MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS...

13
JOURNAL OF BACTERIOLOGY, 0021-9193/99/$04.0010 Feb. 1999, p. 1059–1071 Vol. 181, No. 4 Copyright © 1999, American Society for Microbiology. All Rights Reserved. MINIREVIEW Bacterial Adhesins: Common Themes and Variations in Architecture and Assembly GABRIEL E. SOTO AND SCOTT J. HULTGREN* Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110 INTRODUCTION Among the earliest events in many bacterial infections are the molecular interactions that occur between the pathogen and host cells. These interactions are typically required for extracellular colonization or internalization to occur and may involve a complex cascade of molecular cross talk at the host- pathogen interface. Colonization of host tissues is usually me- diated by adhesins on the surface of the microbe; the adhesins are responsible for recognizing and binding to specific receptor moieties of host cells. The receptor binding event may activate complex signal transduction cascades in the host cell that can have diverse consequences including the activation of innate host defenses or the subversion of cellular processes facilitat- ing bacterial colonization or invasion. In addition, the binding event may also activate the expression of new genes in the microbe that are important in the pathogenic process. In many instances, adhesins are assembled into hair-like appendages called pili or fimbriae that extend out from the bacterial sur- face. In other cases, the adhesins are directly associated with the microbial cell surface (so-called nonpilus adhesins). Col- lectively, these adhesins and related structures are expressed in organisms associated with a broad range of diseases (Table 1). At least four distinct mechanisms have emerged in recent years to account for the assembly of these diverse organelles: (i) the chaperone-usher pathway, (ii) the general secretion pathway, (iii) the extracellular nucleation-precipitation pathway, and (iv) the alternate chaperone pathway. This list is by no means all-inclusive but rather represents some of the best-character- ized systems to date (for a recent review of other systems that do not utilize these pathways, see reference 51). Molecular blueprints of these pathways will ultimately facilitate the un- derstanding of host-pathogen interactions as well as provide a framework for understanding how complex hetero-oligomeric interactions are orchestrated within the bacterial cell. In this minireview, we focus on the molecular architecture of the adhesive organelles assembled by these four principal path- ways and on the coordinated functions of the proteins that constitute their assembly machineries. MOLECULAR STRUCTURES OF FIMBRIAL ADHESINS We begin by looking at the architectural features of various fimbrial organelles assembled by each of the four general as- sembly pathways. We focus on the best-characterized systems in each pathway as prototypes for each assembly classification: P and type 1 pili (chaperone-usher pathway), type IV pili (general secretion pathway), curli (extracellular nucleation- precipitation pathway) and CS1 pili (alternate chaperone path- way). Note that for the purposes of this minireview, the term subunit will apply to the structural proteins that make up these composite organelles, while the term adhesin will be reserved for those subunits with specific receptor binding properties. P pili and type 1 pili. P pili are expressed on the surfaces of uropathogenic strains of Escherichia coli associated with acute pyelonephritis (63). Eleven genes organized in the pap gene cluster are required for the expression and assembly of these organelles (46, 49, 50, 78). Studies of P pili using quick-freeze, deep-etch electron microscopy have shown that P pili are com- posite fibers consisting of flexible fibrillae joined end to end to pilus rods (67). The tip fibrillae are comprised predominantly of PapE subunits. The rod is composed of repeating PapA subunits packed into a right-handed helical assembly, with an external diameter of 68 Å, an axial hole of 15 Å, and a pitch distance of 24.9 Å, with 3.28 subunits per turn of the helical cylinder (14, 37). The adhesin of P pili, PapG, mediates bind- ing to Gala(1,4)Gal moieties present in the globoseries of glycolipids on uroepithelial cells and erythrocytes (71, 111). The adhesin is located at the distal end of the tip and is joined to the PapE fibrillum via a specialized adapter protein, PapF. Another adapter protein, PapK, joins the adhesin-containing tip to the PapA rod. Another minor component, PapH, is located at the base of the PapA rod; its incorporation into the growing organelle is thought to signal the termination of as- sembly. Type 1 pili are important virulence determinants expressed in E. coli as well as in most members of the Enterobacteriaceae family that mediate binding to mannose-oligosaccharides (66). The expression and assembly of type 1 pili typically require at least nine genes that are present in the type 1 gene cluster (46, 50). Like P pili, type 1 pili are also composite structures in which a short tip fibrillar structure containing FimG and the FimH adhesin (and possibly the minor component FimF as well) are joined to a rod comprised predominantly of FimA subunits (58). The overall structure of the type 1 rod is very similar to that of the PapA rod of P pili. The type 1 subunits are arranged in a helix with an external diameter of 6 to 7 nm and an axial hole of 20 to 25 Å, with a pitch distance of 23.1 Å and 3.125 subunits per turn (13). Type IV pili. Type IV pili have been implicated in a variety of functions, including adhesion to host cell surfaces, twitching motility, modulation of target cell specificity, and bacterio- phage adsorption. They are found on such bacteria as Pseudo- monas aeruginosa, pathogenic Neisseria, Moraxella bovis, Dich- elobacter nodosus, Vibrio cholerae, and enteropathogenic E. coli (EPEC) (113). The role of type IV pili in the virulence of EPEC strains has recently been demonstrated by Bieber and * Corresponding author. Mailing address: Department of Molecular Microbiology, Washington University School of Medicine, Campus Box 8230, 660 South Euclid Ave., St. Louis, MO 63110. Phone: (314) 747-3627. Fax: (314) 362-1998. E-mail: [email protected]. 1059 on November 1, 2020 by guest http://jb.asm.org/ Downloaded from

Transcript of MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS...

Page 1: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

JOURNAL OF BACTERIOLOGY,0021-9193/99/$04.0010

Feb. 1999, p. 1059–1071 Vol. 181, No. 4

Copyright © 1999, American Society for Microbiology. All Rights Reserved.

MINIREVIEW

Bacterial Adhesins: Common Themes and Variations inArchitecture and Assembly

GABRIEL E. SOTO AND SCOTT J. HULTGREN*

Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110

INTRODUCTION

Among the earliest events in many bacterial infections arethe molecular interactions that occur between the pathogenand host cells. These interactions are typically required forextracellular colonization or internalization to occur and mayinvolve a complex cascade of molecular cross talk at the host-pathogen interface. Colonization of host tissues is usually me-diated by adhesins on the surface of the microbe; the adhesinsare responsible for recognizing and binding to specific receptormoieties of host cells. The receptor binding event may activatecomplex signal transduction cascades in the host cell that canhave diverse consequences including the activation of innatehost defenses or the subversion of cellular processes facilitat-ing bacterial colonization or invasion. In addition, the bindingevent may also activate the expression of new genes in themicrobe that are important in the pathogenic process. In manyinstances, adhesins are assembled into hair-like appendagescalled pili or fimbriae that extend out from the bacterial sur-face. In other cases, the adhesins are directly associated withthe microbial cell surface (so-called nonpilus adhesins). Col-lectively, these adhesins and related structures are expressed inorganisms associated with a broad range of diseases (Table 1).At least four distinct mechanisms have emerged in recent yearsto account for the assembly of these diverse organelles: (i) thechaperone-usher pathway, (ii) the general secretion pathway,(iii) the extracellular nucleation-precipitation pathway, and(iv) the alternate chaperone pathway. This list is by no meansall-inclusive but rather represents some of the best-character-ized systems to date (for a recent review of other systems thatdo not utilize these pathways, see reference 51). Molecularblueprints of these pathways will ultimately facilitate the un-derstanding of host-pathogen interactions as well as provide aframework for understanding how complex hetero-oligomericinteractions are orchestrated within the bacterial cell. In thisminireview, we focus on the molecular architecture of theadhesive organelles assembled by these four principal path-ways and on the coordinated functions of the proteins thatconstitute their assembly machineries.

MOLECULAR STRUCTURES OF FIMBRIAL ADHESINS

We begin by looking at the architectural features of variousfimbrial organelles assembled by each of the four general as-sembly pathways. We focus on the best-characterized systemsin each pathway as prototypes for each assembly classification:

P and type 1 pili (chaperone-usher pathway), type IV pili(general secretion pathway), curli (extracellular nucleation-precipitation pathway) and CS1 pili (alternate chaperone path-way). Note that for the purposes of this minireview, the termsubunit will apply to the structural proteins that make up thesecomposite organelles, while the term adhesin will be reservedfor those subunits with specific receptor binding properties.

P pili and type 1 pili. P pili are expressed on the surfaces ofuropathogenic strains of Escherichia coli associated with acutepyelonephritis (63). Eleven genes organized in the pap genecluster are required for the expression and assembly of theseorganelles (46, 49, 50, 78). Studies of P pili using quick-freeze,deep-etch electron microscopy have shown that P pili are com-posite fibers consisting of flexible fibrillae joined end to end topilus rods (67). The tip fibrillae are comprised predominantlyof PapE subunits. The rod is composed of repeating PapAsubunits packed into a right-handed helical assembly, with anexternal diameter of 68 Å, an axial hole of 15 Å, and a pitchdistance of 24.9 Å, with 3.28 subunits per turn of the helicalcylinder (14, 37). The adhesin of P pili, PapG, mediates bind-ing to Gala(1,4)Gal moieties present in the globoseries ofglycolipids on uroepithelial cells and erythrocytes (71, 111).The adhesin is located at the distal end of the tip and is joinedto the PapE fibrillum via a specialized adapter protein, PapF.Another adapter protein, PapK, joins the adhesin-containingtip to the PapA rod. Another minor component, PapH, islocated at the base of the PapA rod; its incorporation into thegrowing organelle is thought to signal the termination of as-sembly.

Type 1 pili are important virulence determinants expressedin E. coli as well as in most members of the Enterobacteriaceaefamily that mediate binding to mannose-oligosaccharides (66).The expression and assembly of type 1 pili typically require atleast nine genes that are present in the type 1 gene cluster (46,50). Like P pili, type 1 pili are also composite structures inwhich a short tip fibrillar structure containing FimG and theFimH adhesin (and possibly the minor component FimF aswell) are joined to a rod comprised predominantly of FimAsubunits (58). The overall structure of the type 1 rod is verysimilar to that of the PapA rod of P pili. The type 1 subunitsare arranged in a helix with an external diameter of 6 to 7 nmand an axial hole of 20 to 25 Å, with a pitch distance of 23.1 Åand 3.125 subunits per turn (13).

