocation of protein from the bacterial cytoplasm to targets 
in or beyond the cell wall envelope represent prime examples of this common- 
ality. Not only are the mechanisms for protein secretion conserved across 
species, many seemingly distinct secretion mechanisms share common compo- 
nents. Although protein secretion mechanisms represent only a fraction of the 
virulence strategies employed by bacteria, several of these processes represent 
primary virulence determinants. It appears that several pathogens use a combi- 
nation of secretion mechanisms to establish infection, and the identification of 
mechanisms by analogy has allowed for rapid progress in the classification of 
a particular pathogen arsenal. This of course provides the potential for rapid 
biochemical characterization of secretion systems and theu" protein substrates, 
as well as development and application of therapeutic targets to cover a wide 
range of bacterial species. 



References 

Navarre WW, Schneewind O: Surface proteins of grarn-positive bacteria and mechanisrns of their 
Largeliiig Lo the cell wall cJivelope. Microbiol Mol Biol Rev 1999,63:174-229. 
Bruneteau M, Minka S: Lipopolysaccharides of bacterial pathogens from the genus Yersinia A mini- 
review, Biochimie 2003;85:145-152. 

Berrington AW, Tan YC, Srikhanta Y, Kuipers B, van der Ley P, Peak IR, Jennings MP: Phase 
variation in meningococcal lipooligosaccharide biosynthesis genes. FEMS Immunol Med Microbiol 
2002;34:267-275. 



Bacterial Invasins 



20 



4 Inouye M: Bacterial Outer Membranes: Biogenesis and Functions. Wiley, New York, 1979. 

5 Missiakas D, Rama S: Protein folding in the bacterial periplasm. J Bacteriol 1997; 179:2465-2471, 

6 Driessen AJ, Fekkes P, Van DerWolk JP: The Sec system. Curr Opin Microbiol 1998;1:216-222. 

7 Danese PN, Silhavy TJ: Targeting and assembly of periplasmic and outer-membrane proteins in 
Escherichia coli, Annu Rev Genet 1998;32;59-94, 

8 Randall RR: Peptide binding by chaperone SecB: Implications for recognition of normative staicture. 
Science 1992;257:241-245. 

9 Economou A, Wickner W: SecA promotes preprotein translocation by undergoing ATP-driven 
cycles of membrane insertion and deinsertion. Cell 1992;78:835-843. 

10 Dalbey RE, Lively MO, Bron S, Van DijI JM: The chemistry and enzymology of the type I signal 

peptidases. Protein Sci 1997;6:l 129-1 138. 
1 I Lory S, Strom MS: Structure-function relationship of type-lV prepilin peptidase o^ Pseiidomonas 

aentginosa - A review. Gene 1 997; 1 92: 1 1 7-1 2 1 . 

12 Sankaran K, Wu HC: Bacterial prolipoprotein signal peptidase. Methods Enzymol 1995;248: 
169-180. 

13 Poritz MA, Bernstein HD, Strub l<L, Zopf D, Wilhelm H, Walter P: An £. co/?ribonucJeoprotein con- 
taining 4. 5S RNA resembles mammalian signal recognition particle. Science 1990;250:1 111-1117. 

14 Miller JD, Bernstein HD, Walter P: Interaction of Exoli Ffh/4.5S ribonucleoprotein and FtsY 
mimics that of mammalian signal recognition particle and its receptor. Nature 1994;367:657-659. 

15 Lingappa VR, Katz FN, Lodish HF, Blobel G: A signal sequence for the insertion of a transmembrane 
glycoprotein. Similarities to the signals of secretory proteins in primary structure and function. J Biol 
Chem 1978;253:8667-8670. 

16 Davis NG, Model P: An artificial anchor domain: Hydrophobicity suffices to stop transfer Cell 
1985;41:607-614. 

17 van Wely KLHM, Swaving J, Freudl R, Driessen AJM: Translocation of proteins across the cell 
envelope of gram-positive bacteria. FEMS Microbiol Rev 2001 ;25:437— 454. 

18 Stanley NR, Palmer T, Berks BC:The twin arginine consensus motLf ofTat signal peptides is involved 
in Sec-independent protein targetijig in Escherichia coli. J Biol Chem 2000;275: 1 J 591-1 1 596. 

19 Wu L-F, Ize B, Chanal A,QuintinY, Fichant G: Bacterial twin-arginine signal peptide-dependent pro- 
tein translocation pathway: Evolution and mechanism. J Mol Microbiol Biotechnol 2000;2:179-189. 

20 Santini C-L, Ize B, Chanal A, Muller M, Giordano G, Wu L-F: A novel Sec-independent peri- 
plasmic protein translocation pathway in Escherichia coli. EMBO J 1998;17:101-1 12, 

21 Ochsner UA, Snyder A, Vasil Af, Vasil ML: Effects of the twin-arginine translocase on secretion 
of virulence factors, stress response, and pathogenesis. Proc Natl Acad Sci USA 2002;99: 
8312-8317. 

22 Pradel N, Ye C, Livrelli V, Xu J, Joly B, Wu L-F: Contribution of the twin arginine translocation 
system to the virulence of enterohemorrhagic Escherichia coli 0157:H7. Infect Immun 2003;71: 
4908-4916. 

23 Bolhuis A, Mathers JE, Thomas JD, Barrett CM, Robmson C: TatB and TatC form a functional 
and structural unit of the twin-arginine translocase complex of Escherichia coli. J Biol Chem 
2001;276:20213-20219. 

24 Sargent F, Gohlke U, de Leeuw E, Stanley ^fR, Palmer T, Saibil HR, Berks BC: Purified components 
of the Escherichia coli Tat protein transport system form a double-layered ring structure. Eur J 
Biochem 2001;268:3361-3367. 

25 van Dijl JM, Braun PG, Robinson C, Quax WJ, Antelmann H, Hecker M, Muller J, Tjalsma H, 
Bron S, Jongbloed JDH; Functional genomic analysis of the BaciUus subtilis Tat pathway for protein 
secretion. J Biotechnol 2002;98:243-254. 

26 Ilangovan U Ton-That H, Iwahai*a J, Schneewind O, Clubb RT: Structure of sortase, the trans- 
peptidase that anchors proteins to the cell wall of Staphylococcus aureus. Proc Natl Acad Sci USA 
2001;98:6056-6061. 

27 Mazmanian SK, Liu G, Ton-That H, Schneewind O: Staphylococcus aureus sortase, an enzyme 
that anchors surface proteins to the cell wall. Science 1999;285:760-763. 

28 Mazmanian SK, Schneewind 0: Cell wall-anchored surface proteins and lipoproteins of 
gram-positive bacteria; in Sonenshein AL, Losick R, Hoch JA (eds): BaciUus subtilis and Its 
Closest Relatives: From Genes to Cells. Washington, ASM Press, 2002. 



Cambronne/Schneewind 202 



29 Schneewind 0, Model P, Fischetti VA: Sorting of protein A to the staphylococcal cell walL Cell 
1992;70:267-28L 

30 Navarre WW, Daefler S, Schneewnnd O: Cell wall sorting of lipoproteins in Siaphylococciis aureus. 
JBacteriol I996;178:441^M6. 

31 Schneewind 0, Fowler A, Faull KF; Structure of the cell wall anchor of surface proteins in 
Staphylococcus aureus. Science 1995;268:103-106- 

32 Mazmanian SK, Ton-That H, Su IC, Schneewind 0: An iron-regulated sortase anchors a class of 
surface protein during Staphylococcus aureus pathogenesis. Proc Natl Acad Sci USA 2002;99: 
2293-2298. 

33 Cossart P, Pizarro-Cerda J, Lecuit M: Invasion of mammalian cells by Listeria monocytogenes: 
Functional mimicry to subvert cellular functions. Trends Cell Biol 2003;13:23-31. 

34 Bierne H, Mazmanian SK, Trost M, Pucciarelli G, Liu G, Dehoux P, Jansch L, Garcia-del Porlillo F, 
Schneewind O, Cossait P; European Listeria Genome Consortium: Inactivarion of the sriA gene in 
Listeria monocytogenes inhibits anchoring of surface proteins and affects virulence. Mol Microbiol 
2002;43:869-88 L 

35 Jonquieres R, Fiedler F, Gounon P, Cossart P: Interaction between the protein IniB of Listeria 
monocytogenes and lipoteichoic acid: A novel mechanism of protein association at the surface of 
gram-positive bacteria. Mol Microbiol 1999;34:902-914. 

36 Braun L, Ghebrehiwet B, Cossart P: gClq-R/p32, a Clq-binding protein, is a receptor for the InlB 
invasion protein oi Listeria monocytogenes. EM BO J 2000;19:1458-1466. 

37 Mengaud J, Chenevert J, Geoffroy C, Gail lard JL, Cossart P: E-cadherin is the receptor for inter- 
nalin, a surface protein required for entry of L. monocytogenes into epithelial cells. Cell 1996;84: 
923-932. 

38 Shen Y, Naujokas M, Park M, Ireton K: InlB-dependent internalization oi Listeria is mediated by 
the Met receptor tyrosine kinase. Cell 2000;103:501-510. 

39 Camilli A, Tilney LG, Portnoy DA: Dual roles of pIcA in Listeria monocytogznzs pathogenesis. 
Mol Microbiol 1993;8:143-157. 

40 Gedde MM, Higgins DE, Tilney LG, Portnoy DA: Role of listeriolysin O in cell-to-cell spread of 
Listeria monocytogenes. Infect Immun 2000;68:999-1003. 

41 Kocks C, Gouin E, Tabouret M, Berche P, Ohayon H, Cossart P: L. monocytogenes-\\\d.w<:^A actin 
assembly requires the aciA gene product, a surface protein. Cell 1992;68:521-531. 

42 Pistor S, Chakaraborty T, Niebuhr IC, Domann E, Wehland J: The ActA protein of Listeria 
monocytogenes acts as a nucleator inducing reorganization of the actin cytoskeleton, EMBO J 
1994;13:758-763. 

43 Bear JE, Krause M, Gertler FB: Regulating cellular actin assembly. Curr Opin Cell Biol 2001;13: 
158-166. 

44 Marquis H, Goldfine H, Portnoy DA: Proteolytic pathways of activation and degradation of a bac- 
terial phospholipase C during intracellular infection by Listeria monocytogenes. J Cell Biol 
1997;137:1381-1392. 

45 Isberg RR, Voorhis DL, FaJkow S: Identification of invasin: A protein that allows enteric bacteria 
to penetrate cultured mammalian cells. Cell 1987;50:769-778. 

46 Hamburger ZA, Brown MS, Isberg RR, Bjorkman PJ: Crystal structure of invasin: A bacterial 
integrin-binding protein. Science 1999;286:291-295, 

47 Leong JM, Fournier RS, Isberg RR: Mapping and topographic localization of epitopes of the 
Yersinia pseudotuberculosis invasin protein. Infect Immun 1991;59:3424-3433. 

48 Isberg RR, Leong JM: Multiple P-l chain integrins are receptors for invasin, a protein that 
promotes bacterial penetration into mammalian cells. Cell 1990;60:861-871. 

49 Schulle R, Kerneis S, Klinke S, Bartels H, Preger S, Kraehenbuhl J-P, Pringault E, Autenrieth IB: 
Translocation of Yersinia enlerocolidca across reconstituted intestinal epithelial monolayers is trig- 
gered by Yersinia invasin binding to p-1 integrins apically expressed on M-like cells. Cell Microbiol 
2000;2:173-185. 

50 Lindberg F, Lund B, Johansson L, Normark S: Localization of the receptor-binding protein 
adhesin at the tip of the bacterial pilus. Nature 1987;328:84— 87. 

5 1 Pasloske BL, Finlay BB, Pai"anchych W; Cloning and sequencing of the Pseudomonas aeruginosa 
PAK pilin gene. FEBS Lett 1985; 183:408^ 12. 



Bacterial Invasins 



203 



52 Koronakisy Hughes C: Synthesis, maturation and export of the E, co// hemolysin. Med Microbiol 
Immunol 1996;185:65-71. 

53 Path MJ, Kolier R: ABC transporters: Bacterial exporters. Microbiol Rev 1993;57:995-1 01 7. 

54 Wang RC, Seror SJ, Blight M, Pratt JM, Broome-Smith JK, Holland IB: Analysis of the membrane 
organization of an Escherichia coti protein translocator, HlyB, a member of a lai'ge family of 
prokaryote and eakaryote surface transport proteins. J Mol Biol 1 99 1;2 17:44 1^54. 

55 Stanley P, Packman LC, Koronakis V, Hughes C: Fatty acylation of two internal lysine residues 
required for the toxic activity of Escherichia coli hemolysin. Science 1994;266:1992-1996. 

56 Issartel JP, Koronakis V, Hughes C: Activation of Escherichia coli prohaemolysin to the mature 
toxin by acyl carrier protein-dependent fatty acylation. Nature 1991;351:759-761. 

57 Ludwig A, Vogel M, Goebel W: Mutations affecting activity and transport of haemolysin in 
Escherichia coll. Mol Gen Genet 1987;206:238-245. 

58 ThanabaluT, Koronakis E, Hughes C, Koronakis V: Substrate-induced assembly of a contiguous 
channel for protein export from E. coli: Reversible bridging of an inner-membrane translocase to 
an outer membrane exit pore. EMBOJ 1998;17:6487-6496. 

59 Gray L, Baker K, Kenny B, Mackman N, Haigh R, Holland IB: A novel C-terminal signal sequence 
targets E. coli haemolysin directly to the medium. Mol Gen Genet 1989;205:127-133. 

60 Koronakis V, Koronakis E, Hughes C: Isolation and analysis of the C-terminal signal directing 
export of Escherichia call hemolysin protein across both bacterial membranes. EMBO J 1989;8: 
595^05. 

61 Koronakis V, Sharff A, Koronakis E, Luisi B, Hughes C: Crystal structure of the bacterial 
membrane protein TolC central to multidrug efflux and protein export. Nature 2000;405: 
914-919, 

62 Koronakis V, Hughes C, Koronakis E: ATPase activity and ATP/ADP-induced conformational 
change in the soluble domain of the bacterial protein translocator HlyB. Mol Microbiol 1993;8: 
1163-1175. 

63 Felmlee T, Welch RA: Alterations of amino acid repeats in the Escherichia coli hemolysin affect 
cytolytic activity and secretion. Proc NJatl Acad Sci USA 1988;85:5269-5273. 

64 Stathopoulos C, Hendrixson DR, Thanassi DG, Hultgren SJ, St Geme JW 3rcL Curtiss R 3rd: 
Secretion of virulence determinants by the general secretory pathway in gram-negative pathogens: 
An evolving story. Microbes Infect 2000;2:1061-1072, 

65 Russel M: Filamentous phage assembly. Mol Microbiol 1991;5:1607-1613. 

66 Bender H, Wallenfels K: Pullulanase (an amylopectin and glycogen debranching enzyme) from 
Aerohacier aerogenes . Methods Enzymol 1966;8:555-559. 

67 Pugsley AP, Francetic O, Possot OM, Sauvonnet N, Hardie KR: Recent progress and future direc- 
tions in studies of the main terminal branch of the general secretory pathway in gram-negative 
bacteria - A review. Gene 1997; 192:13-19. 

68 Pugsley AP, Chapon C, Schwartz M: Extracellular pullulanase of Klebsiella pneumoniae is a 
lipoprotein. J Bacteriol 1986;166:1083-1088. 

69 Possot OM, Pugsley AP: The conserved letracysteine motif in the general secretory pathway com- 
ponent PulE is required for efficient pullulanase secretion. Gene 1997;192:45-50. 

70 Sandkvist M, Bagdasarian M, Howard SP, DiRita VJ: Interaction between the autokinase EpsE 
and EpsL in the cytoplasmic membrane is required for extracellular secretion in Vibrio cholerae. 
EMBOJ 1995;14:1664-1673. 

71 Russel M: Macromolecular assembly and secretion across the bacterial cell envelope: Type II pro- 
tein secretion systems. J Mol Biol 1998;279:485^99. 

72 Pugsley AP, Dupuy B: An enzyme with type FV prepilin peptidase activity is required to process 
components of the general extracellular protein secretion pathway of Klebsiella oxyioca. Mol 
Microbiol 1992;6:751-760. 

73 Hobbs M, Mattick JS: Common components in the assembly of type 4 fimbriae, DNA transfer 
systems, filamentous phage and protein-secretion apparatus: A general system for the formation 

of surface-associated protein complexes. Mol Microbiol 1993;10:233-243. 

74 Nouwen N, Ranson N, Saibil H, Wolpensinger B, Engel A, Ghazi A, Pugsley AP: Secretin PulD: 
Association with pilot PulS, structure, and ion-conducting channel formation, Proc Natl Acad Sci 
USA 1999:96:8173-8177. 



Cambronne/Schneewind 204 



15 Schmitt CKL, Meysick KC, 'Brien AD: Bacterial toxins: Friends or foes? Emerg Ijifect Dis 1999; 
5:224-234. 

76 Lonnroth I, Holmgren J: Subunit structure of cholera toxin. J Gen Microbiol 1973;76:4I7^27. 

77 Sandkvist M, Michel LO, Hough LP, Morales VM, Bagdasarian M, Koomey M, DiRita VJ: 
General secretion pathway (eps) genes required for toxin secretion and outer membrane biogenesis 
in Vibrio cholerae. J Bacteriol 1997;179:6994-7003. 

78 Cuatrecasas P: Interaction of Vibrio cholerae enterotoxin with cell membranes. Biochemistry 
1973;12:3547-3558. 

79 Cassel D, Selinger Z: Mechanism of adenylate cyclase activation by cholera toxin: inhibition of 
GTP hydrolysis at the regulatory site. Proc Natl Acad Sci USA 1977;74:3307-331 I. 

80 Rosqvist R, Magnusson K-E, Wolf-Walz H: Target cell contact triggers expression and polarized 
transfer of Yersinia YopE cytotoxin into mammalian cells. EMBO J 1994; 13:964-972. 

81 Hueck CJ: Type HI protein secretion in bacterial pathogens of animals aiid plants. Microbiol Mol 
BiolRe\' I998;62:379^33. 

82 Winstanley C, Hart CA: Type HI secretion systems and pathogenicity islands, J Med Microbiol 
200J;50: 1 16-126, 

83 Fallen MJ, Chaudhuri RR, Henderson ]R: Genomic analysis of secretion systems. Curr Opin 
Microbiol 2003;6:519-527. 

84 Young GM, Schmiel DH, Miller VL: A new pathway for the secretion of virulence factors by bac- 
teria: The flagellar export apparatus functions as a protein secretion system. Proc Natl Acad Sci 
USA 1999;96:6456-6461. 

85 Cornelis GR, Boland A, Boyd AP, Geuijen C, Iriarte M, Neyi C, Sory M-P, Stainier I: The viru- 
lence plasmid of )fe™mfl, an antihost genome. Microbiol Mol Biol Rev 1998;62:1315-1352. 

86 Allaoui A, Schulte R, Cornelis GR: Mutational analysis of the Yersinia enierocolitica v/rCoperon: 
Characterization of yscE, F, C, /, J, K required for Yop secretion and yscH encoding YopR. Mol 
Microbiol 1995;18:343-355. 

87 Allaoui A, Woestyn S, Sluiters C, Cornelis GR: YscU, a Yersinia enierocolitica inner membrane 
protein involved in Yop secretion. J Bacteriol 1994;176:4534-4542. 

88 BergmannT, Hakansson S, Forsberg A, Noriander L, MacellaroA, Backman A, Bolin 1, Wolf-WatzH: 
Analysis of V antigen IcrGVH-yopBD operon of Yersinia pseudotuberculosis'. Evidence for a regula- 
tory role of /cr// and /crK. J Bacteriol 1991;173:1607-1616. 

89 Macnab RM: How bacteria assemble flagella. Annu Rev Microbiol 2003;57:77-100. 

90 Kimbrough TG, Miller SI: Contribution o{ Salmonella typhimurium t)T>e 111 secretion components 
to needle complex formation. Proc Natl Acad Sci USA2000;97:1 1008-11013. 

91 MichielsT, Vanooteghem J-C, Lambert de Rouvroit C, China B, Gustin A, Boudry P, Cornelis GR: 
Analysis of v/rC, aji operon involved in the secretion of Yop proteins by Yersinia enierocolitica. 
J Bacteriol 1991;173:4994-5009. 

92 Woestyn S, Allaoui A, Wattiau P, Cornelis GR: YscN, the putative energizer of the Yersinia Yop 
secretion machinery. J Bacteriol 1994;176:1561-1569. 

93 Genin S, Boucher CA: A superfamily of proteins uwolved in different secretion pathways in gram- 
negative bacteria: Modular structure and specificity of the N-terminal domain. Mol Gen Genet 
1994;243:112-118. 

94 Koster M, Bitter W, de Cock H, AJlaoui A, Cornelis GR, Tommassen J: The outer membrane 
component, YscC, of the Yop secretion machinery of Yersinia enterocoliiica forms a ring-shaped 
multimeric complex. Mol Microbiol 1997;26:789-798. 

95 Edqvist PJ, Olsson J, Lavander M, Sundberg L, Forsberg A, Wolf-Watz H, Lloyd SA: YscP and 
YscU regulate substrate specificity of the Yersinia type III secretion system. J Bacteriol 2003; 
185:2259-2266. 

96 Hoiczyk E, Blobel G: Polymerization of a single protein of the pathogen Yersinia enterocoliiica 
into needles punctures eukat70tic cells. Proc Natl Acad Sci USA 2001;98:4669-4674. 

97 Payne PL, Straley SC: YscO of Yersinia pes tis is a mobile core component of the Yop secretion 
system. J Bacteriol 1998;180:3882-3890. 

98 Kubori T, Shimamoto N, Nakamura D, Uralil J, Lara-Tejero M, Sukhan A, Galan JE, Aizawa SI: 
Supramolecular structure of the Salmonella typhimurium type HI protein secretion system. 
Science 1998;280:602-605. 



Bacterial Invasins 



205 



99 Sekiya K, Ohishi M, Ogino T, Tamano K, Sasakawa C, Abe A: Supermolecular structure of the 
enteropathogenic Escherichia coli type HI secretion system and its direct interaction with the 
EspA-sheath-like structure. Proc Natl Acad Sci USA 2001;98:l 1638-1 1643. 

100 Tamano IC, Aizawa S, Katayama E, Nonaka T, Imajoh-Ohmi S, Kuwae A, Nagai S, Sasakawa C: 
Supramolecular structure of the Shigella type III secretion machinery: The needle part is change- 
able in length and essential tor delivery of effectors. EMBO J 2000;19:3876-3887. 

101 Blocker A, Jouihri N, Larquet E, Gounon P, Ebel F, ParsoL C, Sansonetti P, Allaoui A: Structure and 
composition of the Shigella Jlexneri 'needJe complex', a part of its type HI secretion. Mol Microbiol 
2001;39:652-663. 

102 Cordes FS, Komoriya K, Larquet E, Yang S, Egelman EH, Blocker A, Lea SM: Helical strucoire 
of the needle of the type ]|] secretion system of Shigella Jlexneri. J Biol Chem 2003;278: 
I7I03-I7I07. 

103 HakaJisson S, Bergman T, Vanooteghem J-C, Cornelis G, Wolf-Watz H: YopB and YopD constitute 
a novel class oC Yersinia Yop proteins. Infect Immun 1993;61:71-80, 

104 Nleyt C, Cornelis GR: Insertion of a Yop translocation pore into the macrophage plasma membrane 
by Yersinia enterocoiitica: Requirement for translocators YopB and YopD, but not LcrG, MoJ 
Microbiol 1999;33:971-981. 

105 Persson C, Nordfelth R, Holmstrom A, Hakansson S, Rosqvist R, Wolf-Watz H: Cell-surface- 
bound Yersinia translocate the protein tyrosine phosphatase YopH by a polarized mechanism into 
the target cell. Mol Microbiol 1995;18:135-150. 

106 Sarker MR, Neyt C, Slainier 1, Cornelis GR: The Yersinia yop virulon: LcrV is required for extru- 
sion of the translocators YopB and YopD, J Bacteriol 1998;180:1207-1214. 

107 Anderson DM, Schneewind O: A mRJvJA signal for the type III secretion of Yop proteins by 
Yersinia enterocolitica. Science 1997;278:1 140-1 143, 

108 Anderson DM, Schneewind O: Yersinia enterocoliiica type IFI secretion: An mRNA signal that 
couples translation and secretion ofYopQ. Mol PvLicrobiol 1999;3I:1 139-1 148. 

109 Lloyd SA, Norman M, Rosqvist R, Wolf-Watz H: Yersinia YopE is targeted for type ill secretion 
by N-terminal, not mRNA, signals. Mol Microbiol 2001;39:520— 531. 

I 10 Ramamurthi KS, Schneewind O: Yersinia enterocoliiica type 111 secretion: Mutational analysis of 
xh^yopQ secretion signal. J Bacteriol 2002;184:3321-3328. 

1 1 1 Sory M-P, Boland A, Lambermont 1, Cornelis GR: Identification of the YopE and YopH domains 
required for secretion and internalization into the cytosol of macrophages, using the cyaA gene 
aision approach. Proc Natl Acad Sci USA 1995;92:1 1998-12002. 

1 12 Birtalan SC, Phillips RM, Ghosh P: Three-dimensional secretion signals in chaperone-effector 
complexes of bacterial pathogens. Mol Cell 2002;9:971-980. 

1 13 Cheng LW, Anderson DM, Schneewind 0: Two independent type 111 secretion mechanisms for 
YopE iji Yersinia enterocoliiica. Mol Microbiol 1997;24:757-765. 

1 14 Black DS, Bliska JB: The RhoGAP activity of the Yersinia pseudotuberculosis cytotoxin YopE is 
required for antiphagocytic function and virulence. Mol Microbiol 2000;37:515-527. 

1 15 Von Pawal-Rammingen U, Telepnev MV, Schmidt G, Aktories K, Wolf-Watz H, Rosqvist R: GAP 
activity of the Yersinia YopE cytotoxin specifically targets the Rho pathway: A mechanism for dis- 
ruption of actin microfilament structure. Mol Microbiol 2000;36:737-748. 

1 16 Zhang ZY, Clemens JC, Schubert HL, Stuckey JA, Fischer MW, Hume DM, Saper MA, Dixon JE: 
Expression, purification, and physicochemical characterization of a recombinant Yersinia protein 
tyrosine phosphatase. J Biol Chem 1992;267:23759-23766. 

1 17 Deleuil F, Mogemark L, Francis MS, Wolf-Watz H, Fallman M: Interaction between the Yersinia 
protein tyrosine phosphatase YopH and eukaryotic Cas/Fyb is an important virulence mechanism. 
Cell Microbiol 2003;5:53-64. 

1 18 HamidN, Gustavsson A, Andersson K, McGeeK, Persson C, Rudd CE, FallmenM:YopH dephos- 
phorylates Cas and Fyn-binding protein in macrophages, Microb Pathog 1999;27:231-242, 

1 1 9 Sauvonnet N, Predet-Balade B, Garcia-Sanz JA, Cornelis GR: Regulation of mRMA expression in 
macrophages following Yersinia enterocolitica infection: Role of different Yop effectors. J Biol 
Chem 2002;227:25133-25142. 

120 Skrzypek E, Cowan C, Straley SC: Targeting of the Yersinia pestis YopM protein into HeLa cells 
and intracellular trafficking to the nucleus. Mol Microbiol 1998;30:1051-1065- 



Cambronne/Schneewind 206 



121 Juris SJ, Rudolph AE, HuddJer D, Orth K, Dixon JE: A distinctive role for the Yersinia protein 
kinase: Actin binding, kinase activatiori, and cytoskeleton disruption. Proc Natl Acad Sci USA 
2000;97:9431-9436. 

1 22 Gaiyov BE, Hakansson S, Forsberg A, Wolf-Watz H: A secreted protein kinase of Yersinia pseudo- 
tuberculosis is an indispensable virulence determinant. Nature 1993;361:730-732, 

123 Boland A, Comelis GR: Role of YopP in suppression of tumor necrosis factor alpha release by 
macrophages during Yersinia infection. Infect Lnmun 1998;66:1878-1884. 

1 24 Orth K, Palmer LE, Bao ZQ, Stewart S, Rudolph AE, Bliska JB, Dixon JE: Inhibition of the mitogen- 
activated protein kinase kinase superfamily by a Yersinia effector. Science 1 999;285: 1 920-1 923. 

125 Schesser K, Spiik AK, Dukuzumuremyi JM, Neurath MF, Pettersson S, Wolf-Waiz H: l\\t yopJ 
locus is required for yer^/n/^s-mediated inhibition of NF-kappaB activation and cytokine expres- 
sion: YopJ contains a eukaryotic SH2-like domain that is essential for its repressive activity. Mol 
Microbiol 1998;28:1067-1079, 

126 Orth K, Xu Z, Mudgett MB, Bao ZQ, Palmer LE, Bliska JB, Mangel WF, Staskawicz B, Dixon JE: 
Disruption of signaling by Yersinia effector YopJ, a ubiquitin-like protein protease. Science 
2000;290:1594-1597. 

127 Denecker G, Declercq W, Geuijen CA, Boland A, Benabdillah R, van Gurp M, Sory M-P, 
Vandenabeele P, Cornells GR: Yersinia enlerocoliiica YopP-induced apoptosis of macrophages 
involves the apoptotic signaling cascade upstream of bid. J Biol Chem 2001;276:19706-19714. 

128 Shao F, Merritt PM, Bao Z, Innes RW, Dixon JE: A Yersinia effector and a Pseudomonas aviru- 
lence protein define a family of cysteine proteases functioning in bacterial pathogenesis. Cell 
2002;109:575-588. 

129 Jar\MS KG, Giron JA, Jerse AE, McDaniel TK, Donnenberg MS, Kaper JB: Enteropathogenic 
Escherichia coli contains a putative type III secretion system necessary for the export of proteins 
involved in attaching and effacing lesion formation. Proc Natl Acad Sci USA 1995;92:7996-8000. 

130 Knutton S, Lloyd DR, McNiesh AS: Adhesion of enteropathogenic Escherichia coli to human 
intestinal enterocytes and cultured human intestinal mucosa. Infect Immun 1987;57:69-77. 

131 K_ermy B, DeVinney R, Stein M, Reinscheid DJ, Frey EA, FinJay BB: Enteropathogenic E. coli 
(EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell I997;91:5I 1-520. 

