Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Review TRENDS in Microbiology Vol.12 No.

9 September 2004

Intracellular bacterial communities of


uropathogenic Escherichia coli in
urinary tract pathogenesis
Gregory G. Anderson1, Karen W. Dodson1, Thomas M. Hooton2 and Scott J. Hultgren1
1
Department of Molecular Microbiology, Washington University School of Medicine, Box 8230, 660 S. Euclid Ave., St Louis, MO
63110, USA
2
Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington School of Medicine, Box 359930,
Harborview Medical Center, 325 Ninth Ave., 2WC-516, Seattle, WA 98104, USA

Urinary tract infections in young, healthy women complicating factors, such as diabetes, spinal cord inju-
frequently recur, despite their traditional classification ries, urinary catheters, or hospitalization, can also present
as acute infections. Conventional wisdom dictates that with UTI caused by Klebsiella pneumoniae, Proteus
uropathogens causing recurrent infections in such mirabilis, Enterococcus faecalis, Pseudomonas aerugi-
individuals come from the fecal or vaginal flora, in the nosa, Serratia marcescens and group B streptococci [4].
same manner as the initial infection. However, recent Although traditionally thought of as acute and self-
studies of uropathogenic Escherichia coli have found limiting, 27–44% of women with an initial UTI will
that it can carry out a complex developmental program experience at least one recurrence of symptoms within
within the superficial epithelial cells of the mouse six months, despite antibiotic therapy [5,6]. Although
bladder, forming intracellular bacterial communities these recurrent infections might occasionally be due to a
with many biofilm-like properties. These intracellular persistent focus of infection, the majority have been
biofilms allow the bacteria to outlast a strong host thought to be reinfections caused by the initially infecting
immune response to establish a dormant reservoir of strain persisting in the fecal flora [7]. The bacteria
pathogens inside the bladder cells. Re-emergence of associated with recurrent UTI often appear to be pheno-
bacteria from this reservoir might be the source typically or genetically identical to the bacterial strain
of recurrent infection. that caused the initial infection, suggesting that selected
E. coli strains might become uniquely adapted for
Urinary tract infections (UTIs), considered among the
colonizing and infecting their respective hosts [7]. The
most common of bacterial diseases, afflict a large pro-
steady increase of antibiotic resistance [8] and occurrences
portion of the world population [1]. A majority of these
of clonal outbreaks of UPEC-associated UTIs [9] highlight
infections occur in young, healthy women. In a large
the need for greater understanding of the mechanisms of
prospective study of young, sexually active women, the
UPEC pathogenesis.
incidence of UTI was w0.5 per person-year [2]. More
recently, a population-based survey estimated that as UPEC have proven to be quite stealthy in its patho-
many as 11 million women in the United States had at genesis of the urinary tract, using a complex pathway to
least one presumed UTI treated with an antibiotic in 1995, mediate infection and persistence in the face of a strong
and that the cost for evaluation and management of UTIs innate host immune response. One of the most studied
was 1.6 billion dollars [1]. In this survey, the self-reported abilities of UPEC is binding host tissues and particular
incidence of UTI in women 18 years and older was 10.8%, organ niches. UPEC accomplish this binding by assem-
and the cumulative lifetime risk of UTI was 60%. Even bling several different adhesive organelles on their sur-
episodes of acute, uncomplicated UTI are associated with face, the most understood comprising P pili and type 1 pili
considerable morbidity, including 6.1 days of symptoms, [10]. These pili, assembled by the chaperone–usher path-
2.4 days of restricted activity and 0.4 bed days [1]. Given way, display an adhesin molecule at their distal end [11].
that an estimated 130–175 million cases of UTI occur P pili display the PapG adhesin, which has been shown to
annually worldwide [3], the societal costs associated with be required for pyelonephritic pathogenesis by binding to
UTI are huge. The most common isolates from patients are globoside present on human kidney cells [12]. By contrast,
uropathogenic Escherichia coli (UPEC), accounting for type 1 pili have been found to be vital for attachment to the
w80% of all acute, community-acquired UTIs [4]. Aside bladder epithelium. The FimH adhesin on type 1 pili binds
from UPEC, several other microorganisms can also cause mannose, and in the bladder FimH recognizes mannosy-
UTI, most notably Staphylococcus saprophyticus, impli- lated residues present on uroplakin proteins that line the
cated in most of the remaining UTI cases [5]. Patients with luminal surface of the bladder superficial epithelial cells
Corresponding author: Scott J. Hultgren (hultgren@borcim.wustl.edu).
[13]. FimH-mediated binding of the bladder epithelium is
Available online 24 July 2004 the initial step in an intricate cascade of events leading to
www.sciencedirect.com 0966-842X/$ - see front matter Q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tim.2004.07.005
Review TRENDS in Microbiology Vol.12 No.9 September 2004 425