Type IV pili. Type IV pili have been implicated in a varietyof functions, including adhesion to host cell surfaces, twitchingmotility, modulation of target cell specificity, and bacterio-phage adsorption. They are found on such bacteria as Pseudo-monas aeruginosa, pathogenic Neisseria, Moraxella bovis, Dich-elobacter nodosus, Vibrio cholerae, and enteropathogenic E. coli(EPEC) (113). The role of type IV pili in the virulence ofEPEC strains has recently been demonstrated by Bieber and

* Corresponding author. Mailing address: Department of MolecularMicrobiology, Washington University School of Medicine, CampusBox 8230, 660 South Euclid Ave., St. Louis, MO 63110. Phone: (314)747-3627. Fax: (314) 362-1998. E-mail: [email protected].

1059

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 2: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

TABLE 1. Adhesive structures on the bacterial cell surface and their assembly pathways

Assembly pathway Structure Assembly geneproducts Organism Disease(s) associated with

pilus expression Reference(s)

Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera

P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50Prs pili PrsD/PrsC E. coli Cystitis 77, 112Type 1 pili FimC/FimD E. coli Cystitis 58, 93, 109

Salmonella sp.Klebsiella pneumoniae

S pili SfaE/SfaF E. coli UTIb 84, 105Newborn meningitis

F1C pili FocC/FocD E. coli Cystitis 96Haemophilus

influenzae fimbriaeHifB/HifC Haemophilus influenzae Otitis media 39, 118

MeningitisH. influenzae

biogroup aegyptiusfimbriae

HafB/HafE H. influenzae Brazilian purpuric fever 95

Type 2 and 3 pili FimB/FimC Bordetella pertussis Whooping cough 75, 85, 121MR/P pili MrpD/MrpC Proteus mirabilis Nosocomial UTI 5PMF pili PmfC/PmfD P. mirabilis Nosocomial UTI 6Long polar fimbriae LpfB/LpfC Salmonella typhimurium Gastroenteritis 9Pef pili PefD/PefC S. typhimurium Gastroenteritis 28Ambient-temperature

fimbriaeAftB/AftC P. mirabilis UTI 79, 80

987P fimbriae FasB/FasD E. coli Diarrhea in piglets 15, 25Thin, flexible piliK99 pili FaeE/FaeD E. coli Neonatal diarrhea in calves,

lambs, and piglets7, 107

K88 pili FanE/FanD E. coli Neonatal diarrhea in piglets 7, 26F17 pili F17D/F17papC E. coli Diarrhea 74MR/K pili MrkB/MrkC K. pneumoniae Pneumonia 3, 34REPEC fimbriae RalE/RalD E. coli Diarrhea in rabbits 1Atypical structures FGL chaperone/ushera

CS31A capsule-likeprotein

ClpE/ClpD E. coli Diarrhea 35

Antigen CS6 CssC/CssD E. coli Diarrhea 65, 122Myf fimbriae MyfB/MyfC Yersinia enterocolitica Enterocolitis 54pH 6 antigen PsaB/PsaC Yersinia pestis Plague 73CS3 pili CS3-1/CS3-2 E. coli Diarrhea 57, 72Envelope antigen F1 Caf1M/Caf1A Y. pestis Plague 32, 64Nonfimbrial

adhesins INfaE/NfaC E. coli UTI 2, 36

Newborn meningitisSEF14 fimbriae SefB/SefC S. enteritidis Gastroenteritis 16, 86Agregative adherence

fimbriae IAggD/AggC E. coli Diarrhea 104

AFA-III AfaB/AfaC E. coli Pyelonephritis 33, 70

Extracellular nucleation-precipitation pathway

Curli CsgG/CsgE/CsgF E. coliS. enteritidis

Sepsis 10, 11, 17, 40, 41,76, 94, 106

General secretionpathway

Type 4 pili General secretionapparatus

Neisseria sp. Gonorrhea

4, 12, 23, 24, 31, 42,43, 44, 62, 83, 89,91, 92, 99, 100,113

P. aeruginosaV. cholerae CholeraM. bovisD. nodosusEikenella corrodens

Bundle-forming pili General secretionapparatus

E. coli Diarrhea 12, 21, 22, 108

Alternate chaperonepathway

CS1 pili CooB/CooC E. coli Diarrhea 102CS2 pili CotB/CotC E. coli Diarrhea 30CS4 pili E. coli Diarrhea 115CS14 pili E. coli Diarrhea 81CS17 pili E. coli Diarrhea 82CS19 pili E. coli Diarrhea 38CFA/I pili CfaA/CfaC E. coli Diarrhea 61Cable type II pili B. cepacia Opportunistic infections in

cystic fibrosis patients101

a The chaperone is given before the slash, and the usher is given after the slash.b UTI, urinary tract infection.

]——

1060 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 3: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

colleagues (12). These structures have a diameter of 60 Å andare typically up to 4,000 nm long, with a pitch distance ofapproximately 40 Å and about five subunits per turn (89). Theyare composed predominantly of identical pilin subunits with anumber of distinctive features, including a short (6 to 7 aminoacids), positively charged leader sequence, a modified aminoacid (N-methylphenylalanine) at the amino terminus of themature pilin, and a highly conserved amino-terminal domain(4, 43, 91, 92). A few other proteins also associate with thesestructures, including in the case of Neisseria, the tip-localizedadhesin (100).

The crystal structure of a type IV pilin subunit (PilE) fromNeisseria gonorrhoeae has recently been determined by Pargeand coworkers at an atomic resolution of 2.6 Å (89). It revealsan a-b roll fold with a rather long hydrophobic N-terminala1-helical spine (residues 2 to 54) that gives the molecule anoverall ladle shape (Fig. 1). Other elements of the structureinclude the following: (i) an extended disaccharide-boundsugar loop (residues 55 to 77), with N-acetylglucosamine-a(1,3)-galactose O linked at position Ser-63, (ii) two b-hairpinsforming a four-stranded antiparallel b-sheet (residues 78 to 93and 103 to 122), (iii) a b2-b3 loop connection (residues 94 to102), and (iv) a disulfide-containing region (residues 121 to158), which despite its hypervariable nature, appears to be aregular b-hairpin (b5-b6) followed by a loop connection. Sys-tematic modeling of the pilin monomer within the constraintsimposed by the available biochemical and biophysical data hasled to a three-layered model of the type IV pilus (27, 89). Theoutermost hypervariable layer in the proposed fiber model iscomprised of residues 123 to 143 and 152 to 158, as well as thedisaccharide at Ser-63, from each monomer. The central layeris a continuous 25-stranded b-sheet, made up of the fourstrands from the antiparallel b-sheet as well as the sugar loopfrom each of the five pilin monomers present in each turn. Theinnermost layer is a parallel coiled-coil made up of the highly

conserved N-terminal a1-helices. A key feature of this model isthat essentially only the hypervariable and sugar-binding do-mains of each pilin monomer are exposed in the final assem-bled pilus structure, which may account for the antigenic vari-ation that these pili undergo.

Curli. Many clinical E. coli and Salmonella enteritidis isolatesproduce a class of thin, irregular, and highly aggregated surfacestructures known as curli (17, 94). These organelles mediatebinding to a variety of host proteins, including fibronectin (94),plasminogen (106), and human contact phase proteins (10).Curli are highly stable structures that require extreme chemicaltreatment (e.g., 90% formic acid) to depolymerize them. Themajor component of E. coli curli is a 15.3-kDa protein termedCsgA, which exhibits more than 86% primary sequence simi-larity to its counterpart in S. enteritidis, AgfA. CsgB is a minorcomponent that may be found associated with the outer mem-brane (OM) or distributed along the length of the curli fiber(11). CsgE, CsgF, and CsgG are required assembly factors thatdo not appear to constitute part of the final curli structure andhence may serve as part of the assembly apparatus (40).

CS1 pili. CS1 pili are found on the surface of EPEC and arethought to be involved in the colonization of the host intestine(102). Other pilus structures in this family include CS2 (30),CS4 (115), CS14 (81), CS17 (82), CS19 (38), CFA/I (61), andthe cable type II pili of the cystic fibrosis-associated pathogenBurkholderia cepacia (101). CS1 pili appear to be composedpredominantly of one component, CooA, with a distally lo-cated minor component, CooD. Electron microscopic exami-nation of these structures reveals that they are morphologicallysimilar to P and type 1 pili, although the structural proteins ofCS1-like pili bear no significant sequence similarity to those ofother pilus systems (102).

ASSEMBLY OF FIMBRIAL ADHESINS

The coordinated assembly of complex hetero-oligomeric or-ganelles poses many special challenges to the bacterial cell,including the correct incorporation of individual subunits in apredefined order during biogenesis and the prevention of pre-mature associations between intrinsically aggregative subunits.Details of how these molecular interactions are orchestratedhave been worked out to various degrees in different systemsand have begun to shed light on the similarities and variationsemployed in diverse bacterial species in the assembly of theseorganelles.