1 32 Hueck CJ, Hanlman MJ, Bajaj y Johnston C, Lee CA, Miller SI: Salmonella typhimurium secreted 
invasion determinants are homologous to Shigella Ipa proteins. Mol Microbiol 1995; 18:479^90. 

133 Zhou D, Mooseker MS, Galan JE: Role of the 5. typhimurium actin-binding protein SipA in bac- 
terial internalization. Science 1999;283:2092-2095. 

1 34 Hard! WD, Chen LM, Schuebel KJE, Bustelo WR, Galan JE: Salmonella typhimurium encodes an 
activator of Rho GTPases that induces membrane ruffling and nuclear responses in host cells. Cell 
1998;93:815-826. 

135 Stender S, Friebel A, Linder S, Rhode M, Mirold S, Hardt WD: Identification of SopE2 from 
Salmonella typhimurium, a conserved guanine nucleotide exchange factor for Cdc42 of the host 
cell. Mol Microbiol 2000;36: 1206^1221. 

1 36 Fu Y, Galan JE: A Salmonella protein antagonizes Rac- 1 and Cdc42 to mediate host-cell recovery 
after bacterial invasion. Nature 1999;401:293-297. 

137 Fu Y, Galan JE: The Salmonella typhimurium tyrosine phosphatase SptP is translocated into host 
cells and disrupts the actin cytoskeleton. Mol Microbiol 1998;27:359-368. 

138 Hensel M: Salmonella pathogenicity island 2. Mol Microbiol 2000;36:1015-1023. 

139 Zychlinsky A, Prevost MC, Sansonetti PJ: Shigella Jlexneri induces apoptosis in infected 
macrophages. Nature 1992;358:167-169. 

140 Sansonetti PJ: Microbes and Microbial Toxins: Paradigms for Microbial-Mucosal Interactions HI. 
Shigellosis: From symptoms to moleculai* pathogenesis. Am J Physiol Gastrointesl Liver Physiol 
2001;280:319-323. 

141 Bernardini ML, Mounier J, d'Hauteville H, Coquis-Rondon M, Sansonetti PJ: Identification of 
icsA, a plasmid locus of Shigella Jlexneri which governs bacterial intra- and intercellular spread 
through interaction with F-actin. Proc Natl Acad Sci USA 1989;86:3867-3871. 

142 Egile C, Loisel TP, Laurent V, Rong Li Pantaloni D, Sansonetti PJ, Carlier MF: Activation of the 
CDC42 effector N- WASP by the Shigella icsA protein promotes actin nucleaiion by Arp2/3 com- 
plex and bacterial actin-based motility. J Cell Biol 1999;146:1319-1332. 



Bacterial Invasins 



207 



143 Bums DL: Type IV transporters of pathogenic bacteria. Curr Opin Microbiol 2003;6:29-34. 

144 Cascales E, Christie PJ: The versatile bacterial type IV secretion systems. Nat Rev Microbiol 
2003;1:137-149. 

145 Nicosia A, Perugini M, Franzini C, Casagli MC, Borri MG, Antoni G, Almoni M^ Neri P, Ratti G, 
Rappuoli R: Cloning and sequencing of the pertussis toxin genes: Operon structure aiid gene 
duplication. Proc Natl Acad Sci USA 1986;83:463 1^635. 

146 Pansegrau W, Lanka E: Enzymology of DNA transfer by conjugative mechanisms. Prog Nucleic 
Acid Res Mol Biol 1 996;54 : 1 97-25 1 . 

147 Kuldau GA, De Vos G, Owen J, McCaffrey G, Zambryski P: The virB operon of Agrobactentim 
tumefaciens pTiC58 encodes 1 1 open reading frames. Mol Gen Genet 1990;221:256-266. 

148 Lai EM, Kado CI: Processed VjrB2 is the major subunit of the promiscuous "pWu^ of Agrobacterium 
tumefaciens. J Bacteriol I998;180;27l 1-2717. 

149 Anderson LB, Hertzel AV, Das A: Agrobacierium nmiefaciens VirB7 and VirB9 form a disulfide- 
linked protein complex. Proc Natl Acad Sci USA 1996;93:8889-8894, 

150 Das A, Xie YH: Construction of transposon Tn3phoA: Its apphcation in defining the membrane 
topology of the Agrobaciemwi tumefaciens DNA transfer proteins, Mol Microbiol I998;27:405-^ 14, 

151 Kumar RB, Xie YH, Das A: Subcellular localization of the Agrobacierium tumefaciens T-DNA 
transport pore proteins: VirB8 is essential for the assembly of the transport pore. Mol Microbiol 
2000;36:608-617. 

152 Savvides SN, Yeo HJ, Beck MR, Blaesing F, Lurx R, Lanka E, Buhrdorf R, Fischer W, Haas R, 
Waksman G: VirBl I ATPasesare dynamic hexameric assemblies: New insights into bacterial type 
W secretion, EMBO J 2003;22:1969-1980. 

153 Yeo H-J, Sa\^ides SN, Herr AB, Lanka E, Waksman G: Crystal structure of the hexameric traffic 
ATPase of the Helicobacter pylori type IV secretion system, Moll Cell 2000;6:M61-1472. 

154 Farizo KM, Huang T, Burns DL: Membrane localization of the SI subunit of pertussis toxin in 
Bordetella pertussis and implications for pertussis toxin secretion. Infect Immun 2002;70: 
1193-1201. 

1 55 Covacci A, Telford JL, Del Giudice G, Parsonnet J, Rappuoli R: Helicobacter pylori virulence and 
genetic geography Science 1999;284:1328-1333. 

156 Odenbreit S, Puis J, Sedlmaier B, Gerland E, Fischer W, Haas R: Translocation q{ Helicobacter 
pylori CagA into gastric epithelial cells by type rv secretion. Science 2000;287:1497-1500. 

157 Roy CR: Exploitation of the endoplasmic reticulum by bacterial pathogens. Trends Microbiol 
2002; 10:4 18^24. 

158 Vogel JP, Andrews HL, Wong SK, Isberg RJR: Conjugative transfer by the virulence system of 
Legionella pneumophila. Science 1998;279:873-876. 

159 Nagai H, Kagan JC, Zhu X, Kahn RA, Roy CR: A bacterial guanine nucleotide exchange factor 
activates ARF on Legionella phagosomes. Science 2002;295:679-682. 

1 60 Conover GM, Derre I, Vogel JP, isberg RR: The Legionella pheumophi la Lid A protein: A translo- 
cated substrate of the Dot/lcm system associated v^ith maintenance of bacterial integrity. Mol 
Microbiol 2003;48:305-321. 

161 Halter R, Pohlner J, Meyer TF: IgA protease of Neisseria gonorrhoeae: Isolation and characteri- 
zation of the gene and its extracellular product. EMBO J 1984;3:I595-I60L 

162 Pohlner J, Halter R, Beyreuther K, Meyer TF: Gene structure and extracellular secretion of 
Neisseria gonorrhoeae IgA protease. Nature 1987;325:458^62. 

163 KlauserT, Pohlner J, Meyer TF: Extracellular transport of cholera toxin B subunit using Neisseria 
IgA protease domain: Conformation-dependent outer membrane translocation. EMBO J 1990;9: 
1991-1999. 

164 Egile C, D'Hauteville H, Parsot C, Sansonetti PJ: SopA, the outer membrane protease responsible 
for polar localization of IcsA in Shigella fJexneri, Mol Microbiol 1997;23:1063-1073. 

165 Roggenkamp A, Ackermann N, Jacobi CA, Truelzsch K, Hofftnann H, Heesemann J: Molecular 
analysis of transport and oligomerization of the Yersinia enterocolitica adhesin YadA. J Bacteriol 
2003;185:3735-3744. 

1 66 Schonherr R, Tsolis R, Focareta T, Braun V: Amino acid replacements in the Serratia marcescens 
haemolysin ShlA define sites involved in activation and secretion. Mol Microbiol 1993;9: 
1229-1237. 



Cambronne/Schneewind 208 



167 Konninger UW, Hobble S, Benz R, Braun V: The haemolysin-secrering ShIB protein of the outer 
membrane of Serratia marcescens: Determination of surface-exposed residues and formation of 
ion-permeable pores by ShiB mutants in artificial lipid bilayer membranes. Mol Microbiol 
1999;32:1212-1225. 

168 Jacob-Dubuisson F, Buisine C, Willery E, Renauld-Mongenie G, Locht C: Lack of functional 
complementation between Bordetella pertussis filamentous hemagglutinin and Proteus mirabUis 
HpmA hemolysin secretion machineries. J Bacteriol 1997;179:775-783. 



Eric D, Cambronne 

Section of Microbial Pathogenesis, Yale University School of Medicine 

295 Congress Avenue, New Haven, CT 06536 (USA) 

Tel. + 1 203 737 2404, E-Mail cambronne@yale,edu 



Bacterial Invasins 



209 



Signaling and Gene Regulation 

Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kai'ger, 2005, vol 12, pp 210-233 



Bacterial Iron Transport Related 
to Virulence 



Volkmar Braiin 

Mikrobiologie/Membranphysiologie, Universitat Tiibmgen, 
Tiibingen, Germany 



The Problem of Iron Supply 

Under oxic conditions, iron occurs in the Fe^"^ valence state and forms 
insoluble polymeric hydroxyl-aquo complexes. Therefore, all aerobically livmg 
organisms that contain iron in many cytosolic and membrane-bound redox pro- 
teins, in particular in respiratory chains, have developed means to solubilize Fe^^. 
Bacteria and fiingi synthesize iron-complexing compounds, designated sidero- 
phores, which are secreted, bind extracellular Fe^^, and are transported as Fe^^ 
complexes via specific transport systems into the cells, where Fe^^ is released 
from the complexes, usually by reduction to Fe^^, and then incorporated into 
heme, iron-sulfur proteins, and other forms of protein reaction centers. 

Higher organisms synthesize heme, which is the most abundant form of 
iron-containing compounds. OnJy a small percentage of the heme occurs in free 
form; most of it is incorporated into hemoglobin and bound to hemopexin. 

Important extracellular iron-binding proteins in higher organisms are 
transferrin and lactoferrin and intracellular ferritin. Transferrin is the predomi- 
nant iron carrier that delivers iron to cells. The di-iron complex is taken up by 
transferrin receptors, and the iron is released in endosomes and then further 
metabolized. Lactoferrin is the predominant iron-binding protein in secretory 
fluids. Transferrin and lactoferrin bind Fe^"^ so tightly that the free Fe-^"^ con- 
centration in equilibrium with these proteins is in the order of 1 ion per liter. 
The extreme Jack of iron inhibits growth of microorganisms. However, some 
bacteria synthesize transferrin and lactoferrin receptor proteins exposed at the 
bacterial cell surfaces, which remove the iron from transferrin and lactoferrin 
and transport iron across the outer membrane. 



This short overview focuses on some prominent examples of iron supply 
systems formed by human pathogenic bacteria. The reader is referred to more 
comprehensive reviews on specific aspects [1 24]. 



Overview of Bacterial Iron Transport Systems 

Transport across the Cytoplasmic Membrane 

The design of Fe^"*" transport systems across the cytoplasmic membrane is 
the same for gram-negative and gram-positive bacteria. The systems belong to 
the ATP-binding cassette (ABC) transporters, which consist of a binding protein, 
a permease, and an ATPase (fig. 1). The binding proteins of gram-negative bac- 
teria are located in the periplasm. In gram-positive bacteria, the binding proteins 
are linked by a lipid of the murein-lipoprotein type (triacyl-glyceryl cysteine) to 
the outer surface of the cytoplasmic membrane. The permease consists of one or 
two proteins that are incorporated into the cytoplasmic membrane and trans- 
locate Fe^"*", Fe^"'"-siderophores, or heme across the cytoplasmic membrane. The 
ATPase provides the energy derived from ATP binding and subsequent ATP 
hydrolysis [25]. 

Crystal structures have been determined for two Fe-'^-binding proteins, 
FbpA of Neisseria gonorrhoeae and hFbpA of Haemophilus influenzae [26], 
and for the ferrichxome-binding protein FhuD, which binds structurally related 
siderophores of the hydroxamate type and the antibiotic albomycin [16, 27]. 
The crystal structures of FbpA and hFbpA are similar, but differ from that 
of FhuD. The three proteins are composed of two globular domains; in FbpA 
and hFbpA, these domains are connected by a hinge region that permits closure 
of the globular domains upon binding of Fe^"^ (liice a Venus fly trap). In con- 
trast, the two globular domains of FhuD are connected by a rigid, kinked 
a-helix that allows onJy a slight movement of the globular domains. The crys- 
tal structure of an entire ABC transporter, the vitamin B12 transporter of 
Escherichia coll, has recently been unraveled. The ABC transporter consists of 
the BtuC permease and associated BtuD ATPase [28], and the BtuF-binding 
protein [29]. Since the BtuF structure is similar to FhuD and the transmem- 
brane topology of BtuC is comparable to that of FhuB [15] which transports 
ferrichrome across the cytoplasmic membrane [30], it is predicted that the 
structure of the vitamin B12 transport system is representative for the ferric 
siderophore and heme transport systems. BtuF can be positioned via salt 
bridges on top of the BtuC permease. BtuCD forms a translocation channel that 
is large enough to accommodate vitamin B|2. In the crystal, the channel is open 
to the periplasmic side and closed to the cytoplasmic side. BtuD controls open- 
ing of the BtuC channel. The two BtuD subunits located at the inner side of the 



Iron Transport Related to Virulence 21 1 




n 



Q 

X 

LU 



CD 

i2 



c 



a 



fPhuD 




CQ 




FhuC 





FhuC 




ATP ADP + P| ATP ADP + Pj 





m 



lij 



Q 

.2 



OM 



PP 



CM 





C 



Albomycin 



Rifamycin CGP4832 



Braun 



212 



H 
I 

C- 



O 



H2N-' I 



H 






o 



H,C-C^.^ 



H^C^ o 



N 

I 

O 



Fe 



O" 



H 



0"-N 



\CH,), y 



.r.0 



3* 



o=c 



v./ 



I 



N- 



/ 



H 




CHo-O-C-CHn-C-0, 



CH3O 



I 



H ^C 



NH 
I 



I 



■N' "0 
H 



HOOC-CH I 

ho-(!:h ^° 
.s 





O" 



r 


"A 


N 


( 


V_ 


_y 



CH 



Albomycin 



Rifamycin CGP4832 



/ 



Fig. L Crystal structure of the FhuA outer membrane (OM) transport protein off", coli 
with bound anribiotics albomycin (a) and rifamycin (b) CGP 4832, which are transported by 
FhuA. The structures of the antibiotics derived from the crystal structures (c, d) and the chem- 
ical formula {e,f) are shown, a, A The model illustrates the subcellular location of the proteins 
TonB, ExbB, and ExbD, which form the energy-transducing complex between the cytoplas- 
iTiic membrane and the outer membrane, the transport proteins across the cytoplasmic mein- 
brane, and the interactions of the proteins. This protein arrangement is typical for all transport 
systems of gram-negative bacteria that transport Fe^"^, Fe^"^-siderophores, and heme. For 
further ijiformadon, see the text. PP - Periplasm; CM — cylopJasmic membrane. 



cytoplasmic membrane are in close contact to the two BtuC subunits. Binding 
of ATP moves the two BtuD subunits closer together. This might rearrange the 
two BtuC subunits such that the channel opens to the cytoplasmic side, BtuF 
loaded with vitamin B|2 is bound to BtuC, delivers vitamin B12 to BtuC, and 
triggers ATP hydrolysis. The BtuD molecules move apart, which in tum closes 
the BtuC channel to the cytoplasmic side and opens it to the periplasmic side 
for the next round of vitamin B12 transport. 

Transport across the Outer Membrane 

Gram-negative bacteria contain an outer membrane that forms a perme- 
ability barrier for hydrophilic substrates above a certain molar mass, which in 
E. coli is 600 daltons [31]. The inner diameter of the porins through which the 
substrates diffuse across the outer membrane determines the substrate size. The 
Fe^"^ siderophores usually have a molecular weight greater than 600 and cannot 



Iron Transport Related to Virulence 



213 



difFuse with a sufficient rate through porins. In addition, their concentration is 
too low for diffusion to satisfy the growth requirement - in the order of 10^ 
iron ions per cell per generation. The siderophores, heme, and the iron-binding 
proteins adsorb to outer membrane proteins, which not only serve as receptors 
but also function as transporters across the outer membrane. The iron com- 
pounds are thereby concentrated at the bacterial cell surface and are sub- 
sequently actively transported by an energy-consuming process across the 
outer membrane into the periplasm. There is no energy source in the outer 
membrane to drive active transport. Energy is provided by the cytoplasmic 
membrane through the proton motive force [32]. TonB, ExbB, and ExbD are 
the three known proteins that relay the energy from the cytoplasmic membrane 
into the outer membrane [33, 34], These proteins are located in the cyto- 
plasmic membrane and interact with each other, and TonB interacts with the 
outer membrane transport proteins. It is thought that these three proteins 
respond to the proton motive force of the cytoplasmic membrane (e.g.^ the proton 
gradient)^ react with a conformational change, and store the energy as poten- 
tial energy. Upon interaction of energized TonB with the outer membrane 
transporters^ the bound iron compounds are released from their binding sites 
and a channel is opened through which the iron compounds diffuse into the 
periplasm. 

The crystal stnictiires of three outer membrane iron transporters FhuA [35, 
36], FepA [37], and FecA [38, 39], and the vitamin B|2 transporter BtuB [40] pro- 
vide a conceptual framework of how these transporters might function. The struc- 
tures reveal a p-barrel composed of 22 antiparallel p-strands that form a channel. 
The channel is cjosed by a globular domain, which is designated as the cork, plug, 
or hatch. Binding of the substrates to the transporters occurs at a site well above 
the cell surface. Very strong bmding occurs through approximately ten-amino acid 
side chains with a binding constant in the nanomolar range. Energy input is 
required to release the substrates from their binding sites and to move the cork so 
that a channel is formed through which the substrates gain access to the periplasm. 
The theory is that TonB transfers potential energy to the transporters, which alter 
their confomiation to open a channel, TonB is deenergized, and tlie ti'ansportei's 
close the channels after the iron compounds have passed through by diflflision. The 
genetically and biochemically identified sites of interaction between TonB and the 
transporters are located in the TonB box of the transporters and a region around 
residue 160 of TonB [41, 42]. The crystal structures and electron spin resonance 
determinations of nitroxide-substituted TonB box residues of BtuB demonstrate 
that the TonB box is exposed to the periplasm and moves upon binding of the sub- 
strates to the transporters [43]. The TonB box and the substrate-binding sites are 
far apart, which implies long-range structural transitions throughout the entire 
transporter Transport across the outer membrane is mechanistically not coupled 



Braun 



214 



to transport across the cytoplasmic membrane. The two membrane transport 
processes occur independently of each other. 



Iron Transport Associated with Virulence 

Iron-Controlled Bacterial Functions 

Since iron is an essential element, but available only in growth-Jimiting 
concentrations, those bacteria that multiply in the human body express potent 
iron transport systems. The relationship of iron transport to virulence is usually 
not easy to establish since bacteria normally express several iron transport sys- 
tems. Knocking out one system by mutation might not result in conversion of a 
pathogenic strain to a nonpathogenic strain since other iron transport systems 
take over the iron supply For example, a pathogenic £'. coli strain may transport 
Fe^^ by the siderophores aerobactin, enterobactin, salmochelin, citrate, ferri- 
chrome, and heme, and Fe^"^ via the/eo-encoded transport system. tonB, exbB, 
and exbD are the only genes involved in all energy-coupled outer membrane 
iron transport systems of gram-negative bacteria, lonB mutants are impaired in 
virulence in various animal infection systems [44, 45], However, some bacteria 
contain up to three tonB and exbB, exbD genes, which might participate in dif- 
ferent iron uptake systems (see, for example. Iron Transport of Vibrio cholerae 
Related to Virulence), In addition, it is usually not known which iron transport 
system is important for proliferation at a specific infection site. Moreover, the 
iron limitation usually encountered in the human body could serve as an envi- 
ronmental signal that tells a bacterial strain its location in the human body. This 
could induce expression of genes required for multiplication, but might not be 
directly related to the iron supply Therefore, different approaches are required 
to elucidate a relationship between iron transport and virulence. Such studies 
have involved knocking out a particular iron transport system and a genome- 
wide search for the expression of genes in vivo compared to the expression of 
genes in synthetic media under iron-deplete and iron-replete conditions. Such 
large-scale expression profiles usually reveal genes related to the iron supply. 
These genes encode proteins for siderophore biosynthesis and transport, heme 
transport, hemolysins^ and toxins. The most prominent toxin is the diphtheria 
toxin, which is synthesized under iron-limiting conditions. Other iron-regulated 
toxins are the Shiga toxin of Shigella and E, coli strains, the hemolysins/ 
cytolysins of Serratia marcescens and certain E, coli strains, exotoxin A of 
Pseudomonas aeniginosa, and the tetanus toxin oi Clostridium tetani. By dam- 
aging cells, the toxins can mobilize intracellular iron sources and make them 
available to bacteria. S. marcescens, for example, colonizes the intestine of 
Caenorhabditis elegans and kills the nematode, S, marcescens mutants are 



Iron Transport Relaied to Virulence 215 



impaired in virulence when they carry a transposon in the hemolysin gene or in 
a siderophore biosynthesis gene [46]. 

Stress by Iron Surplus 

Not only iron shortage, but also iron surplus can affect the outcome of a 
bacterial infection. Aerobic metabolism constantly creates hydrogen peroxide 
and superoxide radicals. If too much H2O2 is formed, it might not be completely 
destroyed by catalase and peroxidase. In the Haber- Weiss reaction, the oxygen 
radical reacts with H2O2 to form the highly reactive hydroxyl radical and 
hydroxyl anion. In the Fenton reaction, Fe^"^ converts H2O2 to the hydroxyl 
radical and hydroxide anion. Fe-^^ oxidizes the oxygen radical to oxygen. H2O2, 
the oxygen radicals, and the hydroxyl radicals damage DNA, lipids in mem- 
branes, and proteins. The lack of regulation of iron metabolism could, therefore, 
be deleterious to cells [47]. This has been demonstrated for E. coli, in which 
a mutation in the/wr (iron uptake regulator) gene renders cells sensitive to 
oxygen. An additional mutation in the recA gene, which is involved in DNA 
repair, kills cells when they are cultivated under oxic conditions [48]. The sur- 
plus of reactive intracellular free iron might result from an uncontrolled import 
and the lack of intracellular iron storage proteins. Iron uptake is controlled by 
the/wr gene in most gram-negative bacteria and certain gram-positive bacteria 
with a low GC content and by the dtxR gene in most (GC-rich) gram-positive 
bacteria. When the intracellular iron concentration reaches a certain level, the 
Fur and DtxR proteins are loaded with Fe^"^ and repress transcription of genes 
encoding iron transport proteins and enzymes that synthesize siderophores [7]. 

Two types of iron storage proteins contribute to intracellular iron homeostasis 
in bacteria [22]. Ferritins are also found in eukaryotes, and heme-containing 
bacterioferritins are only found in bacteria. Both types are composed of 24 identi- 
cal subunits that form an almost spherical shell into which more than 2,000 Fe^"^ 
ions can be deposited. The FtnA ferritin of E. coli accumulates iron in the post- 
exponential growth phase in the presence of excess iron in the medium and sup- 
ports subsequent growth under non-deficient conditions. Helicobacter pylori and 
Campylobacter Jejuni express a similar protein that stores iron and protects cells 
against oxygen damage. No physiological role has been ascribed to the Bfr bacte- 
rioferritin of E. coli, but a bfr mutant of/? aeruginosa is sensitive to peroxides. 

Dps is another iron-binding protein that forms a shell, but with 12 sub- 
units. Dps is probably less important for iron storage than for protecting DNA 
against the combined action of iron and H2O2. 

Iron Transport ofE. coli <3«(i Shigella Related to Virulence 
Pathogenic E. coli strains express ten outer membrane proteins that transport 
ferric siderophores and heme (table I). All the ferric hydroxamates (aerobactin, 



Braun 



216 



Table L Iron transport systems o^ E. coli 



Substrate 


Outer membrane 


Peri plasn lie 


Cytoplasiiiic membrane 




protein 


protein 


proteins 


Enterobactin 


FepA 


FepB 


FepD", FepG^ FepC' 


Salmochelin 


IroN 


FepB 


FepDS FepGS FepC'' 


Catecholates 


Cir 


FepB 


FepD", FepG^ FepC' 


Catecho ates 


Fiu 


FepB 


FepD^ FepG^ FepC 


Ferrichrome 


FhuA 


FhuD 


FhuB^ FhuC'' 


Aerobactin 


lutA 


FhuD 


FhuB% FhuC 


Coprogen 


FhuE 


FhuD 


FhuB^ FhuC' 


Citrate 


FecA 


FecB 


Fece, FecD", FecE'' 


Heme 


ChuA 


ChuP 


ChuU^■^ ChuV''^'^ 


Yersiniabactin'' 


FyuA 


NI 


YbtP, YbtP 


Fe^^- 






FeoB 



^Transmembrane transport proteins in the cytoplasmic membrane, 

^ATPase. 

"Designations adapted from S. dysenteriae which is justified by the highly homologous E. coli 
and Shigella genomes. In E. coll K-12 ChuA alone is sufficient to support heme-dependent 
growth but the transport system in the cytoplasmic membrane may increase sensitivity to 
heme and rate of heme uptake. 

^The transport system of yersiniabactin is encoded on pathogenicity islands which occiu" 
in various Enterobacteriaceae. 

The nomenclature of reference 58 was used. For further details, see text and references 8 
and 49-5 L NJ = Not identified. 



ferrichrome, coprogen) for which specific transporters are found in the outer 
membrane are transported by the same transport system across the cytoplasmic 
membrane. The same holds true for the ferric catecholates, including ferric entero- 
bactin and presumably ferric salmochelin^ which are transported across the cyto- 
plasmic membrane by the same system. It is not clear whether or to what extent 
the entile FepBCD transport system is involved in the ferric salmochelin tians- 
port. The heme transport system has been characterized in Shigella dysenteriae 
and its phylogenetic distribution in enteric bacteria has been determined [52]. The 
assignment of the heme genes to functions is based on the fh'st functionally char- 
acterized heme transport system of Yersinia enterocolitica [53]. Heme and aero- 
bactin transport^ as well asTonB are required for vinjience of the uropathogenic 
E. coli strain CFT073 in a mouse model of urinary tract uifection [54]. In addition, 
E. coli strains isolated from patients with an intra-abdominal infection have been 
shown to secrete a protease, Hbp, that degrades hemoglobin. Hbp binds the released 
heme [55] and promotes the growth of Bactero ides fingi lis, which is frequently 



Iron Transport Related to Virulence 



217 



associated with E. coli in intra-abdominal infections. In a mouse infection model, 
Hbp contributes to the pathogenic synergy of these two organisms in abscess 
development. Heme transport systems are widely distributed among gram-positive 
and gram-negative bacteria [10, 11]. 

The Fe-^'^-yersiniabactm transport system is frequently encoded on a 'high 
pathogenicity island', which occurs in several Enterobacteriaceae [56], but is 
also present in strains with less pathogenic potential [57]. The transport system 
of Fe^"^-yersiniabactin across the cytoplasmic membrane is interesting since the 
two permease protems YbtP and YbtQ are each fused with the ATPase [58], as 
is found with human ABC export proteins. Subcutaneous infection by a ybtP 
mutant fails to cause disease \n mice, a route that mimics Yersinia pestis trans- 
mission by fleas causing bubonic plague. 

To date there has been no association reported between virulence and the 
ferric citrate transport system, in which FecB (binding protein), FecCD (perme- 
ase), and FecE (ATPase) catalyze transport across the cytoplasmic membrane. A 
nearly identical transport system is located on a pathogenicity island of Shigella 
flexneri [59]. Coliform isolates oiE. coli and Klebsiella pneumoniae from bovine 
inflammatory infections (mastitis) contain FecA, as evidenced by anti-FecA anti- 
bodies [60], and FecA is being considered as a vaccine component for the treat- 
ment of mastitis. A study of the regulation of the ferric citrate transport proteins 
uncovered a new type of transcription regulation. The inducer of the transcription 
of the transport genes binds to the FecA outer membrane protein and elicits a sig- 
nal that is transmitted by FecA across the outer membrane to the FecR protein, 
which transmits the signal across the cytoplasmic membrane. In the cytoplasm, 
the Feci sigma factor is activated and directs the RNA polymerase specifically to 
the promoter of the^ec transport genes upstream offecA [61, 62]. 

Siderophores like ferrichrome and coprogen, which are not synthesized by 
E. coli or any other bacteria, but which are transported by many bacteria, includ- 
ing E. coli, might be used during coinfection with fungi that synthesize the 
siderophores or during bacterial growth outside the human body. The large vari- 
ety of transport systems for ferric siderophores and heme found in E. coli and 
Shigella are typical for pathogenic bacteria. The systems are distiibuted among 
bacteria by horizontal gene transfer. For example, the aerobactin synthesis genes 
are found on plasmids in E. coli and Salmonella, on pathogenicity islands in 
S. flexneri and Shigella sonnei^ and on the chromosome of Shigella boydil and 
certain E. coli strains [8, 63]. Another example is the recently discovered iroN 
gene, which was originally identified in Salmonella enterica and then shown to 
contribute to the uropathogenicity of £■. coli isolates [64, 65]. iroN is encoded on 
a pathogenicity island on the chromosome [64] and on a transmissible plasmid 
[65]. In a mouse model of ascending urinary tract infection, IroN contributes to 
colonization of the bladder, kidneys, and urine [64]. 



Braun 



218 



In addition to the Fe^"*" transport systems, E. coli also contains an Fe^"*" 
transport system, which is encoded by the feoAB genes [23], This transport sys- 
tem functions Linder anoxic conditions, as found in the colon and in biofihns. 