the myriad of symptoms associated with acute UTI and systems, including aerobactin [24] and the more recently
possible long-term residence of UPEC in the urinary tract. described IroN system [25,26]. These iron siderophores
scavenge available iron and enhance UPEC survival in the
nutrient-limiting bladder environment. Finally, most
UPEC virulence determinants
UPEC strains produce an acidic polysaccharide capsule,
Other than type 1 and P pili, UPEC produce additional
which protects the bacteria from phagocytosis by human
factors that influence disease progression (Table 1). Some
PMNs and inhibits activation of complement [24].
of these virulence determinants are located on one of
several UPEC specific pathogenicity-associated islands
Host response to UPEC infection
[14]. A recent genomic analysis of a UPEC strain revealed
The host counters the actions of UPEC upon the bladder
the presence of genes for ten putative chaperone–usher
by mounting a robust and dramatic response. Bladder
pilus systems, two putative type IV pili and at least seven
superficial epithelial cells express Toll-like receptor 4
putative autotransporter proteins [15]. The large reper-
(TLR-4) on their membrane, which, along with CD14,
toire of pilus systems might confer upon UPEC multiple
recognizes lipopolysaccharide from the bacteria and
binding specificities and the capacity to colonize various
activates an innate immune response [27,28]. A burst of
sites throughout the urinary tract and other environ-
inflammatory cytokines leads to a massive infiltration of
ments. Autotransporter proteins can also have adhesive
neutrophils to fight the infection [29,30]. Further,
properties or they might fill other roles, such as toxins,
increased transcription of inducible nitric oxide synthase
proteases, invasins, serum resistance factors and motility
by PMNs and the bladder epithelium results in high levels
mediators [16]. One UPEC specific autotransporter, Sat,
of nitric oxide and related breakdown products, which
appears to exert a toxic effect upon urinary tract cells
could have toxic effects on the bacteria [31,32]. Upon
in vitro [17]. Incubation of bladder and kidney cells with
infection, the bladder epithelium is triggered to exfoliate
Sat-producing UPEC leads to extreme vacuolation in the
superficial facet cells [33]. This aids clearance of any
cytoplasm of the host cells and possible loosening of
tissue-associated bacteria [34]. The underlying epi-
cellular junctions. In a mouse model of UTI, Sat also
thelium, normally extremely inert [35], rapidly undergoes
induces cytoplasmic vacuolation and severe histological
a renewal process wherein underlying epithelial cells
damage in kidneys of infected laboratory animals [18].
proliferate and differentiate to replace the superficial
Two other UPEC-expressed autotransporter proteins, Pic
layer of cells [36]. Despite the defensive immune arsenal
and Tsh, appear to have serine protease activity [19].
and rapid epithelial turnover, UPEC are able to colonize
These two proteins are expressed during infection in
the bladder and establish a persistent reservoir which can
laboratory mice and appear to be generally associated
last at least several months after infection in laboratory
with pyelonephritis strains. UPEC also express an RTX
mice [37]. Bacteria persist in the bladder notwithstanding
(repeat in toxin) toxin called a-hemolysin (HlyA), often
antibiotic treatment [38], and, periodically, bacteria
linked with the P pilus operon [20]. HlyA forms pores in a
arising from this reservoir are shed in the urine of these
variety of host cell membranes and can induce calcium
mice. Human patients have also been shown to shed
oscillations in proximal tubule cells of infected rat kidneys
bacteria in the urine, even in the absence of active UTI
[21]. These fluctuations might serve as second messengers
symptoms [39,40]. Therefore, UPEC appear to be quite
during immune activation of the host. Another toxin
adept at establishing a protective niche for themselves in
produced by UPEC, cytotoxic necrotizing factor 1 (CNF-1),
the bladder and persisting for extended periods of time.
influences the host cell cytoskeleton by targeting the Rho
family of GTP-binding proteins [22]. CNF-1 has been
Intracellular bacterial communities
shown to kill human bladder epithelial cells in vitro and
Recently, it was discovered that UPEC activate a complex
inhibit phagocytosis of bacteria by human polymorpho-
developmental cascade upon their entry into superficial
nuclear leukocytes (PMNs) [23]. In addition to proteases
bladder cells [41,42]. This cascade was elucidated using
and toxins, UPEC produce several iron acquisition
state of the art microscopy, including scanning and
Table 1. Virulence determinants of uropathogenic Escherichia transmission electron microscopy, immunohistochemistry
coli and time-lapse videomicroscopy. For videomicroscopy,
mouse bladders infected with green fluorescence protein-
Factor Refs expressing UPEC were stretched on an incubation
Adhesins P pili [10] chamber and placed on an epifluorescence microscope
Type 1 pili [10] [42]. Images were captured every 30 seconds to two
S pili [10]
Dr adhesins [10]
minutes, and multiple overlapping time frames were
Toxins HlyA [20] examined to piece together a timeline of UTI progression.
CNF-1 [23] Using these techniques, it was found that, in the early
Sat [18] stages, the bacteria invade the superficial cells and rapidly
Siderophores Aerobactin [24] divide. As the bacteria grow, dramatic phenotypic
IroN [26]
IreA [24]
switches result in the establishment of intracellular
Proteases Pic [19] bacterial communities (IBCs) (Figure 1). IBCs progress
Tsh [19] through several stages, culminating in the formation of
Other Capsule [24] biofilm-like communities inside the superficial cells.
LPS [24]
Eventually, bacteria detach from the biofilm and burst
www.sciencedirect.com
426 Review TRENDS in Microbiology Vol.12 No.9 September 2004