Chaperone-usher pathway. The highly conserved chaper-one-usher pathway is involved in the assembly of more than 25adhesive organelles in gram-negative bacteria. The assemblymachinery is comprised of two specialized classes of proteins,a periplasmic immunoglobulin-like chaperone and an OMusher. The crystal structure of the periplasmic chaperone in-volved in the assembly of P pili, PapD, has been determined toa resolution of 2.0 Å, revealing two immunoglobulin-like do-mains oriented towards one another in such a manner so as togive the molecule an overall boomerang shape (Fig. 2) (45).Hung et al. (52) have shown that the chaperones can be orga-nized into two structurally and functionally distinct subfamilieson the basis of conserved amino acid differences in the chap-erone cleft and the length of the loop that connects the F1 andG1 b-strands of domain 1. The two subfamilies are designatedFGS (for F1-G1 short) and FGL (for F1-G1 long), correspond-ing to loop lengths of #20 amino acids and $21 amino acids,respectively. Interestingly, these two subfamilies assemble piliwith distinct architectures. FGS chaperones, of which PapD isa member, are involved in assembling pili with rod-like archi-tecture. FGL chaperones, on the other hand, mediate the

FIG. 1. Ribbon representation of pilin from N. gonorrhoeae. Colored regionsindicate the secondary structural elements referred to in the text: blue, N-terminal a1-helix (residues 2 to 54); orange, extended disaccharide-bound sugarloop (residues 55 to 77); green, b-hairpins (residues 78 to 93 and 103 to 122);gray, b2-b3 loop (residues 94 to 102); yellow, disulfide bond-containing C-termi-nal region (residues 121 to 158). Also shown are the disulfide bridge (cysteineresidues 121 and 151), signified by a broken line, and Ser-63 with covalentlylinked disaccharide.

VOL. 181, 1999 MINIREVIEW 1061

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 4: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

assembly of very thin or afimbrial adhesive structures on thesurfaces of bacteria.

PapD is known to form periplasmic preassembly complexeswith each of the pilus subunits prior to their incorporation intoa pilus. The relative concentration of each subunit type in theperiplasm is thought to be an important factor in regulating thelength of the tip fibrillum and pilus rod. Overexpression of thePapK adapter, for example, leads to production of pili withshorter tips; similarly, overexpression of PapH leads to short-ened rods (55). Although no crystal structure of any of thesecomplexes has yet been determined, recent studies have iden-tified some of the important determinants for chaperone-sub-unit recognition. Several lines of evidence indicate that chap-erones recognize a highly conserved motif present in the Ctermini of all subunits assembled by PapD-like chaperones(68). This motif is characterized by a series of alternatinghydrophobic residues flanked by a glycine located 14 residuesupstream from the C terminus and by a penultimate tyrosine.Two peptides, corresponding to the C-terminal 19 amino acidsof PapG and PapK, have been cocrystallized with PapD (68,110). Despite significant sequence dissimilarities, both C-ter-minal fragments bound to PapD in a nearly identical mannervia an extended conformation that has been termed a b-zippermotif (68, 110). The results of more recent mutagenesis exper-iments suggested that a highly conserved region found near theN termini of all subunits assembled by PapD-like chaperones isalso recognized by the chaperone (110). This region is alsocharacterized by an alternating pattern of hydrophobic resi-dues, together with a cysteine residue that is involved in anintramolecular disulfide bond. This region is not present at the

N terminus of the PapG adhesin. This is not unexpected, giventhe domain structure of the adhesin. Fimbrial adhesins can bethought of as having a receptor binding domain fused to a pilindomain. In PapG, the receptor binding domain consists of theamino-terminal half of the protein. The C-terminal half of theprotein contains most of the pilin-like features, including theconserved b-zipper motif and two cysteines spaced approxi-mately 40 amino acids apart. Interestingly, the amino-terminalregion of the pilin domain of PapG has recently been shown tocontain a surface that is recognized by the PapD chaperone(110, 123). This region corresponds in approximate sequenceposition (as measured from the COOH terminus) to the highlyconserved N-terminal regions of other pilus subunits.

Initial translocation of P-pilus subunits across the cytoplas-mic membrane occurs via the Sec (general secretion system)machinery, although this pathway itself is not sufficient for theefficient release of subunits into the periplasm (59). Nascentsubunits are retained in the inner cytoplasmic membrane viaan interaction mediated by their hydrophobic C termini. In thepresence of PapD, the subunits are partitioned into theperiplasmic space as chaperone-subunit preassembly com-plexes (Fig. 3A). Based on available crystallographic data, in-variant cleft residues of the chaperone are thought to partici-pate in the b-zippering interaction with a subunit. Mutations inthese invariant cleft residues of the chaperone abolish theability of the chaperone to import subunits and form chaper-one-subunit complexes, underscoring the importance of b-zip-per formation in mediating chaperone function (59, 68, 110).Release of the subunits from the inner membrane is a prereq-uisite for their folding into an assembly-competent conforma-tion, and there is evidence that folding of the subunits with thechaperone serving as a template may occur concomitantly withtheir release from the membrane (59, 110).

Once formed, the chaperone-subunit complexes are tar-geted to the OM PapC usher for assembly. The PapC usherwas purified and shown to form a pore when reconstituted intoliposomes (114). This was confirmed by high-resolution elec-tron microscopy, which showed that PapC assembled into ring-shaped complexes containing central pores of 2 to 3 nm indiameter (114). The PapC complexes consisted of at least sixsubunits. PapC and other usher family members are predictedto have a largely b-sheet secondary structure, typical of bacte-rial OM pore-forming proteins, and likely present large regionsto the periplasm for interaction with chaperone-subunit com-plexes. To facilitate pilus assembly, the usher must be able totranslocate pilus subunits across the OM. The 2-nm-wide lin-ear tip fibrillum would be able to pass through the 2- to 3-nm-diameter usher channel, but the 6.8-nm-wide helical pilus rodwould not be able to fit through the usher. A solution to thisproblem was revealed in experiments that showed that P-pilusrods could be unraveled into linear fibers (114). These unrav-eled rods measure 2 nm in diameter and would therefore benarrow enough to pass through the usher pore. Therefore, itwas proposed that the pilus rod is translocated across the OMin this linear form and adopts its final helical conformationonly upon reaching the external surface. This may be part ofthe mechanism that drives the outward growth of the or-ganelle.

The usher presumably has a more active role in pilus assem-bly than simply functioning as a diffusion pore for the translo-cation of pili across the OM. Dodson et al. (20) showed thatPapC differentially recognized chaperone-subunit complexesdepending upon their final position in the pilus. These studieshave recently been extended by examining the real-time kinet-ics of the interaction of chaperone-subunit complexes with theusher by using surface plasmon resonance technology (103).

FIG. 2. Ribbon representation of the crystal structure of PapD and PapKpeptides. Inset provides a magnified view of the PapD and PapK peptide contactinterface. Note how the conserved alternating hydrophobic residues of the pep-tide interdigitate with the residues along PapD’s G1 b-strand.

1062 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 5: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

FIG. 3. (A) Assembly of P pili from E. coli via the chaperone-usher pathway. Chaperone-mediated extraction of subunits from the inner cytoplasmic membrane(IM) is coupled with their folding into an assembly-competent state. The G1 b-strand of the immunoglobulin-like chaperones, which may serve as a template in thesubunit folding pathway, protects nascently folded subunits from premature oligomerization in the periplasmic space by directly capping the newly formed assemblysurfaces. These interactive surfaces remain protected by the chaperone until delivery of the preassembly complex to the OM assembly site comprised of the usher. PapG(G), PapD (D), PapE (E), PapK (K), PapA (A), and PapH (H) proteins are shown. (B) Assembly of type IV pilus from N. gonorrhoeae via the general secretion pathway.Prepilin is processed by the PilD signal peptidase, which cleaves the positively charged leader sequence from the N terminus of the pilin subunit. The mature PilEsubunit is then assembled by the inner membrane (IM) assembly complex. Translocation of the pilus through the OM is mediated via PilQ, possible with the assistanceof other factors such as PilP. The PilC adhesin, which is thought to ultimately be incorporated at the tip of the growing organelle, also appears to be required fortranslocation through the OM. C, C terminus. (C) Assembly of curli from E. coli via the extracellular nucleation-precipitation pathway. CsgA, the main component ofcurli, is secreted across the OM. Surface-localized CsgB serves to nucleate CsgA assembly. CsgB is also found distributed along the curli fiber, where it may serve toinitiate branching of the fiber. CsgG (G) is an OM-localized lipoprotein that is required for the secretion of CsgA and CsgB, although its function is not known at thistime. (D) Assembly of CS1 pili from E. coli via the alternate chaperone pathway. The CooB (B) chaperone forms periplasmic complexes with the main componentsof the pilus, CooA (A) and CooD (D). It also appears to bind and perhaps stabilize the OM protein CooC (C) in the absence of subunits. CooC may function as anOM channel for passage of the pilin fiber.

VOL. 181, 1999 MINIREVIEW 1063

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 6: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

1064 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 7: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

FIG

.4.

Alig

nmen

tofs

ubun

itsas

sem

bled

byF

GS

chap

eron

es.A

min

oac

idse

quen

ceal

ignm

ento

fstr

uctu

rals

ubun

itsas

sem

bled

bym

embe

rsof

the

FG

Ssu

bfam

ilyof

imm

unog

lobu

lin-li

kech

aper

ones

.Se

quen

ces

have

been

grou

ped

into

thre

ecl

asse

s,ba

sed

upon

whe

ther

they

repr

esen

ta

maj

oror

min

orsu

buni

tan

don

the

mor

phol

ogy

ofth

eas

sem

bled

stru

ctur

es(i

.e.,

thic

kro

dsve

rsus

thin

fimbr

illae

).O

nly

thos

ere

sidu

esth

atar

eco

nser

ved

inat

leas

t90

%of

the

sequ

ence

sw

ithin

acl

ass

orac

ross

clas

ses

are

shad

edan

dco

ded

with

the

follo

win

gco

lors

:pin

k,in

vari

ant;

yello

w,c

onse

rved

hydr

opho

bic

(A,L

,V,I

,P,M

,W,F

,C,Y

,and

G);

purp

le,c

onse

rved

pola

ran

dch

arge

d(N

,Q,S

,T,H

,D,E

,K,a

ndR

).R

esid

ues

mar

ked

byan

aste

risk

appe

arto

beco

nser

ved

with

ina

give

ncl

ass

but

not

acro

sscl

asse

s.