Iron Transport o/ Salmonella Related to Virulence 

S. enterica serovar Typhimurium has h-on transport systems similar to those 
of E. coli and Shigella, but so far no heme or ferric citrate transport system 
has been described. However, a heme transport gene operon similar to that in 
>S'. dysenteriae is encoded on the Salmonella typhimurium genome. The known 
systems include those related to the outer membrane transporters FhuA, FepA, 
FoxA, Cir, and IroN. An additional transport system presumably transports iron, 
as was first demonstrated for the sfuABC iron transport system of 5'. marcescens 
[66] and then for the fbpABC system of A^. gonorrhoeae, hJbpABC (hit ABC) of 
H. influenzae [18], and yfuABC of Y. pestis [67]. sitA encodes a putative peri- 
plasmic permease, sitB an ATPase, and sitCD a permease [68]. However, sitABC 
is not homologous to the sfuABC-ty^iQ transport systems, but is homologous to 
yfeABC o^Y. pestis and it transports Mn'^"'" with a much higher affinity than Fe^"*". 
The Sit system is widely distributed in all S. enterica serovars and is required 
for full virulence of *S. typhimurium [69]; the Yfe system is essential for virulence 
of Y. pestis [70]. Iron transport systems are redundant, depending on the test sys- 
tem, since depleting one system may have no effect on bacterial virulence. The 
S. enterica genome also carries the feoAB genes, which encode an Fe^"^ transport 
system. Single mutations of sitA,feoB, or iucD (Fe^^-aerobactin transport) in 
S.flexneri do not impair the growth of these bacteria on a Henle cell monolayer; 
however, triple mutants do not form plaques [71]. 

A novel siderophore, designated salmochelin, was discovered only recently 
in S. enterica serovar Typhimurium LT2. The iroB gene product, encoded in the 
iron-regulated gene cluster iroNEDCB, shows sequence similarity to glycosyl 
transferases. This fmding prompted a search for the fionction of IroB. Indeed, IroB 
was shown to encode an enzyme that glucosylates enterobactin at the 5' position 
of the benzoyl ring, forming a C-C bond [106]. The published tentative structure 
carries the two glucosyl moieties inserted between two 2,3-dihydroxybenzoylser- 
ine residues [49]. In a Salmonella culture, sahnochelin is more abundant and is 
more soluble than enterobactin. Therefore, it might be less able to elicit antibod- 
ies than enterobactin, which serves, bound to serum albumin, as a hapten. 
Transport of Fe-'^-salmochelin across the outer membrane is mediated by IroN 
and to a lesser extent by the FepA and Cir transporters. 

Iron Transport of?, aeruginosa Related to Virulence 
Pyoverdin and pyochelin are two well-studied siderophores that supply 
iron to P. aeruginosa. A number of indications show a relationship between 



Iron Transport Related to Virulence 2 1 9 



iron supply and virulence of P. aemginosa in animal infection models: 
derepression of siderophore synthesis genes, synthesis of the siderophores 
pyoverdin and pyochelin and the related transport proteins, release of iron 
from the host iron-binding proteins transferrin and lactoferrin, and reduction 
of virulence of mutants deficient in synthesis of siderophores or Fe^"^- 
siderophore transport proteins. In addition, exotoxin A synthesis is controlled 
by the iron supply via the Fur repressor. A tonB mutant devoid of Fe-'^ uptake 
via pyoverdin, pyochelin, and heme grows in the muscles and lungs of 
immunosuppressed mice, but does not kill the animals [72]. Pyoverdin- and 
pyochelin-negative double mutants multiply, but do not kill the mice; however, 
intranasal inoculation of wild-type bacteria results in multiplication and killing 
[73]. PvdS (see below) is an ECF sigma factor synthesized in chronic lung 
infections affiliated with cystic fibrosis and contributes to the synthesis of 
exotoxin A [74]. 

Complex regulatory devices underlie iron-mediated control of gene expres- 
sion in P. aeruginosa. For example, iron-loaded Fur does not bind to the pro- 
moter of the toxA gene of exotoxin A, but acts via the pvf:/,^ gene product, which 
regulates 26 iron-repressible genes. pvdS encodes an ECF sigma factor of the 
Feci type (see Iron Transport of £". coli and Shigella Related to Virulence), and 
its synthesis is repressed by binding of Fe^"^-Fur to the pvdS promoter [75]. The 
activity of PvdS is controlled by pyoverdin secreted in the growth medium; 
pyoverdin (probably Fe-'"'" -pyoverdin) binds to the FpvA protein in the outer 
membrane. FpvA displays several functions: it acts as a signal receiver and as a 
signal transmitter across the outer membrane, and it transports Fe-'^ -pyoverdin 
across the outer membrane. The signal is transmitted by the FpvR protein across 
the cytoplasmic membrane into the cytoplasm, where PvdS is converted into an 
active sigma factor. Since PvdS is active in mutants lacking FpvR and over- 
expression of FpvR inactivates PvdS, FpvR probably fijnctions as an anti-sigma 
factor of PvdS [75]. PvdS directs the RNA polymerase to the promoter of the 
kon-repressible genes, uicluding the pyoverdm synthesis genes. fpvR transcrip- 
tion is repressed by Fe^"^-Fur, as is transcription of a second ECF sigma factor 
gene,^v/. Fpvl synthesis is regulated like PvdS synthesis via Fe^"^ -pyoverdin, 
FpvA, and FpvR, and controls synthesis of FpvA. 

Heme uptake by P. aeruginosa is mediated by two systems, one of which 
is encoded by the phuRSTUVW genes (fig. 2) [76]. This system is very similar 
to the heme transport system of Y. enterocolitica. Heme is bound to the PfuR 
outer membrane protein that transports heme across the outer membrane. 
Further transport into the cytoplasm is achieved by an ABC transporter. The 
other heme transport system is sunilar to the heme transport system of 
S. marcescens and involves a hemophore that is secreted, releases heme from 
hemoglobin, and delivers it to the outer membrane transport protein (fig. 2). 



Braun 



220 



hemP 



R 



S 



T 



U 



V 



Yersinia enterocolitica 




Yersinia pestis 



hmuX Y P R 

phuR 




S 



u 



V 



w 



Pseudomonas aeruginosa 



1=11 




t> 



shuS 



A 



Shigella dysen tehee 






T 



w 



X Y u y 



Vibn'o cholerae 



huLA tonBI exbBI hutB 

exbD1 



D 



hxuC 
Haemophilus Influenzae % i i 



hxuB 



hxuA 



hgpA 





hemO 



hmbR 



Neisseria meningitidis 



hpuA 



hpuB 




has! S 



R 



D 



B 



Serratia marcescens 



hasR 



Yersinia pestis 



Bordetella pertussis 



rhul R 



bhuR 




(ECF*) hasR 

Pseudomonas aeruginosa r[^ — [N n~ 



F 





A 



D 




^=t> 



Outer membranG receptor; perlplasmic binding protein; integrai membrane protein; ATPase subunit; 

i=t> 

Hemophore; hemophore secretion system; protein for heme utiiization 





Fig, 2. Heme transport systems of gram -negative bacteria. The upper panel shows the 
transport genes and some promoters (P). In the lower panel, genes for hemophore synthesis, 
secretion, and regulation, and not the actual heme transport genes are shown for S. marcescens, 
Y. pestis, and F aeruginosa. The HasA hemophores are secreted by the type 1 secretion mecha- 
nism catalyzed by the proteins HasD, HasE, and HasF. HasB is structurally and functionalJy a 
TonB-like protein, hasi and hasS, and rhuJ and rhuR encode a transcription-signaling device of 
the FeclR type in which the ) proteins represent extracytoplasmic membrane (ECF) sigma fac- 
tors that receive signals from outside the cytoplasm and the R or S protein transfers the signals 
across the cytoplasmic membrane. In Bordetella pertussis, rhuIR regulates transcription of the 
bhuRSTUV \\^m^ transport genes [for fijrther mformation, see 10, 62, 77]. 



Iron Transport Related to Virulence 



221 



In S. marcescens, regulation of heme transport gene transcription is mediated 
by a signaling device of the FecIRA type [77]. Heme-loaded hemophore binds 
to the HasR heme transporter and induces transcription of the hasR gene 
via HasI, which functions as an ECF sigma factor, and HasS, which acts as an 
anti-sigma factor. Since P. aeruginosa contains genes homologous to those in 
S. marcescens and arranged similarly, it is likely that the two Has regulatory 
systems function similarly. 

Analysis of the genome of P. aeruginosa predicts nine additional regula- 
tory devices of the FecIRA, HasISR, FpvA/Fpvl, FpvR, and PvdS type. These 
systems usually have the same gene arrangement asfecIRA, and the outer mem- 
brane proteins contain an extended amino-terminus, which in FecA interacts 
with FecR [12-14]. 

In addition to surface signaling elicited by the iron substrates, P. aeitiginosa 
controls iron usage by a number of additional regulatory mechanisms. For 
example, pyochelin synthesis and uptake is repressed by Fe^"^-Fur, which binds 
to promoters of the synthesis and uptake genes. The regulatory protein PchR 
acts as a repressor in the absence of pyochelin and as an activator in the pres- 
ence of pyochelin [78]. Regulation of ferric enterobactin usage is mediated by 
a two-component system consisting of the PfeS signal receiver and the PfeR 
response regulator. Ferric enterobactin in the periplasm binds to the PfeS sensor 
kinase, which is aiitophosphorylated and transfers the phosphate group to the 
receiver domain of PfeR. Phosphorylated PfeR functions as a transcription 
activator of the pfe A gene, which encodes the high-affinity PfeA outer mem- 
brane transporter [79]. In this iron transport system and in all the other iron 
transport systems studied in P aeruginosa, the transported substrate induces 
synthesis of the cognate transport system. This is achieved by various mecha- 
nisms, but always results in the economic adaptation of the cells to the available 
iron source. If only iron depletion of the Fur protein would derepress gene tran- 
scription, many of the approximately 13 iron transport systems would be 
synthesized, even though only the one for the available iron soui'ce would be 
required. 

Iron Transport o/Vibrio cholerae Related to Virulence 
Three heme transport systems have been identified in V cholerae^ repre- 
sented by the outer membrane transporters HutA, HutR, and HasR [80]. A huiA 
hutR double mutant is impaired, but not completely unable to use hemin as an 
iron source. The triple mutant hulA hutR hasR is completely devoid of heme 
utilization. V. cholerae HasR is similar to the HasR proteins of S. marcescens 
and P. aeruginosa^ which receive heme from the hemophore that releases heme 
from hemoglobin. In addition to the use of heme via transporters across the 
outer and cytoplasmic membranes, V. cholerae can use the iron complexes of 



Braun 



222 



the siderophores vibriobactin, enterobactin, and ferrichrome [81], The transporters 
are preferentially coupled to one of the twoTonB proteins present in V choleme 
[82], HasR, VctA, and IrgA, the latter two transport Fe^^-enterobactin [83], are 
only coupled to TonB2, whereas HutA, HutR, ViuA (Fe^^-vibriobactin trans- 
porter) and FhuA (ferrichrome transporter) can use TonBl and TonB2 [80]. \n 
an infant mouse model, the triple mutant competes with the wild-type strain, 
which indicated additional iron sources in vivo [80]. Analysis of gene tran- 
scription in the rabbit ileal loop model have revealed enhanced transcription of 
heme and Fe^^ transport genes and of they^o^^ genes, which encode an Fe^^- 
transport system [84] that may have supplied the necessary iron. 

Functions of Iron in Neisseria Related to Viimlence 

A tonB mutant of Neisseria meningitidis does not actively transport iron 
and is unable to replicate within epithelial cells [85], N, gonorrhoeae and 
N. meningitidis transport iron across the cytoplasmic membrane by an ABC 
transporter encoded by the JbpABC genes [18], which are similar to the sfuABC 
genes of *S. marcescens, the hfbpABC (hitABC) of//, influenzae, and the yf ABC 
genes of Y. pestis (see Iron Transport of Salmonella Related to Virulence). 
No siderophore seems to be involved in iron transport. In A^. gonorrhoeae and 
//. influenzae, the iron might be delivered by the host transferrin and lactoferrin, 
which bind to highly specific outer membrane receptor proteins composed of two 
polypeptides: TbpA and TbpB for the transferrin receptor, and LbpA and LbpB 
for the lactoferrin receptor The B components are lipoproteins and discriminate 
between iron-loaded and iron-unloaded transferrins and lactoferrins. The 
A components are similar to TonB-coupled ferric siderophore and heme 
transporters. TonB is not only required for the transport of iron across the outer 
membrane, but also for the release of Fe^"^ from transferrin and lactoferrin [21], 
The A and B components act in concert and interact with each other Proteolytic 
degradation of TbpB is strongly influenced by coupling of TbpA to TonB. 
A^. gonorrhoeae mutants that lack the transferrin receptor do not elicit symptoms 
of urethritis in human male volunteers [86]. 

Two hemoglobin receptors have been identified in A^. meningitidis: a tu^o- 
component receptor designated HpuAB and a one-component receptor designated 
HpmR. No siderophores have been identified in Neisseria. However, Neisseria can 
utilize Fe-^^-enterobactin taken up via a TonB-coupled transporter across the outer 
membrane and an ABC transporter across the cytoplasmic membrane [5, 87]. 

Iron Transport cj/ Staphylococcus aureus Related to Virulence 
In S. aureus^ several iron transport systems seem to operate. Ferrichrome is 
actively transported [88], and recently heme transport has been correlated with 
proteins (Isd) on the cell surface that are anchored to the murein by Iavo sortases 



Iron Transport Related to Virulence 223 



[89]. S. aureus binds transferrin [90] and haptoglobin-hemoglobin [91]. In certain 
strains, slime production is enhanced by iron limitation [92]. Iron homoeostasis 
is regulated by the Fur repressor, whose synthesis is repressed by a homologous 
protein, PerR, which also regulates synthesis of the iron storage proteins ferritin 
and MrgA, a Dps homolog. PerR is required for full virulence of >S. aureus in a 
murine skin abscess model [93]. The cell wall of 5". aureus and Staphylococcus 
epldermidis contains the Tpn transferrin-binding protein, which is synthesized 
under iron-limiting growth conditions and elicits antibody formation in human 
serum and peritoneum upon staphylococcal infections [94]. The Tpn protein is the 
cell wall glyceraldehyde-3-phosphate dehydrogenase, which also binds plasmin 
[95], It is assumed that the released iron is taken up into the cytoplasm by ABC 
transporters. Two such ABC transporters, encoded by the sir ABC and sstABCD 
genes, have been partially characterized [96]. 



Fe3"^-Siderophores as Antibiotic Carriers 

Multidrug resistance against currently used antibiotics forms an increasing 
problem in the treatment of bacterial diseases. One way out of the resistance 
dilemma is the development of new antibiotics. Since most antibiotics have been 
discovered during the decades of large-scale random screening, new strategies 
will have to be exploited. One possibility is the use of transport systems to trans- 
port antibiotics into cells. There are examples in which active transport, as 
opposed to diflfiision, decreases the minimal inhibitory concentration (MIC) of an 
antibiotic more than 100-fold [97]. 

Antibiotics with F^^-Hydroxamate Carriers 

Most antibiotics diffuse into bacteria, and their rate of diffusion and their 
activity at the target sites determine their efficiency, as measured by the MIC. In 
gram-negative bacteria, the outer membrane forms an additional permeability 
barrier in addition to the cytoplasmic membrane, and renders gram-negative 
bacteria less sensitive to many antibiotics than gram-positive bacteria. However, 
if antibiotics are actively transported across the outer membrane, their MIC 
could be lower in gram-negative than in gram-positive bacteria because the 
antibiotics are accumulated ui the periplasm and form a steep concentration gra- 
dient into the cytoplasm, thereby enhancmg the diffusion rate, or the antibiotic 
might even be actively transported across the cytoplasmic membrane. 

There are naturally occurring antibiotics that consist of an antibiotically 
active moiety and a siderophore carrier The best-studied example is albomycin, 
which is composed of a trihydroxamate that binds Fe^"^, a peptide linker, and a 
thioribosyl pyrimidine moiety that inhibits tRNA^*^"" synthetase [98]. Albomycin is 



Braun 



224 



highly active toward gram-positive and gram-negative bacteria. The MIC against 
an E. coli strain is 200 times lower (0,05 |JLg/mJ) than of ampicillin (12.5 jjug/mJ). 
The high specific activity comes from the active transport across the outer 
membrane and the cytoplasmic membrane into bacteria via the transport system 
of the structural analogue ferrichrome. The ferrichrome analogue serves as carrier 
of the antibiotically active thioribosyl pyrimidine group. After transport into the 
cytoplasm, iron is released from albomycin, and the thioribosyl pyrimidine 
group has to be cleaved from the carrier to be inhibitory. Tn E. coli, this is 
mainly achieved by peptidase N [1, 3, 97], Mutants devoid of peptidase N acti- 
vity are resistant to albomycin, and albomycin then serves as an iron carrier. 
Most of the thioribosyl pyrimidine moiety remains inside the cell, whereas the 
carrier is released into the culture medium. Albomycin is one of the very few 
antibiotics for which transport^ intracellular activation^ and target have all been 
characterized. 

Albomycin has been cocrystallized with FhuA to determine whether it 
binds to the ferrichrome binding site of FhuA and where the bulky side chain is 
located in FhuA (fig. 1). The crystal structure reveals that the Fe-^^-hydroxamate 
portion of albomycin occupies the same site on FhuA and is bound by the same 
amino acid side chains as ferrichrome [99], The thioribosyl pyrimidine moiety 
binds in the external pocket via five residues that are not involved in ferricKrome 
binding. The crystal structure also reveals the hitherto unknown conformation of 
albomycin and the conformation in the transport-competent form. Unexpectedly, 
albomycin assumes two conformations in the crystal - an extended and a com- 
pact conformation. Both conformations fit into the external cavity of FhuA and 
occupy seven different amino acid ligands. The solvent-exposed external cavity 
of FhuA is sufficiently large to accommodate the voluminous side chain of 
albomycin. 

After transport across the outer membrane by FhuA^ albomycin binds to 
FhuD in the periplasm. FhuD subsequently delivers albomycin to the permease 
in the cytoplasmic membrane. Cocrystals of FhuD with bound albomycin have 
been obtained in sufficient quality to determine the structure [100]. In contrast 
to FhuA, where albomycin sits inside the molecule, in FhuD albomycm is 
exposed to the surface of the protein. The thioribosyl moiety is not even seen in 
the crystal since it is not fixed to the protein and is thereby flexible. The fixa- 
tion of albomycin at the surface of FhuD explains the broader substrate speci- 
ficity of FhuD in contrast to FhuA since space is less restricted at the protein 
surface than within a protein. 

Results of studies with albomycin demonstrate that the proteins involved 
in transport across the outer membrane and the cytoplasmic membrane tolerate 
substantial modifications of the substrate. The modular design of albomycin can 
be synthetically mimicked. Antibiotics that are ineffective because of poor entry 



Iron Transport Relaied to Virulence 225 



into the cells can be chemically linked to ferrichrome and then transported into 
cells as ferrichrome derivatives. 

CGP 4832 is a semisynthetic rifamycin derivative with an activity against 
many gram-negative bacteria 200-fold higher than that of unmodified rifamycin 
[101], The reason for the increased activity of CGP 4832 is its energy-coupled 
transport by FhuA across the outer membrane ofE. coli [101]. The use of FhuA 
as transporter is surprising since CGP 4832 does not contain iron and has no 
structural resemblance to ferrichrome or any other hydroxamate. To obtain 
insights into how CGP 4832 is transported by FhuA, the crystal structure of 
FhuA loaded with CGP 4832 was determined [102], CGP 4832 occupies in 
FhuA largely the same site as ferrichrome (fig. 1 ). Nine residues that bind CGP 
4832 also bind ferrichrome. Of 16 amino acid residues that bind CGP 4832, 
5 residues recognize those side chains of CGP 4832 in which it differs from 
unmodified rifamycin. Two additional amino acid residues specifically bind the 
unique CGP 4832 side chains, whereas the other residues bind to sites that CGP 
4832 shares with rifamycin. The crystal structure reveals the conformation of 
CGP 4832, which demonstrates a completely different structure than that of ferri- 
chrome. Unlike albomycin, CGP 4832 is not transported via FhuBCD across 
the cytoplasmic membrane [101]. Rather, its active transport across the outer 
membrane results in an elevated concentration in the periplasm, which facilitates 
diffusion across the cytoplasmic membrane. It is the active transport across the 
outer membrane that reduces the MIC 200-fold. 

Salmycins have been isolated from SU^eptomyces violaceiis 37290 (DSM 
8286) and are highly active against staphylococci and streptococci (MIC 
10 |JLg/ml). Salmycins consist of an Fe-^'^-siderophore with a ferrioxamine group 
and an antibiotically active aminodisaccharide, which in salmycin B consists of 
a 2-ketoglucose linked to the 2-position of a 6-methylaminoheptopyranose 
[103]. It is assumed that the aminodisaccharide is released from the carrier by 
cleavage of the ester bond. Salmycins seem to inhibit protein synthesis by a yet 
unknown mechanism. 

Ferrimycins are among the first sideromycins discovered [97]. The action 
of ferrimycins is antagonized by ferroxamine B, which competes for ferri- 
mycin uptake. Ferrimycin inhibits incorporation of amino acids into proteins 
of ^. aureus SG5I I. Ferrimycin is difficult to isolate and for this reason has 
recently been studied less than aJbomycin and salmycin. 

Antibiotics with Fe^^-Catecholate Carriers 

Enterobactin is the most prominent catecholate siderophore with an 
extremely high Fe-^"^ stability constant. It consists of three dihydroxy benzoyl 
serine residues linked to a cyclic trimer by ester bonds. No natural Fe-^"^- 
catecholates with antibiotic activity are known. However, chemically synthesized 



Braun 



226 



catechol-substituted cephalosporins display MIC values below 1 |jLg/ml [104, 
105], particularly against gram-negative bacteria, including R aeruginosa. Their 
antimicrobial activities can exceed the activity of the unsubstituted 
cephalosporins more than 100-fold, Their high activity is related to their active 
transport into the periplasm^ where the target, the murein transpeptidase, is 
located. They are transported across the outer membrane by the Fe-^"^-catecholate 
transport proteins Fiu and Cir [26]. Iron limitation increases the susceptibility of 
E. coll strains since low iron derepresses Fiu and Cir synthesis. 

Resistance to Fe^^-Siderophore Antibiotics 

Resistant bacteria emerge on every nutrient agar plate containing anti- 
biotics that are carried into the bacteria by active Fe-^"^-siderophore transport 
systems. The higher the number of genes involved in a particular transport system, 
the higher the frequency of resistance. However, when two transport systems 
are used by an antibiotic, for example Cir and Fiu for the cephalosporin cate- 
cholates, the frequency of resistant mutants is low. Although the high resistance 
frequency seems to prevent development of such antibiotics as antibacterial 
drugs, the in vivo situation might be quite different In cases where an iron 
transport system is important for the proliferation of the pathogenic bacteria^ 
loss of the iron transport system is detrimental. Even when several iron trans- 
port systems exist and only one is inactivated by resistance to a particular 
antibiotic, the inactivated system might be the one that is essential for the bac- 
teria to survive and multiply at the site of infection in the human host. Under 
these circumstances, it does not matter whether the number of bacteria is reduced 
by the antibiotic or by loss of the iron supply since under both conditions the 
immune defense system gains time to cope with the infection. 



Concluding Remarks 

Iron deficiency was also designated nutritional immunity which meant that 
growth inhibition by lack of iron prevents bacterial multiplication. Lack of 
growth or growth retardation gives the natural and the adaptive immunity sys- 
tem the chance to cope with an infection. Iron is the only nutrient for which an 
essential role in growth of many bacterial pathogens causing various diseases 
in humans and animals has been demonstrated. There are certainly many more 
nutrients which play a decisive role in extra- and intracellular multiplication of 
bacteria. However, it is difficult to identify these nutrients. Large-scale expres- 
sion profiles of metabolic genes in bacteria isolated from human patients with- 
out further cuJturing and from animal models may indicate metabolic pathways 
from which the nutrients may be derived. From a purely scientific point of view 



Iron Transport Relaied to Virulence 227 



the iron supply systems are of great interest with regard to the various ways 
insoluble Fe^"^ is complexed by siderophores, heme, transferrin, and lactoferrin 
and transported into the bacterial cells by distinct and very sophisticated mech- 
anisms. For the avoidance of iron shortage and iron surplus the transport systems 
are regulated by various means, iron-dependent repression, downregulation by 
small RNAs, transcription enhancement by two-component systems, and tran- 
scription initiation by surface signaling. In the fliture, a detailed knowledge of 
iron uptake and intracellular iron metabolism may be applied to interfere with 
bacterial growth as a means to control bacterial diseases, and siderophore 
antibiotics (sideromycins) may be used when treatment with other antibiotics 
fails because of resistance. 



Acknowledgments 

I would like to thank Klaus Hantke for preparation of figure 2, Michael Braun for 
preparation of figure 1, and Karen A, Brune for critically reading the manuscript. The 
author's work was supported by the Deutsche Forschungsgemeinschaft (Forschergruppe 
^Bakterielle ZeUJiiiUe: Synthese, Funktion und Wijkort', Br 330/14-2) and the Fonds der 
Chemischen Industrie, 



References 

1 Braun V, Hantke K, Koster W: Bacterial iron transport: Mechanisms, genetics, and regulation; in 
Sigel A, Sigel H (eds): Metal Ions in Biological Systems. New York, Marcel Dekker, 1 998, vol 35, 
pp 67-145. 

2 Braun V: Iron uptake mechanisms and their regulation in pathogenic bacteria. Int J Med Microbiol 
2001;291:67-79. 

3 Braun V, Braun M: Active transport of iron and siderophore antibiotics. Curr Opin Microbiol 
2002;5:194-201. 

4 Braun V: hon uptake by Escherichia coll. Front Biosci 2003;1:409-421. 

5 Rhode KH, Dyer DW: Mechanisms of iron acquisition by the human pathogens Neisseria 
meningitidis and Neisseria gonorrhoeae. From Biosci 2003;8: 1 186-1218. 

6 Bullen JJ, Griffith E: Iron and Infection. Molecular, Physiological and Clinical Aspects, New York, 
Wiley, 1999. 

7 Hantke K, Braun V: The art of keeping low and high iron concentrations in balance; in Storz G, 
Hengge-Aronis R (eds): Bacterial Stress Responses. Washington, ASM Press, 2000, pp 275-288. 

8 Crosa JH: The relationship of plasmid-mediated iron transport and bacterial virulence. Annu Rev 
Microbiol 1984;38:69-89. 

9 Crosa JH: Signal transduction and transcripiional and posttranscriptional control of iron-regulated 
genes in bacteria. Microbiol Mol Biol Rev 1997;61:319-336. 

10 Wandersman C, Stojiljkovic I: Bacterial heme sources: The role of heme, hemoprolein receptors 
and hemophores. Curr Opin Microbiol 2000;3:215-220, 

1 1 GencoCA, Dixon DW: Emerging strategies in microbial haem capture. Mol Microbiol 2001;39:1-1 K 

12 Vasil ML, Ochsner UA: The response of Pseudomonas aeruginosa to iron: Genetics, biochemistry 
and virulence. Mol Microbiol 1999;34:399-413. 



Braun 



228 



13 ViscaP, Leono L, Wilson MJ, LamonLlL: Iron transport and regulation, cell signaling and genomics: 
Lessons from Escherichia coli and Pseudomonas aeruginosa. Mol Microbiol 2002;45: 1 177-11 90. 

14 Martinez-Bueno MA, Tobes R, Rey M, Ramos JL: Detection of multiple extracytoplasmic func- 
tion (ECF) sigma factors in the genome of Pseiidomonas putida KT2440 and their counterparts 
in Pseudomonas aeniginosa PAOl. Environ Microbiol 2002;4:842-855. 

15 Koster W: ABC transporter-mediated uptake of iron, siderophores, heme and vitamin B12. Res 
Microbiol 2001;152:292-301. 

16 Clarke TE, Tari LW, Vogel HJ: Structural biology of bacterial iron uptake systems. CurrTop Med 
Chem 2001;1:7-30. 

1 7 Ratledge C, Dover LG: Iron metabolism in pathogenic bacteria. Rev Microbiol 2000;54:88 1-941 . 

18 MietznerTA, Tencza SB, Adhikari P, Vaughan KG, Mowalk AJ: Fe(]ll) periplasm-to-cytosol trans- 
porters of gram-negative pathogens. CurrTop Microbiol Immunol 1998;225:1 13-135. 

19 Woolridge KG, Williams PH: Iron uptake mechanisms of pathogenic bacteria. FEMS Microbiol 
Rev 1993;12:325-348. 

20 Schryvers AB, Bonna R, Yu RH, Wong H, Retzner M: Bacterial lactoferrin receptors. Adv Exp 
MedBioJ 1998;443:123-133. 

21 Cornelissen CN: Transferrin-iron uptake by gram-negative bacteria. Front Biosci 2003;8:836-847. 

22 Andrews SC, Robinson AK, Rodriguez-Quinones F: Bacterial iron homoestasis. FEMS Microbiol 
Lett 2003;27:215-237. 

23 Hantke K: Is the bacterial ferrous iron transporter FeoB a living fossil? Trends Microbiol 
2003;11:192-195. 

24 Perry RD, Abney J, Mier ] Jr, Lee Y, Bearden SW, Felherston JD; Regulation of the Yersinia pesiis 
Yfe and Ybt iron transport systems. Adv Exp Med Biol 2003;529:275-283. 

25 Davidson AL: Mechanism of coupling of transport to hydrolysis in bacterial ATP-binding cassette 
transporters. J Bacteriol 2002;184:1225-1233. 

26 Bruns C, Nowalk A, Arvai A, McTigue M, Vaughan K, Mielzner T, McRee D: Structure of 
Haemophihis influenzae Fe^^-binding protein reveals convergent evolution within a superfamily, 
Nat Stnict Biol 1997;4:919-924. 

27 Clarke TE, Braun V, Winkelmann G, Tari LW, Vogel HJ: X-ray crystallographic structures of the 
Escherichia coli periplasmic protein FhuD bound to hydroxam ate- type siderophores and the 
antibiotic albomycin. J Biol Chem 2002;277:13966-13972. 

28 Locher KJ*, Lee AT, Rees DC: The E. coli BtuCD structure: A framework for ABC transporter 
architecture and mechanism. Science 2002;296:1091-1098, 

29 Borths EL, Locher KJP, Lee AT, Rees DC: The structure 0^ Escherichia coli BtuF and binding to 
its cognate binding cassette transporter. Proc Natl Acad Sci USA 2002;99:16642-16647. 

