Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

REVIEWS

Salmonellae interplay with host cells


Andrea Haraga*, Maikke B. Ohlson‡ and Samuel I. Miller*‡§
Abstract | Salmonellae are important causes of enteric diseases in all vertebrates.
Characterization of the molecular mechanisms that underpin the interactions of
salmonellae with their animal hosts has advanced greatly over the past decade, mainly
through the study of Salmonella enterica serovar Typhimurium in tissue culture and animal
models of infection. Knowledge of these bacterial processes and host responses has
painted a dynamic and complex picture of the interaction between salmonellae and
animal cells. This Review focuses on the molecular mechanisms of these host–pathogen
interactions, in terms of their context, significance and future perspectives.

Pinocytosis Salmonellae are Gram-negative bacterial pathogens multiple mechanisms for crossing the intestinal barrier
A nonspecific process by which that are capable of infecting a wide range of animals, indicates the importance of this strategy to the lifestyle
small volumes of extracellular which can result in several manifestations of disease1. of salmonellae.
fluid are taken up by certain The host specificities and the disease symptoms that are M-cell-mediated uptake also allows salmonellae
eukaryotic cells owing to the
engulfment of fluid in small
caused by some of the thousands of Salmonella serovars to interact with, and possibly enter, intestinal epithe-
membrane vesicles. are listed in TABLE 1. Salmonellae are typically acquired
  lial cells through their basolateral surface. Epithelial
by the oral ingestion of contaminated food or water; turnover and shedding then, in turn, could return
Tight junction however, exposure to pet reptiles and amphibians, which the bacteria to the lumen of the intestine, simply
The connection between two
are often carriers of the bacteria, can also pose a risk for requiring that they survive and, perhaps, replicate
adjacent cells in a monolayer
that is formed by extracellular-
salmonellosis in humans (FIG. 1). Although conditions that
  intracellularly. Such mechanisms are possibly impor-
matrix and protein complexes; increase the gastric pH reduce the infectious dose of the tant for intestinal colonization and even for acute
impermeable to water and bacterium, salmonellae have an adaptive acid-tolerance disease by salmonellae, as a great deal of data, from
other molecules. response that might promote their survival in the low both animal and cell culture models, indicate that the
Macropinocytosis
pH milieu of the stomach2. After entering the small specialized ability to invade and survive in epithe-
Used to refer to the intestine, salmonellae traverse the intestinal mucous lial cells is essential for their pathogenesis 13–17. By
endo­cytosis of large volumes   layer and evade being killed by digestive enzymes, bile contrast, the importance of phagocytosis and traf-
of extracellular fluid and salts, secretory IgA, antimicrobial peptides and other ficking through CD18-positive cells is unknown and
particles by membrane ruffles.
innate immune defences in order to gain access to the might represent another mechanism, redundant to
underlying epithelium3–5. Salmonellae have the ability to M-cell-mediated entrance, for salmonellae to cross
invade the non-phagocytic enterocytes of the intestinal the intestinal barrier without actively interacting with the
epithelium by bacterial-mediated endocytosis6. After epithelial surface. It is interesting to speculate that this
adherence to the apical surface of the cell, using vari- method may be important for typhoid fever, in which so
ous fimbrial adhesins, the bacteria disrupt the epithelial few organisms can cause disease but induce only mini-
brush border and induce membrane ruffles that engulf mal intestinal disturbance in the majority of cases. This
the organisms7,8. However, salmonellae preferentially could allow Salmonella enterica serovar Typhi (S.  typhi)
enter microfold (M) cells, which are specialized epithelial to disseminate systemically in a relatively silent fashion,
*Department of Genome cells that sample intestinal antigens through pinocytosis as opposed to mechanisms that involve bacterial-medi-
Sciences, ‡Department of and transport them to lymphoid cells in the underlying ated endocytosis, which are associated with intestinal
Microbiology, §Department of
Peyer’s patches9,10. Alternatively, they can translocate inflammation and diarrhoea13,14.
Medicine, University of
Washington, Seattle, through the intestinal epithelia after uptake by CD18- Once the epithelial barrier has been breached,
Washington 98195, USA. expressing phagocytes11. Moreover, in vitro, salmonellae Salmonella serotypes that are associated with systemic
Correspondence to S.I.M. are able to disrupt tight junctions, which seal the epithelial illness can enter intestinal macrophages by inducing
e‑mail: millersi@u.washington. cell layer and restrict the paracellular passage of ions, macropinocytosis , sensing the phagosomal environ-
edu
doi:10.1038/nrmicro1788
water and immune cells12. This, in addition to intes- ment and activating various virulence mechanisms
Published online   tinal inflammatory responses, probably contributes in order to survive in the microbicidal environment
19 November 2007 to the induction of diarrhoea. The fact that there are of the macrophage 18–25 . This promotes bacterial