in the bladder might increase the ability of type 1 piliated


organisms to attach and colonize the urinary tract.
After bacterial attachment, UPEC quickly invade the
bound epithelial cell [44,47]. This key event occurs one to
three hours after initial inoculation. It has been shown
using tissue culture models that binding activates the Rho
family of small GTP-binding proteins, specifically RhoA,
Cdc42 and Rac1, which then activate focal adhesin kinase,
phosphoinositide 3-kinase, a-actinin and vinculin. These
processes result in localized actin rearrangements and
membrane extensions around the bacteria [47]. The
membrane engulfs the bacteria, leading to UPEC intern-
alization via zipper-like phagocytosis.
Once intracellular, UPEC rapidly grow and divide,
forming small clusters of bacteria, termed ‘early IBCs’
[42]. Early IBCs have previously been called ‘bacterial
factories’ owing to the ability of UPEC to usurp the
superficial bladder cells and convert them into factories
for bacterial growth [37]. A major benefit that invasion
and establishment of an intracellular community confers
upon UPEC is protection from killing by antibiotics, which
Figure 1. Middle intracellular bacterial communities (IBCs) are seen as ‘pods’ on the has been shown both in vitro and in vivo [37,38]. In this
surface of infected mouse bladder epithelium. (a) Numerous pods cover the way, UPEC form a protective niche in the bladder where
bladder surface in this scanning electron micrograph. Formation of biofilm-like
pods protects the bacteria from immune clearance, thus allowing uropathogenic
they can hide and survive. The bacteria within the early
Escherichia coli (UPEC) to build up to extremely high levels in the bladder. Scale IBC divide rapidly during the first six to eight hours after
bar, 50 mm. (b) Magnification of a pod reveals a smooth shell which is the uroplakin- inoculation, with a doubling time of w30 to 35 minutes.
coated luminal membrane of the bladder superficial epithelial cell. The pod consists
of one cell that is completely filled with intracellular bacteria. Scale bar, 5 mm. (c) An
The bacteria at this stage orient themselves randomly in
IBC is visible inside the bladder epithelium by fluorescent microscopy. Scale bar, the cytoplasm of the host cell, such that they form an
20 mm. The green fluorescence protien-expressing bacteria form a dense sphere amorphous, loosely associated bacterial clump [42].
within the bladder cell. IBC progression can be monitored by time-lapse
videomicroscopy, in which time-lapse images are captured by epifluorescence During this period of quick expansion, UPEC maintain
microscopy [42]. their typical rod morphology, with an average length of
3 mm. This fast growth leads to a surge of microorganisms
out into the bladder lumen. These escaped bacteria then within the bladder early in infection.
rebind to the epithelium and initiate another round of IBC
formation. The downstream effects of this cascade might IBC maturation and middle IBCs
permit UPEC to evade the host immune response and Between six to eight hours postinoculation, early IBCs
persist in the urinary tract. experience a dramatic phenotypic switch [42]. A remark-
able drop in bacterial growth rate, resulting in doubling
Binding and early IBC formation times greater than 60 minutes, takes place concurrent
Type 1 pili, with the FimH adhesin at the distal tip, are with a significant shortening of bacterial morphology to an
crucial for UPEC attachment to the bladder epithelium average 0.7 mm. At the same time, the amorphous clump
[43]. Two lines of evidence confirm the vital role of FimH appears to come together to form a tight, highly organized
for bacterial attachment to the epithelium. First, isogenic sphere inside the host cell. These events represent a
fimH mutants are defective for binding and colonization of maturation of the early IBC into a middle IBC stage with
the bladder [33]. Second, polystyrene latex beads coated many biofilm-like properties [41].
with FimH readily associate with, and are internalized by, Bacterial biofilms are often associated with long-term
human bladder epithelial cells in vitro [44]. FimH binds to persistence of organisms in various environments. Bio-
mannosylated uroplakins on the bladder epithelium via a films are collections of bacteria that are attached to a
deep acidic pocket formed at the tip of the lectin-binding surface, or to each other, and that display community
domain [13]. Four different uroplakin molecules associate behavior [48]. Initial events in biofilm formation involve
into hexameric rings, which are arrayed in crystalline binding of planktonic bacteria to a surface and formation
plaques on the luminal membrane of the bladder super- of small bacterial clusters called microcolonies. As these
ficial epithelial cells. This uroplakin plaque provides a microcolonies grow into mature biofilms, the bacteria
permeability barrier to prevent toxic molecules concen- differentiate, undergo dramatic phenotypic changes and
trated in the urine from diffusing into deeper tissues [35,45]. encase themselves in an extracellular polysaccharide
High resolution freeze-fracture electron microscopy has matrix [49,50]. These actions result in a large three-
revealed that the tips of type 1 pili, which contain the dimensional structure, with subpopulations of bacteria at
adhesin, are buried in the central cavity of the uroplakin different regions of this structure displaying altered gene
hexamers, tethering the bacteria to the epithelium [33]. It transcriptional patterns [48]. It is thought that these
was recently shown that shear force enhances FimH- subpopulations represent a division of labor, where each
mediated binding [46], and such forces when encountered group performs certain specialized functions that benefit
www.sciencedirect.com
Review TRENDS in Microbiology Vol.12 No.9 September 2004 427