VOL. 181, 1999 MINIREVIEW 1065

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 8: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

Chaperone-adhesin complexes from both the P and type 1pilus systems were found to bind tightest and fastest to theirrespective ushers, suggesting that kinetic partitioning of chap-erone-adhesin complexes to the usher is a defining factor in thetip localization of the adhesin. In addition, dissociation ratesfor all of the chaperone-subunit complexes from the usherwere found to be slow, arguing that after association of acomplex with the usher, the subunit is destined for assemblyinto the pilus. A stable usher-chaperone-adhesin complex waspurified from bacteria expressing FimD (type 1 usher), FimC(type 1 chaperone), and FimH (type 1 adhesin) (103). Expres-sion of other combinations of chaperone-subunit complexeswith the usher did not result in formation of a stable ternarycomplex. Formation of the FimDCH complex led to protectionof the usher from degradation by trypsin in vivo, apparentlydue to a conformational change in the usher (103). This con-formational change was maintained during pilus assembly, sug-gesting that interaction of FimCH with FimD stabilizes theusher in an assembly-competent conformation. Since a FimH2

strain is nonpiliated, these data argue that interaction of thechaperone-adhesin complex with the usher is critical to initiatepilus biogenesis, as has been observed in several clinical strains(103).

In addition to preferential interactions of different chaper-one-subunit complexes with the usher, another factor thatplays a key role in dictating the relative order of subunit in-corporation into the growing organelle is subunit-subunit sur-face complementarity. Jacob-Dubuisson et al. (55) demon-strated that the PapF and PapK adapter proteins wererequired for the efficient initiation of tip fibrillae and pilusrods, respectively. Deletion of both the papF and papK genesabolished piliation altogether, suggesting that other pilus sub-units do not possess the structural determinants necessary toinitiate the formation of tip fibrillae and pilus rods. The highlyconserved N- and C-terminal regions of pilus subunits haverecently been identified as serving as the primary assemblysurfaces that mediate subunit-subunit interactions in the qua-ternary structure of the mature pilus. Subtle differences inthese primary assembly regions from one subunit to anothermay be responsible for controlling the order of incorporationof pilus subunits (110).

Additional insight can be gained by grouping the sequencesof individual subunits assembled by FGS chaperones accordingto their known structural roles (Fig. 4). According to thisscheme, there are three basic classes of subunits. Class I sub-units are the major subunits of thick rod-like assemblies. ClassII subunits are minor components of pili, including those thatfunction as adapters and those that assemble into open helicalfibers. Class III subunits are similar to class I subunits in thatthey comprise the major subunit of the respective fiber butdiffer from class I subunits in that they typically assemble intostructures with thin, flexible morphologies.

An alignment of subunits assembled by FGS chaperonesrevealed seven homology regions (HR) consisting of distinctpatterns of residues conserved among all subunits. We furtherdivided class II subunits into two subclasses (II-A and II-B)based upon distinct patterns of conservation, as described be-low. The most extensively conserved regions are the aforemen-tioned N- and C-terminal regions (designated HR-1 and HR-7,respectively). The region (HR-3) near the second cysteine res-idue, which is linked to the cysteine in HR-1 by disulfide bonds,is also highly conserved across all subunits, as is a region nearthe center of all the sequences examined (HR-5). However,there are notable differences between corresponding regionsamong the different classes of subunits. For example, the HR-1motif of class I subunits begins with a highly conserved glycine

that is absent in most class II and class III subunits. Class II-Asubunits appear to be closely related to class I subunits, withonly subtle differences in the HR-1, -2, -4, and -5 motifs. Incontrast, class II-B subunits have characteristic vicinal prolinesimmediately preceding the first cysteine in HR-1, as well asdistinct HR-2, -3, -5, and -6 motifs. The PapF adapter proteinof P pili falls within this class, whereas other minor compo-nents of P pili fall within the class II-A grouping. These dif-ferences may be a determinant in allowing PapF to function asan adapter between the PapG adhesin and the PapE tip fibril-lum-forming subunit. It is intriguing to speculate that theseconserved regions are important structural determinants thatmay dictate subunit function in pilus assembly and that differ-ences in these regions may account in part or in whole fordifferences in function. HR-1 and HR-7 have already beenshown to play a role in mediating subunit-subunit interactionsin the mature pilus (110). HR-3 may also make an importantcontribution to the assembly surface formed by HR-1, as theposition of the conserved alternating pattern of hydrophobicresidues relative to the cysteines suggests these two regionscould be two adjacent parallel b-strands within a sheet. It isinteresting to note that there is no difference in the HR-7 motifamong classes. This likely reflects the importance of this regionas a common recognition motif for the periplasmic chaperones.

General secretion pathway. The formation of type 4 pilirequires the expression of several proteins that are involved inthe assembly of these structures, including the following: (i) aprepilin peptidase that cleaves a short leader peptide from thesubunits; (ii) an integral membrane protein located in theinner cytoplasmic membrane that may serve as a platform forfimbrial assembly; (iii) a hydrophilic nucleotide-binding pro-tein located in the cytoplasm or associated with the cytoplasmicface of the inner membrane that may energize secretion byATP hydrolysis; and (iv) an OM component that forms a chan-nel allowing the translocation of assembled pili through theOM (4). Donnenberg and colleagues (22) have recently iden-tified a total of 14 genes that are sufficient for the biogenesis oftype IV pili in a heterologous E. coli host, including theperiplasmic disulfide-bond oxidoreductase DsbA. This assem-bly system appears to function independently of any chaperoneactivity, which likely reflects the localization of the assemblyplatform within the inner membrane and the absence of a needto transport pilin monomers across the periplasm in a solubleform. It is noteworthy that a number of pilin-like proteins,possessing similar leader peptides and hydrophobic amino ter-mini, are thought to assemble into a pilus-like secretion tubeused in the secretion of a variety of proteases, toxins, and otherextracellular factors across the OM (91, 92). This export ma-chinery is known as the general secretion apparatus and hasbeen shown to be dependent on some of the gene productsinvolved in the assembly of type 4 pili (e.g., signal peptidase),hence, the grouping of the type 4 assembly machinery as partof the general secretion pathway. Our knowledge of type IVpilus biogenesis remains incomplete, but work from severalgroups has laid a solid foundation for understanding how thesecomplex organelles are assembled. We herein present an over-view of our current understanding of type IV pilus biogenesisin N. gonorrheae (Fig. 3B). For discussions describing the as-sembly of these organelles in P. aeruginosa, we refer the readerto the recent reviews by Alm and Mattick (4) and Hahn (42).

Following translocation of the pre-PilE precursor subunitsinto the periplasmic compartment by the general secretionapparatus, these molecules are retained in the inner mem-brane by their hydrophobic N-terminal segments, with theirhydrophilic C-terminal domains oriented towards the peri-plasm (31, 83). The PilD signal peptidase removes the posi-

1066 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 9: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

tively charged leader sequence from the cytoplasmic side of theprepilin to generate mature PilE, which can then undergoassembly as subunits associate with their hydrophobic stems.PilF, PilG, and PilT are among the factors required for thisassembly, although their functions are not well understood. Ithas been suggested based on studies of its homologues thatPilF may function as an ATPase or kinase (116, 117). PilG hasbeen proposed to play a role in the optimal localization orstabilization of PilD and or PilF (116). PilT is a putative nu-cleotide-binding protein that has been postulated to play a rolein twitching motility and pilus retraction (43).

The assembled pili are thought to be translocated across theOM by a gated pore formed by a multimeric form of PilQ (23).A lipoprotein, PilP, appears to function in stabilizing the ex-pression of PilQ as a multimer (24). The PilC adhesin appearsto facilitate passage of the growing organelle through this pore,although the molecular basis for the role of PilC in this processis not well understood (60, 88, 99).

As our understanding of type IV pilus biogenesis continuesto expand, the importance of other players in the assembly andregulation of these organelles will no doubt become apparent.Recent work by Kaiser and colleagues with Myxococcus xan-thus underscores the many subtle factors that influence type IVpilus assembly and function. M. xanthus has two genetic sys-tems, called adventurous (A) and social (S) motility, whichcontrol its gliding motility and swarming behavior, respectively(44). S motility is dependent upon type IV pili, as mutantslacking pili do not display this type of motility (62). It has beenshown that strains with mutations in a particular S motilitygene called tgl (for transient gliding) lack S motility and typeIV pili but that these qualities can be transiently restored bycontact with tgl1 (donor) cells in a process called stimulation(44, 62). Stimulation does not involve a diffusable factor butrather depends upon physical contact between cells. Further-more, stimulation is transient and occurs only phenotypically,as the offspring of stimulated cells remain S2 and lack pili. Thetgl gene product is a putative lipoprotein that appears to belocalized to the periplasm, probably attached to the outermembrane (97, 98), and contains multiple tetratrico peptiderepeat domains that are thought to be important in protein-protein interactions (69). At present, the exact mechanism ofTgl action is unknown. The identification of the proteins thatinteract with Tgl will certainly shed light on the process ofstimulation and how this protein modulates the assembly oftype IV pili.

Extracellular nucleation-precipitation pathway. The forma-tion of curli represents a departure from the chaperone-usherpathway and the general assembly pathway typified by P andtype 4 pili, respectively. Whereas those structures undergoassembly from the base (i.e., the distal end containing theadhesin is assembled first), curli formation occurs from theoutside of the microbe by the precipitation of secreted solublesubunits into thin fibers on the surface of the microbe (41). InE. coli, the products of two divergently transcribed operons arerequired for curli assembly (40). The csgBA operon encodesthe principal fiber-forming subunit, CsgA, which is secreteddirectly into the extracellular milieu as a soluble protein. It alsoencodes CsgB, which is proposed to be a nucleator that inducespolymerization of CsgA on the cell surface (Fig. 3C) (11). Insupport of this model, it has been demonstrated that a CsgA1

CsgB2 donor strain can secrete CsgA subunits that can beassembled into curli on the surface of a CsgA2 CsgB1 recip-ient strain (41). Furthermore, CsgB appears to be distributedalong the length of the curli fiber, where it has been suggestedto be able to initiate branching of the fibrillar structure (11).Interestingly, in the absence of CsgA, overexpressed CsgB

appears to be able to form short polymers on the bacterial cellsurface (11).

