Вы находитесь на странице: 1из 8

REVIEW

10.1111/j.1469-0691.2010.03213.x

Pathogenesis and immunity in enterococcal infections


I. G. Sava1, E. Heikens2 and J. Huebner1
1) Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Germany and 2) Department of Medical Microbiology,
University Medical Center Utrecht, The Netherlands

Abstract
Enterococcus faecalis and Enterococcus faecium have emerged as multi-resistant nosocomial pathogens in immunocompromised and critically ill patients. Multi-resistant strains have acquired virulence genes resulting in hospital-adapted clones. The following review summarizes several proteins and carbohydrate- or glycoconjugates that have been identied as putative virulence factors involved in the
pathogenesis of enterococcal infections and may be used as targets for alternative therapies. Several studies describing the host immune
response against enterococci are also summarized.
Keywords: Enterococcus faecalis, Enterococcus faecium, immunity, review, vaccine, virulence factors
Clin Microbiol Infect 2010; 16: 533540
Corresponding author and reprint requests: J. Huebner, Division
of Infectious Diseases, Department of Medicine, University Medical
Center Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
E-mail: johannes.huebner@uniklinik-freiburg.de

Introduction
Enterococci have until recently been considered as harmless
commensals, especially when compared to other more virulent Gram-positive pathogens. However, there is growing
evidence that these bacteria frequently possess several specic traits that enable them to survive in the hospital environment, colonize patients, and cause infections such as
bacteraemia, peritonitis, endocarditis and urinary tract,
wound, and device-related infections. Several recent studies
have indentied putative virulence factors in E. faecalis, but
also in E. faecium, albeit to a lesser extent (see Table 1). A
better understanding of the factors that enable enterococci
to cause infections and of the host immune response to
these pathogens can contribute to the development of new
therapeutic and prophylactic approaches, which may help to
prevent nosocomial spread of and infections with these
bacteria.

Aggregation Substance
Aggregation substance (AS) is the description used for a
group of surface proteins encoded on pheromone-inducible
conjugative plasmids. AS promotes conjugation by directing

bacterial aggregation, resulting in close cell contact between


donor and recipient [1]. Expression of AS proteins is induced
by a peptide pheromone, which is secreted by plasmid-free
recipient cells [2,3], but can also be induced by a host factor
during in vivo growth [4,5]. Asa1, Asp1, and Acs10 are the
best studied AS proteins and show over 90% amino acid
sequence identity [1,6,7]. AS proteins contain an N-terminal
domain, a variable region, a central domain and two Arg-GlyAsp (RGD) motifs. For Asc10 it has been demonstrated that
both the N-terminal domain and the central domain are
required for aggregation [8]. The N-terminal aggregation
domain promoted binding to cell wall lipoteichoic acid (LTA)
whereas the central aggregation domain did not. Besides its
role in conjugation, AS has also that of a virulence factor in
E. faecalis. Asa1 increased adherence to renal tubular cells
[9] and adherence to and survival in human macrophages
[10]. Asc10 increased internalization [11] and intracellular
survival [12] in polymorphonuclear leukocytes (PMNs). The
RGD motifs probably mediate the interactions with eukaryotic cells [10,11]. Both Asc10 and Asa1 are involved in
adherence to several extracellular matrix proteins [13] and
both increased virulence of E. faecalis in a rabbit endocarditis
model [5,14,15]. For Asc10 it has been demonstrated that
the N-terminal aggregation domain and the RGD motif contribute to virulence during experimental endocarditis [16].
AS did not contribute to virulence in a mouse model of

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases

534

CMI

Clinical Microbiology and Infection, Volume 16 Number 6, June 2010

TABLE 1. Putative virulence factors in E. faecalis and E. faecium

E. faecium (Efm)
Esp

Acm

Pili
PilA
PilB
Scm
EcbA
E. faecalis (Efa)
AS
Asa1
Asp1
Asc10

Esp
Gelatinase/FSR

Pili
ebp locus

bee locus
Ace
cps locus
epa locus

LTA
WTA
Glycolipids

Pathophysiology/Virulence

Epidemiology

References

Biolm formation
Pathogenesis of rat endocarditis
Pathogenesis of mouse UTIa
Antigenic in humans during endocarditis and bacteraemia
Binding to collagen type I, IV
Pathogenesis of rat endocarditis
Antigenic in humans during endocarditis

Specically linked to HAb Efm

Wide spread among Efm

[34,35], [109th General


Meeting of the American
Society of Microbiology,
Posters B-167 and B-211]
[66,67,70]

Not known

Wide spread among Efm

[53]

Binding to collagen type V


Binding to collagen type V
Binding to brinogen

Widespread among Efm


Specically linked to HA Efm

[59]
[68,69]

Promotes conjugation by directing bacterial aggregation


Internalization into intestinal epithetial cells
Binding to renal tubular cells
Binding to and survival in macrophages
Binding to ECM
Pathogenesis of rabbit endocarditis
Internalization into and survival in PMNsc
Binding to ECM
Pathogenesis of rabbit endocarditis
Biolm formation
Pathogenesis of mouse UTI
Biolm formation
Pathogenesis of mouse peritonitis
Caenorhabditis elegans infection
Pathogenesis of rabbit endophthalmitis

Widespread among Efa

[1,1820]

Biolm formation
Pathogenesis of endocarditis
Pathogenesis of UTI
Antigenic in humans during endocarditis
Biolm formation
Binding to collagen type I, IV
Binding to laminin and dentin
Resistance to complement and PMNs-mediated killing
Biolm formation
Enterocyte translocation
Resistance to killing by PMNs
Resistance to infection by phages
Pathogenesis of mouse peritonitis
Pathogenesis of mouse UTI
Target of opsonic antibodies
Not known
Biolm formation
Binding to colonic epithelial cells
Pathogenesis of mouse sepsis

[9,10]
[1315]
[5,1115]

Widespread among Efa

[2427]

Widespread among Efa

[40,4245]

Widespread among Efa

[52,56,58]

Among 5% of Efa
Widespread among Efa

[54]
[6063]

Efa serotype C and D strains


Present in Efa cell wall

[73,8587]
[79,8284]

Present in enterococcal cell wall


Present in enterococcal cell wall
Present in enterococcal cell membrane

[74]
[72]
[89,90]

UTI, urinary tract infection.