30 Groeger W, Koster W: Transmembrane topology of the two FhuB domains representing the 
hydrophobic components of bacterial ABC transporters involved in the uptake of siderophores, 
haem and vitamin B12. Microbiology 1998;144:2759-2769. 

31 Nikaido H: Prevejition of drug access to bacterial taigets: Perjneability barriers and active efifluA. 
Science 1994;264:382-388. 

32 Bradbeer C: The proton motive force drives the outer membrane transport of cobalamin in 
Escherichia coli. ] Bacteriol 1993;175:3146-3150. 

33 Braun V: Energy-coupled transport and signal transduction through the gram-negative outer 
membrane via TonB-ExbB-ExbD-dependent receptor proteins. FEMS Microbiol Rev 1995; 16: 
295-307. 

34 Postle K, Kadner RJ: Touch and go: Tying TonB to transport. Mol Microbiol 2003;49:869-882. 

35 Ferguson AD, Hofmann E, Coulton JW, Diederichs K, Welte W: Structural basis for siderophore- 
mediated iron transport; Crystal structure of FhuA with bound lipopolysaccharide. Science 
l998;282;2215-2220. 

36 Locher ICP, Rees B, Koebnik R, Mit^chler A, Moulinier L, Rosenbusch JP, Moras D: 
Transmembrane signaling across the ligand-gated FhuA receptor: Crystal structures of free and 
ferrichrome-bound states reveal allosteric changes. Cell 1998;95:771-778. 

37 Buchanan SK, Smith BS, Venkatramani L, Xia D, Esser L, Palnitkar M, Chakraborty R, van der 
Helm D, Deisenhofer J: Crystal structure of the outer membrane active transporter FepA from 
Escherichia coli. Nat Struct Biol 1999;6:56-63. 



Iron Transport Related to Virulence 229 



38 Ferguson AD, Chakrabotty R, Smith BS, Esser L, van der Helm D, Deisenhofer J: Structural basis 
of gating by the outer membrane transporter FecA, Science 2002;295:1658-1659. 

39 Wyatt W, Grizot S, Buchanan SK: Structural evidence for iron-free citrate and ferric citrate 
binding to the TonB-dependent outer membrane transporter FecA. J Mol Biol 2003;332: 
353-368, 

40 Merianos HJ, Cadieux N, Lin CH, Kadner RJ, Cafiso DS: Substrate-induced exposure of an energy- 
coupling motif of a membrane transporter. Nat Struct Biol 2000;7:205-209. 

41 Cadieux N, KLadner RJ: Site-directed disulfide bonding reveals an interaction site between energy- 
coupling protein TonB and BtuB, the outer membrane cobalamin transporter. Proc Natl Acad Sci 
USA 1999;96:10673-10678. 

42 Ogiermaa M, Braun V: Interactions between the outer membrane ferric citrate transporter FecA 
and TonB: Studies of the FecA TonB box. J Bacteriol 2003;185:1870-1885. 

43 Cimento DP, Mohanty AK, Kadner RJ, Wiener MC: Substi'ate-induced transmembrane signaling 
in the cobalamin n-ansponer BtuB. Nat Struct Biol 2003; 10:394-^01. 

44 Reeves SA, Torres AG, Payne SM:TonB is required for intracellular growth and virulence of Shigella 
dysenterioe. Infect Jmmun 2000; 1 1:6329-6336. 

45 Torres AG, Redford P, Welch RA, Payne SM: TonB-dependent systems of uropathogenic 
Escherichia coli: Aerobactin and heme transport and TonB are required for virulence in the mouse. 
Infect Immun 2001 ;69:6 1 79-6 1 85. 

46 Kurz CL, Chavet S, Andres E, Aurouze M, Vallet 1, Michel GP, Uh M, Celli J, Filloux A, 
de Bentzmann S, Steinmelz 1, Hoffinann JA, Finlay BB, Gorvel J-P, Ferrandon D, Ewbank JJ: 
Virulence factors of the human opportunistic pathogen Serratia marcescens identified by in vivo 
screening. EMBO J 2003;22: 145 1-1460. 

47 Braun V: Avoidance of iron toxicity through regulation of bacterial iron transport, Biol Chem 
1997;378:779-786. 

48 Touati D, Jacques M, Tardat B, Bouchard L, Despied S: Lethal oxidative damage and mutagenesis 
are generated by iron in/wr mutants of Escherichia coli: Protective role of superoxide dismutase. 
J Bacteriol 1995;I77:2305-23J4. 

49 Hantke K, Nicholson G, Rabsch W, Winkelmann G: Salmochelins, siderophores of Salmonella 
enterica and uropathogenic Escherichia coli strains, are recognized by the outer membrane receptor 
IroN. Proc Natl Acad Sci USA 2003;100:3677-3682, 

50 Schubert S, Rakin A, Karch H, Camiel E, Heesemann J: Prevalence of the 'high-pathogenicity 
island' of Yersinia species among Escherichia coli strains that are pathogenic to humans. Infect 
Immun 1998;66:480-485. 

51 Torres AG, Payne SM: Haem transport in enterohemorrhagic Escherichia coli 0157:H7. Mol 
Microbiol 1997;23:825-833. 

52 Wyckoff EE, Duncan D, Torres AG, Mills M, Maase K, Payne SM: Structure of the Shigella 
dysenteriae haem transport locus and its phylogenetic distribution in enteric bacteria. Mol 
Microbiol 1998;28:1139-1 152. 

53 Stojiljkovic I, Hantke K: Hemin uptake systems of Yersinia enterocoliiica: Similarities with other 
TonB-dependent systems in gram-negative bacteria. EMBO J 1992;1 1:4359-4367. 

54 Torres AG, Redford P, Welch RA, Payne SM: TonB-dependent systems of uropathogenic Escherichia 
coli: Aerobactin and heme transport and TonB are required for virulence in mice. Infect Immun 
2001;69:6179-6185. 

55 Otto BR, van Dooren SJ, Dozois CM, Luirink J, Oudega B: Escherichia coli hemoglobin contributes 
to synergistic abscess formation and heme -dependent growth of Bacieroidesfragilis. Infect Immun 
2002;70:5-10. 

56 Schubert S, Picard B, Gouriou S, Heesemann J, Denamur E: Yersinia high-pathogenicity island 
contributes to virulence in Escherichia coli causing extraintestinal infections. Infect Immun 
2002;70:5335-5337. 

57 Oelschlager TA, Zhang D, Schubert S, Carniel E, Rabsch W^ Karch H, Hacker J: The high- 
pathogenicity island is absent in human pathogens of Salmonella enterica subspecies I but present 
in isolates of subspecies III and VI. J Bacteriol 2003;185:1007-1 111. 

58 Fetherton JD, Bertolino VJ, Perry RD: YbtP and YbiQ: Two ABC transporters required for iron 
uptake in Yersinia pes tis, Mol Microbiol 1999;32:289-299. 



Braun 



230 



59 Luck SN, Turner SA, Rajakumar K^ Sakellaris H, Adler B: Ferric dicitrate transport system (Fee) of 
Shigella JJexneri 2a YSH6000 is encoded on a novel pathogenicity island carrying multiple antibiotic 
resistance genes. Infect Immun 2001;69:6012-6021. 

60 Lin J, Hogan JS, Smith KL: Antigenic homology of the inducible ferric citrate receptor (FecA) of 
coliform bacteria isolated from herds with naturally occurring bovine intramammary infections. 
Clin Diagn Lab Immunol 1999;6:966-969. 

6 1 Braun V: Surface signaling: Novel transcription initiation mechanism starting from the eel I surface. 
Arch Microbiol 1997;237:325-331. 

62 Braun V, Mahren S, Ogiennan M: Regulation of the Feel-type ECF sigma factor by transmembrane 
signalling. CurrOpin Microbiol 2003;6:173-180. 

63 Yokes SA, Reeves SA, Torres AG, Payne SM: The aerobactin iron transport system genes '\n Shigella 
/7ex«^/ are present within a pathogenicity island. Mol Microbiol 1999;33:63-73. 

64 Russo TA, McFadden CD, Carlino-MacDonald UB, Beanaii JM, Barnard TJ, Johnson JR; iroN 
functions as a siderophore receptor and is a urovirulence factor in an extraintestinal pathogenic 
isolate of Escherichia coii. Infect fmmun 2002;70:7 1 56-7 1 60. 

65 Sorsa LJ, Dufke S, Heesemann J, Schubert S: Characterization of an iroBCDEN gene cluster on a 
transmissible plasmid of uropathogenic Escherichia coli: Evidence for horizontal transfer of a 
chi'omosomal virulence factor. Infect Immun 2003;71:3285-3293. 

66 Angerer A, Gaisser S, Braun V: Nucleotide sequences of ihesJiiA.sfiiB, and .s/wC genes oTSerratia 
marcescens suggest a periplasmic-binding-protein-dependent iron transport mechanism. J Bacteriol 
1990;172:572-578. 

67 Gong S, Bearden SW, Geoffroy VA, Fetherston JD, Perry RD: Characterization of the Yersinia 
pestis Yfu ABC inorganic iron transport system. Infect Immun 2001;67:2829-2837. 

68 Zhou D, Hardt WD, Galan JE: Salmonella typhimuhum encodes a putative iron transport system 
within the centisome 63 pathogenicity island. Infect Immun 1999;67:I974— 1981. 

69 Janakiraman A, Slauch JM: The putative iron transport system siiABCD encoded on SPIl 
is required for full virulence of Salmonella lyphimurium. Mol Microbiol 2000;35: 
1146-1155. 

70 Bearden SW, Perry RD: The Yfe system of Yersinia pestis transports iron and manganese and is 
required for full virulence of plague. Mol Microbiol 1999;32:403^I4. 

71 Runyen-Janecky LJ, Reeves SA, Gonzales EG, Payne SM: Contribution of the Shigella Jlexneri 
Sit, luc, and Feo iron acquisition systems to iron acquisition in vitro and in cultured cells. Infect 
Immun 2003;71:1919-1928, 

72 Takase H, Nitanai H, Hoshino K, Otani T: Impact of siderophore production on Pseudomonas 
aeruginosa infections in immunosuppressed mice. Infect Immun 2000;68:1834-1839. 

73 Takase H, Nitanai H, Hoshino K, Otani T: Requirement of the Pseudomonas aemginosa tonB gene 
for high-affinity iron acquisition and infection. Infect Immun 2000;68:4498^504. 

74 Hunt TA, Peng WT, Loubens 1, Storey DC: The Pseudomonas aeruginosa alternative sigma factor 
PvdS controls exotoxin A expression and is expressed in lung infections associated vi'ith cystic 
fibrosis. Microbiology 2002;148:3183-3193. 

75 Beare PA, For RJ, Martin LW, Lamont IL: Siderophore-mediated cell signaling in Pseudomonas 
aeruginosa: Divergent pathways regulate virulence factor production and siderophore receptor 
synthesis, Mol Microbiol 2003;47:195-207. 

76 Ochsner UA, Johnson Z, Vasil ML: Genetics and regulation of two distinct haem uptake systems, 
pflj and has, in Pseudomonas aeruginosa. Microbiology 2000;146:185-198. 

77 Rossi MS, PaquelinA, GhigoJM, Wandersman C: Haemophore-mediated signal transduction across 
the bacterial cell envelope in Setratia marcescens: The inducer and the transported substrate are dif- 
ferent molecules. Mol Microbiol 2003;48:1467-1480. 

78 Heinrichs DE, Poole K: PchR, a regulator of ferric pyochelin receptor gene (JplA) expression in 
Pseudomonas aeruginosa, functions both as an activator and as a repressor. J Bacteriol 1996; 175: 
5882-5889. 

79 Dean CR, Neshat S, Poole K: PfeR, an enterobactin-responsive activator of ferric enterobactin 
receptor gene expression in Pseudomonas aeruginosa. J Bacteriol 1996;178:5361-5369. 

80 Mey AR, Payne SM: Haem utilization in Vibrio cholerae involves multiple TonB-dependent haem 
receptors. Mol Microbiol 2001;42:835-849, 



Iron Transport Related to Virulence 231 



81 Rogers MB, Sexton JA, DeCastro GJ, Calderwood SB: Identification of an operon required for 
ferrichrome iron utilization in Vibrio cholerae, J Bacteriol 2000;182:2350-2353, 

82 Occhino DA, Wyckoff EE, Henderson DP, WronaTJ, Payne SM: ytbho cholerae iron transport: 
Haem transport genes are linked to one of two sets of lonB, exbB, exbD genes. Mol Microbiol 
1998;29;I493-1507. 

83 Mey AR, Wyckoff EE, Oglesby AG, Rab E, Taylor RK, Payne SM: Identification of the Vibrio 
cholerae enterobactin receptors VctA and IrgA: IrgA is not required for virulence. Infect Immun 
2002;70:3419-3426. 

84 Xu Q, Dziejman M, Mekalanos JJ: Determination of the iranscriptome of Vibrio cholerae during 
intraintesiinal growth and nriidexponential phase in vitro. Proc Natl Acad Sci USA 2003; 1 00: 
1286-1291. 

85 Larson JA, Higashi DL, Stojiljkovic I, So M: Replication oi^ Neisseria menigitidis within epithelial 
cells requires TonB-dependent acquisition of host cell iron. Infect Immun 2002;70:1461-1467. 

86 Comelissen CN, Kelley M, Hobbs MM, Anderson JE, Cannon JG, Cohen MS, Sparling RF: The 
transferrin receptor expressed by gonococcal strain FA 1090 is required for the experimental 
infection of human male volunteers. Mol MicrobioJ 1998;27:611-616. 

87 Biegel Carson SD, KJebba PE, Newton SMC, Sparling PF: Ferric enterobactin binding and utilization 
by Neisseria gonorrhoeae. J Bacteriol 1 999; J 8 1 ;2895-290 1 . 

88 Sepulsky MT, Heinrichs DE: Identification and characterization oiffmDl and /hiiD2, tv/o genes 
involved in iron-hydroxamate uptake hy Staphylococcus aureus. J Bacteriol 2001 ;1 83:4994—5000. 

89 Mazmanian SK, Skaar EP, Caspar AH, Humayun M, Gornicki P, Jelenska J, Joachmiak A, 
Missiakas DM, Schneewind 0: Passage of heme-iron across the envelope of Staphylococcus 
aureus. Science 2003;299:906-909. 

90 Taylor JM, Heinrichs DE: Transferrin binding in Staphylococcus aureus: Involvement of a cell 
wall-anchored protein. Mol Microbiol 2002;43:1603-1614. 

91 Dryla A, Gelbmann D, von Gabain A, Nagy E: Identification of a novel iron regulated 
staphylococcal surface protein with haptoglobin-haemoglobtn binding activity, Mol Microbiol 2003; 
49:37-53. 

92 Baldassarri L, Berluccini L, Ammendolia MG, Arciola CR, Montana L: Effect of iron limitation 
on slime production by Staphylococcus aureus. Eur J Clin Microbiol Infect Dis 200l;20: 
343-345. 

93 Horsburgh MJ, Clements MO, Crossley H, Ingham E, Foster SJ: PerR controls oxJdative stress 
resistance and iron storage proteins and is required for virulence in Staphylococcus aureus. Infect 
Immun 2001;69:3744-3754. 

94 Modun BJ, Cockayne A, Finch R, Williams P: The Staphylococcus aureus and Staphylococcvs 
epidermidis transferrin-binding proteins are expressed in vivo during infection. Microbiology 
1998;144:1005-1012. 

95 Modun BJ, Evans RW, Joannou CL, Williams P: Receptor-mediated recognition and uptake of iron 
from human transferrin by Staphylococcus aureus and Staphylococcus epidermidis. Infect Immun 
1998;66:3591-3596. 

96 Morrissey JA, Cockayne A, Hill PJ, Williams P: Molecular cloning and analysis of a putative 
siderophore ABC transporter from Staphylococcus aureus. Infect Immun 2000;68:6281-6288. 

97 Braun V: Active transport of siderophore -mimicking antibacterials across the outer membrane. 
Drug Resist Updat 1999;2:363-369, 

98 Stefanska AL, Pulsion M, Houge-Frydrych CS, Jones JJ, Warr SR: A potent seryl IRNA 
synthetase inhibitor SB-2 17452 isolated from a Streptomyces species, J Antibiot 2000;53: 
1346-1353. 

99 Ferguson AD, Braun V, Fiedler HP, Coulton JW, Diederichs K, Welte W: Crystal structure of the 
antibiotic albomycin in complex with the outer membrane transporter FhuA, Protein Sci 2000;9: 
956-963. 

100 Clarke TE, Braun V, Winkelmann G, Tari LW, Vogel HJ: X-ray crystallographic structures of the 
Escherichia coli periplasmic protein FhuD bound to hydroxamate-type siderophore and the antibiotic 
aJbomycin. J Biol Chem 2002;277:13966-13972. 

101 Pugsley PA, Zimmerman W, Wehrli W: Highly efficient uptake of a rifamycin derivative via FhuA- 
TonB-dependent uptake route in Escherichia coli. J Gen Microbiol 1987;133:3505-351 1. 



Braun 



232 



102 Ferguson AD, Kodding J, Walker G, Bos C, Coulton JW, Diederichs K, Braun y Welte W: Active 
transport of an antibiotic rifamycin derivative by the outer-membrane protein FhuA, Structure 
2001;9:707-716. 

103 Vertesy L, Aretz W, Fehlhaber HW, Kogler H, Salmycins AD: Antibiotics from Strepiomyces 
violaceiis DSM 8286 having a siderophore-aminoglycoside sti'uctui'e. Helv Chim Acta 1995;78: 
46-60. 

104 Dolence EK, Minnick AA, Lin CE, Miller M: Synthesis and siderophore and antibacterial activity of 
N^-acetyl-N^-hydroxy-L-ornithine-derived siderophore-p-lactam conjugates: Iron-transport-mediated 
drug delivery. J Med Chem 1 99 1 ;34:868-978. 

105 Curtiss NAC, Eisenstadt RL, East SJ, Cornford RJ, Walker LA, White AJ: Iron regulated outer 
membrane proteins of Escherichia coli K-12 and mechanisms of action of catechol-substituted 
cephalosporins. Antimicrob Agents Chemother 1 988;32: 1 879- 1 886. 

106 Bister B, BischofT D, Nicholson GJ, Valdebenito M, Schneider K, Winkelmann G, Hantke K, 
Sussmuth RD: The structure of salmochelins: C-glucosyiated enterobaciins of Salmonella enienca. 
BioMetals2004;l7:47l^8l. 



Prof. Volkmar Braun 

Mikrobiologie/Membranphysiologie, Universitat Tubingen 
Auf der Morgenstelle 28, DE-72076 Tubingen (Germany) 
Tel. +49 7071 2972096, Fax +49 7071 2975843 
E-Mail volkmar.braun@mikrobio.uni-tuebingen.de 



Iron Transport Related to Virulence 233 



Signaling and Gene Regulation 

Russell W, Herwald H (eds); Concepts in Bacterial Virulence, 
Contrib Microbiol. Basel, Kai'ger, 2005, vol 12, pp 234-254 



Pathogenicity Islands and Their Role 
in Bacterial Virulence and Survival 



Bianca Hochhiit, Ulrich Dobrindt, Jorg Hacker 

Institut fur molelaiJare Infektionsbiologie, Universitat Wiirzburg, 

Wiirzburg, Germany 



Infections caused by bacterial pathogens are still a significant problem in 
modern medicine. Therefore, the identification of the factors that are related to 
the infections and the understanding of the processes involved in the evolution 
of pathogenic bacteria from their nonpathogenic progenitors is an important 
subject of research. It has long been known that acquisition of virulence deter- 
minants by horizontal gene transfer is one of the major driving forces in the 
emergence and evolution of new pathogens [reviewed in 1^]. Furthermore, our 
knowledge of the organization of the bacterial genome has greatly increased 
within the last few years due to the availability of more than 120 completely 
sequenced eubacterial genomes, including those of almost all pathogenic 
bacteria, which has introduced a new area of pathogen research. It has become 
evident that the typical bacterial genome consists of a conserved 'core gene 
poor comprising genes that encode essential structural features and fundamental 
metabolic pathways, and a 'flexible gene pool' that is more variable and encodes 
functions only advantageous under specific growth conditions. Core genes are 
characterized by a relatively homogenous G + C content and tliey are normally 
encoded in stable regions of the chromosome that are conserved in their orga- 
nization in closely related species. In contrast, the flexible gene pool comprises 
variable regions of the chromosome and various mobile genetic elements such 
as plasmids, bacteriophages, IS elements and transposons, conjugative trans- 
posons, integrons and superintegrons that are transferred between different 
organisms by the means of natural transformation, transduction or conjugation. 
Many of the genes encoding toxins, adhesins, secretion systems, invasins or 
other virulence-associated factors have been found to be encoded by mobile 
genetic elements [overviews in 5^ 6]. Furthermore, the analysis of the genomes 



of closely related species has revealed that the conserved chromosomal 
backbone is interspersed with large regions that exhibit features of former 
mobile genetic elements that have been termed genomic islands (GEls) [7, 8]. 
GEIs are broadly distributed and seem to be a common theme in most bacterial 
genomes. Originally, such elements were identified in uropathogenic 
Escherichia coii strains and were designated 'pathogenicity islands' (PAIs), 
because they encoded key virulence factors of these bacteria [9], However, 
when regions with similar features were also found in nonpathogenic bacteria 
where they encoded other accessory functions, it was recognized that these 
elements are not limited to bacterial pathogens, but are present in most bacteria 
that have been analyzed. In this chapter, the role of GEIs in bacterial virulence 
and survival will be discussed. 



The Concept of GEIs 

Features of GEIs 

A comparative analysis of microbial genome sequences has revealed that 
bacterial genomes can harbor variable and frequently significant amounts of 
foreign DNA [3]. The genome size of different variants of the same species or 
closely related species can vary by more than one megabase, which can be 
accounted for by the acquisition of large blocks of DNA such as plasmids, 
bacteriophages and GEIs, as well as by the acquisition of smaller pieces of foreign 
DNA that have been described as 'islets'. Generally, GEIs represent distinct 
pieces of DNA that have most of the following features in common suggesting 
that they originate from events of lateral gene transfer [10]. 

(1) GEIs are present in the genomes of many bacteria but absent from the 
genomes of closely related strains or species. (2) GEIs occupy relatively large 
regions of the chromosome and can cover between 10 and more than 100 kb, 
which may reflect the introduction of large pieces of DNA into a new host by 
horizontal gene transfer Some strains also carry smaller pieces of DNA (1-1 Okb) 
that have been termed 'genomic islets' in contrast to the larger islands. (3) GEIs 
differ in their G + C content and their codon usage from that of the conserved 
regions of the chromosome. (4) GEIs are often flanked by direct repeats that 
may have been generated during integration of GEl-specific regions into the 
host chromosome via site-specific recombination. (5) GEIs are frequently asso- 
ciated with tRNA loci. The 3' end of tRNA genes have been recognized as pre- 
ferred target sites for the integration of foreign DNA [reviewed in i 1]. (6) GEIs 
often possess functional or cryptic genes coding for factors that are involved in 
genetic mobility such as integrases, transposases, phage genes and origins of repli- 
cation. Furthermore, GEIs normally do not represent homogenous elements but 



Pathogenicity Islands 235 



rather are generated by multistep processes including DNA rearrangements via 
IS elements which is reflected by mosaic-like structures, (7) Some GEls tend to 
be unstable DNA regions due to recombination between the flanking direct 
repeats, between IS elements or between other regions of homologous sequences. 
Generally, little is known about the mechanisms that have led to the acquisition 
of GEIs and there are only few examples of inter- or intracellular mobilization 
of GEls [12-16], 

GEls are prevalently found in organisms that show frequent gene transfer 
by bacteriophages and plasmids which are regarded as possible precursors of 
GEls [8]. However, GEls have also been described in bacteria that exhibit 
natural competence such as Helicobacter pylori. Neisseria gonorrhoeae and 
Streptococcus pneumoniae, and that tend to introduce smaller pieces rather than 
large regions of foreign DNA into their genome [17-19]. 

GEIs Contribute to Bacterial Fitness 

Besides selfish genes such as genes involved in recombination and trans- 
fer or modification of DNA, GEls often carry determinants that are beneficial 
for their host bacterium in certain environments thereby increasing bacterial fit- 
ness and consequently survival. GEls were divided into different subtypes 
reflecting their contribution to the respective microbial lifestyle [8] (table 1), 
GEls that encode virulence traits were defined as 'pathogenicity islands' (PAls). 
The original definition of GEls was based on the characteristics of PAls in patho- 
genic E. coll, but mtensive studies of the genome structure of bacterial pathogens 
resulted in the identification of similar structures in many phylogenetically 
unrelated organisms includmg gram-negative as well as gram-positive bacteria 
(tables 2-3). Typical virulence factors encoded on PAls include toxins, adhesins 
and fimbriae, factors involved in host cell entry, capsules, secretion systems 
and iron uptake systems. Based on the broad distribution of PAls, it can be con- 
cluded that they have contributed significantly to the evolution of virulent vari- 
ants. However, the still growing number of genome sequences has made it clear 
that GEIs are not restricted to pathogenic species. GEIs contributing to the 
adaptation to specific growth conditions or the interaction with a eukaryotic 
host organism have been described in environmental, commensal or symbiotic 
bacteria and have been designated 'symbiosis islands', 'ecological islands' or 
'resistance islands', according to the respective encoded functions. Relatively 
well-studied examples of GEIs include the symbiosis island of Mesorhizobium 
melioti that carries genes required for nitrogen fixation, whereas GEIs such as 
the mec region enhance survival of staphylococci in hospitals where they have 
to face antimicrobial substances. Other islands encode enzymes involved in the 
degradation of phenolic compounds or for uptake and metabolism of certain 
carbohydrates (table I ). Finally, a recently described island in Magnetospirillum 



Hochhut/Dobrindt/Hacker 236 



Table L Examples of GEIs 



Subtype 


of Designation 


Organism 


encoded 
functions 


Reference 


PAJ 


PAI [J536 


Escherichia coli 


P fimbriae, 
a-hemolysin 


26 


PA] 


HPl 


Yersinia spp. 


Iron uptake 


21 


ECl 


HPI 


Fecal Escherichia coli, 
Salmonella enierica 
subgroups 111 + IV 


Iron uptake 


23,24 


FCl 


Clwscr94 


Salmonella 
senfienberg 


Sucrose uptake anc 
metabolism 


3 


ECJ 


etc e ement 


Pseudomonas 
putida 


Degradation of 
pheiiolic compounds 


16 


ECl 


Vlagnetosome 


Magnetospir ilium 


Foimation of 


20 




is and 


gryph is waldense 


magnetosomes 




REI 


STIl 


Salmonella enierica 
DTI 04 


Antibiotic resistance 


76 


\-\ 


mec ocus 


Staphylococcus 
aureus 


Antibiotic resistance 


77 


SYI 




Mesorhizobium 
melioti 


Nitrogen fixation 


78 


SYl 




Sinorhizobiiim 

fredii 


Type 111 secretion 
system 


79 


ECJ = 


- Ego ogical is and; 


REl - resistance is and; SYI - symbiosis is and. 





gryphiswaldense is required for the formation of magnetosomes and the char- 
acteristic magnetotactic phenotype of these bacteria [20]. Interestingly, some 
GEJs have been assigned to different subtypes depending on the habitat and 
genetic background of the respective bacterium. An example is the so-called 
'high pathogenicity island' (HPI) that was originally found in derivatives of 
Yersinia spp. exhibiting increased virulence in mice [21]. As this island and the 
associated iron uptake system have been found in many pathogenic and non- 
pathogenic enterobacteria [22-24], HPI can be considered as a 'broad host 
range GEI'. Whereas it contributes to virulence in pathogenic variants and has 
therefore been defined as a PAI^ it enhances the capability of fecal E. coli, 
Klebsiella spp. and nonpathogenic Salmonella enierica spp. to grow under iron- 
limiting conditions and has therefore been defined as an 'ecological island' in 
these strains. 



Pathogenicity Islands 



237 



Table 2, PAIs of pathogenic Enterobacteriaceae 



Organism 



Designation Encoded traits 



Size, kb Junction 



Integrase Insertion site Reference 



o 

:x 
c 

O 
o 

cr 

'-I 

5" 

a: 
&> 

o 






Escherichia colt 536 
(UPEC) 

Escherichia coli 536 
(UPEC) 

Escherichia coli 536 
(UPEC) 

Escherichia coli 536 
(UPEC) 

Escherichia coli J96 
(UPEC) 

Escherichia coll J96 
(UPEC) 



Escherichia coli 
CFT073 (UPEC) 

Escherichia coli 
CFT073 (UPEC) 

Escherichia coli 
AL862 

Escherichia coli 
Ec222 (APEC) 

Escherichia coli C5 



PAII 



536 



PAIH 



536 



PAIIII 



536 



PAIV 



536 



PAII 



J96 



PAin 



J96 



PAH 



CFT073 



PA I IIcn'07; 



PAI 



ALS62 



VAT^PAl 



PAII 



C5 



a-Hemolysin, put, 
adhesions 

a-Hemolysio, put, 
P fimbriae (Prf), 
adhesion 

S fimbriae (Sfal), iro 
siderophore system, 
hemoglobin protease 

K15 capsule 

ot-Hemolysia, 
P fimbriae (Pap) 

a-HemolySLQ, 
P fimbriae (Prs), 
cytotoxic necrotizing 
factor L (CNFl) 

a-Hemolysin, 
P fimbriae (Pap) 

P fimbriae (Pap), 
iron acquisition 

afa% adhesin 



Vat autotransporter 

ct-Hemolysin, P fimbriae 
(Prs), cytotoxic 
necrotizing factor 1 
(CNFl), heat-resistant 
hemagglutinin 



75.8 



102 



76.8 



>75 



>170 



110 



58 



71 



61 



22 



100 



DR 16 bp 



DR 18 bp 



DR 46 bp 



DR 23 bp 



DR9bp 

NoDR 

DR 14 bp 
or DR 136 bp 
(imperfect) 

No 

DR 18 bp 



CP4-like selC 

(cryptic?) 