The csgDEFG operon encodes a transcriptional activator forcurli production (CsgD) and three putative assembly factors(40). One of these factors, CsgG, has recently been shown to bea lipoprotein that is localized to the OM (76). In its absence,curli assembly does not take place and it appears that CsgAand CsgB are subjected to rapid proteolytic degradation. Theprecise role of CsgG is not known at this time. Loferer andcolleagues (76) have proposed that CsgG might be a chaper-one that works in concert with another, as yet unidentified OMtranslocator to export CsgA and CsgB and protect these sub-units from premature degradation. Alternatively, a multimericform of CsgG itself may function as a Csg-specific channelwithin the OM. The roles of CsgE and CsgF have not beenestablished at this time; however, it has been reported that astrain deficient in these two assembly factors can export as-sembly-competent CsgA, suggesting that expression of CsgG issufficient for production and assembly of CsgA (76).

Alternate chaperone pathway. The operons for CS1 and therelated CS2 and CFA/I fimbrial structures are each composedof four functional genes (29, 30, 61). The pathway for theassembly of these structures also employs a specialized set ofperiplasmic chaperones that appear to be distinct from thoseof the chaperone-usher pathway; hence we term the mode ofassembly for these organelles the alternate chaperone path-way. In the case of CS1 pili, the chaperone CooB has beenshown to form periplasmic complexes with the pilin compo-nents CooA and CooD, which are transported into theperiplasm in a Sec-dependent manner (Fig. 3D). The formerserves as the major pilin component, while the latter is a minorcomponent that appears to be tip localized and may serve toinitiate the assembly of CooA (102). Both subunits appear toshare a conserved sequence motif near their C termini, whichmay function as a chaperone recognition motif (102, 120).Note that this motif shares no homology with the conservedC-terminal motif of alternating hydrophobic residues found insubunits assembled by the chaperone-usher pathway.

CooC is an OM protein that may function as an OM channelfor passage of the pilin fiber. Interestingly, CooB also appearsto stabilize CooC in the OM and is able to bind CooC in theabsence of the other pilin subunits (120). Despite apparentfunctional similarities, CS1 and related structures do not ap-pear to be related to those assembled by the classic chaperone-usher pathway, suggesting that these two systems arose inde-pendently through convergent evolution.

LINKS BETWEEN PILUS BIOGENESIS ANDHOST PATHOGENESIS

Colonization is not a single event but rather a dynamicprocess that involves a panoply of changes in both the bacte-rium and host alike as a result of attachment. Mulvey andcolleagues (87) have recently used scanning and high-resolu-tion transmission electron microscopy in a murine cystitismodel to investigate the structural basis and consequences ofin vivo interactions between type 1-piliated E. coli and hostsuperficial bladder cells. These studies revealed that type 1pilus tips interacted directly with a class of integral membraneglycoproteins known as uroplakins that are situated on theluminal surface of the bladder epithelial cells (Fig. 5). At-tached pili were shortened to an average length of 0.12 6 0.01mm. In contrast, type 1 pili present on bacteria in broth cultureare typically 1 to 2 mm long (13). The mechanism by which thisapparent shortening occurs remains to be elucidated, but re-traction of the pilus upon attachment has been suggested as

VOL. 181, 1999 MINIREVIEW 1067

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 10: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

one possible means (87). Alternatively, contact of the type 1pilus tips with the host epithelium could impede the growth ofnascent pili (87). Either pilus retraction or a hindrance of pilusgrowth mechanism would likely result in a buildup of unas-sembled pilin subunits in the periplasm.

The consequences of such a buildup can be inferred fromstudies showing that the expression of pilus subunits in theabsence of the chaperone is toxic in E. coli strains lacking theDegP periplasmic protease (59). Toxicity presumably resultsfrom the formation of subunit aggregates in the periplasm thatthe DegP protease normally breaks down. By using lacZ fu-sions to degP and cpxP, it was demonstrated that expression ofsubunits in the absence of PapD activates the CpxA-CpxRtwo-component system in which CpxA is the membrane-boundsensor/kinase and CpxR is the DNA binding response regula-tor (18, 19, 59, 90). This pathway up-regulates degP transcrip-tion as well as a number of other chaperone-like proteins, suchas the disulfide isomerase DsbA and cis-trans prolyl isomer-ases. These factors facilitate subunit folding: DsbA is requiredfor pilus biogenesis (56). These studies suggested that Cpxmonitors pilus biogenesis and responds by controlling the ex-pression of factors that facilitate pilus biogenesis. It is intrigu-ing to speculate that activation of the CpxA-CpxR pathway inresponse to pilus-mediated attachment leads to the expressionof an array of virulence genes necessary for establishing aninfection. Hung and colleagues (53) refer to this state as theattached phenotype.

PERSPECTIVE AND FUTURE DIRECTIONS

Bacteria have developed a number of distinct mechanismsfor the assembly of a diverse range of adhesive organelles.

Despite the variations, several common themes do emergefrom the study of these assembly pathways. For example, theinner membrane appears to have the capacity to function as atemporary reservoir for nascently translocated subunits assem-bled by the chaperone-usher pathway and the general secretionpathway. Those pathways that require subunits to be trans-ported through the periplasm prior to their assembly appear torequire the function of a periplasmic chaperone to preventpremature subunit oligomerization. In the case of the chaper-one-usher and alternate chaperone pathways, the correspond-ing chaperones appear to interact with their target proteinseither immediately or shortly following subunit translocationinto the periplasm. In the case of the extracellular nucleation-precipitation pathway, an OM-localized protein may functionas a chaperone and/or usher to transform curlin monomersinto an assembly-competent conformation just before theirexport, thereby perhaps minimizing the chances for prematureassociations in the periplasmic compartment. In contrast, mo-lecular chaperones appear to be absent in the general secretionpathway, as the pilin subunits assembled by this route areassembled directly on the inner membrane. For all four path-ways, it appears that specific OM channels are involved in theexport of pilin subunits either in an assembled or nonas-sembled state.

Understanding the molecular events involved in the biogen-esis of these organelles will be crucial for the development ofnovel therapeutic strategies. Elucidating common themes inthese pathways will be a prerequisite for any efforts targetedtowards developing a therapeutic strategy with broad-spectrumactivity. The identification of those processes that occur fol-lowing attachment will undoubtedly open up further avenues

FIG. 5. Type 1 pilus-mediated bacterial adherence to the mouse bladder epithelium at 2 h postinfection. (A) Scanning electron micrograph (EM) of bacteria onsurface of bladder epithelial cells. The bacteria often appear to be situated in grooves and niches formed by the apical membrane of the superficial cells. (B) ScanningEM of a bacterium being enveloped by the membrane of bladder epithelial cells. (C) High-resolution, freeze-fracture, deep-etch EM of infected bladder epithelia,showing a centrally located bacterium making intimate contact with the luminal surface of the epithelia. Type 1 pili can be seen radiating out from the organisms andspanning the distance between the outer membrane and host cell surface. The FimH adhesin at the tips of these pili mediates contact with the hexagonal uroplakinplaques embedded in the epithelial cell membrane.

1068 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 11: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

of therapeutic possibilities, as we come closer to understandinghow host-pathogen interactions lead to the expression of bac-terial genes that are important in pathogenesis.

ACKNOWLEDGMENTS

We gratefully acknowledge our ongoing collaboration with the lab ofT. Silhavy. It is largely through their enthusiastic sharing of results andideas that our labs jointly formed the current model concerning therole of Cpx in pilus biogenesis. We thank M. Mulvey for kindly pro-viding us with electron micrographs of bacteria expressing type 1 piliinteracting with mouse bladder epithelial cells.

Some of the work described was supported by National Institutes ofHealth grants R01AI29549 and R01DK51406.

REFERENCES

1. Adams, L. M., C. P. Simmons, L. Rezmann, R. A. Strugnell, and R. M.Robins-Browne. 1997. Identification and characterization of a K88- andCS31A-like operon of a rabbit enteropathogenic Escherichia coli strainwhich encodes fimbriae involved in the colonization of rabbit intestine.Infect. Immun. 65:5222–5230.

2. Ahrens, R., M. Ott, A. Ritter, H. Hoschutzkky, T. Buhler, F. Lottspeich,G. J. Boulnois, K. Jann, and J. Hacker. 1993. Genetic analysis of the genecluster encoding nonfimbrial adhesin I from an Escherichia coli uropatho-gen. Infect. Immun. 61:2505–2512.

3. Allen, B. L., G. F. Gerlach, and S. Clegg. 1991. Nucleotide sequence andfunctions of mrk determinants necessary for expression of type 3 fimbriae inKlebsiella pneumoniae. J. Bacteriol. 173:916–920.

4. Alm, R. A., and J. S. Mattick. 1997. Genes involved in the biogenesis andfunction of type-4 fimbriae in Pseudomonas aeruginosa. Gene 192:89–98.

5. Bahrani, F. K., D. E. Johnson, D. Robbins, and H. L. T. Mobley. 1991.Proteus mirabilis flagella and MR/P fimbriae: isolation, purification, N-terminal analysis, and serum antibody response following experimentalurinary tract infection. Infect. Immun. 59:3574–3580.

6. Bahrani, F. K., S. Cook, R. Hull, G. Massad, and H. L. T. Mobley. 1993.Proteus mirabilis fimbriae: N-terminal amino acid sequence of a majorfimbrial subunit and nucleotide sequences of the genes from two strains.Infect. Immun. 61:884–891.

7. Bakker, D., C. E. Vader, B. Roosendaal, F. R. Mooi, B. Oudega, and F. K.de Graff. 1991. Structure and function of periplasmic chaperone-like pro-teins involved in the biosynthesis of K88 and K99 fimbriae in enterotoxi-genic Escherichia coli. Mol. Microbiol. 5:875–886.

8. Balley, M., A. Filloux, M. Akrim, G. Ball, A. Lazdunski, and J. Tommassen.1992. Protein secretion in Pseudomonas aeruginosa: characterization ofseven xcp genes and processing of secretory apparatus by prepilin pepti-dase. Mol. Microbiol. 6:1121–1131.