HA, hospital-associated.
PMNs, polymorphonuclear leukocytes.

b
c

urinary tract infection [17]. However, AS proteins did facilitate bacterial internalization by different cultured intestinal
epithelial cells [1820], indicating that they might be involved
in the translocation of E. faecalis through the intestinal epithelia, leading to systemic infection.

Enterococcal Surface Protein


One putative virulence determinant is the enterococcal surface protein gene esp located on a pathogenicity island (PAI)
in both E. faecalis and E. faecium [21,22]. Esp proteins are
expressed in both species at the surface of the bacterium.
They contain an N-terminal signal sequence and an N-termi-

nal domain followed by three repeat domains designated A,


B, and C, with the number of repeats varying among strains
[21,23]. The C-terminal end of Esp consists of a (Y/F)PxTG
motif, which is presumably responsible for cell-wall anchoring.
In E. faecalis, Esp has been identied as a putative virulence factor involved in colonization of the urinary tract
[24]. Conicting results have been reported about the role
of E. faecalis Esp in biolm formation, ranging from signicant loss of biolm formation in E. faecalis Esp-decient
mutants [25,26] to no apparent effect [27]. Also, no correlation was found in different studies between the presence
or absence of esp in clinical isolates and biolm formation
[2831].

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

CMI

Sava et al. Pathogenesis and immunity in enterococcal infections

Recently, more information has been presented about the


function of E. faecium Esp. Interestingly, in contrast to E. faecalis esp, which is widely spread among strains, E. faecium esp
is predominantly present in hospital-acquired isolates [21,32],
suggesting a role in virulence. It has been shown that esp
expression is affected by changes in environmental conditions, being highest at 37C and under anaerobiosis [33].
Furthermore, expression of esp was correlated with initial
adherence to polystyrene and biolm formation [33]. By constructing an esp-decient mutant and by using different animal models, Heikens et al. [34] demonstrated that Esp of
E. faecium is involved in biolm formation and contributes to
the pathogenesis of experimental endocarditis [109th General Meeting of the American Society of Microbiology, Poster
B-167], urinary tract infection (UTI) [35], and bacteraemia
[109th General Meeting of the American Society of Microbiology, Poster B-211]. No specic role for Esp was found in
peritonitis [35] and colonization of the gastro-intestinal tract
[36]. The importance of E. faecium Esp in UTI and endocarditis probably results from the fact that these models represent typical biolm-associated infection models indicating a
specic role of Esp in the pathogenesis of E. faecium infections.

Gelatinase and fsr Two-Component System


One well-studied virulence factor of E. faecalis is a secreted
bacterial protease called gelatinase which is controlled by the
fsr two-component system comprised of four genes (fsrA,
fsrB, fsrD, and fsrC) [37]. The fsr system responds to the
extracellular accumulation of a peptide lactone, [38] which is
encoded by frsD. It is believed that this peptide is processed
from the FsrD protein and is exported to the extracellular
space by the gene product of fsrB [37]. Extracellular accumulation of the peptide is probably sensed by the FsrC histidine
kinase, subsequently leading to activation of the response
regulator and transcription factor FsrA by a phosphoryl
group. Phosphorylated FsrA activates expression of two
cotranscribed protease genes, gelE and sprE, located downstream of the fsr locus and encoding gelatinase and serine
protease [39]. Microarray results demonstrated that the fsr
locus activates expression of a variety of other genes potentially involved in virulence and metabolic pathways [40].
Mutations in the fsr operon and inactivation of the fsr-controlled gene gelE demonstrated that gelatinase has an important role in biolm formation by E. faecalis, and that the fsr
system controls biolm formation through production of
gelatinase [29]. It was also demonstrated that gelatinase
enzymatic activity is required for biolm formation [29]

535

which is probably established by C-terminal processing of


the gelatinase protein [41]. Besides their role in biolm formation, gelatinase and the fsr system have been implicated in
virulence in different animal infection models, including
mouse peritonitis [42,43], Caenorhabditis elegans infection
[44] and rabbit endophthalmitis [45]. Epidemiological data
indicate involvement of gelatinase and the fsr locus in virulence [4648]. Together these data suggest an important role
of the fsr system in the virulence of E. faecalis.

Pili
Pili of Gram-positive bacteria have been implicated in adhesion to multiple types of human cells and in biolm formation, two processes critical in the pathogenesis of many
bacterial diseases [4952]. E. faecalis and E. faecium harbour
pilin gene clusters (PGCs), comprising genes encoding surface proteins with LPxTG-like motifs and sortases, which are
required for pilus assembly [52,53]. E. faecalis harbors two
PGCs, designated as the ebp locus (endocarditis and biolmassociated pili) [52] and the bee locus (biolm enhancer in
enterococci) [54].
Ebp pili seem to be involved in initial adherence and biolm formation [52]. They contribute to the pathogenesis of
experimental endocarditis [52] and urinary tract infection
[55,56] which are both biolm-associated infections. In addition, it was found that Ebp pili are antigenic in humans during
endocarditis [57]. The bee locus is located on a conjugative
plasmid and was only detected in 5% of the E. faecalis isolates, whereas the ebp operon was found in almost all of the
isolates [58]. E. faecium harbours four PGCs, designated
PGC-1 to -4 [53]. Expression of PilA and PilB was demonstrated in a hospital-acquired E. faecium isolate, proving that
E. faecium can express two different types of pili at the surface of a single cell [53]. It remains to be determined
whether these pili are implicated in virulence of E. faecium;
however, their genes display high similarity with genes of the
ebp operon of E. faecalis, suggesting a possible role of these
pili in interactions with the mammalian host.

