You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7072059

Listeria monocytogenes: a multifaceted model. Nat Rev Microbiol 4, 423-434

Article  in  Nature Reviews Microbiology · July 2006


DOI: 10.1038/nrmicro1413 · Source: PubMed

CITATIONS READS

431 1,281

3 authors, including:

Mélanie Anne Hamon Helene Bierne


Institut Pasteur French National Institute for Agricultural Research
34 PUBLICATIONS   3,074 CITATIONS    71 PUBLICATIONS   4,208 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Host/Pathogen Interactions View project

Listeria persistence View project

All content following this page was uploaded by Helene Bierne on 09 May 2014.

The user has requested enhancement of the downloaded file.


REVIEWS
Listeria monocytogenes:
a multifaceted model
Mélanie Hamon*‡§, Hélène Bierne*‡§ and Pascale Cossart*‡§
Abstract | The opportunistic intracellular pathogen Listeria monocytogenes has become
a paradigm for the study of host–pathogen interactions and bacterial adaptation to
mammalian hosts. Analysis of L. monocytogenes infection has provided considerable insight
into how bacteria invade cells, move intracellularly, and disseminate in tissues, as well as
tools to address fundamental processes in cell biology. Moreover, the vast amount of
knowledge that has been gathered through in-depth comparative genomic analyses and
in vivo studies makes L. monocytogenes one of the most well-studied bacterial pathogens.

The Gram-positive bacterium Listeria monocytogenes is has made it an exceptional tool for the study of cellular
a ubiquitous pathogen that thrives in diverse environ- processes such as actin-based motility, growth-factor-
ments such as soil, water, various food products, humans mediated signalling, endocytosis and cellular adhesion.
and animals. The disease caused by this bacterium, Furthermore, available animal models, genetic tools and
listeriosis, is acquired by ingesting contaminated food genomics have facilitated the compilation of informa-
products and mainly affects immunocompromised tion on different aspects of L. monocytogenes biology and
individuals, pregnant women and newborns. Listeriosis have made this bacterium one of the most useful model
manifests as gastroenteritis, meningitis, encephalitis, organisms for the study of bacterial pathogenesis and
mother-to-fetus infections and septicaemia, resulting in pathophysiology.
death in 25–30% of cases. The diverse clinical manifesta-
tions of infection with L. monocytogenes reflect its ability An intracellular bacterium and a cell biologist
to cross three tight barriers in the human host. Following L. monocytogenes is a facultative intracellular bacterium. Its
ingestion, L. monocytogenes crosses the intestinal barrier life cycle reflects its remarkable adaptation to intracellular
by invading the intestinal epithelium, thereby gaining survival and multiplication in macrophages and other cell
access to internal organs. During severe infections, types1,3,4 (FIG. 1). Similar to the situation for most pathogens,
crossing the blood–brain barrier results in infection of the invasion of macrophages by L. monocytogenes is a pas-
the meninges and the brain, and in pregnant women, sive process, but entry into non-professional phagocytes
crossing the fetoplacental barrier leads to infection of is induced by binding of the bacterial surface proteins
the fetus1. internalin A (InlA) and InlB to receptors on the host cell.
L. monocytogenes infection has been a useful model Both of these invasins are necessary and sufficient for bac-
for evaluation of the cellular interactions that are crucial for terial entry into cell types such as enterocytes, hepatocytes,
the initiation of the host T-cell response. However, this fibroblasts, epithelial cells and endothelial cells, but InlA-
aspect of listerial biology is well documented and has mediated entry is restricted to the smaller number of cell
recently been reviewed elsewhere2. This Review highlights types that express its receptor. Entry of L. monocytogenes
*Institut Pasteur, Unité des the many ways in which L. monocytogenes manipulates its into mammalian cells is a dynamic process that requires
Interactions Bactéries mammalian host, and it focuses on the lessons this bact- actin polymerization and membrane remodelling, and
Cellules, Paris 75015, France. erium has taught us in cell biology, bacterial pathophysiol- is an excellent example of how a bacterium can manipu-

Institut National de la Santé
ogy, virulence-factor regulation, and bacterial adaptation late host-cell signalling and endocytic pathways to its
et de la Recherche Médicale,
U604, Paris 75015, France. to the host cytosol. advantage. L. monocytogenes can also harness the actin-
§
Institut National de la Indeed, the ability of L. monocytogenes to invade polymerization machinery in the cytoplasm to facilitate
Recherche Agronomique, and replicate in different cell types has been extensively intracellular and intercellular movement. New mecha-
USC2020, Paris 75015, studied and has revealed the sophisticated relationship nisms by which L. monocytogenes manipulates the host cell
France.
Correspondence to P.C.
between the bacterium and its host. The breadth of are emerging through the use of microarray analyses that
e-mail: pcossart@pasteur.fr information gathered on the elaborate mimicries that aim to determine the genes that are specifically activated
doi:10.1038/nrmicro1413 are used by L. monocytogenes to subvert host processes by bacterial entry into the host cell5,6.

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 423


© 2006 Nature Publishing Group
REVIEWS

Listeria monocytogenes
a

b a

b
c

0.5 µm
c Phagosome

Lysis of
phagosome
and replication d
in cytosol d
0.5 µm
F-actin

0.5 µm

e
e

Double-membraned
vacuole

f 0.5 µm
f

Lysis of vacuole
0.5 µm

Figure 1 | Schematic representation and electron micrographs of the Listeria monocytogenes life cycle.
a | L. monocytogenes induces its entry into a non-professional phagocyte. b | Bacteria are internalized in a vacuole (also
known as a phagosome). c,d | The membrane of the vacuole is disrupted by the secretion of two phospholipases, PlcA and
PlcB, and the pore-forming toxin listeriolysin O. Bacteria are released into the cytoplasm, where they multiply and start to
polymerize actin, as observed by the presence of the characteristic actin tails (see Supplementary information S3 (figure)).
e | Actin polymerization allows bacteria to pass into a neighbouring cell by forming protrusions in the plasma membrane.
f | On entry into the neighbouring cell, bacteria are present in a double-membraned vacuole, from which they can escape to
perpetuate the cycle. F-actin, filamentous actin. Electron micrographs a–c,e–f are reproduced with permission from REF. 113
 (1998) European Molecular Biology Organization, and d is reproduced with permission from REF. 30  (1992) Elsevier.

InlB: subverting cellular-signalling and endocytic the protein-tyrosine-kinase activity of Met, as well
pathways. Binding of InlB to its cellular receptor as the phosphatidylinositol 3-kinase (PI3K) and
Met results in the entry of L. monocytogenes into the Ras–mitogen-activated protein kinase (MAPK)
different cell types. Met is a protein tyrosine kinase, pathways, all of which are required for the uptake
and the endogenous ligand of this receptor is hepato- process7–9. Interestingly, although InlB and HGF both
cyte growth factor (HGF) 7 (FIG. 2) . In vivo, Met is bind and activate Met, InlB does not strictly mimic
expressed mainly by cells of epithelial origin, whereas HGF. Indeed, the kinetics of InlB-induced signalling
HGF is produced mainly by fibroblasts and stromal are different from those of HGF-induced signalling7,
cells. The binding of HGF to Met activates cellular and at an equal concentration, InlB seems to induce
survival and proliferation signals, and it induces a more potent activation of the Ras–MAPK pathway
cytoskeletal rearrangements that function in cellular than does HGF10. Differences in signalling might be
motility and differentiation. Binding of InlB activates explained by the finding that InlB also binds gC1qR

424 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
REVIEWS

a HGF-mediated signalling b InlB-mediated signalling


Listeria monocytogenes
InlB
HGF
GAGs
Met
Out gC1qR Met
Out
PtdIns(4,5)P2 PtdIns(3,4,5)P3
PtdIns(4,5)P2 PtdIns(3,4,5)P3
In
Recycling Cbl Akt PLCγ In Dynamin
PI3K Akt PLCγ
Rac1

Cortactin
Ub CD2AP PI3K ABI
Clathrin- Cbl
mediated Survival CIN85 Cbl Cbl CDC42
GAB1 Rac1

WAVE
endocytosis EPS15 GAB1
SHC CDC42 Ub
SHC
Ras

SP
GRB2

HR
A3

A
GRB2

N-W
GG
SOS
Cytoskeletal
rearrangements F-actin
Clathrin Ras
Degradation
Arp2/3

Nucleus Proliferation

Figure 2 | Met signalling induced by hepatocyte growth factor (HGF) and internalin B (InlB). a | Phosphorylation
of Met leads to the recruitment and activation of many transducers, which in turn recruit cytosolic signalling proteins.
Signalling mediated by HGF activates survival and proliferation signals, and it induces cytoskeletal rearrangements that
are important for cellular motility and differentiation. On stimulation with HGF, the endocytosis of Met, similar to most
signalling receptors, is an important regulatory mechanism that downregulates the cell-surface expression of the activated
receptor. b | Met signalling mediated by the Listeria monocytogenes protein InlB induces cytoskeletal rearrangements that
are important for bacterial entry into non-phagocytic cells. Clathrin components of the endocytic machinery are also
recruited to the site of entry. The link between the cytoskeletal machinery (shown on the right) and the endocytic
machinery (shown on the left) is still unclear. InlB, through the GW repeats at its C terminus, also binds gC1qR (the receptor
for the globular part of complement component C1q) and glucosaminoglycans (GAGs), which are negatively charged
polysaccharides that are present at cell surfaces. Both components might contribute to entry of L. monocytogenes by
modulating the interaction of InlB with Met. ABI, Abl interactor 1; Arp, actin-related protein; CD2AP, CD2-associated protein;
CIN85, Cbl-interacting protein of 85 kDa; EPS15, epidermal-growth-factor-receptor substrate 15; F-actin, filamentous
actin; GAB1, GRB2-associated binding protein 1; GGA3, Golgi-localized, γ-ear-containing, ADP-ribosylation-factor-binding
protein 3; GRB2, growth-factor-receptor-bound protein 2; HRS, HGF-regulated tyrosine-kinase substrate; N-WASP, neural
Wiskott–Aldrich syndrome protein; PI3K, phosphatidylinositol 3-kinase; PLCγ, phospholipase C-γ; PtdIns(3,4,5)P3,
phosphatidylinositol-3,4,5-trisphosphate; PtdIns(4,5)P2, phosphatidylinositol-4,5-bisphosphate; SHC, SRC-homology-2-
domain-containing transforming protein C; SOS, son of sevenless; Ub, ubiquitin; WAVE, Wiskott–Aldrich syndrome protein
(WASP)-family verprolin homologous protein.