the whole community [51]. As the biofilm ages, a subset of surrounding matrix, creating individualized compart-
bacteria detach from the biofilm and revert to a planktonic ments for each bacterium that establish a spatial
state, which allows for spread of organisms to other niches orientation of microorganisms inside the IBC. UPEC
[48]. A biofilm lifestyle benefits the bacteria by providing express several factors in the middle IBC which are
protection from environmental insults and changes [49]. important for in vitro biofilm formation, including type
Bacteria in biofilms display dramatically increased resist- 1 pili and antigen 43 [41]. Similar to biofilms,
ance to antibiotics because of the inability of antibiotics to expression of these factors is heterogeneous, with the
penetrate the biofilm matrix, the slow growth-rate of the bacteria setting up subpopulations with differential
bacteria in the biofilm, physiological changes in the gene expression. Polysaccharides are also present
bacteria and biofilm-state expression of factors that throughout the middle IBC.
directly inhibit antibiotic activity [48,52]. In addition, By differentiating into an intracellular biofilm, UPEC
biofilms confer increased resistance to host immune effects construct a safe haven where they can hide and wait out
upon their constituent bacteria by concealing the bacteria the tide of the host immune attack. PMNs recruited to the
inside the biofilm matrix. For instance, bacteria in bladder are targeted to the IBCs and attempt to clear the
biofilms are protected from opsonizing antibodies [48]. bacteria [29,42]. However, the IBC remains largely
Phenotypic variation and differentiation might also unaffected. The uroplakin shell covering the IBC appears
enhance bacterial resistance to PMNs and immune to protect UPEC by denying the PMNs access to the
effector molecules [48]. Antibiotic and immune resistances bacteria. The structural organization of the pod might also
facilitate bacterial biofilm survival in vivo during many protect the bacteria from antibiotic susceptibility. The
disease conditions, including native valve endocarditis, matrix surrounding the bacteria inside the cell might also
otitis media, chronic bacterial prostatitis, periodontitis add an additional layer of protection from antibiotics and
and cystic fibrosis. Bacterial biofilms have also been the immune system. It is interesting that in vitro biofilm-
described on inert medical devices, such as prosthetic state UPEC have been found to be resistant, even when
heart valves, central venous catheters, contact lenses, antibiotics reach the bacteria [59], suggesting that the
intrauterine devices, urinary catheters and dental unit physiological state of the microbes in biofilms enhances
water lines [48]. The presence of biofilms in disease or on bacterial survival. In this manner, UPEC are able to
medical devices leads to chronic symptoms, owing to replicate to large numbers, unperturbed by the host and
persistence of bacteria, despite antimicrobial therapy. other insults.
An important factor in E. coli biofilm formation is type 1
pili, which aids stable attachment of the bacteria to a Late IBCs and fluxing
surface [53]. Flagella also assist biofilm formation by Eventually, the IBC progresses to a late stage [42].
mediating initial weak surface contact and influencing Starting around 12 hours postinoculation, bacteria on
expansion of biofilms across a surface [53]. Antigen 43, an the edge of the intracellular biofilm detach from the group
outer membrane autotransporter protein, promotes auto- and differentiate into a typical rod morphology, with an
aggregation of E. coli and can assist interbacterial average length of 2 mm. At this stage of the maturation
interactions during microcolony formation [54]. This cycle, the bacteria become highly motile. These motile
might also be the function of secreted amyloid fibers bacteria swim toward the edge of the cell, burst out into
called curli [55,56]. The regulator of the curli operon, the bladder lumen in a process termed ‘fluxing’ and spread
CsgD, might also regulate several metabolic and other out over the epithelium. Fluxing bacteria have also been
factors that influence formation of mature biofilms [57]. observed erupting en masse from the epithelial cell, the
A key process during biofilm maturation, following entire community simultaneously emerging from its
attachment and microcolony formation, is secretion of intracellular hiding place [37]. It is possible that fluxing
colanic acid, an exopolysaccharide that creates a poly- initiates with a few bacteria, and as the host cell
saccharide matrix consisting of a complex three-dimen- membrane integrity decreases, some signal leads to the
sional architecture [58]. concomitant bursting of the entire IBC. From the volume
The incredible organization and structure produced by of mature IBCs and the size of the bacteria, it is estimated
UPEC in the middle IBC are highly reminiscent of that an average IBC contains in the order of 105
bacterial biofilms [49]. As middle IBCs grow, the bacterial organisms. Fluxing of multiple IBCs, therefore, might
mass pushes against the epithelial cell membrane, result in the release of hundreds of thousands or millions
creating a podlike protrusion on the surface of infected of UPEC back into the bladder lumen, leading to spread
bladder epithelium (Figure 1) [41]. Pods correspond to a within the bladder as well as dissemination back into the
middle IBC. The bacteria in the middle IBC, although free environment [42]. Escaped bacteria often filament, reach-
in the cytoplasm, stay together as a spherical community, ing up to 70 mm or greater in length [37,42]. Although the
filling up much of the intracellular volume of a superficial mechanisms and purpose behind filamentation remain
umbrella cell. The surface of the pod is the luminal unclear, PMNs appear to have little effect upon the
membrane of the bladder cell, which is stretched tight, filaments [42]. This allows UPEC to survive long enough
appearing smooth by scanning electron microscopy, and is in the inflamed bladder to reattach to the epithelium.
coated with an impermeable shell of uroplakin. The
bacteria inside the pod are highly organized and sur- UPEC persistence in the bladder
rounded by numerous fibers emanating from the bacterial A second round of IBC formation occurs as escaped
surface. These fibers make extensive contacts with the bacteria rebind and reinvade the superficial cells [42].
www.sciencedirect.com
428 Review TRENDS in Microbiology Vol.12 No.9 September 2004