Enterococcal MSCRAMMs
Colonization of human tissues is assumed to occur via interactions between specic proteins in the extracellular matrix
and the so-called MSCRAMMs (microbial surface components recognizing adhesive matrix molecules). The publicly
available genome sequences of E. faecalis V583 and E. faecium
TX0016 revealed the presence of 17 and 15 MSCRAMMs,

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

536

Clinical Microbiology and Infection, Volume 16 Number 6, June 2010

respectively [57,59]. So far, seven enterococcal MSCRAMMs


have been characterized in detail: Ace (adhesion of collagen
of E. faecalis), Fss1, Fss2, and Fss3 (E. faecalis surface protein)
of E. faecalis and Acm (adhesin of collagen of E. faecium),
Scm (second collagen adhesin of E. faecium) and EcbA (E. faecium collagen binding protein A) of E. faecium. Ace was
shown to bind to collagen types I and IV, laminin and dentin
[6062]. Furthermore, Ace was shown to be involved in the
pathogenesis of experimental endocarditis. It was demonstrated that rats immunized against recombinant Ace (rAce)
or given specic anti-rAce antibodies were less likely to
develop E. faecalis endocarditis [63]. Fss1, Fss2, and Fss3 bind
to brinogen [64,65], targeting however different brinogen
polypeptide chains.
Acm binds to collagen type I and to a lesser extent to collagen type IV [66], contributing to the pathogenesis of experimental endocarditis and being antigenic in humans during
endocarditis [67]. Both Scm and EcbA bind to collagen type
V [59,68] and EcbA also binds to brinogen [68]. The presence of ecbA is associated with hospital-acquired E. faecium
isolates [69]. The other MSCRAMMs are widely found
among clinical and non-clinical isolates. Interestingly, a functional acm gene is predominantly present in clinical isolates,
whereas a non-functional acm gene, containing an insertion
element, is mainly present in non-clinical isolates [70].

Enterococcal Cell Wall and Capsular


Polysaccharides
The bacterial cell wall represents a complex structure allowing bacteria to interact with their environment and providing
the factors needed for growth in their ecological niche. In
the infected host it separates the interior of the bacteria
from harmful inuences exerted by the surrounding milieu
[71]. The enterococcal cell wall contains peptidoglycan, proteins, polysaccharides, lipids, lipoproteins and several glycoconjugates. The most abundant glycopolymers are wall
teichoic acid (WTA) and lipoteichoic acid (LTA). WTA is a
polymer usually consisting of a glycosylated and D-alaninesubstituted alditol phosphate repeating unit covalently
attached to the peptidoglycan via a linkage unit [72]. So far,
the contribution of this molecule to enterococcal pathogenesis has not been investigated.
Hufnagel et al. [73] grouped E. faecalis strains into four
capsular serotypes (AD). It was recently shown that the
antibodies used to classify serotype A strains actually recognize LTA [74]. Unpublished observations (Sava, personal
observations) based on cross-reactivity in ELISA and opsonophagocytosis assays showed that the predominant antigen of

CMI

serotype B strains is also LTA. Investigations of the role of


LTA in enterococcal pathogenicity were hampered by the
fact that mutants lacking LTA are lethal [75].
However, information obtained from mutants decient in
alanination of LTA indicated that esterication of LTA with
D-alanine changes the net charge of this polymer, the defect
in D-alanination being associated in E. faecalis with higher
susceptibility to antimicrobial peptides and with reduced ability to form biolm and to adhere to eukaryotic cells [76].
Several studies have demonstrated the presence of an
enterococcal polysaccharide antigen, designated Epa, in the
E. faecalis cell wall that seems to be recognized by sera from
patients with systemic enterococcal infections [7780]. The
epa locus consists of 16 open reading frames, and the polysaccharide produced by the enzymes encoded by this locus
is thought to be a rhamnose-containing polysaccharide that is
widespread among E. faecalis strains. The structure of the
Epa polysaccharide has not yet been elucidated and preliminary information indicates that its localization might be inside
the bacterial cell wall [80]. This carbohydrate seems to play
an important role in enterococcal pathogenicity, since disruption of different genes of the epa locus results in decreased
biolm formation [81] and enterocyte translocation [82],
lower resistance to killing by PMNs [83], higher bacterial
susceptibility to infection by phages [84] and reduced virulence in mouse peritonitis [79] and urinary tract infection
[84] models.
In the serotyping system described by Hufnagel et al. [73]
it was shown that serotype C and D strains are resistant to
opsonophagocytosis by antiserum directed against the serotype A strains (i.e. anti-LTA serum), suggesting the presence
of a capsule that masks LTA and thereby prevents opsonisation by anti-LTA antibodies. Bacterial capsules are a major
determinant of virulence for many pathogens, contributing to
evasion of the hosts immune system by preventing complement activation and/or killing by phagocytes. Several studies
[8587] conrmed the ndings of Hufnagel et al. regarding
the synthesis of a capsule by E. faecalis serotype C and D
strains, but not by serotypes A and B strains. A capsule locus
in E. faecalis, designated cps, contains nine genes (cpsC
cpsK). CpsF encodes a putative glucosyltransferase which is
probably responsible for the serodiversity between serotype
C and D strains, modifying the ratio between glucose and
galactose in the two types of capsules [86]. It was shown in
a recent study that the polysaccharides produced by the
enzymes encoded by the cps locus contribute to enterococcal virulence by conferring resistance to complementmediated opsonophagocytosis of serotype C and D strains [87].
However, to date, the chemical structures of these capsular
polysaccharides have not been published.