(the receptor for the globular part of complement Arp2/3 activation seems to be cell-type dependent, but
component C1q), which might therefore function as it involves a combination of the small GTPases Rac and
a co-receptor for InlB11. Furthermore, InlB and HGF CDC42 and proteins of the Wiskott–Aldrich syndrome
do not compete for binding to Met7, indicating that protein (WASP) family, which includes neural WASP
they might bind distinct sites on Met. These results (N-WASP) and WAVE 12. Proteins of the Ena/VASP
are consistent with the fact that InlB and HGF have family (enabled homologue/vasodilator-stimulated-
no sequence homology and are structurally unrelated. phosphoprotein family), which promote actin-filament
Crystal structures of InlB bound to Met could provide elongation, are also central to the process. In addition,
valuable information about the molecular mechanism cofilin, which is essential for depolymerization of actin
that underlies the activation of Met by InlB. filaments, functions successively as a stimulator and
The signalling pathways that are activated by InlB a downregulator of actin rearrangements that occur
ultimately lead to cytoskeletal rearrangements and during the internalization process 12,13. All of these
entry of L. monocytogenes. How activation of the components that are recruited by the binding of InlB
PI3K and the Ras–MAPK pathways leads to cytoskel- to Met also have a role in growth-factor-receptor
etal rearrangements has been extensively studied, and activation. Therefore, although there are differences
many of the proteins that are crucial for invagination between the kinetics of signalling mediated by InlB
and internalization have been identified. Local actin and HGF, the machinery that is recruited to the site
remodelling at the site of InlB attachment is mediated of activation seems to be the same for both molecules,
by the recruitment and activation of the actin-nucleation showing the utility of L. monocytogenes as a tool to
complex, Arp2/3, which promotes actin nucleation and study cellular signalling by Met or other growth-factor
polymerization (discussed later). The mechanism of receptors.

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 425


© 2006 Nature Publishing Group
REVIEWS

Recently, the study of InlB-induced internaliza- domain of E-cadherin, but it is the intracellular domain
tion revealed an unexpected mechanism used by of E-cadherin that is essential for the cytoskeletal
L. monocytogenes during host-cell entry. L. monocytogenes rearrangements that are required for bacterial entry21.
invades epithelial cells by subverting clathrin-mediated Because all of the components of the endogenous
endocytosis14 (FIG. 2b; see Supplementary information machinery of cell–cell junctions seem to be recruited
S1 (figure)), a process that is used by mammalian cells on binding of InlA, L. monocytogenes is a good system
to take up nutrients and to recycle membrane proteins. for the study of cellular adhesion and identification of
Owing to their size, which is usually 1–3 µm, bacteria components that are involved in this process.
were thought to enter cells through a mechanism related Although it is known that assembly and attachment
to phagocytosis (that is, an actin-dependent mecha- of the E-cadherin, α-catenin and β-catenin complex
nism), which differs from ‘endocytosis’, a process that to the cytoskeleton is central to both intercellular
is thought to internalize particles no larger than 120 nm adhesion and L. monocytogenes entry21, neither the
and that was considered to be actin independent until mechanisms that hold cells together nor the molecu-
recently. Therefore, the finding that L. monocytogenes lar mechanisms that are required for InlA-dependent
can induce its internalization by using clathrin-depend- entry are as yet fully understood. Until recently, the
ent machinery was surprising and indicated that clath- accepted model of intercellular adhesion proposed
rin-coated structures can engulf much larger particles that α-catenin anchors the E-cadherin–β-catenin
than previously thought. Cell-surface expression of complex to the actin cytoskeleton, providing a stable
Met is downregulated by HGF, and similarly, soluble structure that maintains tissue integrity. However, it
InlB induces the degradation of Met, through monou- has become apparent that the dynamics of this process
biquitylation and clathrin-mediated endo cytosis 14. are more complex. As was recently shown, α-catenin
Furthermore, as shown using RNA-interference- cannot simultaneously interact with actin filaments
mediated knockdown, important components of the and the E-cadherin–β-catenin complex, indicating
endocytic machinery are required for internalization that α-catenin is not an actin anchor but is, instead,
of L. monocytogenes. Although the underlying molecu- an actin-filament organizer22,23. Further investigation is
lar mechanisms have not been defined, other invasive required to understand this mechanism fully; however,
bacteria had previously been reported to use clathrin- the dynamic interactions between the cadherin–catenin
mediated entry, implying that this mechanism is not complex and the underlying actin cytoskeleton are
restricted to Listeria species and could be a commonly consistent with the findings that L. monocytogenes can
used mechanism for bacterial entry 15–17. Although regulate and rearrange actin structures at intercellular
the endocytic machinery is important for entry of junctions through adhesion to E-cadherin, and further
L. monocytogenes, this bacterium might exploit other emphasize the validity of the listerial model for analysing
mechanisms, because inhibitors of endocytosis reduce events at intercellular junctions.
bacterial entry but do not completely abolish it14. The validity of using L. monocytogenes as a tool to
Plasma-membrane microdomains known as lipid rafts study cell–cell junction formation was shown by a
have also been shown to be important for the entry of recent study of InlA-dependent entry that identified
L. monocytogenes18. Because lipid rafts are usually asso- the protein ARHGAP10 (Rho GTPase-activating pro-
ciated with clathrin-independent endocytic pathways, tein 10) as a novel cellular component that is involved
this indicates either that L. monocytogenes exploits in the recruitment of α-catenin to cell–cell junctions.
more than one endocytic mechanism or that the sepa- This study also showed that ARHGAP10 was essential
ration among the classes of endocytosis is not as well for listerial entry (see Supplementary information S2
demarcated as conventionally thought. Further work is (figure))24. Overexpression of ARHGAP10 disrupted
necessary to decipher how lipid rafts, clathrin-mediated the cytoskeleton and increased the local concentration
endocytosis and actin-mediated phagocytosis combine of α-catenin, indicating that ARHGAP10 has a direct
to enable listerial entry and cellular invasion. role in regulation of the dynamics of cell–cell junction
formation. Furthermore, ARHGAP10 was shown to
InlA: exploiting intercellular junctions. Similar to InlB, control the activity of RhoA and CDC42, two proteins
InlA induces local cytoskeletal rearrangements in the that regulate cell–cell junction formation.
host cell to stimulate uptake of L. monocytogenes by Components that generate the tension that is required
epithelial cells. InlA is a covalently linked bacterial to hold neighbouring cells together, that is, myosin VIIA
cell-wall protein that binds the host epithelial-cell and its ligand vezatin25, have been found to be impor-
protein E-cadherin19 (FIG. 3). E-cadherin is a transmem- tant for L. monocytogenes entry, indicating that these
brane protein that belongs to a large family of cell–cell components might generate the force that is neces-
adhesion molecules that are required for the correct sary for engulfment of the bacterium through phago-
Adherens junctions formation of adherens junctions between epithelial cytosis26. These findings also indicate that the tension
Together with tight junctions cells. E-cadherin is localized at these cellular junc- that holds two cells together could be similar to the
and desmosomes, these are tions, where its intracellular domain forms a complex tension that is exerted during phagocytosis, as if each
specialized structures that with the cytoskeleton through the catenins (which cell is attempting to engulf its neighbour. An analogous
allow epithelial cells to adhere
to each other, and they have
are cadherin-binding proteins), and its extracellular process known as ‘frustrated phagocytosis’ occurs
epithelial cadherin (E-cadherin) domain is in contact with E-cadherin molecules on when macrophages adhere to immune-complex-coated
as a major component. neighbouring cells 20. InlA binds the extracellular surfaces27.