Binding and invasion Early IBC Middle IBC Late IBC


TRENDS in Microbiology

Figure 2. Schematic representation of the intracellular bacterial community (IBC) maturation program of uropathogenic Escherichia coli (UPEC) within the superficial
epithelium of the mouse bladder. Soon after inoculation, UPEC (green) bind and invade the superficial epithelial cells of the bladder in a process mediated by type 1 pili (red).
Rapid multiplication leads to formation of a disorganized bacterial clump within the cell cytoplasm, known as an ‘early IBC’. Activation of developmental cascades results in
maturation to a biofilm-like middle IBC. An impermeable uroplakin shell and the biofilm-like environment protect UPEC in the middle IBC from immune attack. Eventually,
UPEC detach from the intracellular biofilm, become motile (yellow flagella) and flux out of the cell into the bladder lumen. This presumably facilitates bacterial persistence
within the host and transmission to the environment. Fluxed UPEC can rebind to the epithelium to form additional generations of IBCs. The time scale for IBC progression is
dependent upon the bacterial strain and the host strain used. During infection with UPEC strain UTI89 in C3H/HeN mice, the first round of IBC development generally
completes within 24 hours after inoculation [42]. After several days, a quiescent bacterial reservoir develops in the bladder, which could be a seed for urinary tract infection
recurrence. Reproduced, with permission, from Ref. [42].

However, this second round takes much longer to transmission back into the environment, subsequent IBC
progress, with doubling times in the early IBC of greater cycles and establishment of a chronic bacterial reservoir.
than 60 minutes. Multiple IBC cycles probably occur but Alternatively, formation of the reservoir might occur
after several days, only a quiescent reservoir of small through an entirely separate route. Continuing studies
intracellular bacterial clusters (doublets, quadruplets, will provide answers to this and other fundamental
small groups) remains in the bladder. The bladder questions relating to UPEC pathogenesis and persistence
epithelium is intact at this stage, with the bacteria (Box 1). The elucidation of the IBC program was carried
residing inside superficial cells which appear smaller out using a mouse model of infection. However, UPEC also
than normal. These smaller superficial cells might result bind, invade and replicate inside human epithelial cells
from growth and differentiation of the underlying epi- in vitro [37,44,47], and it will be vital for future studies to
thelial cells following exfoliation of the original superficial establish whether IBCs and quiescent reservoirs of
cells. As infection progresses, a greater number of fluxed bacteria form in human patients. As more is understood
UPEC might bind and invade the underlying epithelium, about the factors that are expressed during UTI, and their
exposed during exfoliation. Physiological differences regulation, it will be essential to determine their role in
between the superficial and underlying cells influence the IBC program. Such knowledge will provide a compre-
whether IBCs form. Indeed, IBCs have never been seen hensive understanding of UTI progression and lead to new
within the underlying bladder cells. During this reservoir clues about how to abrogate UPEC infection. Further,
stage, no apparent actions are taken either by the host or because many different microorganisms invade host
the bacteria, and UPEC can persist in this state in the tissues and multiply inside host cells, it is possible that
bladder for months after an initial infection [37,38]. an IBC-like developmental program is important during
Periodically during this persistent stretch, large numbers various stages of other chronic recurrent infections.
of organisms are seen in the urine [38]. It is thought that A greater understanding of the nature of intracellular
reactivation of UPEC from their dormant reservoir state, bacterial communities in chronic or recurrent infections
and subsequent multiplication inside the bladder epi- will aid the development of new and more effective
thelium to re-form IBCs, lead to this bacterial spike treatments for these problematic diseases.
in the urine.