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

CMI

Sava et al. Pathogenesis and immunity in enterococcal infections

Regarding E. faecium, published information about capsular


polysaccharides is completely lacking. In a study by Huebner
et al. [88] about one-third of the clinical isolates tested were
efciently killed by anti-LTA antibodies, suggesting that these
strains do not express a capsule.

Glycolipids
Recent studies demonstrated an important role of enterococcal membrane glycolipids in bacterial virulence [89,90]. These
molecules are involved in the formation of a permeable layer
between the cytoplasm and the environment. In many
Gram-positive bacteria they also function as anchor of LTA,
representing the hydrophobic part of this amphiphilic polymer
[91,92]. A specic glycolipid, a-diglycosyl-diacylglycerol (DGlcDAG), is the most abundant in E. faecalis, accounting for 8
37% of all polar lipids of the cell membrane. Other glycolipids
are mostly a-monoglycosyl-diacylglycerol (MGlcDAG) and
small traces of a-triglycosyl-diacylglycerol (TGlcDAG) [91,92].
DGlcDAG, either by itself or as part of the LTA molecule, has
recently been demonstrated to specically recognize and bind
the glycosaminoglycans heparin and heparan sulfate present
on the surface and/or in the extracellular matrix of human
colonic cells, representing an important molecule involved in
bacterial adhesion to host tissues [89].
An E. faecalis mutant unable to produce DGlcDAG,
through a deletion in the biolm-associated glycolipid synthesis A gene (bgsA) encoding a putative glucosyltransferase,
showed reduced ability to adhere to enterocytes and to
form biolm on plastic surfaces [90].
The DGIcDAG-decient mutant was cleared more rapidly
from the bloodstream of mice compared to wild-type bacteria
in a bacteraemia model. Since both strains are complementresistant, differ very little in their sensitivity to opsonophagcytic killing and are equally susceptible to antimicrobial peptides,
this suggests that persistence of E. faecalis in the blood stream
might be related to its ability to form biolm [90], conrming
previous studies using a mouse bacteraemia model. Deletion
mutants of the sugar-binding transcriptional regulator bopD
[93] and of the D-alanine-poly(phosphoribitol) ligase dltA
(unpublished observations) genes in E. faecalis, which are also
defective in biolm formation, were also less virulent in the
mouse bacteraemia model.

Host Immune Response Against Enterococci


Several recent studies investigated the host immune
response, in particular to E. faecium. Using a non-lethal

537

peritonitis model in mice it was determined that the normal


immune system reacts to E. faecium infection with a rapid
neutrophil inux into the peritoneal cavity causing a consecutive rapid decline in peritoneal and systemic enterococcal
load. Recognition by TLR2 (Toll-like receptor 2) and subsequent signaling through MyD88 [94], peritoneal macrophages
[95], neutrophil inux [96] and opsonization of bacteria by
complement [97] were all found to be important in the early
containment of E. faecium peritonitis. Depletion of these cells
and molecules caused prolonged high peritoneal loads of
E. faecium with subsequent more serious systemic infection.
Interestingly, eventually all mice were able to clear the bacteria. Apparently, in the healthy host, deciency of one part of
the immune system is counterbalanced by other components
of the immune system. This parallels the clinical situation
where especially severely ill, immunocompromised patients
are vulnerable to infections with enterococci.
To mimic this scenario, mice were rendered immunocompromised by inducing a sterile acute-phase response (APR)
or a sepsis before causing E. faecium peritonitis.
Both APR and sepsis impaired the host defence against
E. faecium suggesting that these conditions may contribute to
the increased vulnerability of critically ill patients to enterococcal infections [98,99].

Conclusions
In this review an overview was given describing cell surface
determinants and structures potentially important in enterococcal pathogenicity and immunity.
Most putative virulence factors found so far in E. faecalis
and E. faecium are involved in adherence to extracellular
structures and biolm formation, important processes in initiating colonization of and infection in the host. In healthy
mice, different components of the normal immune system,
including TLRs, neutrophils, macrophages, and the complement system were found to be important in containing
E. faecium infections. Deciency of one of these components
did not necessarily result in inability to clear the infection,
suggesting compensation of one component by the other.
This parallels the clinical situation demonstrating the moderate virulent nature of E. faecium which is usually not capable
to cause overwhelming infections in the healthy host.
Different surface structures have been identied in E. faecalis and E. faecium that may be promising candidates for the
development of alternative treatment and prevention strategies against enterococcal infections. Only few of them have
been tested in appropriate animal models. Cell-wall carbohydrate antigens (such as LTA and capsular polysaccharides)

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

538

CMI

Clinical Microbiology and Infection, Volume 16 Number 6, June 2010

and the enterococcal MSCRAMM Ace have been shown to


be a target of protective antibodies. Several additional protein antigens could also be targets of a protective immune
response, although appropriate protection studies have not
been published so far. Interference with the expression of
specic virulence factors could be exploited for novel treatments, although, to date, no precedent of such a therapeutic
approach has been tested clinically. Some of these new
developments may eventually offer the prospect of replacing
the increasingly obsolete classes of antibiotics against which
enterococci have, so successfully, developed resistances.

11.

12.

13.

14.

Transparency Declaration
The authors have declared that no conict of interest exists.