9. Baumler, A. J., and F. Heffron. 1995. Identification and sequence analysis oflpfABCDE, a putative fimbrial operon of Salmonella typhimurium. J. Bac-teriol. 177:2087–2097.

10. Ben Nasr, A., A. Olsen, U. Sjobring, W. Muller-Esterl, and L. Bjork. 1996.Assembly of human contact phase proteins and release of bradykinin at thesurface of curli-expressing Escherichia coli. Mol. Microbiol. 20:927–935.

11. Bian, Z., and S. Normark. 1997. Nucleator function of CsgB for the assem-bly of adhesive surface organelles in Escherichia coli. EMBO J. 16:5827–5836.

12. Bieber, D., S. W. Ramer, C.-Y. Wu, W. J. Murray, T. Tobe, R. Fernandez,and G. K. Schoolnik. 1998. Type IV pili, transient bacterial aggregates, andvirulence of enteropathogenic Escherichia coli. Science 280:2114–2118.

13. Brinton, C. C., Jr. 1965. The structure, function, synthesis, and geneticcontrol of bacterial pili and a model for DNA and RNA transport in gramnegative bacteria. Trans. N. Y. Acad. Sci. 27:1003–1065.

14. Bullitt, E., and L. Makowski. 1995. Structural polymorphism of bacterialadhesion pili. Nature 373:164–167.

15. Cao, J., A. S. Chan, M. E. Bayer, and D. M. Schifferli. 1995. Orderedtranslocation of 987P fimbrial subunits through the outer membrane ofEscherichia coli. J. Bacteriol. 177:3704–3713.

16. Clouthier, S. C., K. H. Muller, J. L. Doran, S. K. Collinson, and W. W. Kay.1993. Characterization of three fimbrial genes, sefABC, of Salmonella en-teriditis. J. Bacteriol. 175:2523–2533.

17. Collinson, S. K., L. Emody, K.-H. Muller, T. J. Trust, and W. W. Kay. 1991.Purification and characterization of thin, aggregative fimbriae from Salmo-nella enteritidis. J. Bacteriol. 173:4773–4781.

18. Danese, P. N., and T. J. Silhavy. 1997. The sE and the Cpx signal trans-duction systems control the synthesis of periplasmic protein-folding systemsin Escherichia coli. Genes Dev. 11:1183–1193.

19. De Las Penas, A., L. Connolly, and C. A. Gross. 1997. The sE-mediatedresponse to extracytoplasmic stress in Escherichia coli is transduced byRseA and RseB, two negative regulators of sE. Mol. Microbiol. 24:373–385.

20. Dodson, K. W., F. Jacob-Dubuisson, R. T. Striker, and S. J. Hultgren. 1993.Outer-membrane PapC molecular usher discriminately recognizes periplas-

mic chaperone-pilus subunit complexes. Proc. Natl. Acad. Sci. USA 90:3670–3674.

21. Donnenberg, M., J. Giron, J. Nataro, and J. Kaper. 1992. A plasmidencoded type IV fimbrial gene of enteropathogenic Escherichia coli asso-ciated with localized adherence. Mol. Microbiol. 6:3427–3437.

22. Donnenberg, M. S., H.-Z. Zhang, and K. D. Stone. 1997. Biogenesis of thebundle-forming pilus of enteropathogenic Escherichia coli: reconstitution offimbriae in recombinant E. coli and role of DsbA in pilin stability. Gene192:33–38.

23. Drake, S. L., and M. Koomey. 1995. The product of the pilQ gene isessential for the biogenesis of type IV pili in Neisseria gonorrhoeae. Mol.Microbiol. 18:975–986.

24. Drake, S. L., S. A. Sandstedt, and M. Koomey. 1997. PilP, a pilus biogenesislipoprotein in Neisseria gonorrhoeae, affects expression of PilQ as a high-molecular weight multimer. Mol. Microbiol. 23:657–668.

25. Edwards, R. A., J. Cao, and D. M. Schifferli. 1996. Identification of majorand minor chaperone proteins involved in the export of 987P fimbriae. J.Bacteriol. 178:3426–3433.

26. Erickson, A. K., J. A. Willgohs, S. Y. McFarland, D. A. Benfield, and D. H.Francis. 1992. Identification of two porcine brush border glycoproteins thatbind the K88ac adhesin of Escherichia coli and correlation of these glyco-proteins with the adhesive phenotype. Infect. Immun. 60:983–988.

27. Forest, K. T., and J. A. Tainer. 1997. Type-4 pilus-structure: outside toinside and top to bottom. Gene 192:165–169.

28. Friedich, M. J., N. E. Kinsey, J. Vila, and R. J. Kadner. 1993. Nucleotidesequence of a 13.9 kb segment of the 90 kb virulence plasmid of Salmonellatyphimurium: the presence of a fimbrial biosynthetic gene. Mol. Microbiol.8:543–558.

29. Froehlich, B. J., A. Karakashian, L. R. Melsen, J. C. Wakefield, and J. R.Scott. 1994. CooC and CooD are required for assembly of CS1 pili. Mol.Microbiol. 12:387–401.

30. Froehlich, B. J., A. Karakashian, H. Sakellaris, and J. R. Scott. 1995.Genes for CS2 pili of enterotoxigenic Escherichia coli and their inter-changeability with those for CS1 pili. Infect. Immun. 63:4849–4856.

31. Fussenegger, M., T. Rudel, R. Barten, R. Ryll, and T. F. Meyer. 1997.Transformation competence and type-4 pilus biogenesis in Neisseria gon-orrhoeae. Gene 192:125–134.

32. Galyov, E. E., A. V. Karlishev, T. V. Chernovskaya, D. A. Dolgikh, O. Y.Smirnov, K. I. Volkovoy, V. M. Abramov, and V. P. Zav*yalov. 1991. Ex-pression of the envelope antigen F1 of Yersinia pestis is mediated by theproduct of cal1M gene having homology with the chaperone protein PapDof Escherichia coli. FEBS Lett. 286:79–82.

33. Garcia, M. I., A. Labigne, and C. LeBouguenec. 1994. Nucleotide sequenceof the afimbrial-adhesin-encoding afa-3 gene cluster and its translocationvia flanking IS1 insertion sequences. J. Bacteriol. 176:7601–7613.

34. Gerlach, G. F., B. L. Allen, and S. Clegg. 1988. Molecular characterizationof the type 3 (MR/K) fimbriae of Klebsiella pneumoniae. J. Bacteriol. 170:3547–3553.

35. Girdeau, J. P., M. D. Vartanian, J. L. Ollier, and M. Contrepois. 1988.CS31A, a new K88-related fimbrial antigen on bovine enteropathogenicand septicemic Escherichia coli strains. Infect. Immun. 56:2180–2188.

36. Goldhar, J., R. Perry, J. R. Golecki, H. Hoschutzky, B. Jann, and K. Jann.1987. Nonfimbrial, mannose-resistant adhesins from uropathogenic Esche-richia coli O83:K1:H4 and O14:K?:H11. Infect. Immun. 55:1837–1842.

37. Gong, M., and L. Makowski. 1992. Helical structure of P pili from Esche-richia coli. J. Mol. Biol. 228:735–742.

38. Grewal, H. M., H. Valvatne, M. K. Bhan, L. van Dijk, W. Gaastra, and H.Sommerfelt. 1997. A new putative fimbrial colonization factor, CS19, ofhuman enterotoxigenic Escherichia coli. Infect. Immun. 65:507–513.

39. Guerina, N. G., S. Langerman, H. W. Clegg, T. W. Kessler, D. A. Goldman,and J. R. Gilsdorf. 1982. Adherence of piliated Haemophilus influenzae tohuman oropharyngeal cells. J. Infect. Dis. 146:564.

40. Hammar, M., A. Arnqvist, Z. Bian, A. Olsen, and S. Normark. 1995.Expression of two csg operons is required for production of fibronectin- andcongo red-binding curli polymers in Escherichia coli K-12. Mol. Microbiol.18:661–670.

41. Hammar, M., Z. Bian, and S. Normark. 1996. Nucleator-dependent inter-cellular assembly of adhesive curli organelles in Escherichia coli. Proc. Natl.Acad. Sci. USA 93:6562–6566.

42. Hahn, H. P. 1997. The type-4 pilus is the major virulence-associated adhesinof Pseudomonas aeruginosa. Gene 192:99–108.

43. Hobbs, M., and J. S. Mattick. 1993. Common components in the assemblyof type 4 fimbriae, DNA transfer systems, filamentous phage and protein-secretion apparatus: a general system for the formation of surface-associ-ated protein complexes. Mol. Microbiol. 1:233–243.

44. Hodgkin, J., and D. Kaiser. 1979. Genetics of gliding motility in Myxococcusxanthus (Myxobacterales): two gene systems control movement. Mol. Gen.Genet. 171:177–191.

45. Holmgren, A., and C. I. Branden. 1989. Crystal structure of chaperoneprotein PapD reveals an immunoglobulin fold. Nature 342:248–251.

46. Hull, R. A., R. E. Gill, P. Hsu, B. H. Minshaw, and S. Falkow. 1981.Construction and expression of recombinant plasmids encoding type 1 and

VOL. 181, 1999 MINIREVIEW 1069

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 12: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

D-mannose-resistant pili from a urinary tract infection Escherichia coli iso-late. Infect. Immun. 33:933–938.

47. Hull, R. A., S. I. Hull, and S. Falkow. 1984. Frequency of gene sequencesnecessary for pyelonephritis-associated pili expression among isolates ofEnterobacteriaceae from human extraintestinal infections. Infect. Immun.43:1064–1067.

48. Hull, R. A., and S. I. Hull. 1994. Adherence mechanisms in urinary tractinfections, p. 79–90. In V. L. Miller, J. B. Kaper, D. A. Portnoy, and R. R.Isberg (ed.), Molecular genetics of bacterial pathogenesis. ASM Press,Washington, D.C.

49. Hultgren, S. J., and S. Normark. 1991. Biogenesis of the bacterial pilus.Curr. Opin. Genet. Dev. 1:313–318.

50. Hultgren, S. J., S. Normark, and S. N. Abraham. 1991. Chaperone-assistedassembly and molecular architecture of adhesive pili. Annu. Rev. Micro-biol. 45:383–415.