Box 1. Intriguing questions for future study


Clinical impact † Do intracellular bacterial communities (IBCs) form in human
The discovery of the IBC maturation cascade suggests patients with cystitis?
that UPEC possess a program that enables the bacteria to † What genetic mechanisms do uropathogenic Escherichia coli
evade early innate immune defenses. Considering that (UPEC) use to form IBCs?
† What accounts for polymorphonuclear leukocyte resistance by the
extracellular bacteria are readily cleared by PMNs [42], filaments?
the formation of intracellular communities of bacteria † Why do IBCs form only in the superficial epithelial cells of the
inside the superficial cells of the bladder appears key to bladder?
the survival of UPEC in the urinary tract during the acute † What induces UPEC to become quiescent?
phases of infection (Figure 2). IBCs appear to permit † Can stimulation of the dormant reservoir lead to a urinary tract
infection recurrence?
UPEC to survive the immune onslaught that occurs at † What signals activate the dormant reservoir?
these early stages. In this haven, the bacteria can safely † How and when do known virulence determinants influence the IBC
multiply, creating an overpowering legion which then cascade?
escapes into the bladder lumen. Presumably, the sheer † What bacterial and/or host signals regulate IBC processes?
† How do IBCs relate to the chronic phase of infection?
numbers of bacteria that escape overwhelm the immune
† Are IBCs formed in other chronic recurrent infections?
system, leading to several potential downstream effects:
www.sciencedirect.com
Review TRENDS in Microbiology Vol.12 No.9 September 2004 429

Acknowledgements 24 Johnson, J.R. (2003) Microbial virulence determinants and the