References
1. Olmsted SB, Kao SM, van Putte LJ, Gallo JC, Dunny GM. Role of the
pheromone-inducible surface protein Asc10 in mating aggregate formation and conjugal transfer of the Enterococcus faecalis plasmid
pcf10. J Bacteriol 1991; 173: 76657672.
2. Dunny GM, Leonard BA, Hedberg PJ. Pheromone-inducible conjugation in Enterococcus faecalis: Interbacterial and host-parasite chemical
communication. J Bacteriol 1995; 177: 871876.
3. Leonard BA, Podbielski A, Hedberg PJ, Dunny GM. Enterococcus
faecalis pheromone binding protein, Prgz, recruits a chromosomal
oligopeptide permease system to import sex pheromone Ccf10 for
induction of conjugation. Proc Natl Acad Sci USA 1996; 93: 260
264.
4. Chandler JR, Hirt H, Dunny GM. A paracrine peptide sex pheromone
also acts as an autocrine signal to induce plasmid transfer and virulence factor expression in vivo. Proc Natl Acad Sci USA 2005; 102:
1561715622.
5. Hirt H, Schlievert PM, Dunny GM. In vivo induction of virulence and
antibiotic resistance transfer in Enterococcus faecalis mediated by the
sex pheromone-sensing system of pCF10. Infect Immun 2002; 70:
716723.
6. Galli D, Lottspeich F, Wirth R. Sequence analysis of Enterococcus faecalis aggregation substance encoded by the sex pheromone plasmid
pAD1. Mol Microbiol 1990; 4: 895904.
7. Galli D, Friesenegger A, Wirth R. Transcriptional control of sexpheromone-inducible genes on plasmid pAD1 of Enterococcus faecalis
and sequence analysis of a third structural gene for (pPD1-encoded)
aggregation substance. Mol Microbiol 1992; 6: 12971308.
8. Waters CM, Hirt H, McCormick JK, Schlievert PM, Wells CL, Dunny
GM. An amino-terminal domain of Enterococcus faecalis aggregation
substance is required for aggregation, bacterial internalization by epithelial cells and binding to lipoteichoic acid. Mol Microbiol 2004; 52:
11591171.
9. Kreft B, Marre R, Schramm U, Wirth R. Aggregation substance of
Enterococcus faecalis mediates adhesion to cultured renal tubular cells.
Infect Immun 1992; 60: 2530.
10. Sussmuth SD, Muscholl-Silberhorn A, Wirth R, Susa M, Marre R,
Rozdzinski E. Aggregation substance promotes adherence, phagocytosis, and intracellular survival of Enterococcus faecalis within human

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

macrophages and suppresses respiratory burst. Infect Immun 2000;


68: 49004906.
Vanek NN, Simon SI, Jacques-Palaz K, Mariscalco MM, Dunny GM,
Rakita RM. Enterococcus faecalis aggregation substance promotes
opsonin-independent binding to human neutrophils via a complement
receptor type 3-mediated mechanism. FEMS Immunol Med Microbiol
1999; 26: 4960.
Rakita RM, Vanek NN, Jacques-Palaz K et al. Enterococcus faecalis
bearing aggregation substance is resistant to killing by human neutrophils despite phagocytosis and neutrophil activation. Infect Immun
1999; 67: 60676075.
Rozdzinski E, Marre R, Susa M, Wirth R, Muscholl-Silberhorn A.
Aggregation substance-mediated adherence of Enterococcus faecalis to
immobilized extracellular matrix proteins. Microb Pathog 2001; 30:
211220.
Chow JW, Thal LA, Perri MB et al. Plasmid-associated hemolysin and
aggregation substance production contribute to virulence in experimental enterococcal endocarditis. Antimicrob Agents Chemother 1993;
37: 24742477.
Schlievert PM, Gahr PJ, Assimacopoulos AP et al. Aggregation and
binding substances enhance pathogenicity in rabbit models of Enterococcus faecalis endocarditis. Infect Immun 1998; 66: 218223.
Chuang ON, Schlievert PM, Wells CL, Manias DA, Tripp TJ, Dunny
GM. Multiple functional domains of Enterococcus faecalis aggregation
substance Asc10 contribute to endocarditis virulence. Infect Immun
2009; 77: 539548.
Johnson JR, Clabots C, Hirt H, Waters C, Dunny G. Enterococcal
aggregation substance and binding substance are not major contributors to urinary tract colonization by Enterococcus faecalis in a mouse
model of ascending unobstructed urinary tract infection. Infect Immun
2004; 72: 24452448.
Waters CM, Wells CL, Dunny GM. The aggregation domain of
aggregation substance, not the RGD motifs, is critical for efcient
internalization by HT-29 enterocytes. Infect Immun 2003; 71: 5682
5689.
Sartingen S, Rozdzinski E, Muscholl-Silberhorn A, Marre R. Aggregation substance increases adherence and internalization, but not translocation, of Enterococcus faecalis through different intestinal epithelial
cells in vitro. Infect Immun 2000; 68: 60446047.
Wells CL, Moore EA, Hoag JA, Hirt H, Dunny GM, Erlandsen
SL. Inducible expression of Enterococcus faecalis aggregation substance
surface protein facilitates bacterial internalization by cultured
enterocytes. Infect Immun 2000; 68: 71907194.
Leavis H, Top J, Shankar N et al. A novel putative enterococcal
pathogenicity island linked to the esp virulence gene of Enterococcus
faecium and associated with epidemicity. J Bacteriol 2004; 186: 672
682.
Shankar N, Baghdayan AS, Gilmore MS. Modulation of virulence
within a pathogenicity island in vancomycin-resistant Enterococcus faecalis. Nature 2002; 417: 746750.
Eaton TJ, Gasson MJ. A variant enterococcal surface protein Esp(fm) in
Enterococcus faecium; distribution among food, commensal, medical, and
environmental isolates. FEMS Microbiol Lett 2002; 216: 269275.
Shankar N, Lockatell CV, Baghdayan AS, Drachenberg C, Gilmore
MS, Johnson DE. Role of Enterococcus faecalis surface protein Esp in
the pathogenesis of ascending urinary tract infection. Infect Immun
2001; 69: 43664372.
Tendolkar PM, Baghdayan AS, Gilmore MS, Shankar N. Enterococcal
surface protein, Esp, enhances biolm formation by Enterococcus faecalis. Infect Immun 2004; 72: 60326039.
Tendolkar PM, Baghdayan AS, Shankar N. The N-terminal domain of
enterococcal surface protein, Esp, is sufcient for Esp-mediated biolm enhancement in Enterococcus faecalis. J Bacteriol 2005; 187: 6213
6222.