426 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
REVIEWS

a Intercellular junctions b InlA-induced entry


F-actin
Arp2/3

α G-actin α Formin
α
β β β
Listeria monocytogenes

In
InlA

Out E-cadherin
E-cadherin

Out

In
Out Vezatin
Catenins
β β
In Myosin α β α G-actin
ARHG
VII AP10 α
ARF6

β β β
Catenins
α α α Arp2/3
Formin F-actin

Figure 3 | Adherens junction and internalin A (InlA)-induced bacterial entry. a | Adherens junctions hold adjacent
cells together through the transmembrane protein epithelial cadherin (E-cadherin). The intracellular domain of E-cadherin
recruits α-catenin and β-catenin, and α-catenin bridges the actin cytoskeleton and E-cadherin. Formins, which interact
directly with α-catenin, are also essential for forming actin cables at cell–cell junctions, although the mechanism by which
they achieve this is not understood. b | The receptor for the Listeria monocytogenes protein InlA is E-cadherin. Many
components that are important for adherens junctions are recruited to the site of bacterial entry, where the cytoskeletal
rearrangements that are required for invasion occur. ARF6, ADP-ribosylation factor 6; ARHGAP10, Rho GTPase-activating
protein 10; Arp, actin-related protein; F-actin, filamentous actin; G-actin, globular actin.

Harnessing the actin-polymerization machinery. information S3 (figure)). Polymerization of host actin is


Following internalization into a host-cell vacuole, mediated by the bacterial surface protein ActA, the first
L. monocytogenes lyses the membrane-bound phago- protein that was identified to have functions that promote
some (discussed later) and escapes into the cytoplasm, actin nucleation30–32. It was later found that ActA mimics
where it can polymerize host actin and propel itself its eukaryotic counterparts, proteins of the WASP family
through the cell and into neighbouring cells28. The abil- (which includes N-WASP and WAVE)33. The N-terminal
ity to spread from cell to cell without coming in contact region of ActA, which is essential for actin-based motil-
with the extracellular milieu allows the bacterium to ity 34, has homology to the C-terminal region of WASP,
propagate through tissues and avoid contact with cir- which binds the actin-nucleation complex Arp2/3
culating antibodies or other extracellular bactericidal (REF. 35). Accordingly, both ActA and WASP-family pro-
compounds. At the leading edge of moving cells, the teins function as nucleation-promoting factors (NPFs)
control of actin polymerization is a complex mechanism for the Arp2/3 complex and are involved in forming a
triggered by intricate signalling pathways that take place ternary complex that is composed of Arp2/3, an NPF and
at the plasma membrane. L. monocytogenes bypasses actin. The Arp2/3 complex consists of seven proteins,
these signalling pathways and directly nucleates actin including two proteins that are related to actin, Arp2 and
constitutively, making it a simple and effective model Arp3, which (as a result of their structural similarity to
for studying the dynamics of actin polymerization. actin) are thought to function as a template for polymeri-
L. monocytogenes polymerizes actin asymmetrically zation. Discovery of the Arp2/3 complex ensued from
along its surface, producing an actin tail that propels the studies of ActA partners, thus the role of Arp2/3 as a key
bacterium through the cytoplasm28,29 (see Supplementary actin-nucleating complex was consequently revealed.

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 427


© 2006 Nature Publishing Group
REVIEWS

a b N
used to bypass stringent host species specificity and gen-
Listeria erate relevant model systems to study infection in vivo.
monocytogenes In this respect, L. monocytogenes is a good example of
InlB InlA how in vitro studies can help to generate animal models
Pro16 that more closely reflect human infection.
For many years, the animal model used to study
L. monocytogenes infection was intravenous infection
of mice. This model provides a dose-dependent infec-
tion with dissemination of the bacteria into organs and
was crucial in the discovery of cell-mediated immu-
C nity 3. However, recent molecular evidence showed that
Out the mouse model is inadequate to study the crossing of
barriers that is characteristic of listeriosis. It had long
In Met E-cadherin Met E-cadherin Met E-cadherin been known that oral inoculation of mice (rather than
Guinea pig Mouse Human intravenous infection), which most closely reflects the
and rabbit human mode of infection, is not efficient because only
Figure 4 | Host specificity of Listeria monocytogenes proteins internalin A (InlA) small numbers of L. monocytogenes cross the mouse
and InlB. a | InlA and InlB can bind and induce entry of L. monocytogenes into human intestinal barrier. The reason for this was uncovered
cells that express the respective cell-surface receptors, epithelial cadherin (E-cadherin) by molecular in vitro studies that showed that a single
or Met. However, a single amino-acid change in E-cadherin (at position 16; see b) amino-acid difference in the mouse cellular recep-
prevents InlA from binding mouse E-cadherin, and for unknown reasons, InlB cannot tor for InlA, E-cadherin, prevented it from binding
recognize or activate guinea pig or rabbit Met. b | A diagrammatic representation of the InlA, thereby showing the inadequacy of the mouse
crystal structure of the leucine-rich-repeat region of InlA (purple) bound to E-cadherin
model for study of the invasive role of InlA45 (FIG. 4).
(green) is shown. The position of the crucial proline residue at position 16 of E-cadherin is
Consequently, a novel animal system was developed to
indicated. It is this residue that determines intermolecular recognition and therefore
host specificity. The crystal-structure representation is reproduced with permission from study the crossing of the intestinal barrier: a transgenic
REF. 114  (2002) Elsevier. mouse that expresses human E-cadherin on the surface
of enterocytes46. This model showed that InlA has a
key role in disease, because it is essential for crossing
The L. monocytogenes model has also been useful for of the intestinal barrier. In this model system, InlA
understanding the importance and the function of VASP, could interact with E-cadherin, and a wild-type strain
the other main ligand of ActA. VASP binds the central of L. monocytogenes was able to cause disease through
proline-rich domain of ActA and promotes efficient oral inoculation.
actin-based motility34,36–38, highlighting the importance So far, E-cadherin has been found only at cell–cell
of this protein in actin polymerization. Only recently, junctions and on the basolateral face of epithelial cells,
however, have the complex molecular mechanisms of so the mechanism by which L. monocytogenes gains
VASP activity begun to emerge. VASP seems to promote access to E-cadherin was enigmatic. Two hypotheses
listerial motility by recruiting the actin-binding protein were put forward to explain how InlA could target
profilin, which promotes polymerization at actin-filament E-cadherin 46. The first hypothesis proposed that
barbed ends39. VASP also seems to induce faster growth L. monocytogenes could gain access to E-cadherin
of the actin network at the bacterial surface by causing at the tips of intestinal microvilli, where apoptotic
the release of Arp2/3 from ActA40. In addition, VASP epithelial cells slough off. The second hypothesis pro-
seems to decrease Y-branch formation, thereby increas- posed a synergy between InlA- and InlB-dependent
ing parallel alignment of actin filaments41. Although their internalization, because activation of Met by HGF has
mechanistic role is not fully elucidated, proteins of the been shown to stimulate the disassembly of junctions
Ena/VASP family are important in the formation of actin between epithelial cells46. Recent results have shown
fibres, filopodial tips and the lamellipodial leading edge42. that L. monocytogenes invades the intestinal epithelium
Intracellular and intercellular movement using actin at sites of cell extrusion at the tips of villi47 and that the
polymerization is not restricted to L. monocytogenes. contribution of InlB to crossing of the intestinal bar-
A growing number of intracellular pathogens, includ- rier is insignificant in vivo48. Whether the mechanism
ing Rickettsia species, Shigella species, mycobacteria, of intestinal invasion is used to cross other barriers is
Filopodia Burkholderia pseudomallei and vaccinia virus, show this unknown. Synergy between InlA and InlB could still be
Rod-like cell-surface feature during infection (see Supplementary information important for crossing of the placental barrier49.
projections that are composed S3 (figure), and for recent reviews, see REFS 43,44). Because the transgenic mice described here express
of actin filaments. They are
human E-cadherin only on enterocytes, it is not pos-
found on various cell types and
have sensory or exploratory A paradigm in pathophysiology sible to study the role of InlA in deeper tissues. The
functions. As a pathogen that displays such interesting features generation of transgenic mice that express E-cadherin
as strong T-cell activation, a sophisticated relation- on all cells is in progress. At present, the role of InlA
Lamellipodia ship with its host and crossing of protective barriers, can also be studied in guinea pigs or rabbits, because
Thin actin-rich structures that
form protrusions at the edge of
L. monocytogenes has more recently also emerged as a E-cadherin is recognized by InlA in these animals.
the cell and are essential for model to study the pathophysiology of a complex bac- However, recent studies show a species specificity for
cellular motility. terial infection. Findings from in vitro work have been InlB: InlB does not recognize or activate guinea-pig