We thank S. Justice, C. Hung and J. Palermo for helpful discussions. This pathogenesis of urinary tract infection. Infect. Dis. Clin. North Am.
work was supported by National Institutes of Health grants AI29549, 17, 261–278
DK51406 and AI48689, and by ORWH SCOR grant DK64540 in 25 Russo, T.A. et al. (1999) Identification of genes in an extraintestinal
partnership with DHHS, NIDDK and the FDA. isolate of Escherichia coli with increased expression after exposure to
human urine. Infect. Immun. 67, 5306–5314
26 Sorsa, L.J. et al. (2003) Characterization of an iroBCDEN gene cluster
on a transmissible plasmid of uropathogenic Escherichia coli:
References evidence for horizontal transfer of a chromosomal virulence factor.
1 Foxman, B. (2002) Epidemiology of urinary tract infections: incidence, Infect. Immun. 71, 3285–3293
morbidity, and economic costs. Am. J. Med. 113(Suppl. 1), 5S–13S 27 Schilling, J.D. et al. (2003) Toll-like receptor 4 on stromal and
2 Hooton, T.M. et al. (1996) A prospective study of risk factors for hematopoietic cells mediates innate resistance to uropathogenic
symptomatic urinary tract infection in young women. N. Engl. J. Med. Escherichia coli. Proc. Natl. Acad. Sci. U. S. A. 100, 4203–4208
335, 468–474 28 Schilling, J.D. et al. (2003) CD14- and Toll-like receptor-dependent
3 Russo, T.A. and Johnson, J.R. (2003) Medical and economic impact of activation of bladder epithelial cells by lipopolysaccharide and type 1
extraintestinal infections due to Escherichia coli: focus on an piliated Escherichia coli. Infect. Immun. 71, 1470–1480
increasingly important endemic problem. Microbes Infect. 5, 449–456 29 Schilling, J.D. et al. (2001) Bacterial invasion augments epithelial
4 Ronald, A. (2002) The etiology of urinary tract infection: traditional cytokine responses to Escherichia coli through a lipopolysaccharide-
and emerging pathogens. Am. J. Med. 113(Suppl. 1), 14S–19S dependent mechanism. J. Immunol. 166, 1148–1155
5 Hooton, T.M. and Stamm, W.E. (1997) Diagnosis and treatment of 30 Haraoka, M. et al. (1999) Neutrophil recruitment and resistance to
uncomplicated urinary tract infection. Infect. Dis. Clin. North Am. 11, urinary tract infection. J. Infect. Dis. 180, 1220–1229
551–581 31 Poljakovic, M. and Persson, K. (2003) Urinary tract infection in iNOS-
6 Ikaheimo, R. et al. (1996) Recurrence of urinary tract infection in a deficient mice with focus on bacterial sensitivity to nitric oxide. Am.
primary care setting: analysis of a 1-year follow-up of 179 women. J. Physiol. Renal Physiol. 284, F22–F31
Clin. Infect. Dis. 22, 91–99 32 Poljakovic, M. et al. (2001) Escherichia coli-induced inducible nitric
7 Stapleton, A. and Stamm, W.E. (1997) Prevention of urinary tract oxide synthase and cyclooxygenase expression in the mouse bladder
infection. Infect. Dis. Clin. North Am. 11, 719–733 and kidney. Kidney Int. 59, 893–904
8 Stamm, W.E. (2002) Scientific and clinical challenges in the manage- 33 Mulvey, M.A. et al. (2000) Bad bugs and beleaguered bladders:
ment of urinary tract infections. Am. J. Med. 113(Suppl. 1), 1S–4S interplay between uropathogenic Escherichia coli and innate host
9 Manges, A.R. et al. (2001) Widespread distribution of urinary tract defenses. Proc. Natl. Acad. Sci. U. S. A. 97, 8829–8835
infections caused by a multidrug-resistant Escherichia coli clonal 34 Gunther, N.W.T. et al. (2001) In vivo dynamics of type 1 fimbria
group. N. Engl. J. Med. 345, 1007–1013 regulation in uropathogenic Escherichia coli during experimental
10 Mulvey, M.A. (2002) Adhesion and entry of uropathogenic Escherichia urinary tract infection. Infect. Immun. 69, 2838–2846
coli. Cell. Microbiol. 4, 257–271 35 Southgate, J. et al. (1999) Urothelial tissue regulation. Unraveling the
11 Sauer, F.G. et al. (2000) Chaperone-assisted pilus assembly and role of the stroma. Adv. Exp. Med. Biol. 462, 19–30
bacterial attachment. Curr. Opin. Struct. Biol. 10, 548–556 36 Mysorekar, I.U. et al. (2002) Molecular regulation of urothelial
12 Dodson, K.W. et al. (2001) Structural basis of the interaction of the renewal and host defenses during infection with uropathogenic
pyelonephritic E. coli adhesin to its human kidney receptor. Cell 105, Escherichia coli. J. Biol. Chem. 277, 7412–7419
733–743 37 Mulvey, M.A. et al. (2001) Establishment of a persistent Escherichia
13 Hung, C.S. et al. (2002) Structural basis of tropism of Escherichia coli reservoir during the acute phase of a bladder infection. Infect.
coli to the bladder during urinary tract infection. Mol. Microbiol. Immun. 69, 4572–4579
44, 903–915 38 Schilling, J.D. et al. (2002) Effect of trimethoprim-sulfamethoxazole
14 Hacker, J. et al. (1999) Pathogenicity islands of extraintestinal on recurrent bacteriuria and bacterial persistence in mice infected
Escherichia coli. In Pathogenicity Islands and Other Mobile Virulence with uropathogenic Escherichia coli. Infect. Immun. 70, 7042–7049
Elements (Kaper, J.B. and Hacker, J. eds), pp. 59–75, American 39 Anderson, M. et al. (2004) Viable but nonculturable bacteria are
Society for Microbiology present in mouse and human urine specimens. J. Clin. Microbiol. 42,
15 Welch, R.A. et al. (2002) Extensive mosaic structure revealed by the 753–758
complete genome sequence of uropathogenic Escherichia coli. Proc. 40 Hooton, T.M. et al. (2000) A prospective study of asymptomatic
Natl. Acad. Sci. U. S. A. 99, 17020–17024 bacteriuria in sexually active young women. N. Engl. J. Med. 343,
16 Henderson, I.R. and Nataro, J.P. (2001) Virulence functions of 992–997
autotransporter proteins. Infect. Immun. 69, 1231–1243 41 Anderson, G.G. et al. (2003) Intracellular bacterial biofilm-like pods in
17 Guyer, D.M. et al. (2000) Identification of sat, an autotransporter toxin urinary tract infections. Science 301, 105–107
produced by uropathogenic Escherichia coli. Mol. Microbiol. 38, 53–66 42 Justice, S.S. et al. (2004) Differentiation and developmental pathways
18 Guyer, D.M. et al. (2002) Sat, the secreted autotransporter toxin of of uropathogenic Escherichia coli in urinary tract pathogenesis. Proc.
uropathogenic Escherichia coli, is a vacuolating cytotoxin for bladder Natl. Acad. Sci. U. S. A. 101, 1333–1338
and kidney epithelial cells. Infect. Immun. 70, 4539–4546 43 Gunther, I.N. et al. (2002) Assessment of virulence of uropathogenic
19 Heimer, S.R. et al. (2004) Autotransporter genes pic and tsh are Escherichia coli type 1 fimbrial mutants in which the invertible
associated with Escherichia coli strains that cause acute pyelone- element is phase-locked on or off. Infect. Immun. 70, 3344–3354
phritis and are expressed during urinary tract infection. Infect. 44 Martinez, J.J. et al. (2000) Type 1 pilus-mediated bacterial invasion of
Immun. 72, 593–597 bladder epithelial cells. EMBO J. 19, 2803–2812
20 Stanley, P. et al. (1998) Acylation of Escherichia coli hemolysin: a 45 Lewis, S.A. (2000) Everything you wanted to know about the bladder
unique protein lipidation mechanism underlying toxin function. epithelium but were afraid to ask. Am. J. Physiol. Renal Physiol. 278,
Microbiol. Mol. Biol. Rev. 62, 309–333 F867–F874
21 Uhlen, P. et al. (2000) Alpha-haemolysin of uropathogenic E. coli 46 Thomas, W.E. et al. (2002) Bacterial adhesion to target cells enhanced
induces Ca2C oscillations in renal epithelial cells. Nature 405, by shear force. Cell 109, 913–923
694–697 47 Martinez, J.J. and Hultgren, S.J. (2002) Requirement of Rho-family
22 Mills, M. et al. (2000) Cytotoxic necrotizing factor type 1 of GTPases in the invasion of Type 1-piliated uropathogenic Escherichia
uropathogenic Escherichia coli kills cultured human uroepithelial coli. Cell. Microbiol. 4, 19–28
5637 cells by an apoptotic mechanism. Infect. Immun. 68, 5869–5880 48 Donlan, R.M. and Costerton, J.W. (2002) Biofilms: survival mechan-
23 Hofman, P. et al. (2000) Escherichia coli cytotoxic necrotizing factor-1 isms of clinically relevant microorganisms. Clin. Microbiol. Rev. 15,
(CNF-1) increases the adherence to epithelia and the oxidative burst 167–193
of human polymorphonuclear leukocytes but decreases bacteria 49 Dunne, W.M., Jr. (2002) Bacterial adhesion: seen any good biofilms
phagocytosis. J. Leukoc. Biol. 68, 522–528 lately? Clin. Microbiol. Rev. 15, 155–166
www.sciencedirect.com
430 Review TRENDS in Microbiology Vol.12 No.9 September 2004