nature reviews | microbiology volume 6 | january 2008 | 53


© 2008 Nature Publishing Group
REVIEWS

Table 1 | Examples of Salmonella enterica serovars; their hosts and diseases and a hollow needle29,30. Another set of proteins comprise
the translocon, which is thought to be involved in the
Salmonella Host Disease and symptoms translocation of effectors by forming a translocation pore
enterica serovar specificity
in the host cellular membrane31. The effector proteins
Typhoid have been shown to contain specific targeting signals
Typhi; Human- Enteric fever: fever; abdominal pain; transient located in their amino termini that route them to the
Paratyphi restricted diarrhoea or constipation; and a salmon- T3SS32–35. In addition to the short secretion signal, many
coloured maculopapular rash on the trunk effectors have a binding site for a specific chaperone,
Non-typhoid the function of which is thought to be to stabilize and
Typhimurium; Broad-range Gastroenteritis: abdominal pain; vomiting; and target its cognate substrate to the translocation appara-
Enteriditis inflammatory diarrhoea tus34,36,37. Most chaperones are specific for a single effec-
tor, but some can facilitate the secretion of more than one
effector protein38–41. An ATPase that is located in the base
replication and subsequent dissemination throughout of the NC not only drives the export process but also
the reticulo­endothelial system (RES). Infection with facilitates the release of effectors from chaperones before
non‑typhoidal strains in healthy human adults, however, transport42. Although much of the T3SS is similar among
is usually limited to the intestine, where the bacteria Gram-negative bacteria, the set of effector proteins is
induce an early inflammatory response that results in the unique to each species. For a complete list of Salmonella
infiltration of polymorphonuclear leukocytes (PMNs) spp. effectors, their host-cell functions and binding
into the intestinal lumen1,26. Production of the potent partners, see Table 2.
PMN chemokine interleukin (IL)‑8, and, perhaps, other Salmonellae encode two distinct virulence­­‑associated
similar molecules, by the infected epithelia is thought T3SSs within Salmonella pathogenicity islands 1 and 2
to be required for this process27. The release of cyto- (SPI1 and SPI2) that function at different times during
toxic granules by PMNs, as well as the effects of various infection28. Whereas the SPI1-encoded T3SS is active
bacterial molecules in stimulating innate immune on contact with the host cell and translocates bacterial
inflammatory responses and manipulating host- proteins across the plasma membrane, the SPI2 T3SS is
cell processes, may then result in the destruction or expressed within the phagosome and translocates effectors
turnover of the intestinal mucosa, which contributes across the vacuolar membrane. The SPI1 system has been
to inflammatory diarrhoea. shown to be required for invasion of non-phagocytic
Recent progress has been made in our understanding cells, induction of intestinal inflammatory responses
of the molecular mechanisms by which salmonel- and diarrhoea, as well as colonization of the intestine.
lae interact with host cells and manipulate various host The SPI2 T3SS, by contrast, has an important role in
responses that result in the induction of intestinal bacterial survival in macrophages and establishment of
inflammatory responses and systemic illness. Specifically, systemic disease. Interestingly, Brown and colleagues43
the identification of bacterial and host proteins that are have recently shown by RIVET (recombination-based
involved in such processes as the invasion of epithelial in vivo expression technology) that expression of the
cells, stimulation and repression of signalling cascades, SPI2 T3SS in mice might begin in the early stages of
sensing of the intracellular environment and establishment Salmonella enterica serovar Typhimurium (S. typhimu-
of a niche for intracellular replication have contributed to rium) infection, before intestinal penetration. As there
insights into the complex pathogenesis of this bacterium, is currently no evidence that this T3SS is also involved
and will be discussed here in detail. in intestinal colonization, the authors speculated that its
early induction in the intestinal lumen might prepare
Reticuloendothelial system The two virulence-associated T3SSs the bacterium for the inhospitable intracellular environ-
(RES). The meshwork of A specialized apparatus, named the type III secretion ment of the macrophage and ease the transition to the
connective tissue that contains
system (T3SS), is essential to salmonellae pathogen- later systemic phase of the disease. However, it is also
immune cells, such as
macrophages, and surrounds esis and the colonization of host tissues. The T3SS possible that early expression of this T3SS is required for
tissues that are associated mediates the transfer of bacterial virulence proteins, the optimal function of the invasion-associated T3SS, as
with the immune system, such known as effectors, from the bacterial cell into the mutations in the SPI2 apparatus can cause a significant
as the spleen and lymph host-cell cytoplasm28. Once inside the eukaryotic cell, reduction in the expression of several SPI1 T3SS genes
nodes. Immune cells in the RES
provide surveillance of antigens
these effectors can alter host cellular functions, such as and impair the ability of the bacterium to invade epi-
that the body encounters and cytoskeletal architecture, membrane trafficking, signal trans- thelial cells44,45. Conversely, there is mounting evidence
can be quickly recruited to duction and cytokine gene expression, in order to promote that some of the effectors of the SPI1 T3SS are expressed,
sites of infection. bacterial survival and colonization. The T3SS is or persist within, host cells long after bacterial inter-
evolutionarily related to the flagellar export system and nalization and contribute to events that were previously
Pathogenicity island
A large region of genomic DNA is present in multiple Gram-negative animal and plant attributed exclusively to effectors of the SPI2 T3SS46–51.
that encodes genes that are pathogens. It comprises more than 20 proteins, some of Although it is unknown whether mutations in the SPI1
associated with virulence. A which form a supramolecular structure that is known as T3SS could cause pleiotropic effects, these findings indi-
pathogenicity island is typically the needle complex (NC) (FIG. 2). Electron micrographs of cate that the two Salmonella T3SSs do not operate in an
transferred horizontally
between bacterial strains and
purified T3SSs revealed that the NC consists of a multi- independent manner, as previously thought, and pos-
is often inserted into tRNA ring base that spans the inner and outer membranes of sibly cooperate to facilitate the intracellular lifestyle of
genes within the genome. the bacterial envelope, an inner rod that joins the rings the bacteria. This is particularly intriguing, because the