P4-liJce leiiX 



Sfx-like 



P4-Iike 



7 



DR 135 bp P4-lLke 



P4-like 



P4-like 



P4-like 
P4-like 



thrW 



pheV 



pheV 



pheU 



pheV 

pheU 

pheU 
pheV 



Sfil-like ihrW/yagV 

(truncated) 

? leuX 



26 



26 



26 



Unpublished 



32 



32 



29 



29 



80 



81 



82 





Escherichia coli 
E2348/69 (EPEC) 


FspC-PAI 


Autotransporter/ 
eaterotoxin 




Escherichia coli 


LEE 


^ ype in secretion, 




F2348/69 (EPEC) 




invasion 


o 

JO 


Escherichia coli 
EDL933 (EHEC) 


LEE 


lype III secretion, 

invasion 


3 


Escherichia coli 
RW 1374 (SI EC) 


LEE 


Type III secretion, 

invasion, parts of the 
she PAI (S. /Jexneri 2a) 


CI. 


Escherichia coli 


LEE 


Type HI secretion, 




RDEC-1 (REPEC) 




invasion, put. adhesion 




Escherichia coli 
83/39 (RFPEC) 


LEE 


Type 111 secretion, 
invasion, put. adhesin, 

enterotoxin 




Escherichia coli 


LEE 


lype 111 secretion, 




84/110-1 (REPEC) 




invasion 




Escherichia coli 


FPEC 


Diffuse adherence 




135/12 


Afa-PAI 


adhesin 




Escherichia coli 
(FHFC) 


LPA 


Serine protease (Espl), 
vitamin R|2 1'^ceptor 
(BtuB), adhesion 




Escherichia coli 


IPAl'l 


Invasion 




10407 (ETEC) 








Pathogenic 


HPI 


Yersiniabactin 




Escherichia coli, 

nonpathogenic 

Salmonella 


(PAi rv53o) 


synthesis, transport 




Yersinia enterocolytica 


HPI 


Yersiniabactin 


to 


Ye8081 




synthesis, transport 


^o 


Yersinia 


HPI 


Yersiniabactin 




pseudotuberculosis 




synthesis, transport 




Yersinia peslis 


HPI (pgm 
locus) 


Yersiniabactin synthesis, 
transport, herriin uptake 



15.2 



35 



43 



>80 



59.5 



85 



>11 



33 



46 



31^3 



45 



36 



102 



NoDR 



NoDR 



NoDR 



9 



NoDR 



No DR 



DR 23 bp 
(imperfect) 



9 



No DR 



DR 25 bp 



NoDR 



NoDR 

DR 17 bp 

ISlOO 
DR 17 bp 



No 



No 



CP4-like 



No 



P4-like 



P4-like 



P4-like 



P4-like 



CP4-l)ke 



Yes 



P4-like 



P4-)ike 



P4-like 



P4-)ike 



ssrA 



selC 



selC 



pheV 



pheU 



pheU 



pheV 



pheV 



selC 



selC 



asnT 



asnT 



asnT, U, W 



asnT 



83 



84 



27 



85 



37 



61 



61 



86 



87 



88 



23,24 



21 



12 



89 



Table 2. (continued) 



Organism 



Designation Encoded traits 



Size, kb Junction 



Integrase Insertion site Reference 





Sh i gel la jlexn eri 


SH[-1 
{she) 


Enterotoxin (Set), 
protease (Pic) 


X 

o 

o 

nr 


Shigella flexneri 


SHl-2 


Aerobactin synthesis, 
CO icin V uniiiunity 


c 
o 

a. 


Shigella flexneri 


SRL 


Ferric diciLiate 
transport, antibiotic 
resistances 


£0 


Shigella flexneri 


Shi-0 


Genes involved in 


O 






serotype conversion 


^^ 


Salmonella enterica 


SPl-1 


Type III secretion, 




sv yphimuriiim 




invasion into epithe ial 
ce s, apoptosis 




Salmonella enterica 


SPI-2 


"ype III secretion, 
invasion into 
monocytes 




Salmonella enterica 


SPl-3 


Invasion, survival 
in macrophages 




Salmonella enterica 


SPl-4 


Invasion, siu'viva 
in monocytes 




Salmonella enterica 


SPI-5 


SPI-1 effector 
protein (SopB) 




Salmonella enterica 
sv iyphi 


SPl-7 


Vi exopolysaccharide 
production 




Erwinia amylovora 
Ea321 


hrp PAl 


Type III secretion, 
effectors 



46.6 



23-30 



66 



11 



40 



40 



17 



25 



7 



134 



60 



DR 22 bp 
(imperfect) 



DR 14 bp 



NoDR 



NoDR 



NoDR 



NoDR 



NoDR 



NoDR 



DR 55 bp 



P4-like 



CP4-]ike 



Yes 



Yes 



No 



No 



No 



No 



No 



Yes 



Yes 



pheV 



selC 



serX 



ihrW 

Between 
JhlAlmuiS 

valV 



selC 

Putative 
tRNA gene 

serT 

pheU 

pheV 



68 



90,91 



92 



93 



41 



43 



44 



45 



46,94 



47 



56 






DR ^ Direct repeat; APEC ~ avian pathogenic £. coli\ REPEC = rabbit enteropathogenic E. coli\ ETEC = enterotoxigenic E. coli\ 
LPA = locus of proteolysis activity; SRL = Shigella resistance locus; put, = putative; STEC = Shiga toxin-producing E. cod. 



Table 3. Examples of PAIs of gram-positive bacteria 



o 






Organism 



Designation 



Encoded traits 



Size, kb Junction Integrase 



Insertion site Reference 



Staphylococcus 
aureus RM4282 

Staphylococcus 
aureus RN3984 

Staphylococcus 
aureus COL 

Staphylococcus 
aureus RF122 

Staphylococcus 
aureus T^ 1 1 4 



SaPU 



SaPI2 



SaPr3 



SaPIbov 



etd PAI 



Enterococcus faecaiis Enterococcus 

faecalis PAI 



Toxic shock syndrome 
toxin-] (TSST-1) 

Toxic shock syndrome 
toxin- 1 (TSST-1) 

Enterotoxin 
serotypes B, K, Q 

Toxic shock syndrome toxin- 1 
(TSST-1), enterotoxin C 

Exfoliative toxin D, 
glutamyl endopeptidase 

Cytolysin, surface proteiji 
(Esp), aggregation 
substance 





Clostridium difficile 


Pathogenicity 


Enterotoxin (TcdA), 






locus (PaLoc) 


cytotoxin (TcdB) 




Streptococcus 


PPII 


Iron uptake system 




pneumoniae 








Pathogenic Listeria 


LIPI-1 


PrfA-dependent virulence 
gene cluster (phospholipases, 
listerio ysin, ActA) 




Listeria ivanovii 


LIPI-2 


Internalins, 
sphingomye inase C 











15 



16 



16 



15 



19 



27 



22 



DR 1 7 bp Yes 



DR 1 7 bp Yes 



DR 74 bp ? 



DR 5 bp No 



150 DR 10 bp Yes 



NoDR 



NoDR 



NoDR 



No 



No 



No 



Near tyrB 

Near trp 
gene cluster 

■7 



LQtergenic 



Intergenic 



No DR Recombinase ye/A 



14 



14 



95 



96 



Intergenic 97 



60 



Intergenic 58 



18 



Lntergenic 57 



Intergenic 57 



PAIS Contribute to Virulence of Bacterial Pathogens 

PAIs of Enterobacterial Pathogens 

Most of the characterized GEIs so far have been found m members of the 
Enterobacteriaceae (table 2), which may in part be explained by the fact that 
this family has been intensively studied, but also mdicates that PAJs have played 
a pivotal role in the evolution of enterobacterial pathogens. E. coli normally 
lives as a harmless commensal in the bowels of humans or animaJs, but some 
variants have the potential to cause gastrointestinal as well as extraintestinal 
infections [25]. Pathogenic E. coli can be linked to a variety of quite diverse 
symptoms that include enteric diseases that range from cholera-like diarrhea to 
severe dysentery and hemorrhagic colitis, cystitis or pyelonephritis, septicemia 
and meningitis. Based on their mode of pathogenesis, virulent E. coli have been 
classified into different pathotypes such as uropathogenic E. coli (UPEC), 
enteropathogenic E. coli (EPEC), enterohemorrhagic E. coli (EHEC), entero- 
toxigenic E. coli or enteroinvasive E. coli and the pathogenetically related Shigella 
species. They are characterized by the expression of specific virulence factors 
that enable them to exploit new niches in their host and to disrupt the normal 
host physiology. In pathogenic E. coli and Shigella spp., many of these key 
virulence factors are encoded on PAJs, which underlines their importance in the 
formation of the various pathotypes. The diversity of diseases that are associated 
with E. coli infections is also reflected by the structural and functional varieties 
in PAIs (see table 2, fig. 1). Whereas some PAIs are widely distributed among 
different enterobacterial species [e.g. HPI and the Jocus of enterocyte efface- 
ment (LEE)], others are closely related to a specific pathotype. Furthermore, 
most strains carry multiple PAIs that can cover more than 5% of the genome. 
For example, at least five PAIs (PAI \^2t to PAI V535) have been identified in the 
chromosome of the uropathogenic isolate E. coli 536 (table 2) [26]. 

Besides PAIs that have been identified on the basis of functional studies, a 
still increasing number of putative GEIs have been detected in the completely 
sequenced genomes of pathogenic E. coli and Shigella flexneri. However, it has 
yet to be investigated whether the encoded factors contribute to virulence or fit- 
ness of the respective pathogen [27-29]. Most PAIs of pathogenic E. coli exhibit 
a mosaic-like modular structure and although some PAIs show similarities in 
respect to the presence and linkage of certain virulence determinants, there is 
also a great variability in regard to size, organization and chromosomal local- 
ization even among strains of the same patho- or serotype [26, 30]. Interestingly, 
some tRNA genes seem to represent hot spots for the integration of foreign 
DNA including PAIs. The majority of PAIs in enterobacteria is linked to either 
selC, the gene for a selenocysteine-specific tRNA, or one of two genes for a 
phenylalanine-tRNA, p/?eP^ or p/?ei7. Whereas the associated integrase genes 



Hochhut/Dobrindt/Hacker 242 



^ 



\A^ 



seIC int 

-► 
DR 



» I I 








F17-like fimbriae? 











CS12-like fimbriae? a-Hemolysin 



m 




my 



PAN 



536 



DR 



m^mm^mrm 




(\ 




:i 



sefC int Secreted Intimin Tir Type III secretion system 

Prophage 933L Proteins 



^ 




se/C int 



^ 



selC int 




sefC 



■_^»<hHOH 



Protease B12receptor 

Adhesin 







Aerobactin synthesis 






■-/ 




Survival in macrophages 



EDL933 



LPA 



SHI-2 



SPI-3 



Fig. i. Examples of jvWC-associated PAls of Enterobacteriaceae. The orgardzarion of 
^^/C-associated PAls is shown. Known or putative virulence genes are shown as gray arrows 
and their (predicted) function is given, ORFs with similarity to transposases are indicated by 
hatched arrows. Also shown are genes for a CP4-Hke integrase (int). With the exception of 
SPI-3 from S, enterica, highly similar genes are present in all islands. Finally, direct repeat 
sequences in PAI ]^26 ^nd the prophage sequence present m EPEC EDL933 are also shown. 



are well conserved and seem to be specific for the linked tRNA gene, the further 
structural organization and nucleotide sequence of islands that are integrated 
into identical sites in their respective bacterial host are not necessarily closely 
related but rather encode functions that determine the lifestyle and pathotype of 
the bacterium (fig. 1). 

The best-studied PAls of £. coli belong to the LEE island and PAls of 
uropathogenic strains. One important trait of UPEC isolates is the presence of 
adhesins that enables them to adhere to uroepithelial cells [31], Besides type I 
fimbriae, PAls often carry genes that are specific for P fimbriae that bind to the 
Gal a(l-4)Gal moieties of glycoproteins and S-fimbrial adhesions. 
Furthermore, UPEC produce the pore-forming toxin oc-hemolysin, several iron 
uptake systems, as well as capsules that function as a protection against the host 



Pathogenicity Islands 



243 



defense. All these traits are encoded by PAJs that are similar to each other, but 
not identical in the different isolates [26, 30, 32]. Interestingly, adhesin and 
toxin gene clusters are often linked with each other, suggesting a coevolution of 
these factors [33]. 

The LEE island encodes the outer membrane adhesion protein intimin, a 
type III secretion system and several secreted effector proteins. One of these 
secreted proteins is Tir (translocated intimin receptor) that is inserted into the 
eukaryotic host cell membrane where it serves as a receptor for intimin to mediate 
binding of the bacterium to the host cell [34]. Strains carrying the LEE locus 
cause characteristic attaching and effacing lesions. When LEE is transferred to 
E. coli K-12, it exhibits the same phenotype which indicates the potential of 
LEE to transform a nonpathogenic strain into a more virulent variant [35]. The 
LEE island has been identified in E. coli isolates of humans and many animals 
as well as in Cilrobacter wdenthim [36]. Sunilar to PAI Igj^ of UPEC strain 
536, the LEE locus is located next to selC in some EHEC and EPEC strains 
(fig. 1), but can also be associated w<f'\X\\ pheV or pheil in other isolates. When 
LEE sequences of EHEC, EPEC and the rabbit isolate RDEC-1 were compared, 
it became evident that the esc genes encoding the secretion apparatus were 
highly conserved, whereas the other genes were less similar than it would have 
been expected from clonal lineages. This may reflect the differences in interactions 
with the specific host but also suggests that the LEE locus has been acquired 
more than once during the evolution of £'. coli [36, 37]. 

Similar to pathogenic E. coli, PAls have played a fundamental role in the 
evolution of the genus Salmonella. Five PAJs (SPI-1 to SPL5) have been iden- 
tified in a range of serovars of 5'. enterica and were characterized in more detail. 
Furthermore, additional chromosomal regions that exhibit features of GEls 
have been found in the genomic sequences of serovars Typhimurium and Typhi 
[38, 39]. SPI-1 is regarded as a very ancient island which was already intro- 
duced into the genome of a common ancestor of S. enterica and Salmonella 
bongon [40]. Consequently, it has become a stable part of the chromosome and 
lacks most of the typical traits of PAls. Similar to the LEE locus, SPl-1 encodes 
a type III secretion system including the components of the secretion appara- 
tus, effector proteins, specific chaperones, and virulence gene regulators [41]. 
SPl-1 mediates invasion of host cells and induction of macrophage apoptosis 
[reviewed in 42]. SPI-2 (located next to valV) encodes a second type III system 
that is required for systemic infections and replication within macrophages 
[43], Similarly, SPI-3 and SPl-4 have also been shown to be involved in intra- 
macrophage survival [44, 45]. As for PAI I535 and LEE, SPI-3 is associated with 
selC, but seems to have lost the corresponding integrase gene (fig. 1). The 
mglBC operon of SPI-3 is not only required for replication m macrophages, but 
also for in vitro growth under low Mg^"^ conditions. SPI-5 encodes an effector 



Hochhut/Dobrindt/Hacker 244 



protein (SopB) that acts as a substrate for the SPI-1 -encoded secretion 
apparatus. This is an example of a tight connection between different PATs 
of one strain [46]. Finally, the so-called SPI-7 is only present in a subset of 
S. enterica isolates including S. enterica serovar Typhi CT18 that produce the 
Vi capsular polysaccharide. The corresponding genes reside on a 134-kb island 
that seems to have evolved from several independent insertion events and 
carries a region with similarity to the pilus genes of the conjugative plasmid 
R46 [47] . 

PA Is of Other Gram-Negative Pathogens 

Besides in Enterobacteriaceae, PAJs are also present in the genomes of 
several other gram-negative bacterial pathogens and can contribute significantly 
to the viiulence potential of their host bacterium. In the case of Vibrio cholerae, 
two PAIs have been described [48, 49]. The first Vibrio pathogenicity island 
VPI-1 is present in all epidemic and pandemic strains, but absent in most 
nonpathogenic strains. The 39.5-kb island encodes a type IV pilus, the toxin- 
coregulated pilus (TCP) that functions as an essential intestinal colonization 
factor in humans and animal models [50]. Besides its role as an adhesion factor, 
TCP also functions as the receptor for the cholera toxin encoding filamentous 
phage CTXcp [51]. Therefore, acquisition of VPT-1 seems to be a prerequisite 
for the emergence of highly pathogenic V. cholerae variants. Furthermore, VPI- 1 
is linked to cholera toxin production because toxT, the gene for a transcriptional 
activator of the AraC family, also resides on the island. ToxT is involved in both 
activation of the top gene cluster and the toxin genes. A second pathogenicity 
island, VPI-2, has recently been found to be present in the majority of toxigenic 
(CTXcl)-positive) strains, but absent from nontoxigenic isolates [49]. VPI-2 
encodes a neuramidase and a putative metabolic pathway for amino sugars. The 
role of these determinants for either virulence or fitness of V. cholerae has yet 
to be elucidated. Similarly, the impact of a putative pathogenicity island in the 
genome of Legionella pneumophila serogroup 1 Philadelphia- 1 (LpPI-I) that 
carries genes for a type IV secretion system is still unclear [52]. In contrast, the 
role of the cag island in the virulence of//, pylori has been intensively studied. 
This island is only present in H. pylori strains that are associated with severe 
forms of gastroduodenal disease (type 1 strains) suggesting that acquisition of 
this region has been an important event in the evolution of more virulent forms 
of//, pylori [19]. Like LpPI-1, the cag island encodes a type IV secretion sys- 
tem that resembles other toxin secretion systems as well as transport systems 
that are required for transfer of DNA. It has been shown that Cag A is delivered 
by the island-encoded secretion apparatus into host cells where it induces 
cellular growth changes that are specific for infections with type I strains of 
//. pylori [53]. 



Pathogenicity Islands 245 



Finally, PAJs have also been identified in animal and plant pathogens; 
however, they have not been as extensively studied as in human pathogens. Tn 
Dichelobacter nodosus, the causative agent of foot rot in sheep, two chromo- 
somal regions with PAI-typical features have been described [54, 55]. However, 
their role in virulence is still unclear. 

Similar to many enterobacterial pathogens of humans and animals, several 
gram-negative plant pathogens also use type IIJ secretion systems to inject 
effector proteins into plant cells that induce a plant tissue defense line includ- 
ing programmed cell death. The corresponding genes have been designated hrp 
(hypersensitivity response) or hrc (hypersensitivity response and conserved) 
and form PAJ-like regions that can be located either on the chromosome or on 
plasmids [reviewed in 56]. This reveals common themes in virulence of human, 
animal or plant pathogens. 

PA Is in Gram-Positive Pathogens 

Chromosomal regions with the typical features of PAIs in gram-negative 
bacteria are less frequently found in gram-positive pathogens. However, a few 
regions that exhibit some of the characteristics of PAIs have also been identi- 
fied in gram-positive bacteria (table 3). For example, virulence gene clusters in 
Listeria spp. or Clostridium difficile are not flanked by direct repeats or linked 
to mobility genes, but have been described as PAI-like elements [57, 58]. In 
S. pneumoniae, the characterization of an iron uptake system that is required for 
full vu-ulence revealed that the corresponding genes for an ABC transporter are 
linked to a recombinase gene in a 27-kb region designated as PPll [18]. 
Furthermore, GEls seem to be crucial elements for genetic exchange in staphylo- 
cocci. Besides the above-mentioned methicillin resistance islands, staphylo- 
coccal pathogenicity islands (SaPIs) contribute to horizontal transmission of 
resistance and toxin genes in Staphylococciis aureus [reviewed in 59]. PAIs of 
S. aureus share many of the criteria of PAIs of enterobacteria and are mobiliz- 
able by phage transducdon (see also below). SaPIs of human as well as animal 
isolates have been distinguished by the different toxin types they encode such 
as superanligen LoxJns (e.g. LoxJc shock syndrome toxin), exotoxins and enlero- 
toxins. Finally, a large chromosomal island has recently been identified in the 
genome of an Enterococcus faecalis isolate that caused an infectious outbreak 
[60]. This island encompasses more than 150kb, is flanked by direct repeat 
sequences and exhibits a lower G + C content than the rest of the genome. It 
encodes several putative virulence factors including a cytolysin and a surface 
protein that contributes to the colonization of the bladder. A closer investigation 
o^E. faecalis isolates revealed that structural variations of the island occur with 
relatively high frequencies thereby enabling strains harboring this island to 
modulate their virulence potential. 



Hochhut/Dobrindt/Hacker 246 



PAIS and Genome Plasticity 

The relatively high genetic flexibility of a bacterium is thought to facilitate 
the access to new ecological niches and may represent an advantage over organ- 
isms with less flexible genomes. As already discussed, PAIs have contributed to 
the long-term evolution of many bacterial pathogens, but beyond that, they may 
also be involved in relatively recent changes within the genetic information of 
an organism thereby modulating the virulence potential of a strain. 

Intact or rudimentary mobility genes on PAIs give evidence that they 
have been acquired by means of horizontal gene transfer such as transduction or 
conjugation; however, so far only little is known about the actual mechanisms 
that have been involved. It has been assumed that integration into the recipient's 
chromosome, at least in some cases, was mediated by site-specific recombina- 
tion similar to the integration mechanism of several bacteriophages. This is 
supported by the findings that some island-encoded integrases still have the 
potential to carry out these reactions [61, 62]. Most PAIs have undergone modi- 
fications such as deletions and mutations within the direct repeat sequences or 
mobility genes (fig, 2), Often, these processes have resulted in a relatively sta- 
ble integration of PAIs in the bacterial chromosome which has been designated 
as 'homing'. Examples of PAIs that have become locked in the chromosome are 
some of the islands specific to S. enterica (also see above). This may at least 
partly reflect the fact that the encoded traits have become indispensable for the 
host bacterium and are becoming part of the core chromosome. 

In contrast to such presumably very ancient elements, other islands are still 
mobilizable and can be transferred from one bacterium to another, at least in 
laboratory settings. VPl-l has been transmitted among V choleme strains by a 
transducing vibriophage, CP-Tl [15] and there is also one report that VPl-1 
itself may correspond to a functional prophage [63]. Furthermore, almost iden- 
tical regions to VPI have been found in the chromosome of some Vibrio mimicus 
isolates, which suggests a relatively recent gene transfer between these two 
species [64], General transduction also plays a role for transfer of SaPlI and 
related islands in S. aureus. Even though these PAIs are not self-transferable, 
they can be propagated by staphylococcal phages such as c(>80a and c|>13 [14] 
in a mechanism reminiscent of the relationship between the defective coli- 
phage P4 and its helper phage P2 [65]. Besides mobilization by transduction, 
conjugation may have played a role for the acquisition of PAIs and it is not 
unlikely that PAIs may have been derived from conjugative plasmids. In the 
last few years a number of elements termed 'integrating conjugative elements 
(ICEs)' or 'conjugative transposons' have been reported to be normally 
integrated in the chromosome, but can excise in a precise manner to be subse- 
quently transferred by close cell-to-cell contact [66, 67]. Similar to PAIs, these 



Pathogenicity Islands 247 



E 



virC 




Horizontal gene transfer 



tRNAgene 



I I ! I I rT~r 



DR ini 



virA virB virC 



e 



I I— 1 1 I 



IS 




Acquisition of new PAIs 



S E 



PAH 



PAI2 



Increased virulence potential 



Chronnosome 



Recombination, integration 



Chromosome H- pre-PAl 



mob DR 



Deletions, point mutations, 
DNA rearrangements 



Chromosome + PAI 



Amob 




Deletion of PAIs/ 
parts of PAIs 



E 



Decreased virulence potential 



Fig. 2. PAJs and genome plasticity. Evolution of PAIs is based on acquisition of novel 
DNA by horizontal gene transfer followed by point mutations, recombination and deletion 
events that can render the PAl inunobile. The viruJence potential of a bacterium can subse- 
quently be either increased by introduction of new PAIs or decreased by partly or complete 
deletions of PAIs. int ^ Integrase gene; wr ^ virulence-associated gene; mob ^ mobility 
genes; Amo6 = truncated mobility genes; DR = direct repeat sequences. 



elements encode site-specific recombinases and lack the ability to replicate 
autonomously. 

Finally, several islands seem no longer mobilizable, but have a tendency to 
delete from the chromosome either as a complete unit or in parts [12, 19, 61, 
68, 69]. Precise deletion from the chromosome has been observed for PAIs of 
the UPEC isolate 536 and requires functional integrase genes [70] (our unpubl. 
results). Furthermore^ an increase of deletion incidences was observed under 
certain environmental conditions [71]. It has been speculated that loss of virulence 
determinants may play a crucial role during the transition fi^om an acute state of 
disease to chronic infections [8, 72]; therefore it v^ill be interesting to further 



Hochhut/Dobrindt/Hacker 



248 



investigate how the content of virulence genes can be modulated by environ- 
mental, bacterial or eukaryotic host factors. 

Similarly, rearrangements or deletions within islands are often mediated 
by coresiding transposons or IS elements (fig. 2). This is especially true for 
H. pylori where an ongoing adaptation between bacterium and host based on 
1^605 mediated DNA rearrangements within the cag island (see also PAIs of 
Other Gram-Negative Pathogens) has been described [reviewed in 73]. Full 
virulence of//, pylori depends on an intact cag island, whereas deletions within 
the island render the bacterium less pathogenic. The interaction of//, pylori 
with epithelial cells results in an elevated production of cytokines such as 
interleukin-8 (IL-8). This induction of IL-8 production correlates with the 
presence of a complete cag island, whereas //. pylori strains carrying only parts 
of the island induce lL-8 at significantly lower levels [74]. 



Conclusions 

GEIs contribute to virulence and survival of pathogens in several ways. 
First, the acquisition of GEIs has been described as 'evolution in quantum 
leaps', because they often carry more than one virulence or fitness determinant 
[75]. These GEI-encoded factors enable the bacterium to colonize novel niches 
in the eukaryotic host and facilitate the adaptation to the respective environ- 
mental conditions. This increase of fitness gives an advantage over coresiding 
bacteria. Furthermore, the genome of many bacterial pathogens contains more 
than one GEI that encode important virulence factors, thereby determining the 
capability to cause disease. In addition, to ensure coordinated expression of 
virulence or virulence-related genes that are located on GEIs, a tight connection 
to regulatory networks of the bacterium has evolved, as well as a link of island- 
encoded regulators to genes encoded elsewhere in the genome. Finally, an 
ongoing mobilization and transfer of GEIs as well as reorganization, partial or 
complete deletion of existing GEIs affect long-term (macro-) as well as short-term 
(micro-) evolution of pathogenic bacteria. 



References 

Dobrindt U, ReidI J: Pathogenicity islands and phage conversion: Evolutionary aspects of bacterial 
pathogenesis, liu J Med Microbiol 2000;290:519-527. 

Davis BM, Waldor MK: Mobile genetic elements and bacterial pathogenesis; in Craig NL, 
Craigie R, Gellert M, Lambowitz AM (eds): Mobile DNA W, Washington, ASM Press, 2002, 
pp 1040-1059. 

Ochman H, Lawrence JG, Groisman EA: Lateral gene transfer and the nature of bacterial innova- 
tion. Nature 2000;405:299-304. 



Pathogenicity Islands 249 



4 Hacker J, Hentschel U Dobrindt U: Prokaryotic chromosomes and disease. Science 2003;301: 
790-793, 

5 Cheetham BF, Katz ME: A role for bacteriophages in the evolution and transfer of bacterial virulence 
determinants. Mol Microbiol 1995;18:201-208. 

6 Boyd EF, Brussow H: Common themes among bacteriophage-encoded virulence factors and 
diversity among the bacteriophages involved. Trends Microbiol 2002;10:521-529. 

7 Hacker J, Carniel E: Ecological fitness, genomic islands and bacterial pathogenicity. EMBO Rep 
200I;2:376-38L 

8 Hacker J, Kaper JB: Pathogenicity islands and the evolution of microbes. Annu Rev Microbiol 
2000;54:641-679. 

9 Hacker J, Bender L, OttM, Wingender J, Lund B, Marre R, Goebel W: Deletions of chromosomal 
regions coding for fimbriae and hemolysins occur in vitro and in vivo in various extraintestinal 
Escherichia coli isolates, Microb Pathog 1990;8:213-225, 

10 Hentschel U, Hacker J: Pathogenicity islands: The tip of the iceberg. Microbes Infect 2001;3: 

545-548, 
I I Williams KP: integration sites for genetic elements in prokaryotic tRNA and tmRNA genes: 

Sublocation preference of integrase subfamilies. Nucleic Acids Res 2002;30:866-875. 

12 Buchrieser C, Brosch R, Bach S, Guiyoule A, Carniel E:The high-pathogenicity \s\and of Yersinia 
pseudotuberculosis can be inserted into any of the three chromosomal asn tRNA genes. Mol 
Microbiol 1998;30:965-978. 

13 Hochhut B, Jalireis K, Lengeler JW, Schmid K: CTY\scr94, a conjugalive transposon found in 
enterobacteria, J Bacteriol 1997;179:2097-2102. 

14 Lindsay JA, Ruzin A, Ross HF, Kurepina N, Novick RP: The gene for toxic shock toxin is carried 
by a family of mobile pathogenicity islands in Staphylococcus aureus, Mol Microbiol 1998;29: 
527-543. 

15 O'Shea YA, Boyd EF: Mobilization of the Vibrio pathogenicity island between Vibrio 
cholerae isolates mediated by CP-Tl generalized transduction, FEMS Microbiol Lett 2002;214: 
153-157. 

16 van der Meer JR, Ravatn R, Sentchilo V: The clc element of Pseudomonas sp. strain B13 and 
other mobile degradative elements employing phage-like integrases. Arch Microbiol 2001 ;1 75: 
79-85. 

17 Dillard JP, Seifert HS: A variable genetic island specific for Neisseria gonorrhoeae is involved in 
providing DNA for natural transformation and is found more often in disseminated infection 
isolates. Mol Microbiol 2001;41:263-277. 

18 Brown JS, Gilliland SM, Holden DW; A Streptococcus pneumoniae pathogenicity island encoding 
an ABC transporter involved in iron uptake and virulence, Mol Microbiol 2001;40:572-585. 

19 Censini S, Lange C, Xiang Z, Crabtree IE, Ghiara P, Borodovsky M, Rappuoli R, Covacci A: cag, 
a pathogenicity island of Helicobacter pylori, encodes type-1 specific and disease-associated 
virulence factors. Proc Natl Acad Sci USA 1996;93:14648-14653. 