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

CMI

Sava et al. Pathogenesis and immunity in enterococcal infections

27. Toledo-Arana A, Valle J, Solano C et al. The enterococcal surface


protein, Esp, is involved in Enterococcus faecalis biolm formation. Appl
Environ Microbiol 2001; 67: 45384545.
28. Di Rosa R, Creti R, Venditti M et al. Relationship between biolm
formation, the enterococcal surface protein (Esp) and gelatinase in
clinical isolates of Enterococcus faecalis and Enterococcus faecium. FEMS
Microbiol Lett 2006; 256: 145150.
29. Hancock LE, Perego M. Systematic inactivation and phenotypic characterization of two-component signal transduction systems of Enterococcus faecalis V583. J Bacteriol 2004; 186: 79517958.
30. Kristich CJ, Li YH, Cvitkovitch DG, Dunny GM. Esp-independent
biolm formation by Enterococcus faecalis. J Bacteriol 2004; 186: 154
163.
31. Raad II, Hanna HA, Boktour M et al. Vancomycin-resistant Enterococcus faecium: catheter colonization, esp gene, and decreased susceptibility to antibiotics in biolm. Antimicrob Agents Chemother 2005; 49:
50465050.
32. Willems RJ, Homan W, Top J et al. Variant esp gene as a marker of a
distinct genetic lineage of vancomycin-resistant Enterococcus faecium
spreading in hospitals. Lancet 2001; 357: 853855.
33. Van Wamel WJ, Hendrickx AP, Bonten MJ, Top J, Posthuma G, Willems RJ. Growth condition-dependent Esp expression by Enterococcus
faecium affects initial adherence and biolm formation. Infect Immun
2007; 75: 924931.
34. Heikens E, Bonten MJ, Willems RJ. Enterococcal surface protein Esp
is important for biolm formation of Enterococcus faecium E1162. J Bacteriol 2007; 189: 82338240.
35. Leendertse M, Heikens E, Wijnands LM et al. Enterococcal surface
protein transiently aggravates Enterococcus faecium-induced urinary
tract infection in mice. J Infect Dis 2009; 200: 11621165.
36. Heikens E, Leendertse M, Wijnands LM et al. Enterococcal surface
protein Esp is not essential for cell adhesion and intestinal colonization of Enterococcus faecium in mice. BMC Microbiol 2009; 9: 19.
37. Nakayama J, Chen S, Oyama N et al. Revised model for Enterococcus
faecalis fsr quorum-sensing system: the small open reading frame fsrD
encodes the gelatinase biosynthesis-activating pheromone propeptide
corresponding to staphylococcal agrd. J Bacteriol 2006; 188: 8321
8326.
38. Nakayama J, Cao Y, Horii T et al. Gelatinase biosynthesis-activating
pheromone: a peptide lactone that mediates a quorum sensing in
Enterococcus faecalis. Mol Microbiol 2001; 41: 145154.
39. Qin X, Singh KV, Weinstock GM, Murray BE. Characterization of fsr,
a regulator controlling expression of gelatinase and serine protease
in Enterococcus faecalis OG1RF. J Bacteriol 2001; 183: 33723382.
40. Bourgogne A, Hilsenbeck SG, Dunny GM, Murray BE. Comparison of
OG1RF and an isogenic fsrB deletion mutant by transcriptional analysis:
the fsr system of Enterococcus faecalis is more than the activator of
gelatinase and serine protease. J Bacteriol 2006; 188: 28752884.
41. Del Papa MF, Hancock LE, Thomas VC, Perego M. Full activation of
Enterococcus faecalis gelatinase by a C-terminal proteolytic cleavage.
J Bacteriol 2007; 189: 88358843.
42. Qin X, Singh KV, Weinstock GM, Murray BE. Effects of Enterococcus
faecalis fsr genes on production of gelatinase and a serine protease
and virulence. Infect Immun 2000; 68: 25792586.
43. Singh KV, Qin X, Weinstock GM, Murray BE. Generation and testing
of mutants of Enterococcus faecalis in a mouse peritonitis model.
J Infect Dis 1998; 178: 14161420.
44. Sifri CD, Mylonakis E, Singh KV et al. Virulence effect of Enterococcus
faecalis protease genes and the quorum-sensing locus fsr in
Caenorhabditis elegans and mice. Infect Immun 2002; 70: 56475650.
45. Mylonakis E, Engelbert M, Qin X et al. The Enterococcus faecalis fsrB
gene, a key component of the fsr quorum-sensing system, is associated with virulence in the rabbit endophthalmitis model. Infect Immun
2002; 70: 46784681.