428 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
REVIEWS

or rabbit Met. Therefore, the role of InlA and InlB in Novel regulatory mechanisms
infection cannot be studied using these animal mod- L. monocytogenes is a facultative intracellular pathogen
els48. A human model remains the best model system that can live both inside and outside its host. This bac-
for studying listerial infection, and human explants have terium has therefore evolved sophisticated regulatory
been successfully used to determine the mechanism by mechanisms to ensure that virulence factors are opti-
which L. monocytogenes crosses the maternofetal bar- mally expressed when they are required. These regula-
rier49. Human placental villus explants, together with tory mechanisms might prove to be general mechanisms
primary or immortalized trophoblastic cells, were used by other bacteria.
used to show that InlA is a key bacterial protein that is
required for crossing of the human maternofetal bar- PrfA: a tightly regulated protein. Most of the virulence
rier, although InlA is not essential for this role in the proteins that have been identified in L. monocytogenes
pregnant guinea-pig model49,50. Crossing of the blood– are under the control of one transcriptional regulator,
brain barrier is still poorly understood. However, the PrfA, which itself is tightly regulated by environmen-
optimization of animal models should help to decipher tal conditions. During exponential growth, prfA is
this crucial step. mainly transcribed as a bicistronic mRNA from the
Other model systems are being developed to identify plcA promoter. By contrast, during stationary phase,
the host factors that are required for the intracellular sur- a monocistronic mRNA is preferentially transcribed
vival of L. monocytogenes and possibly of other intracel- from a promoter upstream of prfA55–57 (FIG. 5a). Similar
lular pathogens. Drosophila melanogaster has attracted to many other pathogens, L. monocytogenes can sense
attention as a model because of the many genetic and conditions in the mammalian host and respond by
immunological studies that have been carried out using expressing virulence genes. A novel regulation of PrfA
this organism, and it has been successfully used to test by temperature, owing to the structure of the upstream
L. monocytogenes virulence51. Moreover, genome-wide untranslated region of the prfA mRNA, was recently dis-
RNA-interference screens in D. melanogaster S2 cells covered58 (FIG. 5b). At low temperature (30°C), the prfA
(which are macrophage-like cells) have revealed many leader transcript controls translation of the downstream
new host factors that are important for entry into the mRNA by forming a secondary structure that masks the
host cell, escape from the vacuole and intracellular ribosome-binding site. At mammalian host tempera-
growth of L. monocytogenes52,53. Another noteworthy ture (37°C), this structure partially melts to expose the
organism that has been shown to support a listerial ribosome-binding site, thereby allowing translation to
infection is Caenorhabditis elegans54. occur. Fusion of the prfA leader transcript to the gene
that encodes green fluorescent protein (GFP) also
a resulted in thermoregulation of GFP. This mechanism
might be used by other bacteria, as has previously been
plcA prfA suggested for the Yersinia pestis activator protein LcrF59.
P P1 and P2
Such post-transcriptional regulation of prfA allows
b rapid expression of the encoded transcription factor and
therefore efficient transcription of virulence factors as
Ribosome soon as the bacterium enters the host.
subunits Other environmental conditions — such as osmol-
arity, iron concentrations, pH, the presence of fermentable
sugars, stress (through σB), and conditions in the host-cell
intracellular compartment — have been shown to regu-
late prfA and PrfA-controlled genes through mechanisms
SD that are not completely understood60,61. Post-translational
High
temperature regulation of PrfA by a putative cofactor is suggested by
prfA ATG
mRNA prfA its structure, which resembles that of the cyclic-AMP
Low receptor62,63. The number of mechanisms that regulate
temperature PrfA is probably indicative of the importance of this
No PrfA PrfA crucial virulence factor during infection.

Virulence-gene Listeriolysin O: a pH-sensing protein. L. monocytogenes


expression
thrives in the cytoplasm of numerous cell types. Following
Figure 5 | The PrfA regulator. a | Schematic representation of the prfA region. internalization, bacteria escape from membrane-bound
During exponential phase, prfA is mostly transcribed as a bicistronic product from phagosomes by secreting two phospholipases, PlcA and
the promoter (P) upstream of plcA. By contrast, during stationary phase, prfA is mostly PlcB, and the pore-forming toxin listeriolysin O (LLO),
transcribed as a monocistronic product from P1 and/or P2. b | Mechanism that controls
thereby gaining access to the cytoplasm (FIG. 1).
the thermoregulated expression of PrfA in the promoter upstream of prfA. At low
temperatures (≤30°C), a secondary structure forms in the untranslated region of prfA, Although LLO is a member of a large family of
and this prevents ribosome binding and therefore expression of PrfA. At high cholesterol-dependent cytolysins that are secreted by
temperatures (≥37°C), melting of the prfA untranslated region allows ribosomes numerous Gram-positive bacteria, L. monocytogenes
to bind and PrfA expression to occur. SD, Shine–Dalgarno sequence. This figure is is the only pathogen that secretes this type of toxin
modified with permission from REF. 58  (2002) Elsevier. inside the host cell. Therefore, secretion of LLO must

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 429


© 2006 Nature Publishing Group
REVIEWS

Premature In addition to its pore-forming ability, LLO has the


unfolding surprising ability to induce potent signalling in the host
cell. It has been shown to activate NF-κB67, MAPK68,
phosphatidylinositol 69,70, calcium 71–73 and protein-
Neutral kinase-C 74 signalling pathways. As a pore-forming
pH toxin with cholesterol as the only identified ligand, this
signalling function is puzzling. LLO could have a signal-
Soluble ling function at neutral pH, as has been suggested for
monomer
Formation of another member of the cholesterol-dependent-cytolysin
D1 Membrane-bound Pre-pore the pre-insertion family, Streptococcus intermedius intermedilysin, which
monomers oligomer β-sheet binds the host cellular receptor CD59 (REF. 75). However,
it has been difficult to separate the signalling function of
D2 D3 Pore
oligomer LLO from its pore-forming ability. For example, MAPK
activation occurs on treatment of host cells with LLO,
Low but it also occurs after exposure to a mild detergent,
D4 pH indicating that signalling could be the result of mem-
brane damage rather than a specific activity of LLO68.
Clearly, the signalling activity of LLO requires further
Out investigation.

A model of bacterial adaptation


In One particularly interesting feature of L. monocytogenes
is its capacity to thrive both inside and outside the host.
This ability to colonize a broad range of ecosystems is
Figure 6 | Listeriolysin O (LLO) pore-forming mechanism. At low (acidic) pH, the reflected in the genome of the organism, which contains an
soluble LLO monomer interacts with the host-cell plasma membrane, presumably by unusually large number of regulatory and transport pro-
binding cholesterol. On contact with the membrane, structural rearrangements in one teins (11.6% of all predicted genes)76. With one-quarter
monomer expose residues that can form hydrogen bonds with other monomers, thereby
of its transport proteins dedicated to carbohydrate
allowing oligomerization into a pre-pore complex. Following oligomerization, two
α-helical bundles from each monomer extend to form transmembrane β-hairpins (red) transport, L. monocytogenes has the largest number of
that punch through the membrane. Pores formed by LLO and other cholesterol-binding phosphotransferase systems that have been described
proteins can be 250–300 Å in diameter. At neutral pH, domain 3 (D3) of the monomer for bacteria so far. Furthermore, the genome contains 16
prematurely unfolds, rendering the protein unable to form pores. This figure is modified putative two-component regulatory systems, more than are
with permission from REF. 115  (2005) American Society for Microbiology. found in other pathogenic bacteria and comparable to
the number found in ubiquitous bacteria such as Bacillus
subtilis. Inactivation of several of these two-component
be tightly regulated, because the bacterium needs to regulatory systems has revealed their importance for
balance efficient escape from the vacuole against pre- the resistance of L. monocytogenes to various stresses
vention of host-cell damage to allow its intracellular and for survival in the host77–82. In addition to these
survival. conventional bacterial two-component regulatory sys-
Unlike other pore-forming toxins, the activity of tems, which phosphorylate histidine and aspartic-acid
LLO is optimal at acidic pH (<6). So, LLO is fully active residues, L. monocytogenes also uses a eukaryotic-like
in the acidic environment of the phagosome, but it is serine/threonine protein phosphatase system, known as
less active at the neutral pH of the host-cell cytoplasm. Stp, to regulate the translation elongation factor EF-Tu
An L. monocytogenes strain that was constructed to and, consequently, virulence83.
express the pore-forming toxin perfringolysin O, which Outside the host, L. monocytogenes is particularly
is active at neutral pH, can also escape the vacuole well adapted to grow at low temperatures, making it
but is toxic to the host cell and is avirulent in mice, difficult for the food industry to rely on refrigeration
Two-component regulatory highlighting the importance of pH regulation of LLO to control listerial contamination84–87. More remark-
system activity 64. The molecular mechanism that underlies able is the ability of L. monocytogenes to survive in
A two-protein signal-
transduction system that is
the optimum pH for LLO activity was only recently the host. The resistance of L. monocytogenes to acidic
important for the bacterial identified: LLO was found to be stable at acidic pH, conditions88,89 and to bile salts90–92 makes this patho-
response to environmental and to unfold and aggregate at neutral pH65 (FIG. 6). At gen particularly adept at infecting the gastrointestinal
changes. It consists of a acidic pH, pore formation occurs when LLO monomers tract. Consistent with the many bile-resistance genes
membrane-bound sensor
oligomerize into large complexes that penetrate the in the L. monocytogenes genome, a recent study using
protein kinase and a
transcriptional-response membrane by extending transmembrane β-hairpins66. in vivo bioluminescence imaging (a non-invasive pro-
regulator. At neutral pH, acidic amino-acid residues in the trans- cedure that allows bioluminescent L. monocytogenes
membrane domain initiate irreversible denaturation of to be traced through the body of a mouse) has shown
Elongation factor the β-hairpins of LLO, leading to inactivation of the an intriguing new reservoir for L. monocytogenes, the
A protein that allows tRNAs
to bind the ribosome and is
pore-forming function of LLO65. The elaborate struc- gall bladder, where bile is stored and concentrated93.
essential for elongation of the ture of LLO therefore allows this protein to sense and Whether this organ is indeed colonized during human
polypeptide chain. respond to its environment. listeriosis is unknown.