20 Schubbe S, Kube M, Scheffel A, Wawer C, Heyen U, Meyerdierks A, MadJtour MH, Mayer F, 
Reinhardt R, Schuler D: Characterization of a spontaneous nonmagnetic mutant of Magneiospirillum 
gryphiswaldense reveals a large deletion comprising a putative magnetosome island. J Bacteriol 
2003;185:5779-5790. 

21 Carniel E, Guilvout 1, Prentice M: Characterization of a large chromosomal 'high-pathogenicity 
island' in biolype IB Yersinia enferocolitica. J Bacteriol 1996;178:6743-6751. 

22 Bach S, de AJmeida A, Carniel E: The Yersinia high-pathogenicity island is present in different 
members of the family Enterobacteriaceae. FEMS Microbiol Lett 2000;183:289-294. 

23 Olschlager TA, Zhang D, Schubert S, Carniel E, Rabsch W, Karch H, Hacker J: The high patho- 
genicity island is absent in human pathogens of Salmonella enterica subspecies 1 but present in 
isolates of subspecies HI and VK J Bacteriol 2003; 185:1 107-1 1 1 h 

24 Schubert S, Rakin A, Karch H, Carniel E, Heesemarm J: Prevalence of the 'high-pathogenicity 
island' of Yersinia species among Escherichia coli strains that are pathogenic to humans. Infect 
Immun 1998;66:480-485. 

25 Sussman M: Escherichia coli and human disease; in Sussman M (ed); Escherichia coli: Mechanisms 
of Virulence. Cambridge, Cambridge University Press, 1997, pp 3^8. 



Hochhut/Dobrindt/Hacker 250 



26 Dobrindt U, Blum-Oehler G, Nagy G, Schneider G, Johann A, Gottschalk G, Hacker J: Genetic 
structure and distribution of four pathogenicity islands (PAJ Igj^^-PAJ IV^j^) of uropathogenic 
Escherichia coli strain 536. Infect Immun 2002;70:6365-6372. 

27 Pema NT, Plunkett G 3rd, Burland V, Mau B, Glasner JD, Rose DJ, Mayhew GF, Evans PS, Gregor J, 
Kirkpatrick HA, Posfai G, Hackert J, Klink S, Boutin A, Shao Y, Miller L, Grotbeck EJ, Davis >AV, 
Lim A, Dimalania ET, Potamousis KD, Apodaca J, Anantharaman TS, Lin J, Yen G, Schwartz DC, 
Welch RA, Blattner FR: Genome sequence of enterohaemorrhagic Escherichia coli OI57:H7. 
Nature 2001;409:529-533. 

28 Jin Q, Yuan Z, Xu J, Wang Y, Shen Y, Lu W, Wang J, Liu H, Yang J, Yang F, Zhang X, Zhang J, 

Yang G, Wu H, Qu D, Dong J, Sun L, Xue Y, Zhao A, Gao Y, Zhu J, Kan B, Ding K, Chen S, 

Cheng H, Yao Z, He B, Chen R, Ma D, Qiang B, Wen Y, HouY,Yu J: Genome sequence of .%/g^//a 

Jlexneri 2a: Insights into pathogenicity through comparison with genomes oi Escherichia coli K\2 

and 0157, Nucleic Acids Res 2002;30:4432^44l. 

29 Welch RA, Burland V, Plunkett G 3rd, Redford P, Roesch P, Rasko D, Buckles EL, Liou SR, Boutin A, 
Hackett J, Stroud D, Mayhew GF, Rose DJ, Zhou S, Schwartz DC, Pema NT, Mobley HL, 
Donnenberg MS, Blattner FR: Extensive mosaic structure revealed by the complete genome 
sequence of uropathogenic Escherichia coli. Proc Nail Acad Sci USA 2002;99:17020-17024. 

30 Guyer DM, Kao JS, Mobley HL: Genomic analysis of a pathogenicity island in uropathogenic 
Escherichia coli CFT073: Distribution of homologous sequences among isolates from patients 
with pyelonephritis, cystitis, and catheter-associated bacteriuria and fi*om fecal samples. Infect 
Immun I998;66:44I1^MI7. 

31 OlschlagerTA, Dobrindt U, Hacker J: Virulence factors of uropathogensXurrOpinLJrol 2002; 12: 
33-38. 

32 Swenson DL, Bukanov NO, Berg DE, Welch RA: Two pathogenicity islands in uropathogenic 
Escherichia coli J96: Cosmid cloning and sample sequencing. Infect Immun 1996;64: 
3736-3743. 

33 Blum G, Otl M, Lischewski A, Ritter A, Imrich H, Tschape H, Hacker J: Excision of large DNA 
regions termed pathogenicity islands from tRNA-specific loci in the chromosome of an 
Eschen'chia coli y^Wd-typt p2i\hog'&n. Infect Immun 1994;62:606-614. 

34 Kenny B, E>eVinney R, Stein M, Reinscheid DJ, Frey EA, Finlay BB: Enteropathogenic E. coli 
(EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell 1997;91:51 1-520, 

35 McDaniel TK, Kaper JB: A cloned pathogenicity island from enteropathogenic Escherichia 
coli confers the attaching and effacing phenotype on E. coli K-12. Mol Microbiol I997;23: 
399^07, 

36 Deng W, Li Y, Vallance BA, Finlay BB: Locus of enterocyte eflfacement from Citrobacler rodeniium: 
Sequence analysis and evidence for horizontal transfer among attaching and effacing pathogens. 
Infect Immun 2001;69:6323-6335. 

37 Zhu C, Agin TS, Elliott SJ, Johnson LA, Thate TE, Kaper JB, Boedeker EC: Complete nucleotide 
sequence and analysis of the locus of enterocyte effacement from rabbit diarrhea^enic Escherichia 
coli RDEC-1. Infect Immun 2001;69:2107-21 15. 

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

39 Parkhill J, Dougan G, James KD, Thomson NR, Pickard D, Wain J, Churcher C, Mungall KL, 
Bentley SD, Holden MT, Sebaihia M, Baker S, Basham D, Brooks K, Chillingworth T, Connerton P, 
Cronin A, Davis P, Davies RM, Dowd L, White N, Farrar J, Feltwell T, Hamlin N, Haque A, Hien TT, 
Holroyd S, Jagels K, Kj'ogh A, Larsen TS, Leather S, Moule S, O'Gaora P, Pan7 C, Quail M, 
Rutherford K, Simmonds M, Skelton J, Stevens IC, Whitehead S, Barrel! BG: Complete genome 
sequence of a multiple drug rtsisiant Salmonella enferica strov^r Typhi CTI8. Nature 200 1 ;4 13: 
848-852. 

40 Groisman EA, Blanc-Potard A-B, Uchiya K: Pathogenicity islands and the evolution of 
Salmonella virulence; in Kaper JB, Hacker J (eds): Pathogenicity Islands and Other Mobile 
Virulence Elements. Washington, ASM Press, 1999, pp 127-150. 



Pathogenicity Islands 251 



41 Mills DM, Bajaj Y Lee CA: A 40 kb chromosomal fragment encoding Salmonella typhimurium 
invasion genes is absent from the corresponding region of the Escherichia coli K-J 2 chromosome. 
Mol Microbiol 1995;15:749-759. 

42 Zhou D, Galan J: Salmonella entry into host cells: The work in concert of type III secreted effector 
proteins. Microbes Infect 2001 ;3; 1293-1 298, 

43 Hensel M, Nikolaus T, Egelseer C: Molecular and ftinctional analysis indicates a mosaic structure 
oi Salmonella pathogenicity island 2. Mol Microbiol 1 999;3 1:489^98. 

44 Blanc-Potard AB, Groisman EA: The Salmonella selC locus contains a pathogenicity island medi- 
ating intramacrophage survival. EMBO J 1997;16:5376-5385. 

45 Wong KK, McClelland M, Stillwell LC, Sisk EC, Thurston SJ, Saffer JD: Identification and 
sequence analysis of a 27-kilobase chromosomal fragment containing a Salmonella pathogenicity 
island located at 92 minutes on the chromosome map of Salmonella enterlca serovar typhimurium 
LT2. Infect Immun 1998;66:3365-3371, 

46 Wood MW, Jones MA, Watson PR, Hedges S, Wallis TS, Galyov EE: Identification of a 
pathogenicity island required for Salmonella enteropathogenicity, Mol Microbiol I998;29: 
883-891. 

47 Pickard D, Wain J, Baker S, Line A, Chohan S, Fookes M, Barron A, Gaora PO, Chabalgoity JA, 
Thanky N, Scholes C, Thomson N, Quail M, Parkhill J, Dougan G: Composition, acquisition, 
and distribution of the Vi exopolysaccharide-encoding Salmonella enlerica pathogenicity island 
SPT-7. J Bacteriol 2003;185:5055-5065. 

48 Karaolis DKR, Johnson JA, Bailey CC, Boedeker EC, Kaper JB, Reeves PR: A Vibrio cholerae 
pathogenicity island associated with epidemic and pandemic strains, Proc Natl Acad Sci USA 
1998;95:3134-3139. 

49 Jermyn WS, Boyd EF: Characterization of a novel Vibrio cholerae pathogenicity island (VPI-2) 
encoding neuraminidase (nanH) among toxigenic Vibrio cholerae isolates. Microbiology 2002; 
148:3681-3693. 

50 Thelin KH, Taylor RK: Toxin-coregulated pilus, but not mannose-sensitive hemagglutinin, is 
required for colonLzation by Vibrio cholerae 01 El Tor biotype ajid OJ39 strams. Jnfect Jnunun 
1996;64:2853-2856. 

5 1 Waldor MK, Mekalanos JJ: Lysogenic conversion by a filamentous phage encoding cholera toxin. 
Science 1996;272:1910-1914. 

52 Brassinga AK, Hiltz MP, Sisson GR, Morash MG, Hill N, Garduno E, Edelstein PH, Garduno RA, 
Hoffman PS: A 65-kilobase pathogenicity island is unique to Philadelphia-1 %\.v^\r\s of Legionella 
pneumophila. J Bacteriol 2003; 185:4630^-4637, 

53 Selbach M^ Moese S, Hunvitz R, Hauck CR, Meyer TF, Backert S: The Helicobacter pylori CagA 
protein induces cortactin dephosphorylation and actin rearrangement by c-Src inactivation. 
EMBO J 2003;22:515-528. 

54 Billington SJ, Muggins AS, Johanesen PA, Crellin PK, Cheung JK, Katz ME, Wright CL, Haring V, 
Rood Jl: Complete nucleotide sequence of the 27-kilobase virulence related locus {vrl) 
of Dichelobacter nodosus'. Evidence for exlrachromosomal origin. Infect Immun 1999;67: 
1277-1286. 

55 Bloomfield GA, Whittle G, McDonagh MB, Katz ME, Cheetham BF: Analysis of sequences 
flanking the vap region of Dichelobacter nodosus: Evidence for multiple integration events, a 
killer system, and a new genetic element. Microbiology 1997;143:553-562. 

56 Kim JF, Alfano JR: Pathogenicity islands and virulence plasmids of bacterial plant pathogens. 
CurrTop Microbiol Immunol 2002;264/ll:l27-l47. 

57 Kreft J, Vazquez-Boland JA, Altrock S, Dominguez-Bernal G, Goebel W: Pathogenicity 
islands and other virulence elements in Listeria. Curr Top Microbiol Immunol 2002;264/II: 
109-125. 

58 Braun V, Hundsberger T, Leukel R Sauerborn M, von Eichel-Streiber C: Definition of the single 
integration site of the pathogenicity locus in Clostridium difficile. Gene 1996;181:29-38. 

59 Novick RP, Schlievert P, Ruzin A: Pathogenicity and resistance islands of staphylococci. Microbes 
Infect 2001;3:585-594. 

60 ShankaJ" N, Baghdayan AS, Gilmore MS: Modulation of virulence within a pathogenicity island 
in vancomycin-resistant Enterococcus faecalis. Nature 2002;417:746-750. 



Hochhut/Dobrindt/Hacker 252 



61 Tauschek M, Strugnell RA, Robins-Browne RM: Characterization and evidence of mobilization 
of the LEE pathogenicity island of rabbit-specific strams of enteropathogenic Escherichia coli. 
Mol Microbiol 2002;44:1533-1550. 

62 Rakin A, Noelting C, Schropp P, Heesemann J: Integrative module of the high-pathogenicity 
island of Yersinia. Mol Microbiol 200l;39:407^15. 

63 JCaraolis DKR,SomaraS, Maneval DRJr, Johnson JA,KaperJB: A bacteriophage encoding a patho- 
genicity island, a type IV pilus and a phage receptor in cholera bacteria. Nature 1999;399:375-379. 

64 Boyd EF, Moyer KLE, Shi L, Waldor MK: Infectious CTXtp and the vibrio pathogenicity island 
prophage in Vibrio mimicus: Evidence for recent horizontal transfer between V mimicus and 
Vcholerae. Infect Immun 2000;68:1507-1513, 

65 Lindqvist BH, Deho G, Calendar R: Mechanisms of genome propagation and helper exploitation 
by satellite phage P4. Microbiol Rev 1993;57:683-702. 

66 Hochhut B, Waldor MK: Site-specific integration of the conjugal Vibrio cholerae SXT element 
into prfC. Mol Microbiol 1999;32:99-1 10. 

67 Burrus V, Pavlovic G, Decaris B, Gu6don G: Conjugative transposons:The tip of the iceberg. Mol 
Microbiol 2002;46:601-610. 

68 Rajakumar K, Sasakawa C, Adier B: Use of a novel approach, termed island probing, identifies 
ihQ Shigella flexneri she pathogenicity island which encodes a homolog of the immunoglobulin A 
protease-like family of proteins. Infect Immun 1997;65:4606-4614. 

69 Turner SA, Luck SN, Sakellaris H, Rajakumar K, AdIer B: Nested deletions of the SRL patho- 
genicity island of Shigella flexnen 2a. J Bacteriol 2001;183:5535-5543. 

70 Middendorf B, Hochhut B, Leipold K, Dobrindt U, Blum-Oehler G, Hacker J; Instability of 
pathogenicity islands in uropaihogenic Escherichia coli 536. J Bacteriol 2004;186:3086-3096. 

71 Middendorf B, Blum-Oehler G, Dobrindt U, Miihldot^er 1, Sage S, Hacker J: The pathogenicity 
island (PAJs) of the uropathogenic Escherichia coli strain 536:island probing of PAl 11535. J Infect 
Dis2001;183:S17-S20. 

72 Blum G, Ort M, Lischewski A, Ritter A, Imrich H, Tschape H, Hacker J: Excision of large DNA 
regions termed pathogenicity islands fi'om tRJMA-specific loci in the chromosome of an 
foc/renc/;/^ co// wild-type pathogen. Infect Immun 1994;62:606-614. 

73 Odenbreit S, Haas R: Helicobacter pylori: Impact of gene transfer and the role of the cag patho- 
genicity island for host adaptation and virulence, CurrTop Microbiol Immunol 2002;264/II:l-22, 

74 Nilsson C, Sillen A, Eriksson L, Strand ML, Enroth H, Normark S, Falk P, Engstrand L: Correlation 
between cag pathogenicity island composition and Helicobacter pylori -?iSS0c\?i.\t6 gastroduodenal 
disease. Infect Immun 2003;71:6573-6581. 

75 Groisman EA, Ochman H: Pathogenicity islands: Bacterial evolution in quantum leaps. Cell 
1996;87:791-794. 

76 Boyd D, Peters GA, Cloeckaert A, Boumedine KS, Chaslus-Dancla E, Imberechts H, Mulvey MR: 
Complete nucleotide sequence of a 43-kilobase genomic island associated wMth the multidrug 
resistance region of Salmonella enterica serovar Typhimurium DTI04 and its identification in 
phage type DTI 20 and serovar Agona. J Bacteriol 2001;183:5725-5732. 

77 Hiramatsu K, Katayama Y, Yuzawa H, Ito T: Molecular genetics of methicillin-resistant 
Staphylococcus aureus. Int J Med Microbiol 2002;292:67-74, 

78 Sullivan JT, Ronson CW: Evolution of rhizobia by acquisition of a 500 kb symbiosis island that 
integrates into a phe-tRNA gene. Proc Natl Acad Sci USA 1998;95:5145-5149, 

79 Marie C, Broughion WJ, Deakin WJ: Rhizobium type HI secretion systems: Legume charmers or 
alarmers? Curr Opin Plant Biol 2001;4:336-342. 

80 Lalioui L, Le Bouguenec C: afa-8 gene cluster is carried by a pathogenicity island inserted into 
the iRNA(Phe) of human and bovine pathogenic Escherichia coli isolates. Infect Immun 
200l;69;937-948- 

81 Parreira VR, Gyles CL: A novel pathogenicity island integrated adjacent to the thrW tRNA gene 
of avian pathogenic Escherichia coli encodes a vacuolating autotransporter toxin. Infect Immun 
2003;71:5087-5096. 

82 HoudouinV, Bonacorsi S, Brahimi N, Clermont 0, Nassif X, Bingen E: A uropathogenicity island 
contributes to the pathogenicity of Escherichia coli strains that cause neonatal meningitis. Infect 
Immun 2002;70:5865-5869. 



Pathogenicity Islands 253 



83 Mellies JL, Navarro-Garcia F, Okeke I, Frederickson J, Nataro JP, Kaper JB: espC pathogenicity island 
of enteropathogenic Escherichia coli encodes an enterotoxin. Infect Immun 200 1 ;69:3 1 5-324. 

84 Elliott SJ, Wainwright LA, McDaniel TK, Jarvis KG, Deng YK, Lai LC, McNamara BP, 
Donnenberg MS, tCaper JB: The complete sequence of the locus of enterocyte effacement (LEE) 
from enteropathogenic Escherichia coli E2348/69. Mol Microbiol 1998;28;1^, 

85 Jores J, Rumer L, Kiessling S, Kaper JB, Wieler LH: A novel locus of enterocyte effacement 
(LEE) pathogenicity island inserted at pheV in bovine shiga toxin-producing Escherichia coli 
strain 01 03:H2. FEMS Microbiol Lett 200l;20'^:75-79. 

86 Keller R, Ordonez JG, de Oliveira RR, Trabuisi LR, Baldwin TJ, Knutton S: Afa, a diffuse adher- 
ence fibrillar adhesin associated with enteropathogenic Eschenchia coli. Infect Immun 2002;70: 
2681-2689. 

87 Schmidt H, Zhang WL, Hemmrich U, Jelacic S, Brunder W, Tarr PI, Dobrindt U, Hacker J, Karch H: 
Identification and characterization of a novel genomic island integrated at selC in locus of entero- 
cyte efTacement-negative, Shiga toxin-producing Escherichia coli, bifect Immun 2001 ;69: 
6863-6873. 

88 FJeckenstein JM, Kopecko DJ, Warren RJL, Elsinghorst EA: Molecular characterization of the tia 
invasion locus from enterotoxigenic Escherichia coli. Infect Immun 1996;64:2256-2265. 

89 Buchrieser C, Prentice M, Carniel E: The 102-kilobase unstable region of Yersinia pesfis com- 
prises a high-pathogenicity island linked to a pigmentation segment which undergoes mternal 
rearrangement. J Bacteriol 1998;180:2321-2329. 

90 Yokes SA, Reeves SA, Torres AG, Payne SM: The aerobactin iron transport system genes in 
Shigella flexneri are present within a pathogenicity island, Mol Microbiol 1999;33;63-73. 

91 Moss JE, Cardozo TJ, Zychlinsky A, Groisman EA: The 5e/C-associated SHl-2 patiiogenicity 
[shnAoi Shigella Jlexneri. Mol Microbiol 1999;33:74-83, 

92 Luck SM, Turner SA, Rajakumar K, Sakellaris H, Adler B: Ferric dicitrate transport system (Fee) 
oi Shigella flexneri In YSH6000 is encoded on a novel pathogenicity island carrying multiple 
antibiotic resistance genes. Infect Immun 2001;69:6012-6021. 

93 Adhikari P, AJIison G, Whittle B, Verma MK: Serotype la 0-antigen modification: Molecular 
characterization of the genes involved and their novel organization in the Shigella flexneri chromo- 
some. J Bacteriol 1999;I8I:471 1^718. 

94 Hong KH, Miller VL; Identification of a novel Salmonella invasion locus homologous to Shigella 
ipgDE. ]B^atno\ 1998;180:1793-1802. 

95 Yarwood JM, McCormick JK, Paustian ML, Orwin PM, Kapur V, Schlievert PM: Characterization 
and expression analysis of Staphylococcus aureus pathogenicity island 3. Implications for the evo- 
lution of staphylococcal pathogenicity islands. J Biol Chem 2002;277:13138-13147. 

96 Fitzgerald JR, Monday SR, Foster TJ, Bohach GA, Hartigan PJ, Meaney WJ, Smyth CJ: 
Characterization of a putative pathogenicity island from bovine Staphylococcus aureus encoding 
multiple superantigens. J Bacteriol 2001;183:63-70. 

97 Yajnaguchi T, Nishifliji K, Sasaki M, Fudaba Y, Aepfelbacher M, Takata T, Ohara M, 
Komatsuzawa H, Amagai M, Sugai M: Identification of tiie Staphylococcus aureus eld patho- 
genicity island which encodes a novel exfoliative toxin, ETD, and EDJN-B. Infect Immun 
2002;70:5835-5845. 



Jorg Hacker 

Institut fijr molekulare Infektionsbiologie 

Rontgenring 1 1, DE-97070 Wiirzburg (Germany) 

Tel. +49 931 312575, Fax +49 931 312578, E-Mail j\hacker@maiLuni-wuerzburg.de 



Hochhut/Dobrindt/Hacker 254 



Signaling and Gene Regulation 

Russell W, Herwald H (eds): Concepts in Bacterial Virulence.! 
Contrib Microbiol. Basel, Karger, 2005, vol 12, pp 255-271 



Horizontal and Vertical Gene Transfer 
The Life History of Pathogens 



Jejfrey G. Lawrence 

Pittsburgh Bacteriophage Institute and Department of Biological Sciences, 
University of Pittsburgh, Pittsburgh, Pa., USA 



Viewpoints regarding the evolution of pathogenic bacteria have themselves 
evolved over the past two decades. Although it is perhaps extreme to suggest 
different teleological camps have been established, it is fair to say that opinions 
regarding the evolution of pathogens are varied, and the strength of different 
points of view have waxed and waned. Initially, many viewed pathogenic bac- 
teria as being specialized, highly derived bacteria, which evolved complex and 
intimate associations with their hosts. In this way, special evolutionary mecha- 
nisms were perhaps responsible for the origin or persistence of pathogens. 
Gradually, a viewpoint that every microorganism was adapted to a particular 
niche was widely accepted, and pathogenicity represented just another bacterial 
lifestyle; therefore, no special evolutionary forces were at play. The evolution 
of well-studied pathogens could even be used as models for how other bacteria 
adapted to their environment. 

Somewhat surprisingly, perhaps, data collected in the 'genomic era' have 
brought opinion back to the view that the evolution of pathogens indeed may 
encompass evolutionary paths typically not experienced by nonpathogenic bac- 
teria. That is, the association of pathogens with particular hosts results in 
smaller effective population sizes, low genetic diversity, infrequent recombuia- 
tion and other factors influencing their evolution as dictated by their population 
genetics. As a result, pathogens would not serve as good models for the evolution 
of nonpathogenic bacteria that do not share these population genetic constramts. 
As discussed below, both viewpoints are perhaps true, when applied to the differ- 
ent stages of pathogen evolution. At the heart of the difference between the stages 
of pathogen evolution are the relative roles of gene acquisition via horizontal 
gene exchange versus gene loss (genome degradation). Rather than representing 



different paths of pathogen creation or modification, these modes of genomic 
evolution likely represent a continuum or pathway along which a single lineage 
may travel. 



Early Examples of Horizontal Gene Transfer 

Horizontal gene transfer (HGT) is defined as the transfer of genetic material 
between bacterial cells uncoupled with cell division [1-3]. In contrast, vertical 
inheritance is the transmission of genetic material from mother cell to daughter 
cell during cell division. Most often, HGT refers to gene transfer across large 
phylogenetic distances (that is, between otherwise unrelated organisms), 
whereby genes are integrated into a replicon by illegitimate means. On occasion, 
HGT is used to denote allelic exchange among closely related bacterial strains 
where integration occurs via homologous recombination; herein that process 
will be referred to as ' recombination \ 

Some of the eariiest examples of HGT involved the transfer of antibiotic 
resistance genes [4], many times among pathogens, which were often facilitated 
by the localization of these genes on plasmids. Here, virulent strains of bacteria 
could acquire resistance to antibiotics at alarmmgly high rates, ones inconsistent 
with the evolution of such a complex trait from preexisting genetic material via 
random point mutation. Further investigation revealed that the bacteria had 
obtained a gene conferring antibiotic resistance from another bacterium. This 
observation reinforced the idea that the strong selection imposed by the adoption 
of a pathogenic lifestyle allowed investigators to see otherwise rare evolutionary 
events, like horizontal gene exchange. As a result^ this process of gene exchange 
was not considered to be a potent evolutionary force. A cogent model of bacterial 
evolution relied on the systematic periodic selection of random mutations arising 
in the population [5], The exchange of genes among bacterial strains was not 
considered to be important until many years later [6-8], and the potential high 
rates of occurrence were not appreciated until rather recently [9, 10]. 



Pathogenicity Islands 

The special role of HGT in pathogen evolution was reinforced as the| 
sequences of bacterial genes becanue abundant [11], Early analyses suggested 
that the genes encoding virulence fionctions in many pathogens were somehow 
different from other genes in the chromosome; differences often included changes 
in overall nucleotide composition (%GC), codon usage bias, association with 
mobile genetic elements, and association with tRNA genes (frequent sites of 



Lawrence 



256 



bacteriophage integration). The term 'pathogenicity island' was coined to denote 
the distinct evolutionary histories reflected by these bits of genetic material, histo- 
ries that were not shared with the remaining genes in the chromosomes [12-17]. 
Almost uniformly, genes encoding virulence factors mapped to pathogenicity 
islands, thus unplicating HGT in virtually every step of pathogen evolution. 
Functions encoded by pathogenicity islands included those required for adhesion 
and invasion [12, 13], type III secretion systems for altering host cell metabolism 
[18-20], toxin production [21-23] and a host of metabolic capabilities including 
the acquisition of phosphate and iron at low concentrations [24]. 



ing Pathogens by Gene Transfer 

While the role of HGT in pathogen evolution was compelling, it is not the 
only route to pathogen creation. In some cases, pathogens are merely bacteria 
found in the wrong place at the wrong time. For example, Legionella persists in 
macrophages using mechanisms that evolved to allow it to passage through its 
more common Entamoeba host [25, 26]; this strategy may be common among 
pathogens [27, 28]. Clostridium tetanus is just a soil anaerobe delivered unexpect- 
edly hito the human body via a puncture wound [29]; certainly some of the more 
unexpected results of tetanus infection [e.g. autism-like symptoms, 30] are not 
considered traits resulting from strong selection for particular vuTjIence functions. 
Even the well -characterized pathogen Salmonella enterica has been implicated in 
causing disease in nematodes, which may represent their primary host [3 I]. 

Yet other times, pathogens inhabit a somewhat different ecological niche 
than do their nonpathogenic ancestors. For example, the ancestor of 5. enterica 
was likely an intestinal-dwelling bacterium which never invaded epithelial cells. 
Here, new physiological capabilities are required for the pathogen to succeed in 
its new environment, and acquisition of fully functional genes from other 
pathogens is an effective strategy in making this transition [32-35]. Analyses of 
many genomes have demonstrated that similar genes are found in diverse 
organisms, and that their evolutionary histories reflect frequent travel among 
genomes [1, 36-38]. As a whole, one may view pathogen evolution as the gain 
of genes via HGT coupled with the loss of genes (necessary from a population 
genetic point of view, as discussed below), which changes the ecological capa- 
bilities of a bacterial ta^on (fig. 1). 

The power of HGT ui creating pathogens from nonpathogens is strikingly 
demonstrated in the examination of the complete genome sequences of four 
strains of Escherichia coli, including one benign laboratory straui [39], two 
pathogenic strains of £". coli [23, 40] and the phylogenetically very closely related 
strain Shigella flexneri [41] (despite being placed in a different genus, gene 



Horizontal and Vertical Gene Transfer 257 



oidEFG 



oi<i)(m 



o 




y 



2 



Gene loss 



newKLM 



Ancestral DNA retained in 
derived lineage 



QldRST 



oldABC 




newNOP 



newHJK 



Chromosome of 
ancestral lineage 



Introduced 
DNA 



DNA not 
maintained 




newUVW 



Chromosome of 
derived lineage 



Fig. L The role of HGT in changing a bacterial species. Here, an ancestral taxon gains 
(black genes and arrows) and loses (gray genes and arrows) both chromosomal and episomal 
genes. Both classes of events alter the phenotypic capabilities of the bacterium, and both 
classes of events may increase the pathogenicity of the bacterium (see text). 







EDL933(0157:H7) 
enterohemorrhagic 



Fig, 2. Genetic differences between three completely sequenced isolates off, coli. the 
nonpathogenic strain MG1655 (hold line), the uropathogenic strain CFT073 (gray line), and the 
enterohemorrhagic strain EDL933 (thin line). The number of genes shared among genomes, or 
unique to a genome, is shown in the appropriate location in the Venn diagram [after 40]. 



sequences group Shigella within the E. coli complex [42, 43]). Even though high 
sequence identity among genes shared among the four strains place them all in 
the same species [43], Jess than 40% of the collective gene pooJ among the three 
named strains of £. coli is shared (fig. 2), and nearly 47% of the genes are unique 
to one of the three taxa [40]. Each strain has numerous genes found only in that 
genome, the lion's share found in the two disparately pathogenic E. coli strains 



Lawrence 



258 



(fig. 2). In contrast, the Shigella genome has many pseudogenes and prophages, 
but fewer unique genes, implicating large-scale gene transfer as a factor in its 
evolution to a lesser degree [41], Indeed the role of gene loss (the cadA and ompT 
genes) in maintaining virulence has been noted in Shigella [44, 45]. Taken 
together, the variation among these strains shows that gene transfer can act 
quickly to introduce genes that allow for dramatic changes in lifestyle, but, as 
discussed further below, it is not the only route. 