51. Hultgren, S. J., C. H. Jones, and S. Normark. 1996. Bacterial adhesins andtheir assembly, p. 2730–2756. In F. C. Neidhardt, R. Curtiss III, J. L.Ingraham, E. C. C. Lin, K. B. Low, B. Magasanik, W. S. Reznikoff, M.Riley, M. Schaechter, and H. E. Umbarger (ed.), Escherichia coli andSalmonella: cellular and molecular biology. ASM Press, Washington, D.C.

52. Hung, D. L., S. D. Knight, R. M. Woods, J. S. Pinkner, and S. J. Hultgren.1996. Molecular basis of two subfamilies of immunoglobulin-like chaper-ones. EMBO J. 15:3792–3805.

53. Hung, D. L., T. L. Raivio, C. H. Jones, T. J. Silhavy, and S. J. Hultgren.Unpublished data.

54. Iriarte, M., C. Vanooteghem, I. Delor, R. Diaz, S. Knutton, and G. R.Cornelis. 1993. The Myf fibrillae of Yersinia enterocolitica. Mol. Microbiol.9:507–520.

55. Jacob-Dubuisson, F., J. Heuser, K. Dodson, S. Normark, and S. Hultgren.1993. Initiation of assembly and association of the structural elements of abacterial pilus depend on two specialized tip proteins. EMBO J. 12:837–847.

56. Jacob-Dubuisson, F., J. Pinkner, Z. Xu, R. Striker, A. Padmanhaban, andS. J. Hultgren. 1994. PapD chaperone function in pilus biogenesis dependson oxidant and chaperone-like activities of DsbA. Proc. Natl. Acad. Sci.USA 91:11552–11556.

57. Jalajakumari, M. B., C. J. Thomas, R. Halter, and P. A. Manning. 1989.Genes for biosynthesis and assembly of CS3 pili of CFA/II enterotoxigenicEscherichia coli: novel regulation of pilus production by bypassing an ambercodon. Mol. Microbiol. 3:1685–1695.

58. Jones, C. H., J. S. Pinkner, R. Roth, J. Heuser, A. V. Nicholes, S. N.Abraham, and S. J. Hultgren. 1995. FimH adhesin of type 1 pili is assem-bled into a fibrillar tip structure in the Enterobacteriaceae. Proc. Natl. Acad.Sci. USA 92:2081–2085.

59. Jones, C. H., P. N. Danese, J. S. Pinkner, T. J. Silhavy, and S. J. Hultgren.1997. The chaperone-assisted membrane release and folding pathway issensed by two signal transduction systems. EMBO J. 16:6394–6406.

60. Jonsson, A. B., G. Nyberg, and S. Normark. 1991. Phase variation ofgonococcal pili by frameshift mutation in pilC, a novel gene for pilusassembly. EMBO J. 10:477–488.

61. Jordi, B. J. A. M., G. A. Willshaw, B. A. M. van der Zeist, and W. Gaastra.1992. The complete nucleotide sequence of region 1 of the CFA/I fimbrialoperon of human enterotoxigenic Escherichia coli. DNA Sequence 2:257–263.

62. Kaiser, D. 1979. Social gliding is correlated with the presence of pili inMyxococcus xanthus. Proc. Natl. Acad. Sci. USA 76:5952–5956.

63. Kallenius, G., S. B. Svenson, H. Hultberg, R. Molby, I. Helin, B. Cedergen,and J. Windberg. 1981. Occurrence of P fimbriated Escherichia coli inurinary tract infection. Lancet ii:1369–1372.

64. Karlyshev, A., E. Galyov, O. Smirnov, A. Guzayev, V. Abramov, and V.Zav*yalov. 1992. A new gene of the f1 operon of Y. pestis involved in thecapsule biogenesis. FEBS Lett. 297:77–80.

65. Knutton, S., M. M. McConnel, B. Rowe, and A. S. McNeish. 1989. Adhesinand ultrastructural properties of human enterotoxigenic Escherichia coliproducing colonization factor antigens III and IV. Infect. Immun. 57:3364–3371.

66. Krogfelt, K. A., H. Bergmans, and P. Klemm. 1990. Direct evidence that theFimH protein is the mannose specific adhesin of Escherichia coli type 1fimbriae. Infect. Immun. 58:1995–1999.

67. Kuehn, M. J., J. Heuser, S. Normark, and S. J. Hultgren. 1992. P pili inuropathogenic E. coli are composite fibers with distinct fibrillar adhesivetips. Nature 356:252–255.

68. Kuehn, M. J., D. J. Ogg, J. Kihlberg, L. N. Slonim, K. Flemmer, T. Berg-fors, and S. J. Hultgren. 1993. Structural basis of pilus subunit recognitionby the PapD chaperone. Science 262:1234–1241.

69. Lamb, J. R., S. Tugendreich, and P. Hieter. 1995. Tetratrico peptide repeatinteraction: to TPR or not to TPR. Trends Biochem. Sci. 20:257–258.

70. Le Bouguenec, C., M. I. Garcia, V. Oulin, J.-M. Desperrier, P. Gounon, andA. Labigne. 1993. Characterization of plasmid-borne afa-3 gene clustersencoding afimbrial adhesins expressed by Escherichia coli strains associatedwith intestinal or urinary tract infections. Infect. Immun. 61:5106–5514.

71. Leffler, H., and C. Svanborg-Eden. 1980. Chemical identification of a gly-

cosphingolipid receptor for Escherichia coli attaching to human urinarytract epithelial cells and agglutinating human erythrocytes. FEMS Micro-biol. Lett. 8:127–134.

72. Levine, M. M., P. Ristaino, G. Marley, C. Smyth, S. Knutton, E. Boedeker,R. Black, C. Young, M. L. Clements, C. Cheney, and R. Patnaik. 1984. Colisurface antigens 1 and 3 of colonization factor antigen II-positive entero-toxigenic Escherichia coli: morphology, purification, and immune responsesin humans. Infect. Immun. 44:409–429.

73. Lindler, L. E., and B. D. Tall. 1993. Yersinia pestis pH 6 antigen formsfimbriae and is induced by intracellular association with macrophages. Mol.Microbiol. 8:311–324.

74. Lintermans, P. F., P. Pohl, A. Bertels, G. Charlier, J. Vandekerckhove, J.Van Damme, J. Shoup, C. Schlicker, T. Korhonen, H. De Greve, and M.Van Montagu. 1988. Characterization and purification of the F17 adhesinon the surface of bovine enteropathogenic and septicemic Escherichia coli.Am. J. Vet. Res. 49:1794–1799.

75. Locht, C., M.-C. Geoffroy, and G. Renauld. 1992. Common accessory genesfor the Bordetella pertussis filamentous hemagglutinin and fimbriae sharesequence similarities with the papC and papD gene families. EMBO J.11:3175–3183.

76. Loferer, H., M. Hammar, and S. Normark. 1997. Availability of the fibresubunit CsgA and the nucleator protein CsgB during assembly of fibronec-tin-binding curli is limited by the intracellular concentration of the novellipoprotein CsgG. Mol. Microbiol. 26:11–23.

77. Lund, B., B. I. Marklund, N. Stromberg, F. Lindberg, K. A. Karlsson, andS. Normark. 1988. Uropathogenic Escherichia coli can express serologicallyidentical pili of different receptor binding specificities. Mol. Microbiol.2:255–263.

78. Marklund, B. I., J. M. Tennent, E. Garcia, A. Hamers, M. Baga, F. Lind-berg, W. Gaastra, and S. Normark. 1992. Horizontal gene transfer of theEscherichia coli pap and prs pili operons as a mechanism for the develop-ment of tissue-specific adhesive properties. Mol. Microbiol. 6:2225–2242.

79. Massad, G., F. K. Bahrani, and H. L. T. Mobley. 1994. Proteus mirabilisfimbriae: identification, isolation, and characterization of a new ambient-temperature fimbria. Infect. Immun. 62:1989–1994.

80. Massad, G., J. F. Fulkerson, Jr., D. C. Watson, and H. L. T. Mobley. 1996.Proteus mirabilis ambient-temperature fimbriae: cloning and nucleotide se-quence of the aft gene cluster. Infect. Immun. 64:4390–4395.

81. McConnell, M. M., H. Chart, A. M. Field, M. Hibberd, and B. Rowe. 1989.Characterization of a putative colonization factor (PCFO166) of entero-toxigenic Escherichia coli of serogroup O166. J. Gen. Microbiol. 135:1135–1144.

82. McConnell, M. M., M. Hibberd, A. M. Field, H. Chart, and B. Rowe. 1990.Characterization of a new putative colonization factor (CS17) from a hu-man enterotoxigenic Escherichia coli of serogroup O114:H21 which pro-duces only heat-labile enterotoxin. J. Infect. Dis. 161:343–347.

83. Meyer, T. F. 1990. Variation of pilin and opacity-associated protein inpathogenic Neisseria species, p. 137–159. In B. Iglewski and V. Clark (ed.),The bacteria 43—molecular basis of bacterial pathogenesis. AcademicPress, New York, N.Y.

84. Moch, T., H. Hoschutzky, J. Hacker, K.-D. Kroncke, and K. Jann. 1987.Isolation and characterization of the a-sialyl-b-2,3-galactosyl-specific adhe-sin from fimbriated Escherichia coli. Proc. Natl. Acad. Sci. USA 84:3462–3466.

85. Mooi, F. R., W. H. Jansen, H. Brunings, H. Gielen, H. G. J. van der Heide,H. C. Walvoort, and P. A. M. Guniee. 1992. Construction and analysis ofBordetella pertussis mutants defective in the production of fimbriae. Microb.Pathog. 12:127–135.

86. Muller, K. H., S. K. Collinson, T. J. Trust, and W. W. Kay. 1991. Type 1fimbriae of Salmonella enteriditis. J. Bacteriol. 173:454–457.