430 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
REVIEWS

InlG 490 identified: the sugar-uptake system (Hpt) and lipoate


protein ligase A1 (LplA1). Hpt (which is homologous
InlE 499
to the mammalian translocase of glucose-6-phosphate)
Lmo1136 539 is required for optimal intracellular replication 95.
InlH 548 Interestingly, transcription of hpt is regulated by PrfA
Lmo0610 589 and is therefore activated upon entry into the cytosol of
Lmo1289 593 the host cell. Expression of Hpt allows the bacterium to
use glucose-1-phosphate, which is an available carbon
Lmo1290 598
source in the host-cell cytoplasm. Other bacterial patho-
Lmo0514 605 gens such as Escherichia coli, Salmonella enterica, Shigella
Lmo2026 626 flexneri and Chlamydia trachomatis have a homologue
LPXTG

Lmo0331 633 of hpt, indicating that this transporter system could have
Lmo0732 638 a general role in intracellular survival. The other factor
that has been identified to be important for intracellular
Lmo0801 646
growth, LplA1, catalyses the formation of a covalent
InlA 800 link between lipoic acid and specific protein targets96.
InIF 821 A mutant that lacks LplA1 cannot replicate in the cyto-
Lmo0171 832 plasm of macrophages and has a 300-fold decrease in
InlJ 851 virulence96. LplA1 seems to be important for lipoylation
and full activity of the pyruvate-dehydrogenase enzyme
Lmo2396 940
complex in the host-cell cytosol, where lipoic acid is
Lmo0327 1349 mainly absent. LplA1 has therefore been proposed to be
InlI important for the scavenging of lipoyl groups from host
1778 molecules.
GW

InlB 630
Interestingly, several reports indicate that the expres-
sion of many L. monocytogenes genes is upregulated in
InlC 296 the cytosol. A library of Tn917-lacZ L. monocytogenes
mutants was screened for higher lacZ expression when
Lmo2445 300
present in the cytosol of macrophages than when grown
Secreted

Lmo2027 367 in rich broth. This approach allowed the identification


Lmo2470 388 of several L. monocytogenes genes, including plcA and
Lmo0549 673 prfA, that are activated after infection of host cells.
Recently, two transcriptomic studies have advanced
Signal peptide LPXTG motif LRR region Sorting signal the understanding of adaptation to the host-cell cytosol
by showing that L. monocytogenes turns on ~500 genes
Conserved inter-repeat region GW module
for survival in this cellular compartment98,99. Further
Figure 7 | The internalin family of proteins. Homologous regions of internalin-family analysis of the genes that were identified could
members include an N-terminal signal-peptide sequence, followed by several leucine- elucidate the metabolism of this bacterium in the
rich tandem repeats (LRRs). Several internalins contain an inter-repeat region, which is host-cell cytosol.
structurally related to an immunoglobulin-like domain. The internalin family can be
divided into three subfamilies on the basis of their association with bacteria. The first Genomics reveals new virulence factors
subfamily includes internalins that contain an LPXTG amino-acid motif (where X denotes Major listerial virulence factors have been identified
any amino acid), and these are covalently anchored to the cell wall. The second subfamily
by classical genetics; that is, the search for a mutant
includes internalin B (InlB), which is loosely associated with the bacterial surface through
its GW modules (which contain the amino-acid motif GW). The third subfamily contains
with a particular phenotype, followed by gene iden-
five internalins that are predicted to be secreted, because they do not have any surface- tification and characterization 1. The genetic basis
anchoring domains. It should be noted that the number of LRRs present in the proteins of L. monocytogenes virulence can now be deci-
Lmo2445 and Lmo2470 is unclear, because these proteins contain degenerate LRRs. phered by exploitation of the genome sequences
The number of amino acids in each protein is indicated. This figure is modified with of L. monocytogenes and the closely related non-
permission from REF. 101  (2002) Elsevier. pathogenic species Listeria innocua76, using three main
strategies: first, the analysis of genes that are present
in L. monocytogenes and absent from L. innocua;
Survival and replication in the cytosol of many types second, the study of genes that encode potentially
of host cells, including macrophages and epithelial cells, interesting proteins; and third, the use of whole-
is one of the distinguishing features of infection with genome DNA arrays to screen a large population of
L. monocytogenes3,4. Other extracellular bacteria and strains from different Listeria species and to identify
intracellular pathogens that normally reside inside a genes that are present in, and specific to, all virulent
vacuole cannot replicate in the cytosol of the host cell94, L. monocytogenes strains100.
highlighting that L. monocytogenes has evolved specific A first glance at the global genome structure indi-
mechanisms to grow in the host-cell cytosol. Although cates that, in contrast to many other bacterial genomes,
intracytosolic survival remains poorly understood, the L. monocytogenes genome (strain EGD-e) contains
two key proteins for intracellular growth have been only three insertion sequences and five bacteriophages,

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 431


© 2006 Nature Publishing Group
REVIEWS

Chaperone none of which seems to have a role in acquisition of for the infectious process in vivo110. Vip interacts with
A protein that assists other virulence genes. Moreover, except for genes of the the host-cell endoplasmic-reticulum chaperone gp96, a
proteins to fold correctly. major virulence locus, virulence genes seem to be dis- protein that is involved in Toll-like-receptor signalling111.
persed on the chromosome and are not concentrated in The LPXTG protein InlJ, the function of which remains
pathogenicity islands. One of the most striking features to be elucidated, is a leucine-rich-repeat-containing
of the L. monocytogenes genome with respect to virulence protein that is structurally related to InlA and InlB105.
is the exceptionally large number of genes that encode These three proteins are members of the internalin
surface proteins (4.7% of all predicted genes101). These family, which is a large family of proteins that contain
proteins are among the most likely candidates to interact leucine-rich repeats (FIG. 7). The gene that encodes InlJ,
with the host and therefore to be virulence factors. These similar to the genes that encode five other proteins of
proteins can be divided into three families. The largest the internalin family (InlA, InlB, InlE, InlH and InlI),
family is composed of 68 lipoproteins, and the second is conserved in the genomes of pathogenic serovars
largest family contains 41 LPXTG proteins (including of L. monocytogenes (that is, 1/2a, 1/2b, 1/2c and 4b)
InlA), which are anchored to the cell wall by a sortase (an and absent from all other Listeria species100,105, and this
enzyme that is involved in the covalent linkage of Gram- is consistent with a role for InlJ in virulence.
positive bacterial proteins to the bacterial surface)102–104.
Inactivation of sortase A (SrtA) or Lsp, a signal peptidase Conclusions and future perspectives
that is involved in maturation of lipoproteins, attenu- The study of L. monocytogenes infection highlights the
ates virulence, thereby pinpointing the importance of sophisticated relationship between this bacterium and its
these two classes of surface protein in L. monocytogenes host, and reveals their long co-evolution and reciprocal
infection103,105,106. The third family of surface proteins adaptation. For decades, L. monocytogenes has been a tool
includes proteins that are non-covalently attached to for immunologists. It has now also become a paradigm in
the bacterial surface by their C-terminal domains. This bacterial pathogenesis and cellular microbiology.
family includes GW proteins (such as InlB and Ami), The availability of the genome sequence of five
which contain modules of 80 amino acids that contain L. monocytogenes serovars76,100,112 and L. innocua76, and
the dipeptide Gly–Trp (also known as GW modules107) soon of Listeria ivanovii and Listeria welshimeri, will pro-
in their C-terminal region. An important outcome from vide further insight into the molecular basis of the patho-
genome-sequence analysis was the discovery of a SecA2- genesis determinants of Listeria species. Comparative
dependent pathway, which allows proteins that lack a genomics could also reveal genetic loci that confer
typical signal-peptide sequence to be targeted to the specific pathogenic traits to epidemic strains: for exam-
bacterial surface or to be secreted108. ple, loci that facilitate adaptation to different environ-
Comparison of the L. monocytogenes and L. innocua ments and that influence the onset of infection. It should
genomes has proved an efficient approach to identify new be noted that it is often difficult to unravel the function
virulence factors, including bile-salt hydrolase (Bsh)90, of virulence factors that are identified by post-genomic
two LPXTG proteins (Vip and InlJ) and a GW protein methods and reverse genetics. To this end, more sophisti-
(Auto)105,109,110. The GW protein Auto is a novel surface- cated in vivo studies — such as measurement of cytokine
associated cell-wall hydrolase that is required for entry production, non-invasive in vivo imaging techniques or
into eukaryotic cells. Its contribution to listerial infection infection of ex vivo tissue explants — need to be used to
is still unclear but might also be linked to the release of understand the role of these bacterial factors in the inter-
immunologically active cell-wall components that could play between the bacterium and the host. Applications
interact with components of the innate immune sys- of these novel techniques to the study of Listeria species
tem109. The LPXTG protein Vip, which is positively regu- will probably continue to show that this bacterium is an
lated by the transcriptional activator PrfA, is required for invaluable model in the fields of cellular microbiology,
bacterial entry into some eukaryotic cells in vitro and bacterial pathogenesis and cell biology.