539

46. Coque TM, Patterson JE, Steckelberg JM, Murray BE. Incidence of
hemolysin, gelatinase, and aggregation substance among enterococci
isolated from patients with endocarditis and other infections and
from feces of hospitalized and community-based persons. J Infect Dis
1995; 171: 12231229.
47. Garsin DA, Sifri CD, Mylonakis E et al. A simple model host for identifying gram-positive virulence factors. Proc Natl Acad Sci USA 2001;
98: 1089210897.
48. Pillai SK, Sakoulas G, Gold HS et al. Prevalence of the fsr locus in
Enterococcus faecalis infections. J Clin Microbiol 2002; 40: 26512652.
49. Abbot EL, Smith WD, Siou GP et al. Pili mediate specic adhesion of
Streptococcus pyogenes to human tonsil and skin. Cell Microbiol 2007; 9:
18221833.
50. Maisey HC, Hensler M, Nizet V, Doran KS. Group B streptococcal
pilus proteins contribute to adherence to and invasion of brain
microvascular endothelial cells. J Bacteriol 2007; 189: 14641467.
51. Mandlik A, Swierczynski A, Das A, Ton-That H. Corynebacterium
diphtheriae employs specic minor pilins to target human pharyngeal
epithelial cells. Mol Microbiol 2007; 64: 111124.
52. Nallapareddy SR, Singh KV, Sillanpaa J et al. Endocarditis and biolmassociated pili of Enterococcus faecalis. J Clin Invest 2006; 116: 2799
2807.
53. Hendrickx AP, Bonten MJ, van Luit-Asbroek M, Schapendonk CM,
Kragten AH, Willems RJ. Expression of two distinct types of pili by a
hospital-acquired Enterococcus faecium isolate. Microbiology 2008; 154:
32123223.
54. Tendolkar PM, Baghdayan AS, Shankar N. Putative surface proteins
encoded within a novel transferable locus confer a high-biolm
phenotype to Enterococcus faecalis. J Bacteriol 2006; 188: 2063
2072.
55. Kemp KD, Singh KV, Nallapareddy SR, Murray BE. Relative contributions of Enterococcus faecalis OG1RF sortase encoding genes, srtA and
bps (srtC), to biolm formation and a murine model of urinary tract
infection. Infect Immun 2007; 75: 53995404.
56. Singh KV, Nallapareddy SR, Murray BE. Importance of the ebp (endocarditis- and biolm-associated pilus) locus in the pathogenesis of
Enterococcus faecalis ascending urinary tract infection. J Infect Dis
2007; 195: 16711677.
57. Sillanpaa J, Xu Y, Nallapareddy SR, Murray BE, Hook M. A family of
putative MSCRAMMs from Enterococcus faecalis. Microbiology (Reading,
Engl) 2004; 150: 20692078.
58. Cobo Molinos A, Abriouel H, Omar NB, Lopez RL, Galvez A. Detection of ebp (endocarditis- and biolm-associated pilus) genes in
enterococcal isolates from clinical and non-clinical origin. Int J Food
Microbiol 2008; 126: 123126.
59. Sillanpaa J, Nallapareddy SR, Prakash VP et al. Identication and phenotypic characterization of a second collagen adhesin, scm, and genome-based identication and analysis of 13 other predicted
MSCRAMMs, including four distinct pilus loci, in Enterococcus faecium.
Microbiology 2008; 154: 31993211.
60. Rich RL, Kreikemeyer B, Owens RT et al. Ace is a collagen-binding
MSCRAMM from Enterococcus faecalis. J Biol Chem 1999; 274: 26939
26945.
61. Kowalski WJ, Kasper EL, Hatton JF, Murray BE, Nallapareddy SR, Gillespie MJ. Enterococcus faecalis adhesin, Ace, mediates attachment to
particulate dentin. J Endod 2006; 32: 634637.
62. Nallapareddy SR, Qin X, Weinstock GM, Hook M, Murray BE. Enterococcus faecalis adhesin, Ace, mediates attachment to extracellular
matrix proteins collagen type IV and laminin as well as collagen type
I. Infect Immun 2000; 68: 52185224.
63. Singh KV, Nallapareddy SR, Sillanpaa J, Murray BE. Importance of the
collagen adhesin Ace in pathogenesis and protection against Enterococcus faecalis experimental endocarditis. PLoS Pathog 2010; 6:
e1000716.

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

540

Clinical Microbiology and Infection, Volume 16 Number 6, June 2010

64. Sillanpaa J, Nallapareddy SR, Houston J et al. A family of brinogenbinding MSCRAMMs from Enterococcus faecalis. Microbiology 2009;
155: 23902400.
65. Sillanpaa J, Prakash VP, Nallapareddy SR, Murray BE. Distribution of
genes encoding MSCRAMMs and pili in clinical and natural populations of Enterococcus faecium. J Clin Microbiol 2009; 47: 896901.
66. Nallapareddy SR, Weinstock GM, Murray BE. Clinical isolates of
Enterococcus faecium exhibit strain-specic collagen binding mediated
by Acm, a new member of the MSCRAMM family. Mol Microbiol
2003; 47: 17331747.
67. Nallapareddy SR, Singh KV, Murray BE. Contribution of the collagen
adhesin Acm to pathogenesis of Enterococcus faecium in experimental
endocarditis. Infect Immun 2008; 76: 41204128.
68. Hendrickx AP, van Luit-Asbroek M, Schapendonk CM et al. Sgra, a
nidogen-binding LPxTG surface adhesin implicated in biolm formation, and EcbA, a collagen binding MSCRAMM, are two novel adhesins of hospital-acquired Enterococcus faecium. Infect Immun 2009; 77:
50975106.
69. Hendrickx AP, van Wamel WJ, Posthuma G, Bonten MJ, Willems RJ.
Five genes encoding surface-exposed LPxTG proteins are enriched in
hospital-adapted Enterococcus faecium clonal complex 17 isolates.
J Bacteriol 2007; 189: 83218332.
70. Nallapareddy SR, Singh KV, Okhuysen PC, Murray BE. A functional
collagen adhesin gene, acm, in clinical isolates of Enterococcus faecium
correlates with the recent success of this emerging nosocomial pathogen. Infect Immun 2008; 76: 41104119.
71. Seltmann G, Holst O The bacterial cell wall. Berlin, New York:
Springer, 2001.
72. Neuhaus FC, Baddiley J. A continuum of anionic charge: structures
and functions of D-alanyl-teichoic acids in gram-positive bacteria.
Microbiol Mol Biol Rev 2003; 67: 686723.
73. Hufnagel M, Hancock LE, Koch S, Theilacker C, Gilmore MS,
Huebner J. Serological and genetic diversity of capsular polysaccharides in Enterococcus faecalis. J Clin Microbiol 2004; 42: 25482557.
74. Theilacker C, Kaczynski Z, Kropec A et al. Opsonic antibodies to
Enterococcus faecalis strain 12030 are directed against lipoteichoic
acid. Infect Immun 2006; 74: 57035712.
75. Grundling A, Schneewind O. Synthesis of glycerol phosphate lipoteichoic acid in Staphylococcus aureus. Proc Natl Acad Sci U S A 2007;
104: 84788483.
76. Fabretti F, Theilacker C, Baldassarri L et al. Alanine esters of enterococcal lipoteichoic acid play a role in biolm formation and resistance
to antimicrobial peptides. Infect Immun 2006; 74: 41644171.
77. Xu Y, Jiang L, Murray BE, Weinstock GM. Enterococcus faecalis antigens in human infections. Infect Immun 1997; 65: 42074215.
78. Xu Y, Murray BE, Weinstock GM. A cluster of genes involved in
polysaccharide biosynthesis from Enterococcus faecalis OG1RF. Infect
Immun 1998; 66: 43134323.
79. Xu Y, Singh KV, Qin X, Murray BE, Weinstock GM. Analysis of a
gene cluster of Enterococcus faecalis involved in polysaccharide biosynthesis. Infect Immun 2000; 68: 815823.
80. Teng F, Singh KV, Bourgogne A, Zeng J, Murray BE. Further characterization of the epa gene cluster and Epa polysaccharides of Enterococcus faecalis. Infect Immun 2009; 77: 37593767.
81. Mohamed JA, Huang W, Nallapareddy SR, Teng F, Murray BE. Inuence of origin of isolates, especially endocarditis isolates, and various
genes on biolm formation by Enterococcus faecalis. Infect Immun
2004; 72: 36583663.
82. Zeng J, Teng F, Weinstock GM, Murray BE. Translocation of
Enterococcus faecalis strains across a monolayer of polarized human
enterocyte-like T84 cells. J Clin Microbiol 2004; 42: 11491154.