50 O’Toole, G. et al. (2000) Biofilm formation as microbial development. 56 Chapman, M.R. et al. (2002) Role of Escherichia coli curli operons in
Annu. Rev. Microbiol. 54, 49–79 directing amyloid fiber formation. Science 295, 851–855
51 Webb, J.S. et al. (2003) Bacterial biofilms: prokaryotic adventures in 57 Brombacher, E. et al. (2003) The curli biosynthesis regulator CsgD
multicellularity. Curr. Opin. Microbiol. 6, 578–585 co-ordinates the expression of both positive and negative determi-
52 Mah, T.F. et al. (2003) A genetic basis for Pseudomonas aeruginosa nants for biofilm formation in Escherichia coli. Microbiol. 149,
biofilm antibiotic resistance. Nature 426, 306–310 2847–2857
53 Pratt, L.A. and Kolter, R. (1998) Genetic analysis of Escherichia coli 58 Danese, P.N. et al. (2000) Exopolysaccharide production is required for
biofilm formation: roles of flagella, motility, chemotaxis and type I pili. development of Escherichia coli K-12 biofilm architecture.
Mol. Microbiol. 30, 285–293 J. Bacteriol. 182, 3593–3596
54 Danese, P.N. et al. (2000) The outer membrane protein, antigen 43, 59 Stone, G. et al. (2002) Tetracycline rapidly reaches all the constituent
mediates cell-to-cell interactions within Escherichia coli biofilms. Mol. cells of uropathogenic Escherichia coli biofilms. Antimicrob. Agents
Microbiol. 37, 424–432
Chemother. 46, 2458–2461
55 Prigent-Combaret, C. et al. (2001) Complex regulatory network
controls initial adhesion and biofilm formation in Escherichia coli
via regulation of the csgD gene. J. Bacteriol. 183, 7213–7223

Have you contributed to an Elsevier publication?


Did you know that you are entitled to a 30% discount on books?
A 30% discount is available to ALL Elsevier book and journal contributors when ordering books or stand-alone CD-ROMs directly
from us.
To take advantage of your discount:
1. Choose your book(s) from www.elsevier.com or www.books.elsevier.com
2. Place your order
Americas:
TEL: +1 800 782 4927 for US customers
TEL: +1 800 460 3110 for Canada, South & Central America customers
FAX: +1 314 453 4898
E-MAIL: author.contributor@elsevier.com
All other countries:
TEL: +44 1865 474 010
FAX: +44 1865 474 011
E-MAIL: directorders@elsevier.com
You’ll need to provide the name of the Elsevier book or journal to which you have contributed. Shipping is FREE on pre-paid
orders within the US, Canada, and the UK.
If you are faxing your order, please enclose a copy of this page.
3. Make your payment
This discount is only available on prepaid orders. Please note that this offer does not apply to multi-volume reference works or
Elsevier Health Sciences products.

www.books.elsevier.com

www.sciencedirect.com

You might also like