1. Khelef, N. et al. The Prokaryotes: An Evolving 6. Cohen, P. et al. Monitoring cellular responses to 11. Braun, L., Ghebrehiwet, B. & Cossart, P. gC1q-R/p32,
Electronic Resource for the Microbiological Listeria monocytogenes with oligonucleotide arrays. a C1q-binding protein, is a receptor for the InlB
Community [online] (eds Dworkin, M., Falkow, S., J. Biol. Chem. 275, 11181–11190 (2000). invasion protein of Listeria monocytogenes. EMBO J.
Rosenberg, E., Schleifer, K.-H. & Stackebrandt, E.) 7. Shen, Y., Naujokas, M., Park, M. & Ireton, K. InIB- 19, 1458–1466 (2000).
(Springer, New York, 2005) <http://link.springer- dependent internalization of Listeria is mediated by 12. Bierne, H. et al. WASP-related proteins, Abi1 and Ena/
ny.com/link/service/books/10125/>. the Met receptor tyrosine kinase. Cell 103, 501–510 VASP are required for Listeria invasion induced by the
2. Pamer, E. G. Immune responses to Listeria (2000). Met receptor. J. Cell Sci. 118, 1537–1547 (2005).
monocytogenes. Nature Rev. Immunol. 4, 812–823 Identifies the cellular receptor for InlB and 13. Bierne, H. et al. A role for cofilin and LIM kinase in
(2004). illustrates the exploitation of a receptor-tyrosine- Listeria-induced phagocytosis. J. Cell Biol. 155,
3. Mackaness, G. B. Cellular resistance to infection. kinase pathway for bacterial entry. 101–112 (2001).
J. Exp. Med. 116, 381–406 (1962). 8. Tang, P., Sutherland, C. L., Gold, M. R. & Finlay, B. B. 14. Veiga, E. & Cossart, P. Listeria hijacks the clathrin-
First in-depth analysis of the interaction between Listeria monocytogenes invasion of epithelial cells dependent endocytic machinery to invade
L. monocytogenes and its host, both in mice and in requires the MEK-1/ERK-2 mitogen-activated protein mammalian cells. Nature Cell Biol. 7, 894–900
cultured mouse macrophages. kinase pathway. Infect. Immun. 66, 1106–1112 (2005).
4. Racz, P., Tenner, K. & Mero, E. Experimental Listeria (1998). Shows that the endocytic machinery is subverted
enteritis. I. An electron microscopic study of the 9. Ireton, K. et al. A role for phosphoinositide 3-kinase in for L. monocytogenes entry and therefore proposes
epithelial phase in experimental Listeria infection. bacterial invasion. Science 274, 780–782 (1996). that clathrin can help engulf large particles such as
Lab. Invest. 26, 694–700 (1972). 10. Copp, J., Marino, M., Banerjee, M., Ghosh, P. & bacteria.
5. McCaffrey, R. L. et al. A specific gene expression van der Geer, P. Multiple regions of internalin B 15. Harvey, H. A., Jennings, M. P., Campbell, C. A.,
program triggered by Gram-positive bacteria in the contribute to its ability to turn on the Ras–mitogen- Williams, R. & Apicella, M. A. Receptor-mediated
cytosol. Proc. Natl Acad. Sci. USA 101, 11386–11391 activated protein kinase pathway. J. Biol. Chem. 278, endocytosis of Neisseria gonorrhoeae into primary
(2004). 7783–7789 (2003). human urethral epithelial cells: the role of the