Generalized Lifestyle Alteration 

Examination of variation among natural isolates of £. coli shows that 
huge dynamics in gene content are evident even among these nonpathogenic 
isolates [46^8]. Therefore, the lessons imparted by pathogenicity islands are 
extensible to the examination of other, nonpathogenic bacteria. If pathogenicity 
islands can allow for rapid adoption of the pathogenic lifestyle, one could posit 
that the introduction of other genes would allow for similarly effective 
invasion of nonpathogenic niches [32-34]. Examination of the genomes of 
numerous bacteria shows that nonpathogenic bacteria have a great deal of DNA 
that is 'atypical' with respect to the majority of genes in the genome and could 
have been introduced recently by HGT [49]. As seen in figure 3, genomes of 
both pathogenic and nonpathogenic bacteria show abundant signs of recent 
gene acquisition. 

The methods employed to generate figure 3 rely on a simple premise: genes 
introduced into a genome from a donor chromosome all share one characteristic; 
they did not evolve for long periods of time in their current genomic context. 
Each organism experiences a unique set of 'directional' mutation pressures 
[50-52] which impart signature patterns of nucleotide composition [53-55], 
codon usage bias [56], nucleotide strand bias [57-59], dinucleotide signatures 
[60—62] and patterns detected by Markov chain models [63]. In effect, genes 
evolving in the same genome Mook alike' due to the mutational proclivities of the 
DNA polymerase, the composition of the nucleotide pools during replication, the 
nature and efficiencies of the DNA mismatch-repair systems, abundance of tRNA 
species and other factors. As a result, 'atypical' genes are often interpreted as 
having evolved in a different genomic context, their unusual features reflecting 
the different mutational pressures of their parental donor genome. Initially, these 
genes are readily detected as having unusual compositional pattems, but over 
time^ these patterns are erased as genes evolve in their new genomic context 
[54, 55]. 

Alternatively, genes introduced by HGT can be detected since their relation- 
ship to homologues in other bacterial genomes will not be congruent with the 



Horizontal and Vertical Gene Transfer 259 



Pseudomonas aeruginosa 

Escherichia cofi 

Mycobacterium tuberculosis 

Bacillus halodurans 

Vibrio choleras 

Bacillus subtilis 

Syr^echocystis PCC6803 

Deinococcus radiodurans 

Xylella fastidiosa 

Pasteurefia multocfda 

Lactococcus lactis 

Archaeoglobus fulgidus 

Neisseria meningitidis Z2491 

Neisseria meningitidis MC58 

Halobactenum NRC-1 

Thermotoga maritima 

Mycobacterium leprae 

Pyrococcus abyssi 

Pyrococcus horikoshii 

Methanobacterium Ihermoautotrophicum 

Aeropyrum pernix 

Campylobacter jejuni 

Haemophilus inlluenzae 

Helicobacter pyfoh 26695 

Aquifex aeollcus 

Thermoplasma acidophilum 

Methanococcus jannaschii 

Treponema pallidum 

Borrefia burgdorferi 

Rickettsia prowazekii 

Mycoplasma pneumoniae 

Ureaplasma urealyiicum 

Buchnera aphidicola 

Mycoplasma genitalium 




Typical genes 



Atypical genes, with atypical 
phylogeny 







T 
2 



T 
3 



T 
4 



T 
5 



Megabases of protein-coding DNA 



relationships of other genes in the same genome. These 'phylogenetic' methods 
can include the direct comparison ofdendrograms to identify genes with uimsual 
branching patterns [64], or examine relationships en masse to detect genes with 
discordant relationships [65]. Each method has the abiHty to detect different sets 
of genes, since each relies upon different sets of criteria for the identification of 
potentially transferred genes [66—68]. A comparison of these 'parametric' methods 
of identifying genes potentially mtroduced by HGT with phylogenetic methods 
that identify genes unique to a lineage show that the two approaches are, for the 
most part, congruent in their predictions [68]. Therefore, it is fair to say that some 
lessons learned from the evolution of pathogens - that is, rapid adaptation can 
occur via HGT - are extensible to the evolution of nonpathogens. 



6 



Fig, 3. The amount of recently acquired DNA in 34 bacterial and archaeal genomes, 
as inferred from the identification of genes with atypical sequence features, including aberrant 
nucleotide composition, dinucleotide signatures and codon usage bias patterns; these atypi- 
cal genes were confirmed as being horizontally transferred by performing a phylogenetic 
concordance test. (That is, the strongest matches to the gene in the database differed signifi- 
cantly from the set of strongest matches shown by other genes in the chromosome.) 



Lawrence 



260 



Pathogens with Little Foreign DNA 

Examination of figure 3 shows that many pathogens have little recently 
acquired DNA, This observation would seem to conflict with the conclusion 
that gene acquisition plays such a strong ro]e in the evolution of pathogens. Yet 
there is good theoretical and empirical evidence that HGT would be of lesser 
importance in the evolution of virulent or host-restricted pathogens, or special- 
ized bacterial symbionts, like those with small genomes (fig. 3). That is, one 
may consider pathogen evolution to be a two-step process. Fu:st, HGT allows 
the introduction of genes which allow adoption of the pathogenic lifestyle. 
Metabolic and physiological capabilities may be augmented, and pathogenicity 
islands will be detected in the genomes. However, as the pathogen adapts to its 
new role, HGT becomes both less important and less feasible, and further evolu- 
tionary change is accommodated by alteration of existing genes. As existing 
foreign genes ameliorate to their new genomic context [54], few genes will be 
detected as 'foreign' using the methods employed for generating figure 3; 
phylogenetic methods would still detect ancient transfer events, as evidenced by 
the facile detection of the transfer of the phenylalanine tRNA synthase mto the 
ancestor of spirochetes from an archaeal donor [64], 



Factors Reducing Rate of HGT in Pathogens 

Three primary influences lead to the reduction in the rate of HGT into 
pathogen genomes. First, many symbionts and pathogens have a reduced expo- 
sure to the agents facilitating gene transfer: conjugation, transduction and trans- 
formation [69]. With a lower opportunity for exposure to foreign DNA, fewer 
foreign genes would be detected in the genomes of these relatively sheltered 
organisms. Second, fewer genes may be of utility to organisms that have adapted 
to a specialized environment. Here, the pathogen may have no use for the major- 
ity of foreign genes that are introduced into its genome, since few would offer 
functions of utility [70, 71], Lastly, tlie changes m population stmcture coincident 
with specialization - lower population size and rare rates of recombination - raise 
the threshold for an effectively neutral mutation [70]- As a result, fewer genes, 
even those offering a potential benefit, would be retained; the benefits they con- 
fer would differ significantly from selective neutrality (that is, small benefits 
are unable to allow a gene to persist in the face of stochastic changes in gene 
frequency - termed random genetic drift - that dominate the fates of variant 
alleles at low population sizes; this effect whereby genes which would confer a 
benefit in a larger population cannot do so m a small population has been termed 
'effective neutrality' [72, 73]), and the genes would be lost. 



Horizontal and Vertical Gene Transfer 261 



It is this same loss of population size that leads to genome decay in many 
pathogens, like Mycobacterium leprae [74] or Rickettsia prowazekii [75-77]. In 
these cases, the populations are insufficiently large to retain the genes present in 
the ancestral organism, and potentially deleterious mutations - those that elimi- 
nate gene function by producing pseudogenes - accumulate. That is, although 
the genes so mutated may have provided a serviceable function, the losses of the 
genes were uisufficiently detrimental to prevent pseudogene formation; as a 
result, the mutations were effectively neutral, given the population size, structure 
and rate of recombmation. The effects of such population bottle necks are evident 
in many pathogens just beginning this process of genome decay, including 
Salmonella typhi [78, 79] and Mycobacterium tuberculosis [80-83]. 



Correlated Genome Changes 

In addition to pseudogene accumulation and the failure to retain genes 
introduced by HGT, the genomes of pathogens may experience other phenomena 
at abnormally high rates. Again, these events reflect a decreased ability for cells 
containing such deleterious rearrangements to be eliminated from the population, 
not an increased rate of their initial occurrence. For example, inversions that do 
not contain the origin or terminus of replication are rare [84]. However, such 
chromosome rearrangements are common in many bacterial genomes, including 
Bordatella pertussis, Rickettsia, and Salmonella typhi [85-87]. 

The increased numbers of inversions in B. pertussis (and, to a lesser extent, 
in Bordatella parapertussis) are thought to have resulted from an accumulation 
of transposable elements [88]. The IS elements provide sites of DNA identity in 
inverted orientation at which homologous recombination may act, thus creating 
an inversion [89]. Similar recombination between IS elements in direct orientation 
may lead to potentially large chromosome deletions, a phenomenon deduced to 
have occurred in the Buchnera aphidicola genome. A similar accumulation of 
IS insertions, especially of IS/, is seen in Shigella [41, 90]. In both cases, IS 
elements are not more prone to transpose in these genomes; rather, the strains 
carrying large numbers of IS elements are not removed from the population 
since the insertions are insufficiently detrimental. 



Gene Loss during Pathogen Evolution 

As noted above, several gene losses at the ompT [45] and cadA [44] loci 
were critical for the evolution of pathogenicity in Shigella [91]. Similarly, loss 
of genes - especially those involved in the producrion of surface antigens - was 



Lawrence 



262 



important in the evolution of the highly virulent strains B. pertussis and 
B. parapertussis from the relatively broad host-range pathogen Bordatella 
bronchiseptica [88]. Common genes losses were detected [92] when comparing 
the smaller genomes ofM. leprae and Mycobacterium bovis [92] to the larger 
genome of M. tuberculosis, suggesting that adaptation occurred via loss of 
function and not gain of functions by way of horizontally transferred genes; 
indeed no genes are found to be unique to M. bovis [92], unlike the situation 
with pathogenic E. coli [40]. 

These changes reflect more than just the inevitable loss of genes that are no 
longer under selection for function [93]. Rather, gene loss can be beneficial if the 
gene product interferes with the functions of the newly evolving pathogen, either 
by diverting metabolic flux along an unproductive pathway or by actively creat- 
ing substances that attenuate its vinalence. Alternatively, chromosomal deletions 
may be beneficial if the loss of DNA removes potentially problematic DNA 
sequences, like genetic parasites [93], or inverted DNA (as discussed above) that 
may interfere with chromosome replication and segregation [58, 94-97]. 



Gene Modification during Pathogen Evolution 

Although gene gain and gene loss are effective means by which the char- 
acter of a bacterial species may change, we have so far overlooked perhaps the 
most fundamental mode of bacterial evolution: gene alteration by mutational 
processes. Mutation has played a critical role in the origin or maintenance 
of pathogenicity in many organisms. For example, increased virulence of 
B. pertussis is due in part to an increased level of expression of the ptxA gene, 
facilitated by mutations which increased the strength of its promoter sites and 
binding sites for the BvgA regulatory protein [88]. Here, it was not gene gain 
that led to toxin production but an increased level of expression of a preexisting 
toxin gene. 

A different kind of mutational processes, replication slippage, plays an 
LinporlanL role in regulating the expression of antigenic loci in a stochastic fash- 
ion in both Haemophilus influenzae and Neisseria species [98-100]. Here, genes 
can be turned on or off at random via the addition of microsatellite repeats 
embedded within protein coding genes (allowing in-frame translation in only 1 
of 3 slippage states), or genes may be attenuated in expression by the addition or 
subtraction of bases in its promoter region. Lastly, single point mutations can 
bring about enormous changes in virulence. Yersinia pestis, the causative agent 
of bubonic plague, is virtually indistinguishable from its parent strain Yersinia 
pseudotuberculosis [101]. TTiis 'instant species' apparently has recently emerged 
by virtue of only a handful of genomic modifications [102, 103]. 



Horizontal and Vertical Gene Transfer 263 



a 



Recipients of these 
donor fragments 
are counterselected 




Recipients of these 
donor fragments 
are counterselected 



X 



Gene flow between 
ancestral and derived 
taxa is reduced in the 
neighborhood of 
acquired genes 



A' 




B' 




a 



>*^"^^<3 



E' 




F' 



G' 




These fragments are 

poor or nonfunctional donors 



b 



A 




B 




^X^y^AKi 




l_ack of recombination leads to an accumulation of 
differences in the neighbortiood of the acquired genes 



A' 




B^ 




C 



>^"^^<^ 



K^ 




F 



G' 




Fig. 4, The localized decrease in homologous recombination as catalyzed by an HGT 
event that leads to ecologically different lineages of bacteria, a After genes (depicted in 
black) are acquired by lateral transfer, homologous recombination in flanking genes 
decreases, both because recombination events leading to the addition or removal of the 
acquired genes are counterselected, or because fragments with ends within the acquired 
genes serve as poor donors, b Lower rates of recombination lead to increased sequence diver- 
gence (denoted by gray shading) at flanking genes due to their inability to participate in local 
periodic selection events [figure after 35; 1 10]. 



Interplay between HGT, Mutation and Recombination 

Though seemingly distinct processes, genes introduced by HGT or modified 
in an adaptive way by mutational processes affect the process of intraspecific 
recombination (that is, gene exchange among closely related microorganisms) 
in a very particular way. While mutations and HGT events introduce potentially 
important genetic variation into a population, recombination among strains dis- 
seminates this genetic information among closely related strains. Among strams 
of a bacterial 'species' - defined as those which exchange genes at high fre- 
quency by homologous recombination [6] - strains can show dramatic differ- 
ences in the environments they inhabit. This phenomenon has been shown for 



Lawrence 



264 



Free-living, benign ancestor 

Gain of pathogenicity islands 

Broad host range pathogen 

Host specialization 

Virulent, narrow host range pathogen 



am 

i 



Escherichia coli 

Escherichia coli 0157:H7 
Salmonella typhimurium 
Bordatella brochiseptica 

Salmonella typhi 



Population crasli, loss 
of recombination 



I 



Virulent, host-dependent pathogen 



Massive gene loss, 

pseudogene accumulation, 

genome decay 

Obligate, host-dependent pathogen 



I 



Mycobacterium tuberculosis 
Bordatella pertussis 

Mycobacterium leprae 

Ricf^ettsia 
Mycoplasma 



Fig. 5. The pathway to pathogenicity (see text). Steps in the evolution of pathogens are 
depicted on the left, and examples of bacteria exhibiting these properties are depicted on the 
right. 



natural isolates of E. coli and other enteric bacteria [104-106], and is also 
evident in the strong genotypic and phenotypic differences between pathogenic 
and nonpathogenic strains of E. coli [40]. Therefore, differences that are adap- 
tive for one strain may not be adaptive for other strains; as a result, recombina- 
tion events which introduce genes into a nonadaptive strain background, or 
remove important genes that were not present in the donor taxa, will be coun- 
terselected [35], In effect, the events which cause phenotypic differentiation 
among strains lead to genetic isolation of these strains (fig. 4); therefore^ 
the processes of pathogenicity island acquisition may contribute to the lack 
of recombination among strains, therefore catalyzing subsequent genome 
reduction. 



Genome Evolution and the Progression of Pathogenicity 

As detailed above, pathogens may evolve through several distinct phases, 
each of which is characterized by different evolutionary mechanisms acting to 
shape the content and composition of their genomes. This process is outlined in 
figure 5. At the start, a presumably benign, free-living ancestor adopts a patho- 
genic lifestyle after acquiring virulence factors by HGT Here, HGT acts as it 
does for many bacterial Imeages in providing genetic modules for rapid and 
effective exploitation of a new environmental niche. At this point, populations 
of broad host-range pathogens may be similar to their nonpathogenic sisters in 



Horizontal and Vertical Gene Transfer 



265 



terms of population size and structure. This stage may be typified by pathogenic 
strains of £. coli, for example. 

However, host specialization (as seen in S. typhi, for example) leads to 
lower population sizes, lower rates of recombination, and eventual gene loss. 
B. pertussis shows an intermediate phenotype, whereby many genes have been 
lost, IS elements are accumulating, and pseudogenes are evident. M. leprae rep- 
resents a genome in massive decay, wherein the pseudogenes abnost outnumber 
functional genes. Eventually, this period of genome instability passes; pathogens 
with extremely small genomes (e.g., Rickettsia or Mycoplasma) remain as the 
result. In endosymbionts which experience similar processes of genome decay 
[107, 108] - this period of stability can last for millions of years [109]. 



Conclusions 

While the introduction of pathogenicity islands by HGT is considered a hall- 
mark in the evolution of pathogenic bacteria, this process represents only one step 
in a multifaceted and complex evolutionary process. Some of the principles of 
pathogen evolution are widely applicable to the evolution of nonpathogen organ- 
isms (e.g., adaptation via the acquisition of foreign gene), while others are not. 



References 

1 Syvanen M, Kado CI: Horizontal Gene Transfer. London, Chapman and Hall, 1998. 

2 Syvanen M: Horizontal gene flow: Evidence and possible consequences. Annu Rev Genet 1994; 
28:237-261. 

3 Syvanen M: Migrant DNA in the bacterial world. Cell 1990;60:7-8. 

4 Davies J: Origins and evolution of antibiotic resistance. Microbiologia 1996;12:9-16. 

5 Levin B: Periodic selection, infectious gene exchange, and the genetic structun; off". coH populations. 
Genetics 198i;99:l-23. 

6 Dykhuizen DE, Green L: Recombination in Escherichia coli and the definition of biological 
species. J Bacteriol 1991;173:7257-7268. 

7 Guttman DS, Dykhuizen DE: Clonal divergence in Escherichia coli as a result of recombination, 
not mutation. Science 1994;266:1380-1383. 

8 Guttman DS, Dykhuizen DE; Detecting selective sweeps in naturally occurring Escherichia coli. 
Genetics 1994;138:993-1003. 

9 Feil EJ, Holmes EC, Bessen DE, Chan MS, Day NP, Enright MC, Goldstein R, Hood DW, Kalia A, 
Moore CE, Zhou J, Spratt BG: Recombination within natural populations of pathogenic bacteria: 
Short-term empirical estimates and long-term phylogenetic consequences. Proc Natl Acad Sci USA 
2001;98:182-187. 

10 Feil EJ, Sjiiitii JM, Eiiurighl MC, Spralt BG: Eiiti mating recorribi national paranieters in Sireptucuccus 
pneumoniae from multilocus sequence typing data. Genetics 2000; 154: 1439 1450. 

1 1 Barinaga M: A shared strategy for virulence. Science 1996;272: 1261 1263. 

12 Galan JE, Curtiss R 3rd: Distribution of the invA, -B, -C, and -D genes of Salmonella lyphimiirium 
among other Salmonella serovars: invA mutants of Salmonella typhi are deficient for entry into 
mammalian cells, bifecl immun 1991;59:2901-2908. 



Lawrence 



266 



13 Galan J, Girmochio C, Costeas P: Molecular and functional characterization of the Salmonella 
invasion gene invA: Homology of InvA to members of a new protein family, J Bacteriol 1992; 174: 
4338^349. 

14 Groisman EA, Ochman H: Pathogenicity islands: Bacterial evolution in quantum leaps. Cell 1996; 
87:791-794. 

15 Groisman EA, Ochman H: How to become a pathogen. Trends Microbiol 1994;2:289-294. 

16 Ochman H, Soncini FC, Solomon F, Groisman EA: Identification of a pathogenicity island 
required for Salmonella survival in host cells. Proc Natl Acad Sci USA 1996;93:7800-7804. 

17 Ochman H, Groisman EA: Distribution of pathogenicity islands in Salmonella spp. Infect Tmmun 
I996;64:54I0^54I2. 

18 Sukhan A, Kubori T, Galan JE: Synthesis and localization of the Salmonella SP]-1 type 111 secre- 
tion needle complex proteins PrgI and PrgJ. J Bacteriol 2003;185:3480-3483. 

19 Russmann H, KuboriT, Sauer J, Galan JE: Molecular and functional analysis of the type 111 secre- 
tion signal of the Salmonella enterica InvJ protein. Mol Microbiol 2002;46:769-779. 

20 Kubori T, Galan JE: Salmonella type HI secretion-associated protein [nvE controls translocation 
of effector proteins into host cells. J Bacteriol 2002; 1 84:4699^708. 

21 Waldor MK, Mekalanos JJ: Lysogenic conversion by a filamentous phage encoding cholera toxin. 
Science 1996;272:1910-1914. 

22 Waldor MK: Bacteriophage biology and bacterial virulence. Trends Microbiol 1998;6:295-297. 

23 Pema NT, Plunkett G, Burland V, Mau B, Glasner JD, Rose DJ, Mayhew GF, Evans PS, Gregor J, 
Kirkpatrick HA, Posfai G, Hackett J, Klink S, Boutin A, Shao Y, Miller U Grotbeck EJ, Davis NW, 
Lim A, Dimalanta ET, Potamousis ICD, Apodaca J, Anantharaman TS, Lin J, Yen G, Schwartz DC, 
Welch RA, Blattner PR: Genome sequence of enterohaemorrhagic Escherichia coli 0157:H7. 
Nature 2001;409:529-533. 

24 Cerdeno-Tarraga AM, Efstraliou A, Dover LG, Holden MT, Fallen M, Bentley SD, Besra GS, 
Churcher C, James KX), De Zoysa A, Chillingworlh T, Cronin A, Dowd L, Fellwell T, Hamlin N, 
Holroyd S, Jagels K, Moule S, Quail MA, Rabbinowitsch E, Rutherford KM, Thomson NR, 
Unwin L, Whitehead S, BarreJl BG, Parkhill J: The complete genome sequence and analysis of 
Cofynebacterium diphtheriae NCTCI3129. Nucleic Acids Res 2003;31:6516-6523. 

25 Harb OS, Gao LY, Abu Kwaik Y: From protozoa to mammalian cells: A new paradigm in the life 
cycle of intracellular bacterial pathogens. Environ Microbiol 2000;2:251-265. 

26 Swanson MS, Hammer BK: Legionella pneumophila pathogenesis: A fateful journey from amoebae 
to macrophages. Annu Rev Microbiol 2000;54:567-613. 

27 Skriwan C, Fajardo M, Hagele S, Horn M, Wagner M, Michel R, Krohne G, Schleicher M, Hacker J, 
Steinert M: Various bacterial pathogens and symbionts infect the amoeba Dictyos(elium discoideum. 
Int J Med Microbiol 2002;291:615-624. 

28 Marrie TJ, Raoult D, La Scola B, Birtles RJ, de Carolis E: Legion ella-Wkt and other amoebal 
pathogens as agents of community-acquired pneumonia. Emerg Infect Dis 2001;7:1026-1029. 

29 Ernsl ME, Klepser ME, Fouts M, Marangos MN: Tetanus: Pathophysiology and management. Ann 
Pharmacother 1997;31:1507-1513. 

30 Bolte ER: Autism and Closiridiim ieiani, Med Hypotheses 1998;51:133-144. 

3 1 Labrousse A, Chauvet S, Couillault C, Kurz CL, Ewbank JJ: Caenorhabdiiis elegans is a model host 
for Salmonella typhimurium. Curr Biol 2000; 1 0: 1 543-1 545, 

32 Lawrence JG: Selfish operons and speciation by gene transfer Trends Microbiol 1997;5:355-359. 

33 Lawrence JG: Gene n-ansfer, speciation, and the evolution of bacterial genomes. Curr Opin Microbiol 
1999;2:519-523, 

34 Lawrence JG: Selfish operons: The evolutionary impact of gene clustering in the prokaryotes and 
eukaryotes. Curr Opin Genet Dev 1999;9:642-648. 

35 Lawrence JG: Gene transfer in bacteria: Speciation without species? Theor Popul Biol 2002;61: 
449^60. 

36 Doolittle WF: Lateral genomics. Trends Cell Biol 1999;9:M5-M8. 

37 Doolittle WF: The nature of the universal ancestor and the evolution of the proteome. Curr Opin 
Struct Biol 2000;10:355-358. 

38 Gogarten JP, Doolittle WF, Lawrence JG: Prokaryotic evolution in light of gene transfer, Mol Biol 
Evol 2002; 19:2226^2238. 



Horizontal and Vertical Gene Transfer 267 



39 Blattner FR, Plunkett GR, Bloch CA, Perna NT, Burland Y Riley M, Collado-Vides J, Glasner JD, 

Rode CK, Mayhew GF, Gregor J, Davis N\V, Kirkpatrick HA, Goeden MA, Rose DJ, Mau B, 

Shao Y: The complete genome sequence of Escherichia coli K-12. Science 1997;277: 
1453-1474. 

40 Welch RA, Burland V, Plunkett G 3rd, Redford P, Roesch P, Rasko D, Buckles EL, Liou SR, 
Boutiji A, Hackett J, Stroud D, Mayhew GF, Rose DJ, Zhou S, Schwartz DC, Perna NT, Mobley HL, 
Donnenberg MS, Blattner FR: Extensive mosaic structure revealed by the complete genome 
sequence of uropathogenic Escherichia coli. Proc Natl Acad Sci USA 2002;99:17020-17024. 

41 Wei J, Goldberg MB, Burland V, Venkaiesan MM, Deng W, Fournier G, Mayhew GF, Plunkett G 3rd, 
Rose DJ, Darling A, Mau B, Perna NT, Payne SM, Runyen-Janecky LJ, Zhou S, Schwartz DC, 
Blattner FR: Complete genome sequence and comparative genomics of Shigella Jlexneri serotype 
2a strain 2457T Infect Immun 2003;7 1 :2775-2786. 

42 Escobar-Paramo P, Giudicelli C, Parsot C, Denamur E: The evolutionary history of Shigella and 
enteroinvasive Escherichia co// revised. J Mol Evol 2003;57:I40^148. 

43 Lan R, Reeves PR: Escherichia coli in disguise: Molecular origins of Shigella. Microbes Infect 
2002;4:1125-1132. 

44 Day WA Jr, Fernandez RE, Maurelli AT: Pathoadaptive mutations that enhance virulence: Genetic 
organization of the cadA regions of Shigella spp. Infect Immun 2001;69:7471-7480. 

45 Nakata N, Tobe T, Fukuda 1, Suzuki T, Komatsu IC, Yoshikawa M, Sasakawa C: The absence of a 
surface protease, OmpT, determines the intercellular spreading ability of Shigella: The relation- 
ship between the ompT and kcpA loci. Mol Microbiol 1993;9:459-468. 

46 Ochman H, Jones IB: Evolutionary dynamics of fijll genome content in Escherichia coli. EMBO J 
2000;19:6637-6643. 

47 Bergthorsson U, Ochman H: Heterogeneity of genome size among natural isolates of Escherichia 
coli. J Bacteriol 1995; 1 77:578^5789. 

48 Bergthorsson U, Ochman H; Distribution of chromosome length variation in natural isolates of 
Escherichia coli. Mol Biol Evol 1998;15:6-16. 

49 Ochman H, Lawrence JG, Groisman E: Lateral gene transfer and the nature of bacterial innovation. 
Nature 2000;405:299-304. 

50 Sueoka N: Directional mutation pressure and neutral molecular evolution. Proc Natl Acad Sci 
USA 1988;85:2653-2657. 

51 Sueoka N: Directional mutation pressure, selective constraints, and genetic equilibria. J Mol Evol 
1992;34:95-114. 

52 Sueoka N: Directional mutation pressure, mutator mutations, and dynamics of molecular evolution. 
J Mol Evol 1993;37:137-153. 

53 Ochman H, Lawrence JG: Phylogenetics and the amelioration of bacterial genomes; in Neidhardt PC, 
Curtiss R 3rd, Ingraham JL, Lin ECC, Low KB, Magasanik B, Reznikoff WS, Riley M, 
Schaechter M, Umbai'ger HE (eds): Escherichia coli and Salmonella typhimiirium: Cellulai' and 
Molecular Biology, ed 2. Washington, American Society for Microbiology, 1996, pp 2627-2637. 

54 Lawrence JG, Ochman H: Amelioration of bacterial genomes: Rates of change and exchange. J Mol 
Evol 1997;44;383-397, 

55 Lawrence JG, Ochman H: Molecular archaeology of the Escherichia coli genome. Proc Natl Acad 
Sci USA 1998;95:9413-9417. 

56 M6digue C, Rouxel T, Vigier P, H6naut A, Danchin A: Evidence of horizontal gene transfer in 
Escherichia coli speciation. J Mol Biol 1991;222:851-856. 

57 Lobry JR: Asymetric substitution patterns in the two DNA strands of bacteria. Mol Biol Evol 
1996;13:660-665. 

58 Lobry JR, Louarn JM: Polarisation of prokaryotic chromosomes. Curr Opin Microbiol 2003;6: 
101-108. 

59 Lobry JR, Sueoka N: Asymmetric directional mutation pressures in bacteria. Genome Biol 2002; 
3:RESEARCH0058. 

60 Karlin S, Burge C: Dinucleotide relative abundance extremes: A genomic signature. Trends Genet 
1995;11:283-290. 

61 Karlin S, Mrazek J, Campbell AM; Codon usages in different gene classes of the Escherichia coli 
genome. Mol Microbiol 1998;29:1341-1355. 



Lawrence 



268 



62 Mrazek J, Karlin S: Detecting alien genes in bacterial genomes. Ann NY Acad Sci 1999;870: 
314-329, 

63 Hayes WS, Borodovsky M: How to interpret an anonyn^ous bacterial genome: Machine learning 
approach to gene identification. Genome Res 1998;8:1 154—1 171. 

64 Woese CR, Olsen GJ, Ibba M, Soil D: Aminoacyl-tRNA synthetases, the genetic code, aiid the 
evolutionary process. Microbiol Mol Biol Rev 2000;64:202-236. 

65 Clarke GD, Beiko RG, Ragan \4A, Charlebois RL: Inferring genome trees by using a filter to 
eliminate phylogenetically discordant sequences and a distance matrix based on mean normalized 
BLASTP scores. J Bacteriol 2002;184:2072-2080. 

66 Ragan MA: On surrogate methods for delecting lateral gene transfer. FEMS Microbiol Lett 2001; 
201:187-191. 

67 Ragan MA; Detection of lateral gene transfer among microbial genomes. Curr Opin Genet Dev 
2001;n:620-626. 

68 Lawrence JG, Ochman H: Reconciling the many faces of gene transfer. Trends Microbiol 2002; 
10:1^. 

69 Moran NA: Microbial minimalism: Genome reduction in bacterial pathogens. CeJI 2002; 108: 
583-586. 

70 Lawrence JG: Catalyzing bacterial speciation: Correlating lateral transfer with genetic headroom. 
SystBiol200l;50:479^96. 

71 Lawrence JG, Roth JR: Selfish operons: Horizontal transfer may drive the evolution of gene clusters. 
Genetics 1996;143:1843-1860. 