CMI

83. Teng F, Jacques-Palaz KD, Weinstock GM, Murray BE. Evidence


that the enterococcal polysaccharide antigen gene (epa) cluster is
widespread in Enterococcus faecalis and inuences resistance to
phagocytic killing of Enterococcus faecalis. Infect Immun 2002; 70:
20102015.
84. Singh KV, Lewis RJ, Murray BE. Importance of the epa locus of
Enterococcus faecalis OG1RF in a mouse model of ascending urinary
tract infection. J Infect Dis 2009; 200: 417420.
85. Hancock LE, Gilmore MS. The capsular polysaccharide of Enterococcus
faecalis and its relationship to other polysaccharides in the cell wall.
Proc Natl Acad Sci U S A 2002; 99: 15741579.
86. Thurlow LR, Thomas VC, Hancock LE. Capsular polysaccharide
production in Enterococcus faecalis and the contribution of CpsF to
capsule serospecicity. J Bacteriol 2009; 191: 62036210.
87. Thurlow LR, Thomas VC, Fleming SD, Hancock LE. Enterococcus
faecalis capsular polysaccharide serotypes C and D and their contributions to host innate immune evasion. Infect Immun 2009; 77:
55515557.
88. Huebner J, Wang Y, Krueger WA et al. Isolation and chemical characterization of a capsular polysaccharide antigen shared by clinical
isolates of Enterococcus faecalis and vancomycin-resistant Enterococcus
faecium. Infect Immun 1999; 67: 12131219.
89. Sava IG, Zhang F, Toma I et al. Novel interactions of glycosaminoglycans and bacterial glycolipids mediate binding of enterococci to
human cells. J Biol Chem 2009; 284: 1819418201.
90. Theilacker C, Sanchez-Carballo P, Toma I et al. Glycolipids are
involved in biolm accumulation and prolonged bacteraemia in Enterococcus faecalis. Mol Microbiol 2009; 71: 10551069.
91. Fischer W, Laine RA, Nakano M. On the relationship between glycerophosphoglycolipids and lipoteichoic acids in gram-positive bacteria
Ii. Structures of glycerophosphoglycolipids. Biochim Biophys Acta 1978;
528: 298308.
92. Fischer W, Nakano M, Laine RA, Bohrer W. On the relationship
between glycerophosphoglycolipids and lipoteichoic acids in grampositive bacteria I. The occurrence of phosphoglycolipids. Biochim Biophys Acta 1978; 528: 288297.
93. Hufnagel M, Koch S, Creti R, Baldassarri L, Huebner J. A putative
sugar-binding transcriptional regulator in a novel gene locus in Enterococcus faecalis contributes to production of biolm and prolonged
bacteremia in mice. J Infect Dis 2004; 189: 420430.
94. Leendertse M, Willems RJ, Giebelen IA et al. TLR2-dependent
MyD88 signaling contributes to early host defense in murine Enterococcus faecium peritonitis. J Immunol 2008; 180: 48654874.
95. Leendertse M, Willems RJ, Giebelen IA et al. Peritoneal macrophages
are important for the early containment of Enterococcus faecium
peritonitis in mice. Innate Immun 2009; 15: 312.
96. Leendertse M, Willems RJ, Giebelen IA, Roelofs JJ, Bonten MJ, van
der Poll T. Neutrophils are essential for rapid clearance of Enterococcus faecium in mice. Infect Immun 2009; 77: 485491.
97. Leendertse M, Willems RJ, Flierman R, de Vos AF, Bonten MJ, van
der Poll T. The complement system facilitates clearance of Enterococcus faecium during murine peritonitis. J Infect Dis 2010; 201: 544
552.
98. Leendertse M, Willems RJ, Giebelen IA et al. Cecal ligation and puncture induced sepsis impairs host defense against Enterococcus faecium
peritonitis. Intensive Care Med 2009; 35: 924932.
99. Leendertse M, Willems RJ, Giebelen IA, van den Pangaart PS, Bonten
MJ, van der Poll T. The acute-phase response impairs host defence
against Enterococcus faecium peritonitis. Immunology 2009; 128: e335
e342.

2010 The Authors


Journal Compilation 2010 European Society of Clinical Microbiology and Infectious Diseases, CMI, 16, 533540

Вам также может понравиться