432 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
REVIEWS

asialoglycoprotein receptor. Mol. Microbiol. 42, 36. Smith, G. A., Theriot, J. A. & Portnoy, D. A. The that positively regulates expression of listeriolysin,
659–672 (2001). tandem repeat domain in the Listeria monocytogenes the major virulence factor of Listeria
16. Van Nhieu, G. T., Krukonis, E. S., Reszka, A. A., ActA protein controls the rate of actin-based motility, monocytogenes. Proc. Natl Acad. Sci. USA 87,
Horwitz, A. F. & Isberg, R. R. Mutations in the the percentage of moving bacteria, and the 8336–8340 (1990).
cytoplasmic domain of the integrin β1 chain indicate a localization of vasodilator-stimulated phosphoprotein 58. Johansson, J. et al. An RNA thermosensor controls
role for endocytosis factors in bacterial internalization. and profilin. J. Cell Biol. 135, 647–660 (1996). expression of virulence genes in Listeria
J. Biol. Chem. 271, 7665–7672 (1996). 37. Niebuhr, K. et al. A novel proline-rich motif present in monocytogenes. Cell 110, 551–561 (2002).
17. Wyrick, P. B. et al. Entry of genital Chlamydia ActA of Listeria monocytogenes and cytoskeletal Characterizes a novel regulatory mechanism that is
trachomatis into polarized human epithelial cells. proteins is the ligand for the EVH1 domain, a protein encoded by the upstream untranslated region of
Infect. Immun. 57, 2378–2389 (1989). module present in the Ena/VASP family. EMBO J. 16, prfA mRNA and that controls the expression of this
18. Seveau, S., Bierne, H., Giroux, S., Prevost, M. C. & 5433–5444 (1997). important virulence transcription factor according
Cossart, P. Role of lipid rafts in E-cadherin- and 38. Loisel, T. P., Boujemaa, R., Pantaloni, D. & Carlier, M. F. to the temperature.
HGF-R/Met-mediated entry of Listeria Reconstitution of actin-based motility of Listeria and 59. Hoe, N. P. & Goguen, J. D. Temperature sensing in
monocytogenes into host cells. J. Cell Biol. 166, Shigella using pure proteins. Nature 401, 613–616 Yersinia pestis: translation of the LcrF activator
743–753 (2004). (1999). protein is thermally regulated. J. Bacteriol. 175,
19. Mengaud, J., Ohayon, H., Gounon, P., Mege, R. M. & 39. Geese, M. et al. Contribution of Ena/VASP proteins 7901–7909 (1993).
Cossart, P. E-cadherin is the receptor for internalin, a to intracellular motility of Listeria requires 60. Schwab, U., Bowen, B., Nadon, C., Wiedmann, M. &
surface protein required for entry of L. monocytogenes phosphorylation and proline-rich core but not F-actin Boor, K. J. The Listeria monocytogenes prfAP2
into epithelial cells. Cell 84, 923–932 (1996). binding or multimerization. Mol. Biol. Cell 13, promoter is regulated by σB in a growth phase
Identifies E-cadherin as the first cellular receptor 2383–2396 (2002). dependent manner. FEMS Microbiol. Lett. 245,
for L. monocytogenes and reveals a novel 40. Samarin, S. et al. How VASP enhances actin-based 329–336 (2005).
heterophilic interaction with E-cadherin. motility. J. Cell Biol. 163, 131–142 (2003). 61. Kreft, J. & Vazquez-Boland, J. A. Regulation of
20. Perez-Moreno, M., Jamora, C. & Fuchs, E. Sticky 41. Plastino, J., Olivier, S. & Sykes, C. Actin filaments align virulence genes in Listeria. Int. J. Med. Microbiol.
business: orchestrating cellular signals at adherens into hollow comets for rapid VASP-mediated 291, 145–157 (2001).
junctions. Cell 112, 535–548 (2003). propulsion. Curr. Biol. 14, 1766–1771 (2004). 62. Ripio, M. T., Dominguez-Bernal, G., Lara, M.,
21. Lecuit, M. et al. A role for α- and β-catenins in 42. Krause, M., Bear, J. E., Loureiro, J. J. & Gertler, F. B. Suarez, M. & Vazquez-Boland, J. A. A Gly145Ser
bacterial uptake. Proc. Natl Acad. Sci. USA 97, The Ena/VASP enigma. J. Cell Sci. 115, 4721–4726 substitution in the transcriptional activator PrfA
10008–10013 (2000). (2002). causes constitutive overexpression of virulence
22. Drees, F., Pokutta, S., Yamada, S., Nelson, W. J. & 43. Gouin, E., Welch, M. D. & Cossart, P. Actin-based factors in Listeria monocytogenes. J. Bacteriol. 179,
Weis, W. I. α-Catenin is a molecular switch that binds motility of intracellular pathogens. Curr. Opin. 1533–1540 (1997).
E-cadherin–β-catenin and regulates actin-filament Microbiol. 8, 35–45 (2005). 63. Eiting, M., Hageluken, G., Schubert, W. D. &
assembly. Cell 123, 903–915 (2005). 44. Stevens, J. M., Galyov, E. E. & Stevens, M. P. Actin- Heinz, D. W. The mutation G145S in PrfA, a key
23. Yamada, S., Pokutta, S., Drees, F., Weis, W. I. & dependent movement of bacterial pathogens. Nature virulence regulator of Listeria monocytogenes,
Nelson, W. J. Deconstructing the cadherin–catenin– Rev. Microbiol. 4, 91–101 (2006). increases DNA-binding affinity by stabilizing the HTH
actin complex. Cell 123, 889–901 (2005). 45. Lecuit, M. et al. A single amino acid in E-cadherin motif. Mol. Microbiol. 56, 433–446 (2005).
24. Sousa, S. et al. ARHGAP10 is necessary for α-catenin responsible for host specificity towards the human 64. Jones, S. & Portnoy, D. A. Characterization of Listeria
recruitment at adherens junctions and for Listeria pathogen Listeria monocytogenes. EMBO J. 18, monocytogenes pathogenesis in a strain expressing
invasion. Nature Cell Biol. 7, 954–960 (2005). 3956–3963 (1999). perfringolysin O in place of listeriolysin O. Infect.
25. Kussel-Andermann, P. et al. Vezatin, a novel 46. Lecuit, M. et al. A transgenic model for listeriosis: role Immun. 62, 5608–5613 (1994).
transmembrane protein, bridges myosin VIIA of internalin in crossing the intestinal barrier. Science 65. Schuerch, D. W., Wilson-Kubalek, E. M. & Tweten, R. K.
to the cadherin–catenins complex. EMBO J. 19, 292, 1722–1725 (2001). Molecular basis of listeriolysin O pH dependence.
6020–6029 (2000). 47. Pentecost, M., Otto, G., Theriot, J. A. & Amieva, M. R. Proc. Natl Acad. Sci. USA 102, 12537–12542
26. Sousa, S. et al. Unconventional myosin VIIa and Listeria monocytogenes invades the epithelial junctions (2005).
vezatin, two proteins crucial for Listeria entry into at sites of cell extrusion. PLoS Pathog. 2, e3 (2006). Structural studies of the L. monocytogenes toxin
epithelial cells. J. Cell Sci. 117, 2121–2130 (2004). 48. Khelef, N., Lecuit, M., Bierne, H. & Cossart, P. Species LLO reveal the molecular mechanism that controls
27. Takemura, R., Stenberg, P. E., Bainton, D. F. & Werb, Z. specificity of the Listeria monocytogenes InlB protein. the optimal pH for its activity.
Rapid redistribution of clathrin onto macrophage Cell. Microbiol. 8, 457–470 (2006). 66. Shatursky, O. et al. The mechanism of membrane
plasma membranes in response to Fc receptor–ligand Shows host specificity for InlB, thereby challenging insertion for a cholesterol-dependent cytolysin: a novel
interaction during frustrated phagocytosis. J. Cell Biol. the validity of some of the animal models that are paradigm for pore-forming toxins. Cell 99, 293–299
102, 55–69 (1986). used to study listerial infection and emphasizing (1999).
28. Tilney, L. G. & Portnoy, D. A. Actin filaments and the the need to develop new models. 67. Kayal, S. et al. Listeriolysin O-dependent activation of
growth, movement, and spread of the intracellular 49. Lecuit, M. et al. Targeting and crossing of the endothelial cells during infection with Listeria
bacterial parasite, Listeria monocytogenes. J. Cell human maternofetal barrier by Listeria monocytogenes: activation of NF-κB and upregulation
Biol. 109, 1597–1608 (1989). monocytogenes: role of internalin interaction of adhesion molecules and chemokines. Mol. Microbiol.
First characterization of actin nucleation by with trophoblast E-cadherin. Proc. Natl Acad. Sci. 31, 1709–1722 (1999).
L. monocytogenes and its importance for cell–cell USA 101, 6152–6157 (2004). 68. Tang, P., Rosenshine, I., Cossart, P. & Finlay, B. B.
spread during infection. 50. Bakardjiev, A. I., Stacy, B. A., Fisher, S. J. & Listeriolysin O activates mitogen-activated protein
29. Dabiri, G. A., Sanger, J. M., Portnoy, D. A. & Portnoy, D. A. Listeriosis in the pregnant guinea pig: kinase in eucaryotic cells. Infect. Immun. 64,
Southwick, F. S. Listeria monocytogenes moves rapidly a model of vertical transmission. Infect. Immun. 72, 2359–2361 (1996).
through the host-cell cytoplasm by inducing directional 489–497 (2004). 69. Sibelius, U. et al. Listeriolysin is a potent inducer of
actin assembly. Proc. Natl Acad. Sci. USA 87, 51. Mansfield, B. E., Dionne, M. S., Schneider, D. S. & the phosphatidylinositol response and lipid mediator
6068–6072 (1990). Freitag, N. E. Exploration of host–pathogen generation in human endothelial cells. Infect. Immun.
30. Kocks, C. et al. L. monocytogenes-induced actin interactions using Listeria monocytogenes and 64, 674–676 (1996).
assembly requires the actA gene product, a surface Drosophila melanogaster. Cell. Microbiol. 5, 901–911 70. Sibelius, U. et al. The listerial exotoxins listeriolysin
protein. Cell 68, 521–531 (1992). (2003). and phosphatidylinositol-specific phospholipase C
31. Domann, E. et al. A novel bacterial virulence gene 52. Cheng, L. W. et al. Use of RNA interference in synergize to elicit endothelial cell phosphoinositide
in Listeria monocytogenes required for host cell Drosophila S2 cells to identify host pathways metabolism. J. Immunol. 157, 4055–4060
microfilament interaction with homology to the controlling compartmentalization of an intracellular (1996).
proline-rich region of vinculin. EMBO J. 11, pathogen. Proc. Natl Acad. Sci. USA 102, 71. Dramsi, S. & Cossart, P. Listeriolysin O-mediated
1981–1990 (1992). 13646–13651 (2005). calcium influx potentiates entry of Listeria
References 30 and 31 identify ActA as the 53. Agaisse, H. et al. Genome-wide RNAi screen for host monocytogenes into the human Hep-2 epithelial cell
bacterial protein that is required for actin factors required for intracellular bacterial infection. line. Infect. Immun. 71, 3614–3618 (2003).
polymerization by L. monocytogenes. Science 309, 1248–1251 (2005). 72. Tsuchiya, K. et al. Listeriolysin O-induced membrane
32. Welch, M. D., Iwamatsu, A. & Mitchison, T. J. Actin References 52 and 53 use a novel system to permeation mediates persistent interleukin-6
polymerization is induced by Arp2/3 protein complex identify, on a large scale, host factors that are production in Caco-2 cells during Listeria
at the surface of Listeria monocytogenes. Nature 385, important for supporting a listerial infection. monocytogenes infection in vitro. Infect. Immun.
265–269 (1997). 54. Thomsen, L. E., Slutz, S. S., Tan, M. W. & Ingmer, H. 73, 3869–3877 (2005).
Shows that the Arp2/3 actin-nucleation complex Caenorhabditis elegans is a model host for Listeria 73. Wadsworth, S. J. & Goldfine, H. Listeria
initiates ActA-dependent polymerization. monocytogenes. Appl. Environ. Microbiol. 72, monocytogenes phospholipase C-dependent calcium
33. Machesky, L. M. et al. Scar, a WASp-related protein, 1700–1701 (2006). signaling modulates bacterial entry into J774
activates nucleation of actin filaments by the Arp2/3 55. Mengaud, J. et al. Pleiotropic control of Listeria macrophage-like cells. Infect. Immun. 67, 1770–1778
complex. Proc. Natl Acad. Sci. USA 96, 3739–3744 monocytogenes virulence factors by a gene that is (1999).
(1999). autoregulated. Mol. Microbiol. 5, 2273–2283 74. Wadsworth, S. J. & Goldfine, H. Mobilization of protein
34. Lasa, I., David, V., Gouin, E., Marchand, J. B. & (1991). kinase C in macrophages induced by Listeria
Cossart, P. The amino-terminal part of ActA is critical 56. Freitag, N. E., Rong, L. & Portnoy, D. A. Regulation monocytogenes affects its internalization and escape
for the actin-based motility of Listeria monocytogenes; of the prfA transcriptional activator of Listeria from the phagosome. Infect. Immun. 70, 4650–4660
the central proline-rich region acts as a stimulator. monocytogenes: multiple promoter elements (2002).
Mol. Microbiol. 18, 425–436 (1995). contribute to intracellular growth and cell-to-cell 75. Giddings, K. S., Zhao, J., Sims, P. J. & Tweten, R. K.
35. May, R. C. et al. The Arp2/3 complex is essential for spread. Infect. Immun. 61, 2537–2544 (1993). Human CD59 is a receptor for the cholesterol-
the actin-based motility of Listeria monocytogenes. 57. Leimeister-Wachter, M., Haffner, C., Domann, E., dependent cytolysin intermedilysin. Nature Struct.
Curr. Biol. 9, 759–762 (1999). Goebel, W. & Chakraborty, T. Identification of a gene Mol. Biol. 11, 1173–1178 (2004).