72 Ohia T; Role of very slightly deleterious mutations in molecular evolution and polymorphism. 
Theor Popul Biol 1976;10:25^275. 

73 OhtaT: Slightly deleterious mutant substitutions in evolution. Nature 1973;264:96-98. 

74 Cole ST, Eiglmeier K, Parkhill J, James KD, Thomson "NR, Wheeler PR, Honore N, Gamier T, 
Churcher C, Harris D, Mungall K, Basham D, Brown D, Chillingworth T, Connor R, Davies RM, 
Devlin K, Duthoy S, Feltwell T, Fraser A, Hamlin N, Holroyd S, Hornsby T, Jagels K, Lacroix C, 
Maclean J, Moule S, Murphy L, Oliver K, Quail MA, Rajandream MA, Rutherford KM, Rutter S, 
Seeger K, Simon S, Simmonds M, Skelton J, Squares R, Squares S, Stevens K, Taylor K, 
Whitehead S, Woodward JR, Barrell BG: Massive gene decay in the leprosy bacillus. Nature 2001 ; 
409:1007-1011. 

75 Andersson SG, Zomorodipour A, Andersson JO, Sicheritz-Ponten T, AJsmark UC, Podowski RM, 
Naslund AK, Eriksson AS, Winkler HH, Kurland CG: The genome sequence of Rickeltsia 
prowazekii and the origin of mitochondria. Nature 1998;396:133-140. 

76 Andersson JO, Andersson SG: Insights into the evolutionary process of genome degradation. Curr 
Opin Genet Dev 1999;9:664-671, 

77 Andersson JO, Andersson SG: Genome degradation is an ongoing process in Rickettsia. Mol Biol 
Evol 1999;16:1178-1191. 

78 Deng W, Liou SR, Plunkett G 3rd, Mayhew GF, Rose DJ, Burland V, Kodoyianni V, Schwartz DC, 
Blattner FR: Comparative genomics o^ Salmonella enltrica serovar Typhi strains Ty2 and CT18. 
J Bacteriol 2003;185:2330-2337. 

79 Parkhill J, Dougan G, James KD, Thomson NR, Pickard D, Wain J, Churcher C, Mungall KL, 
Bentley SD, Holden MT, Sebaihia M, Baker S, Basham D, Brooks K, Chillingworth T, Connerton P, 
Cronin A, Davis P, Davies RM, DowdL, White N, Farrar J, Feltwell T, Hamlin N, Haque A, HienTT, 
Holroyd S, Jagels K, Krogh A, Larsen TS, Leather S, Moule S, O'Gaora P, Parry C, Quail M, 
Rutherford K, Simmonds M, Skelton J^ Stevens K, Whitehead S, Barrell BG: Complete genome 
sequence of a multiple drug resistant Salmonella enterica serovar Typhi CTI8. Nature 200 1 ;4 1 3: 
848-852. 

80 Sreevatsan S, Pan X, Stockbauer ICE, Connell ND, Kreiswirih BN, Whittam TS, Musser JM: 
Restricted structural gene polymorphism in the Mycobacterium tuberculosis complex indicates 
evoluuonarily recent global dissemination. Proc Natl Acad Sci USA 1997;94:9869-9874. 

81 Musser JM, Amin A, Ramasv^^my S: Negligible genetic diversity of Mycobacterium tuberculosis host 
immune system protein targets: Evidence of limited selective pressure. Genetics 2000;155:7-16. 

82 Frothingham R, Meeker-O'Cormell WA: Genetic diversity in the Mycobacterium tuberculosis 
complex based on variable numbers of tandem DNA repeats. Microbiology 1998;144:1 189-1 196. 



Horizontal and Vertical Gene Transfer 269 



83 Frothingham R: Evolutionary bottlenecks in the agents of tuberculosis, leprosy, and paratuberculosis. 
Med Hypotheses 1999;52:95-99, 

84 Eisen JA, Heidelberg JF, White O, Salzberg SL: Evidence for symn^etric chromosomal inversions 
around the repHcation origin in bacteria. Genome Biol 2000;1:1-1 1. 

85 Liu SL, Hessel A, Sanderson JCE; The XbaJ-Blnl-CeuI genomic cleavage map of Salmonella 
enteriiidis shows an inversion relative to Salmonella typhimurium LT2. Mol Microbiol 1993; 10: 
655-664. 

86 Liu SL, Sanderson KE: Highly plastic chromosomal organization in Salmonella typhi. Proc Natl 
Acad Sci USA 1996;93:10303-10308. 

87 Liu SL, Sanderson KE: Homologous recombination between rrn operons rearranges the chromo- 
some in host-specialized species of Salmonella. FEMS Microbiol Lett 1998;164:275-281. 

88 Parkhill J, Sebaihia M, Preston A, Murphy LD/fhomson N, Harris DE, Holden MT, Churcher CM, 
Benlley SD, Mungall ICL, Cerdeno-Tarraga AM, Temple L, James K, Harris B, Quail MA, 
Achtman M, Aikin R, Baker S, Basham D, Bason N, Cherevach I, Chillingwonh T, Collins M, 
Cronin A, Davis P, Doggett J, Feltwell T, Goble A, Hamlin N, Hauser H, Holroyd S, Jagels K, 
Leather S, Moule S, Norberczak H, O'Neil S, Ormond D, Price C, Rabbinowitsch E, Rutter S, 
Sanders M, Saunders D, Seeger K, Sharp S, Simmonds M, Skelton J, Squares R, Squares S, 
Stevens K, Unwin L, Whitehead S, Barrell BG, Maskell DJ: Comparative analysis of the genome 
sequences of Bordeiella perftissis, Bordeiella parapertussis and Bordetella bronchiseptica . Nat 
Genet 2003;35:32^0. 

89 Roth J, Benson N, Galitski T, Haack K, Lawrence J, Miesel L: Rearrangements of the bacterial 
chromosome- Formation and applications; inNeidhardtFC, CurtissR 3rd, Ingraham JL, Lin ECC, 
Low KB, Magasanik B, ReznikoffWS, Riley M, Schaechter M, Umbarger HE (eds): Escherichia 
coll and Salmonella'. Cellular and Molecular Biology, ed 2. Washington, American Society for 
Microbiology, 1996, pp 2256-2276. 

90 Ohtsubo H, Nyman K, Doroszkiewicz W, Ohtsubo E: Multiple copies of iso-insertion sequences 
of IS/ in Shigella dysenieriae chromosome. Natuj'e 1981;292:640^643. 

91 Maurelli AT, Fem.'indez RE, Bloch CA, Rode CK, Fasano A: *Black holes' and bacterial patho- 
genicity: A large genomic deletion that enhances the virulence of Shigella spp. and enteroinvasive 
Escherichia coli. Proc Natl Acad Sci USA 1998;95:3943-3948. 

92 Gamier T, Eiglmeier K, Camus JC, Medina N, Mansoor H, Pryor M, Duthoy S, Grondin S, 
Lacroix C, Monsempe C, Simon S, Harris B, Atkin R, Doggett J, Mayes R, Keating L, Wheeler PR, 
Parkhill J, Barrell BG, Cole ST, Gordon SV, Hewinson RG: The complete genome sequence of 
Mycobacterium bovis. Proc Natl Acad Sci USA 2003; 100:7877-7882. 

93 Lawrence JG, Hendrix RW, Casjens S: Where are the pseudogenes in bacterial genomes? Trends 
Microbiol 2001;9:535-540. 

94 Lawrence JG, Hendrickson H: Lateral gene transfer: When will adolescence end? Mol Microbiol 
2003;50:739-749. 

95 Capiaux H, Cornet F, Corre J, Guijo M, Perals K, Rebollo JE, Louarn J: Polarization of the 
Escherichia coli chj'omosome. A view from the terminus. Biochimie 2001;83:161-170. 

96 Louarn J, Cornet F, Fancois V, Patte J, Louarn J-M: Hyperrecombination in the terminus region of 
the Escherichia coli chromosome: Possible relation to nucleoid organization. J Bacteriol 1994; 
176:7524-7531. 

97 Perals K, Comet F, Merlet Y, Delon I, Louarn JM: Functional polarization of the Escherichia coli 
chromosome terminus: The t/?/site acts in chromosome dimer resolution only when located 
between long stretches of opposite polarity, Mol Microbiol 2000;36;33^3. 

98 Hood DW, Deadman ME, Jennings MP, Bisercic M, Fleischmann RD, Venter JC, Moxon ER: 
DNA repeats identify novel virulence genes in Haemophilus influenzae. Proc Natl Acad Sci USA 
1996;93:11121-11125. 

99 Jennings MP, Hood DW, Peak IRA, Virji M, Moxon ER: Molecular analysis of a locus for the 
biosynthesis and phase-variable expression of the lacto-N-neotetraose terminal lipopolysaccharide 
structure in Neisseria meningitidis. Mol Microbiol 1995;18:729-740. 

100 Sarkari J, Pandit N, Moxon ER, Achtman M: Variable expression of the Ope outer membrane protein 
in Neisseria meningitidis is caused by size variation of a promoter containing poly-cytidine, Mol 
Microbiol 1994;13:207-217. 



Lawrence 



270 



101 Achtman M, Zurth K, Morelli G, Torrea G, Guiyoule A, Carniel E: Yersinia pestis, the cause of 
plague, is a recently emerged clone of Yersinia pseudombercvlosi^. Proc Natl Acad Sci USA 1999; 
96:14043-14048. 

102 Dykhuizen DE: Yersinia pestis: An instant species? Trends Microbiol 2000;8:296-298. 

103 Parkhill J, Wren BW, Thomson NR, Titball RW, Holden MTG, Prentice MB, Sebaihia M, James KD, 
Churcher C, Mungall KL, Baker S, Basham D, Bentley SD, Brooks K, Cerdeno-Tarraga AM, 
ChiNingworth T, Cronin A, Davies RM, Davis P, Dougan G, Feltwell T, Hamlin N, Holroyd S, 
Jagels fC, Leather S, Karlyshev AY Moule S, Oyston PCF, Quail M, Rutherford K, Simmonds M, 
Skelton J, Stevens K, Whitehead S, Barrel I BG: Genome sequence of Yersinia pestis, the causative 
agent of plague. Nature 200 1 ;4 1 3:523-527. 

1 04 Gordon DM, Bauer S, Johnson JR: The genetic struclure of Escherichia coti populations in primary 
and secondary habitats. Microbiology 2002;148:1513-1522. 

105 Gordon DM: Geographical structure and host specificity in bacteria and the implications for tracing 
the source of colifomi contamination. Microbiology 2001;147:1079-1085, 

106 Okada S, Gordon DM: Host and geographical factors influence the thermal niche of enteric bacteria 
isolated from native Ausnna I ian mammals. Mol Ecol 2001;10:2499-2513. 

107 Moran NA, Mira A: Tlie process of genome shrinkage in the obligate symbiont Buchnera aphidicola. 
Genome Biol 200I;2:RESEARCH0054. 

108 Mira A, Ochman H, Moran NA: Deletional bias and the evolution of bacterial genomes. Trends 
Genet 2001;17:589-596. 

1 09 Tamas 1, Klasson L, Canback B, Naslund K, Eriksson A-S, Wemegreen JJ, Sandstrom JP, Moran NA, 
Andersson SGE: Fifty million years of genomic stasis in endosymbiotic bacteria. Science 
2002:296:2376^2379. 

1 10 Majewski J, Cohan FM: Adapt globally, act locally: The effect of selective sweeps on bacterial 
sequence diversity. Genetics 1999;152:1459-1474. 



Jeffrey G, Lawrence 

Pittsburgh Bacteriophage Institute and Department of Biological Sciences 

University of Pittsburgh, Pittsburgh, PA 15260, USA 

Tel. +1 4 12 624 4204, Fax +1 412 624 4759, E-Mail jlawrenc@pitt,edu 



Horizontal and Vertical Gene Transfer 271 



Subject Index 



ActAJnvasion, role 187 
Actin-ADP-ribosylating toxins 
families 43 

mechanism of action 43, 44 
species distribution 43 
AdbesijQS 

extracellular matrix binding 
collagen 91 
elastin 93 
fibrinogen 94 
fibronectin 91, 92 
glycosaminoglycans 94 
laminin 92, 93 
overview 90, 91 
vitronectiji 93 
Listeria monocytogenes 104, 105 
miscellaneous gram-positive bacteria 

adhesins 104, 105 
Staphylococcus aureus 1 1 - 1 04 
Streptococcus agalactiae 96, 99 
Streptococcus pneumoniae 96, 100, 101 
Streptococcus pyogenes 94—99 
Agrobacterium tumefaciens, type IV 

secretion 197, 198 
Z)-Alanyl-g]ycyl endopeptidase, functions 

149 
Albomycin, iioa siderophore carriers 224, 

225 
Anthrax toxin 
edema toxin 
components 38 
mechanism of action 38,39 



lethal factor metallopeptidase 164 

lethal toxin 41 
Antibiotics, iron siderophore carriers 

iron catecholate carriers 226, 227 

iron hydroxamate carriers 224-226 

resistance 227 
L-Ara4N, synthesis and lipid A 

modification 14, 15 
Arg-C, functions 167 

Bacillus anthraciSy see Anthrax toxin 
Bacteriocin-processing peptidases, 

functions 148, 149 
Bacteroides fragilis, enterotoxin 3 1 
Bbp, adhesin functions J 03 
Biofilm 

development cycle 114, 115 
Escherichia coll 

gastrointestinal biofilms 118, 119 
indwelling device colonization 120, 

121 
in vitro iiludit;s \ I6-I i8 
pathogenesis, role 116 
urinary tract infection 119, 120 
prospects for study 127,128 
Pseudomonas aeruginosa 
antibiotic resistance 126,127 
cystic fibrosis chronic lung infections 

123-126 
in vitro studies 122, 123 
signaling 1 15, 1 16 
BlaRl, functions 164, 165 



272 



BoNT, see Botulinum toxin 

Bordetella pertussis, see also Cya; Pertussis 

toxin 

type rv secretion 198, 199 
Botulinum toxin (BoNT), mechanism of 

action 41,42 
Btu proteins, structures 211,213 

C3 exoenzyme, mechanism of action 46 
C5a peptidase, functions 167 
Calpain, bacterial functions 142 
Camelysin, functions 165 
CAMPs 

hpid A modifications, protection 12, 13 
mechanism of action 1 1, 12 
signaHng pathway activation 14 
Capsular polysaccharide (CP) 
attachment 55 
Escherichia coli 

classification of capsules 59,60 
genetic orgaruzation/regulation, group 
2 capsule gene clusters 60-64 
fujictions 
adherence 56, 57 
desiccation resistance 56 
host immune resistance 57-59 
structures 55, 56 
Caspases, bacterial functions 152 
Cathelicidins, see CAMPs 
Cell wall, components 1 82 
CGP 4832, iron siderophore carriers 226 
Cholera toxin (CT) 

mechanism of action 36 
secretion 192 
structure 36 
trafficking 36 
ClfA, adhesui functions 103 
ClfB, adhesin functions 103 
Clostridium 
botulinum toxin, ^ee Botulinum toxin 
C3 exoenzyme 46 
Clostridium perfringes a- toxin 
mechanisms 30, 31 
phospho I ipase C activity 30 
structure 30 
enterotoxin 32, 33 
glucosylating toxins 47, 48 



neurotoxin mechanisms 42, 43 
Clostripain, bacterial functions 151 
Clp, functions 168 
Cna, adhesin functions 1 02, 1 03, 1 06 
Collagen, features and adhesin binding 

91 
Collagenase, bacterial functions 162 
Corynebacteriiim diphlheriae, see also 

Diphtheria toxin sortases 139 
Curli organelles, assembly 73,74 
Cya 

adenylate cyclase activity 37 

cell internalization 38 

structure 37 
Cytolethal distending toxins (CDTs) 

mechanism of action 39 

species distribution 39 
Cytoplasmic membrane, iron transport 

211,213 
Cytotoxic necrotizing factors (CNFs) 

mechanism of action 45 

structure 45 

types 44,45 

Defensins, see CAMPs 

DegP, functions 166 

Dichelobacter nodosus, pathogenicity 

islands 246 
Wpeptidyl aminopeptidase, bacterial 

functions 167, 168 
Diphtheria toxin (DT) 

elongation factor 2 inhibition 34 

receptor 33, 34 

structure 33 

transport 34 

Ebh, adhesin functions 102 
EbpS, adhesin fiinctions 104 
Elastin, features and adhesin binding 93 
Elongation factor 2 (EF2), inliibition by 

exotoxins 34, 35 
Emp, adhesin functions 104 
Endotoxin, see also Lipopolysaccharide 

definition 1 

history of study 1 
Enterobactin, iron siderophore carriers 

226, 227 



Subject Index 



273 



En lerococcus faecal is ^ pathogenicity 

islands 246 
EptA, pEtn modification of lipid A 15, 16 
Escherichia coli 
biofilms 
gastrointestinal biofilms 1 18, 1 19 
indwelling device colonization 120, 

121 
in vitro studies 116-118 
pathogenesis, role 116 
urinary tract infection 119, 120 
capsular polysaccharide 

classification of capsules 59,60 
genetic organization/regulation, group 
2 capsule gene clusters 60-64 
fimbriae, urinary tract infection 74-76 
horizontal gene transfer 257-259 
iron transport related to virulence 

216-219 
pathogenicity islands 242-244 
Exotoxin 

Cell-surface-active toxins 

Bacteroides fragilis enterotoxin 31 
phospholipases 30, 31 
pore-forming toxins 31-33 
signal transduction pathway 

modulation 28-30 
supcrantigens 33 
definition 28 

intracellularly active toxins 
actin alterations 43-49 
apoptosis induction 39^1 
cell cycle arrest 39 
celJ homeostasis alterations 36-39 
protein synthesis inhibition 33-36 
trafficking alterations 41^3 
overview of mechanisms 28,49 
therapeutic application prospects 50 
Extracellular polysaccharide, sloughing 55 

Fba, adhesin functions 97 

FBP54, adhesin fixnctions 97 

FbpA, structure 21 1 

FecA, structure 214 

FepA, structure 214 

Ferrimycins, iron siderophore carriers 226 

FhuA 



antibiotic carrier 225 
structure 214 
FhuD 

antibiotic carrier 225 
structure 21 1 
Fibrinogen, features and adhesin binding 

94 
Fibronectin, features and adhesin binding 

91,92 
Fimbriae 

adhesive functions 67 
biosynthesis 
chaperone/usher pathway 69-71 
extracellular nucleator pathway 73 
classification 68-74 
CSl fimbria! family 71,72 
pathogenesis, roles 

Escherichia coli urinary tract infection 

74-76 
extracellular component binding 77, 
78 
Flagella 

functions 67, 68 
virulence factors 80,81 
FnbpA, adhesin functions 101 
FnbpB, adhesin functions 101 
F2/PFBP, adhesin ftjnctions 97 
FtsH, therapeutic targeting 158,159 



Genome evolution, see also Horizonta 
gene transfer; Pathogenicity islands 
gene loss during pathogen evolution 

262, 263 
gene modification during pathogen 

evolution 263 
horizontal gene transfer interplay with 

mutation and recombination 

264, 265 
pathogenicity progression 265,266 
Genomic islands (GEIs), see also 
Pathogenicity islands 
bacterial fitness, role 236,237 
definition 235 
features 235, 236 
genome plasticity, role 247-249 
high-pathogenicity island 237 
Gingipains, bacterial functions 1 52-1 55 



Subject Index 



274 



Glycosaminoglycans, features and adhesin 

binding 94 
Gsp proteins, functions 191, 192 
Guanylate cyclase, STa binding 29, 30 

Helicobacter pylori, see also Vac A 
pathogenicity islands 245 
type IV secretion 199 
Hly transporters, functions 190, 191 
Horizontal gene transfer (HGT) 
correlated genome changes 262 
definition 256 
examples 256 
gene detection and abundance 259, 

260 
interplay with mutation and 

recombination 264, 265 
pathogen creation from nonpathogens 

257-259 
pathogenicity islands 256, 257, 259 
pathogenicity progression and genome 

evolution 265, 266 
pathogens with little foreign DNA 261 
rate reduction factors in pathogens 261, 

262 

IdeS peptidase, functions 140, 141 
Immune inhibitor A, bacterial functions 

161 
Lmmunoglobulin-A] -specific peptidases 

metallopeptidases 163 

serine peptidases 1 67 
Liner membrane (IM) 

CAMP permeability 11 J2 

structure, gram-negative bacteria 2, 3 
Lnvasins 

defuiiiions 1 81 

gram-negative pathogen invasive 
strategies 188-201 

gram-positive pathogen invasive 
strategies 184-187 
Iron 

bacterial function, virulence 215, 
216 

binding proteijis 210,211,216 

siderophores as antibiotic carriers 
iron catecholate carriers 226, 227 



iron hydroxamate carriers 224-226 

resistance 227 
surplus and stress 216 
transport related to virulence 

Escherichia coli 2 1 6—2 1 9 

Neisseria 223 

Pseudomonas aeruginosa 2 1 9, 220, 
222 

Salmonella 219 

Shigella 218 

Staphylococcus aureus 223, 224 

Vibrio cholerae 222, 223 
transport systems 

cytoplasmic membrane 211,213 

outer membrane 213-215 

prospects for study 227, 228 

Kdo transferase, lipid A biosynthesis 9 
Klebsiella oxytoca, type II secretion 
pathway 191, 192 

Laminin^ features and adhesin binding 92, 

93 
Lbp, adhesm functions 99 
Legionella pneumophila, type IV secretion 

199,200 
Legumain, bacterial functions 151, 

152 
Lethal toxin (LT) 
metallopeptidase 1 64 
structure 36 
Lipid A 
biosynthesis 7-1 0, 20, 2 1 
modifications 

l-Ara4N synthesis and modification 

14, 15 
CAMP counteraction, role 1 2, 13 
EptA in pEtn modification 15, 16 
LpxO in hydroxylation 18, 19 
magnesium effects on covalent 

structure 12 
PagL deacylation 19, 20 
PagP in palmitoylation 1 6 
prospects for study 21 
Rhizobium features 19, 20 
TolJ-like receptor 4 binding 1 
Lipid X, structure 7 



Subject Index 



275 



Lipid Y, structure 7 

Lipopolysaccharide (LPS), see also Lipid 

A; 0-antigen 

assembly 10, 1 1 

cation binding 1 1 

components 2, 3 

definition 3, 5 

structure 4 

transport 10, 1 1 
Lipoteichoic acid (LTA), adhesin functions 

97 
Listeria monocytogenes 

adhesins 104, 105 

invasion strategies 187 
Lmb, adhesin functions 100 
LpxA, lipid A biosynthesis 8,9 
LpxD, lipid A biosynthesis 9 
LpxO, lipid A hydroxylation 18, 19 
LysC, functions 167 



Magnesium 

lipid A structure modulation 12 

lipopolysaccharide permeability, role 
12 

PhoP/PhoQ signaling, limited 
environments 13, 14 
Map/Eap, adhesin functions 104 
M proteins, adhesin functions 97-99, 

106 
Murein, gram-negative bacteria 2 
MyD88, Toll-like receptor signaling 5 

Neisseria, iron transport related to 

virulence 223 
Nuclear factor-KB (NF-kB), Toll-like 

receptor signaling 5,6 

0-antigen 

assembly 10 

rough versus smooth 3 
OmpT, functions ] 37 
Omptins, functions 137 
Outer membrane (OM) 

CAMP permeability 1 1 

iron transport 213-215 

lipopolysaccharide transport 10,11 

structure, gram-negative bacteria 2, 3 



11 



PagL, lipid A deacylation 19, 20 
PagP 

catalytic mechanism 17, 18 
lipid A palmitoylation 16 
outer membrane localization 16, 18 
palmitoyl donors 17 
species distribution 17 
structure 17 
Papain, bacterial functions 141,142 
Pathogenicity islands (PAls) 

enterobacterial pathogens 242-245 

examples (table) 238-241 

genes 257 

genome plasticity, role 247-249, 256, 

257, 259 
gram-negative pathogens 245, 246 
gram-positive pathogens 246 
high-pathogenicity island 237 
PavA, adhesin functions 101 
Peptidases 
aspartic peptidases 1 35-1 37 
classification 1 33-1 35 
cysteine peptidases 

D-alanyl-glycyl endopeptidase 149 

bacteriocin-processing peptidase 148, 
149 

calpain family 142 

caspase family 152 

clan CA peptidases 141 

clan CD peptidases 1 51 

clan CE peptidases 1 55 

clan CF peptidases 157 

clostripain family 151 

gingipain family 152-155 

IdeS peptidase 140, 141 

legumain family 151, 152 

papain faniily 141, 142 

pyroglutamyl-peptidase I family 157 

sortases 138-140 

staphopain family 146-148 

streptopain family 142-146 

Ulpl endopeptidase family 155, 156 

YopJ pepidase family 156,157 

YopT peptidase family 149-151 
gene abundance, bacteria 1 32 
metallopeptidases 

anthrax lethal factor 164 



Subject Index 



276 



BlaRl 164, 165 

camelysin 165 

collagenase 162 

immune inhibitor A family 161 

immunoglobulin-Al -specific 

peptidases 163 
MIO family 162, 163 
sequence conservation 158 
StcE 165 

tentoxilysin 163, )64 
therapeutic targeting 158, 159 
thermolysin family 160, 161 
serine peptidase families, functions 
165-168 
Pertussis toxin 
mechanism of action 37 
structure 37 
PhoP/PhoQ 

CAMP activation 14 
signaling, magnesium-limited 
environments 1 3, 14 
Phospholipases, exotoxins 30,31 
PI A, adhesin functions 104 
Pili 

adhesive functions 67 
Neisseria pathogenesis, role 76, 77 
phase variation of structures 79, 80 
twitching motility, role 78,79 
type IV pili 72,73,76,77 
Pla peptidase, functions 136, 137 
PmrA/PmrB 

CAiMP activation 14 
signaling, lipid A modification 14 
Pore-forming toxins (PFTs) 

cholesterol-binding cytolysins 32 

classification 31 

Clostridium perfringens cutcrotoxiu 

32,33 
RTX toxins 31,32 
Staphylococcus aureus 32 
Prepelin peptidase, functions 135, 

136 
Proteases, see Peptidases 
Protein H, adhesin functions 97 
Pseudomonas aeruginosa 
biofilms 

antibiotic resistance 126,127 



cystic fibrosis chronic lung infections 

123-126 
in vitro studies 122,123 
iron transport related to virulence 219, 
220, 222 
Pseudomonas exotoxin A, elongation factor 

2 inhibition 35 
Pyroglutamyl-peptidase I family, functions 
157 

Raetz pathway, lipid A biosynthesis 7-10, 

20,21 
Re endotoxin, assembly 1 
Rhizobiiim, lipid A features 19, 20 
Rho GT Pases 

activators 

cytotoxic necrotizing factors 45 
SopE 46 

glucosylation 47, 48 

toxins, GAP activity 48 

YopT proteolysis 48 

Salmonella 

iron transport related to vhulence 219 

pathogenicity islands 244, 245 
Salmycins, iron siderophore carriers 226 
SasG, adhesin functions 103 
ScpB, adhesin functions 100 
Secretion pathways 

Sec pathway 183, 184 

TAT pathway 1 84 

type I 188-191 

type II 188, 189, 191, 192 

type 11 [ 188, 189, 192-197 

typelV 188, 189, 197-200 

typeV 188, 189,200,201 
Sllil, adhesijj Functions 95 
Shiga toxin 

ribosomal RMA inactivation 35, 36 

species distribution 35 

structure 35 
Shigella 

horizontal gene transfer 257-259 

iron transport related to virulence 218 

pathogenicity islands 242 

type III secretion 197 
Shi proteins, functions 201 



Subject Index 



277 



Siderophores, antibiotic carriers 

iron catecholate carriers 226, 227 

iron hydroxamate carriers 224—226 

resistance 227 
Signal peptidase II (SpPase II), functions 

135 
Signal recognition particle (SRP), functions 

183 
SNARE, neurotoxin targeting 42, 43 
SopE, mechanism of action 46 
Sortase A (SrtA) 

functions 138, 139, 187 

structure 139, 140 
Sortase B (SrtB) 

functions 139, 187 

structure 139, 140 
SpeB, adhesin functions 99 
SpsA, adhesin functions 100 
SptP, mechanism of action 48 
STa 

guanylate cyclase binding 29, 30 

structure 28, 29 
Staphopain, bacterial functions 146-148 
StaphylococcaJ protein A (Spa), functions 

104, 185, 186 
Staphylococcus aureus 

adhesins 101-104 

iron transport related to virulence 223, 
224 

pathogenicity islands 246 

pore-forming toxins 32 

sortases 138, 139 
STb 

mechanism of action 28,29 

structure 28 
StcE, functions 165 

Streptococcus ugulucliuc, adhesms 96, 99 
Streptococcus pneumoniae, 2idhQs\ns 96, 

100, 101 
Streptococcus pyogenes y adhesins 94-99 
Streptopain, bacterial functions 142-146 
Superantigcns, immune response 33 

TAT system, overview 1 84 

Tentoxilysin, functions 163,164 



Tetanus toxin (TeTx), mechanism of action 

41,42 
TTiermolysin, bacterial functions 160, 161 
Toll-like receptor 4 (TLR4) 

fimbria interactions 82 

lipid A binding 1 

signaling 5-7 
TonB, iron transport, role 214 
TRAF-6, Toll-like receptor signaling 5, 6 
Type III secretion system dependent ADP- 

ribosylating toxins 

mechanism of action 44 

species distribution 44 

Ulpl endopeptidase family, functions 155, 
156 

VacA 

mechanism of action 39,41 

structure 39 
Vibrio cholerae 

iron transport related to vimlence 222, 
223 

pathogenicity islands 245 
Vitronectin, features and adhesin binding 

93 
V8 protease, functions 1 66 
vWbp, adhesin functions 1 04 

Yersinia, invasion strategies 
surface proteins 188-190 
type III secretion 193-196 
YopE 

ftmctions 195, 196 

mechanism of action 49 
YopH, functions 195, 196 
YopJ, funciions 156, 157 
YopM, functions 195, 196 
YopO, functions 195, 196 
YopP, functions 195, 196 
YopQ, functions 195 
YopT 

funcrions 1 49-151, 195, 196 

mechanism of action 48 
Ysc proteins, functions 193,194 



Subject Index 



278