87. Mulvey, M. A., Y. S. Lopez-Boado, C. L. Wilson, R. Roth, W. C. Parks,J. Heuser, and S. J. Hultgren. 1998. Induction and evasion of host defensesby type 1-piliated uropathogenic Escherichia coli. Science 282:1494–1497.

88. Nassif, X., J. L. Beretti, J. Lowy, P. Stenberg, P. O’Gaora, J. Pfeifer, S.Normark, and M. So. 1994. Roles of pilin and PilC in adhesin of Neisseriameningitidis to human epithelial and endothelial cells. Proc. Natl. Acad. Sci.USA 91:3769–3773.

89. Parge, H. E., K. T. Forest, M. J. Hickey, D. A. Christensen, E. D. Getzoff,and J. A. Tainer. 1995. Structure of the fibre-forming protein pilin at 2.6 Åresolution. Nature 378:32–38.

90. Pogliano, J., A. S. Lynch, D. Berlin, E. C. C. Lin, and J. Beckwith. 1997.Regulation of Escherichia coli cell envelope proteins involved in proteinfolding and degradation by the Cpx two-component system. Genes Dev.11:1169–1182.

91. Pugsley, A. P. 1992. Protein traffic in bacteria, p. 461–479. In J. Lederberg(ed.), Encyclopedia of microbiology, vol. 3. Academic Press, New York,N.Y.

92. Pugsley, A. P. 1993. The complete general secretory pathway in gram-negative bacteria. Microbiol. Rev. 57:50–108.

93. Ofek, I., D. Mirelman, and S. Sharon. 1977. Adherence of Escherichia colito human mucosal cells mediated by mannose receptors. Nature 265:623–625.

1070 MINIREVIEW J. BACTERIOL.

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from

Page 13: MINIREVIEW - Journal of Bacteriology · Reference(s) Chaperone-usher pathway Thick, rigid pili FGS chaperone/ushera P pili PapD/PapC E. coli Pyelonephritis or cystitis 47, 48, 50

94. Olsen, A., A. Jonsson, and S. Normark. 1989. Fibronectin binding mediatedby a novel class of surface organelles on Escherichia coli. Nature 338:652–655.

95. Read, T. D., M. Dowdell, S. W. Satola, and M. M. Farley. 1996. Duplicationof pilus gene complexes of Haemophilus influenzae biogroup aegyptius. J.Bacteriol. 178:6564–6570.

96. Riegman, N., R. Kusters, H. van Veggel, H. Bergmans, P. Van Bergen EnHenegouwen, J. Hacker, and I. Van Die. 1990. F1C fimbriae of a uropatho-genic Escherichia coli strain: genetic and functional organization of the focgene cluster and identification of minor subunits. J. Bacteriol. 172:1114–1120.

97. Rodriguez-Soto, J. P., and D. Kaiser. 1997. The tgl gene: social motility andstimulation in Myxococcus xanthus. J. Bacteriol. 179:4361–4371.

98. Rodriguez-Soto, J. P., and D. Kaiser. 1997. Identification and localizationof the Tgl protein, which is required for Myxococcus xanthus social motility.J. Bacteriol. 179:4372–4381.

99. Rudel, T., B. Hans-Jurgen, and T. F. Meyer. 1995. Pilus biogenesis andepithelial cell adherence of Neisseria gonorrhoeae pilC double knock-outmutants. Mol. Microbiol. 17:1057–1071.

100. Rudel, T., I. Scheuerpflug, and T. F. Meyer. 1995. Neisseria PilC proteinidentified as type 4 pilus tip-located adhesin. Nature 373:357–359.

101. Sajjan, U. S., L. Sun, R. Goldstein, and J. F. Forstner. 1995. Cable (Cbl)type II pili of cystic fibrosis-associated Burkholderia (Pseudomonas) cepacia:nucleotide sequence of the cblA major subunit pilin gene and novel mor-phology of the assembled appendage fibers. J. Bacteriol. 177:1030–1038.

102. Sakellaris, H., D. P. Balding, and J. R. Scott. 1996. Assembly proteins ofCS1 pili of enterotoxigenic Escherichia coli. Mol. Microbiol. 21:529–541.

103. Saulino, E. T., D. G. Thanassi, J. Pinkner, and S. J. Hultgren. 1998.Ramifications of kinetic partitioning on usher-mediated pilus biogenesis.EMBO J. 17:2177–2185.

104. Savarino, S., P. Fox, D. Yikang, and J. P. Nataro. 1994. Identification andcharacterization of a gene cluster mediating enteroaggregative Escherichiacoli aggregative adherence fimbria I biogenesis. J. Bacteriol. 176:4949–4957.

105. Schmoll, T., J. Morschhauser, M. Ott, B. Luwig, I. van Die, and J. Hacker.1990. Complete genetic organization and functional aspects of the Esche-richia coli S fimbrial adhesin determinant: nucleotide sequence of the genessfa B, C, D, E, F. Microb. Pathog. 9:331–343.

106. Sjobring, U., G. Pohl, and A. Olsen. 1994. Plasminogen, absorbed by Esch-erichia coli expressing curli or by Salmonella enteritidis expressing thinaggregative fimbriae, can be activated by simultaneously captured tissue-type plasminogen activator (t-PA). Mol. Microbiol. 14:443–452.

107. Smit, H., W. Gaastra, J. P. Kamerling, J. F. G. Vliegenthart, and F. K. deGraaf. 1984. Isolation and structural characterization of the equine eryth-rocyte receptor for enterotoxigenic Escherichia coli K99 fimbrial adhesin.Infect. Immun. 46:578–584.

108. Sohel, I., J. L. Puente, W. J. Murray, J. Vuopio-Varkila, and G. K.Schoolnik. 1993. Cloning and characterization of the bundle-forming pilingene of enteropathogenic Escherichia coli and its distribution in Salmonellaserotypes. Mol. Microbiol. 7:563–575.

109. Sokurenko, E. V., H. S. Courtney, S. N. Abraham, P. Klemm, and D. L.

Hasty. 1992. Functional heterogeneity of type 1 fimbriae of Escherichia coli.Infect. Immun. 60:4709–4719.

110. Soto, G. E., K. W. Dodson, D. Ogg, C. Liu, J. Heuser, S. Knight, J. Kihlberg,C. H. Jones, and S. J. Hultgren. 1998. Periplasmic chaperone recognitionmotif of subunits mediates quaternary interactions in the pilus. EMBO J.17:6155–6167.

111. Striker, R., U. Nilsson, A. Stonecipher, G. Magnusson, and S. J. Hultgren.1995. Structural requirements for the glycolipid receptor of human uro-pathogenic E. coli. Mol. Microbiol. 16:1021–1029.

112. Stromberg, N., B. I. Marklund, B. Lund, D. Ilver, A. Hamers, W. Gaastra,K. A. Karlsson, and S. Normark. 1990. Host-specificity of uropathogenicEscherichia coli depends on differences in binding specificity to Gala(1-4)Gal-containing isoreceptors. EMBO J. 9:2001–2010.

113. Tennent, J. M., and J. S. Mattick. 1994. Type 4 fimbriae, p. 127–146. In P.Klemm (ed.), Fimbriae: adhesion, genetics, biogenesis, and vaccines. CRCPress, Ann Arbor, Mich.

114. Thanassi, D. G., E. T. Saulino, M. J. Lombardo, R. Roth, J. Heuser, andS. J. Hultgren. 1998. The PapC usher forms an oligomeric channel: impli-cations for pilus biogenesis across the outer membrane. Proc. Natl. Acad.Sci. USA 95:3146–3151.

115. Thomas, L. V., M. M. McConnell, B. Rowe, and A. M. Field. 1985. Thepossession of three novel coli surface antigens by enterotoxigenic Esche-richia coli strains positive for the putative colonization factor PCF8775.J. Gen. Microbiol. 131:2319–2326.

116. Tønjum, T., and M. Koomey. 1997. The pilus colonization factor of patho-genic neisserial species: organelle biogenesis and structure/function rela-tionships. Gene 192:155–163.

117. Turner, L. R., J. C. Lara, D. N. Nunn, and S. Lory. 1993. Mutations in theconsensus ATP-binding sites of XcpR and PilB eliminate extracellularprotein secretion and pilus biogenesis in Pseudomonas aeruginosa. J. Bac-teriol. 175:4962–4969.

118. van Alphen, L., L. Geelen-van Den Broek, L. Blaas, M. van Ham, and J.Dankert. 1991. Blocking of fimbria-mediated adherence of Haemophilusinfluenzae by sialyl gangliosides. Infect. Immun. 59:4473–4477.

119. van der Woude, M., B. Braaten, and D. Low. 1996. Epigenetic phasevariation of the pap operon in Escherichia coli. Trends Microbiol. 4:5–9.

120. Voegele, K., H. Sakellaris, and J. R. Scott. 1997. CooB plays a chaperone-like role for the proteins involved in formation of CS1 pili of enterotoxi-genic Escherichia coli. Proc. Natl. Acad. Sci. USA 94:13257–13261.

121. Willems, R. J. L., H. G. J. van der Heide, and F. R. Mooi. 1992. Charac-terization of a Bordetella pertussis fimbrial gene cluster which is locateddirectly downstream of the filamentous haemagglutinin gene. Mol. Micro-biol. 6:2661–2671.

122. Wolf, M. K., G. P. Andrews, B. D. Tall, M. M. McConnel, M. M. Levine, andE. C. Boedeker. 1989. Characterization of CS4 and CS6 antigenic compo-nents of PCF8775, a putative colonization factor complex from enterotoxi-genic Escherichia coli E8775. Infect. Immun. 57:164–173.

123. Xu, Z., C. H. Jones, D. Haslem, J. S. Pinkner, K. Dodson, J. Kihlberg, andS. J. Hultgren. 1995. Molecular dissection of PapD interaction with PapGreveals two chaperone-binding sites. Mol. Microbiol. 16:1011–1020.

VOL. 181, 1999 MINIREVIEW 1071

on Novem

ber 1, 2020 by guesthttp://jb.asm

.org/D

ownloaded from