NATURE REVIEWS | MICROBIOLOGY VOLUME 4 | JUNE 2006 | 433


© 2006 Nature Publishing Group
REVIEWS

76. Glaser, P. et al. Comparative genomics of Listeria tolerance of Listeria monocytogenes. Infect. Immun. monocytogenes required for entry into eukaryotic
species. Science 294, 849–852 (2001). 73, 894–904 (2005). cells and virulence. Mol. Microbiol. 51, 1601–1614
Comparative analysis of the genome sequences 92. Sleator, R. D., Wemekamp-Kamphuis, H. H., (2004).
of L. monocytogenes (which is pathogenic) and Gahan, C. G., Abee, T. & Hill, C. A PrfA-regulated 110. Cabanes, D. et al. Gp96 is a receptor for a novel
L. innocua (which is non-pathogenic), greatly bile exclusion system (BilE) is a novel virulence factor Listeria monocytogenes virulence factor, Vip, a surface
advancing the understanding of Listeria species in Listeria monocytogenes. Mol. Microbiol. 55, protein. EMBO J. 24, 2827–2838 (2005).
pathogenicity. 1183–1195 (2005). 111. Li, Z., Dai, J., Zheng, H., Liu, B. & Caudill, M. An
77. Williams, T., Bauer, S., Beier, D. & Kuhn, M. 93. Hardy, J. et al. Extracellular replication of Listeria integrated view of the roles and mechanisms of heat
Construction and characterization of Listeria monocytogenes in the murine gall bladder. Science shock protein gp96–peptide complex in eliciting
monocytogenes mutants with in-frame deletions in the 303, 851–853 (2004). immune response. Front. Biosci. 7, d731–d751 (2002).
response regulator genes identified in the genome 94. Goetz, M. et al. Microinjection and growth of bacteria 112. Nelson, K. E. et al. Whole genome comparisons of
sequence. Infect. Immun. 73, 3152–3159 (2005). in the cytosol of mammalian host cells. Proc. Natl serotype 4b and 1/2a strains of the food-borne
78. Autret, N., Raynaud, C., Dubail, I., Berche, P. & Acad. Sci. USA 98, 12221–12226 (2001). pathogen Listeria monocytogenes reveal new insights
Charbit, A. Identification of the agr locus of Listeria 95. Chico-Calero, I. et al. Hpt, a bacterial homolog of the into the core genome components of this species.
monocytogenes: role in bacterial virulence. Infect. microsomal glucose-6-phosphate translocase, Nucleic Acids Res. 32, 2386–2395 (2004).
Immun. 71, 4463–4471 (2003). mediates rapid intracellular proliferation in Listeria. 113. Cossart, P. & Lecuit, M. Interactions of Listeria
79. Mandin, P. et al. VirR, a response regulator critical for Proc. Natl Acad. Sci. USA 99, 431–436 (2002). monocytogenes with mammalian cells during entry
Listeria monocytogenes virulence. Mol. Microbiol. 57, 96. O’Riordan, M., Moors, M. A. & Portnoy, D. A. Listeria and actin-based movement: bacterial factors, cellular
1367–1380 (2005). intracellular growth and virulence require host-derived ligands and signaling. EMBO J. 17, 3797–3806
80. Cotter, P. D., Emerson, N., Gahan, C. G. & Hill, C. lipoic acid. Science 302, 462–464 (2003). (1998).
Identification and disruption of lisRK, a genetic locus 97. Klarsfeld, A. D., Goossens, P. L. & Cossart, P. Five 114. Schubert, W. D. et al. Structure of internalin, a major
encoding a two-component signal transduction system Listeria monocytogenes genes preferentially invasion protein of Listeria monocytogenes, in
involved in stress tolerance and virulence in Listeria expressed in infected mammalian cells: plcA, purH, complex with its human receptor E-cadherin. Cell 111,
monocytogenes. J. Bacteriol. 181, 6840–6843 purD, pyrE and an arginine ABC transporter gene, 825–836 (2002).
(1999). arpJ. Mol. Microbiol. 13, 585–597 (1994). 115. Tweten, R. K. Cholesterol-dependent cytolysins, a
81. Dons, L. et al. Role of flagellin and the two-component 98. Chatterjee, S. S. et al. Intracellular gene expression family of versatile pore-forming toxins. Infect. Immun.
CheA/CheY system of Listeria monocytogenes in host profile of Listeria monocytogenes. Infect. Immun. 74, 73, 6199–6209 (2005).
cell invasion and virulence. Infect. Immun. 72, 1323–1338 (2006).
3237–3244 (2004). 99. Joseph, B. et al. Identification of Listeria Acknowledgements
82. Christiansen, J. K., Larsen, M. H., Ingmer, H., monocytogenes genes contributing to intracellular Our sincere apologies to colleagues whose work has not been
Sogaard-Andersen, L. & Kallipolitis, B. H. The RNA- replication by expression profiling and mutant cited in this manuscript because of space constraints and
binding protein Hfq of Listeria monocytogenes: role in screening. J. Bacteriol. 188, 556–568 (2006). because of the focus of this Review, which does not claim to
stress tolerance and virulence. J. Bacteriol. 186, An important study that identifies the listerial be comprehensive. We thank E. Gouin for critical assessment
3355–3362 (2004). genes that are required to sustain growth in the of the manuscript and E. Veiga for help with FIG. 2. Work in
83. Archambaud, C., Gouin, E., Pizarro-Cerda, J., cytoplasm of the host cell. the laboratory of P.C. is supported by the Institut National de
Cossart, P. & Dussurget, O. Translation elongation 100. Doumith, M. et al. New aspects regarding evolution la Recherche Agronomique, the Institut Pasteur, the Institut
factor EF-Tu is a target for Stp, a serine–threonine and virulence of Listeria monocytogenes revealed by National de la Santé et de la Recherche Médicale, the
phosphatase involved in virulence of Listeria comparative genomics and DNA arrays. Infect. Immun. Ministère de l’Education Nationale et de la Recherche
monocytogenes. Mol. Microbiol. 56, 383–396 72, 1072–1083 (2004). Scientifique et Technique, and the Association par la
(2005). 101. Cabanes, D., Dehoux, P., Dussurget, O., Frangeul, L. & Recherche sur le Cancer. M.H. is supported by a Pasteur
84. Annous, B. A., Becker, L. A., Bayles, D. O., Labeda, D. P. Cossart, P. Surface proteins and the pathogenic Foundation fellowship. P.C. is an international research
& Wilkinson, B. J. Critical role of anteiso-C15:0 fatty potential of Listeria monocytogenes. Trends Microbiol. scholar of the Howard Hughes Medical Institute.
acid in the growth of Listeria monocytogenes at low 10, 238–245 (2002).
temperatures. Appl. Environ. Microbiol. 63, 102. Dhar, G., Faull, K. F. & Schneewind, O. Anchor structure Competing interests statement
3887–3894 (1997). of cell wall surface proteins in Listeria monocytogenes. The authors declare no competing financial interests.
85. Angelidis, A. S. & Smith, G. M. Role of the glycine Biochemistry 39, 3725–3733 (2000).
betaine and carnitine transporters in adaptation of 103. Bierne, H. et al. Inactivation of the srtA gene in
Listeria monocytogenes to chill stress in defined Listeria monocytogenes inhibits anchoring of surface
DATABASES
medium. Appl. Environ. Microbiol. 69, 7492–7498 proteins and affects virulence. Mol. Microbiol. 43,
The following terms in this article are linked online to:
(2003). 869–881 (2002).
Entrez Genome Project: http://www.ncbi.nlm.nih.gov/
86. Ko, R., Smith, L. T. & Smith, G. M. Glycine betaine 104. Pucciarelli, M. G. et al. Identification of substrates of
entrez/query.fcgi?db=genomeprj
confers enhanced osmotolerance and cryotolerance on the Listeria monocytogenes sortases A and B by a non-
Bacillus subtilis | Burkholderia pseudomallei | Chlamydia
Listeria monocytogenes. J. Bacteriol. 176, 426–431 gel proteomic analysis. Proteomics 5, 4808–4817
trachomatis | Escherichia coli | Listeria innocua | Listeria
(1994). (2005).
ivanovii | Listeria monocytogenes | Listeria welshimeri | Shigella
87. Junttila, J. R., Niemela, S. I. & Hirn, J. Minimum 105. Sabet, C., Lecuit, M., Cabanes, D., Cossart, P. &
flexneri | Yersinia pestis
growth temperatures of Listeria monocytogenes and Bierne, H. LPXTG protein InlJ, a newly identified
UniProtKB: http://ca.expasy.org/sprot
non-haemolytic Listeria. J. Appl. Bacteriol. 65, internalin involved in Listeria monocytogenes
InlA | InlB | LLO | PrfA
321–327 (1988). virulence. Infect. Immun. 73, 6912–6922 (2005).
88. Cotter, P. D., Gahan, C. G. & Hill, C. Analysis of the 106. Reglier-Poupet, H. et al. Maturation of lipoproteins by FURTHER INFORMATION
role of the Listeria monocytogenes F0F1-ATPase type II signal peptidase is required for phagosomal Bacteria–Cells Interactions Unit, Institut Pasteur:
operon in the acid tolerance response. Int. J. Food escape of Listeria monocytogenes. J. Biol. Chem. 278, http://www.pasteur.fr/recherche/unites/uibc/welcome.html
Microbiol. 60, 137–146 (2000). 49469–49477 (2003). ListiList: http://genolist.pasteur.fr/ListiList/
89. Cotter, P. D., Gahan, C. G. & Hill, C. A glutamate 107. Braun, L. et al. InlB: an invasion protein of Listeria Microbiology in Motion:
decarboxylase system protects Listeria monocytogenes monocytogenes with a novel type of surface http://www.nature.com/nrmicro/animation/index.html
in gastric fluid. Mol. Microbiol. 40, 465–475 (2001). association. Mol. Microbiol. 25, 285–294 (1997). The Prokaryotes: An Evolving Electronic Resource for the
90. Dussurget, O. et al. Listeria monocytogenes bile salt 108. Lenz, L. L., Mohammadi, S., Geissler, A. & Microbiological Community:
hydrolase is a PrfA-regulated virulence factor involved Portnoy, D. A. SecA2-dependent secretion of http://link.springer-ny.com/link/service/books/10125/
in the intestinal and hepatic phases of listeriosis. Mol. autolytic enzymes promotes Listeria monocytogenes
Microbiol. 45, 1095–1106 (2002). pathogenesis. Proc. Natl Acad. Sci. USA 100, SUPPLEMENTARY INFORMATION
91. Begley, M., Sleator, R. D., Gahan, C. G. & Hill, C. 12432–12437 (2003). See online article: S1 (figure) | S2 (figure) | S3 (figure)
Contribution of three bile-associated loci, bsh, pva, 109. Cabanes, D., Dussurget, O., Dehoux, P. & Cossart, P. Access to this links box is available online.
and btlB, to gastrointestinal persistence and bile Auto, a surface associated autolysin of Listeria

434 | JUNE 2006 | VOLUME 4 www.nature.com/reviews/micro


© 2006 Nature Publishing Group
View publication stats

You might also like