54 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS

older data that had suggested a role for the SPI1 T3SS
during systemic disease17,63. This, combined with the
Salmonella spp. previously mentioned putative function of the SPI2 T3SS
prior to intestinal penetration, further demonstrates that
M cell the dogmatic compartmentalization of the roles of the two
Epithelial cell T3SSs is simplistic. In addition, the recent resurrection of
the streptomycin-treated mouse model of acute intestinal
infection with inflammation and diarrhoea might enable
dissection of the roles of the host and bacteria in acute
intestinal disease64. It is likely that, in the near future,
models of salmonellae-induced chronic intestinal disease
Macrophage at the intestinal mucosal surface will be further devel-
T cell B cell oped and studied. In fact, our group has demonstrated
Gastroenteritis: that the A/J mouse strain develops this type of disease
PMN influx (L. Peiser, K. Smith and S.I.M., unpublished observa-
tions). These animal models should be useful in under-
standing the contributions of mucosal immune defences,
as well as bacterial factors such as T3SS effectors, to
Enteric fever: dissemination chronic intestinal disease. However, these experimental
to lymph nodes, liver and spleen systems also have limitations, as specific disease outcomes
may be different in different hosts for broad host-range
Figure 1 | Biology of salmonellae infection. Orally ingested salmonellae
Nature Reviews |survive at the
Microbiology salmonellae. A future challenge in understanding these
low pH of the stomach and evade the multiple defences of the small intestine in order to animal models will be to determine the effects of specific
gain access to the epithelium. Salmonellae preferentially enter M cells, which transport effectors in both the model and natural hosts. In addi-
them to the lymphoid cells (T and B) in the underlying Peyer’s patches. Once across the
tion, the diversity and evolution of the effector content of
epithelium, Salmonella serotypes that are associated with systemic illness enter intestinal
macrophages and disseminate throughout the reticuloendothelial system. By contrast, different Salmonella strains isolated from natural sources
non-typhoidal Salmonella strains induce an early local inflammatory response, which might also reveal different functions or roles that cannot
results in the infiltration of PMNs (polymorphonuclear leukocytes) into the intestinal be explored by these model systems. In this regard, many
lumen and diarrhoea. S. typhimurium effectors are present in only a percentage
of strains, and in some serotypes, such as Typhi, are only
present as pseudogenes. Selection for the presence or
sequence information and genetic organization of the loss of effectors in specific animal populations or human
two Salmonella T3SSs, as well as their phylogenetic disease settings might, ultimately, be more informative of
distribution among Salmonella species, indicate that the actual natural disease or colonization than the phe-
they were acquired independently, at different periods notypes of deletion mutants in model systems. Therefore,
and from different sources44,52–57. Therefore, it is likely the acceptance of the limitations of these animal mod-
that selection pressure from animal environments led els is important to prevent dogmatic viewpoints about
to the cooperation between the two T3SSs, to opti- the roles of such mechanisms or specific T3SS effector
mize colonization, invasion and intracellular survival proteins. This is particularly relevant to the study of effec-
of salmonellae in animals. tor proteins with redundant functions, as the necessity
The most extensively used animal model for investi- for their similar tasks during pathogenesis might not be
gating the contributions of the two T3SSs to salmonellae apparent in model systems. A complete picture will prob-
virulence is the highly sensitive natural-resistance- ably require the integration of animal model information
associated macrophage protein 1 (Nramp1)‑null mouse, with real world information about effector content and
which is susceptible to mortal infection if inoculated with disease course in nature.
as few as ten bacteria58. Nramp1 is a macrophage-specific
ion exporter that removes ions from the Salmonella- Bacterial-mediated endocytosis
containing vacuole (SCV) and, thus, restricts bacterial The concerted function of at least five SPI1 T3SS effec-
replication59. The Nramp1-null mouse model system tors is required for efficient invasion of cultured epithe-
has been used successfully to identify many important lial cells, although optimal invasion in animal tissues
genes that are required for T3SS-associated virulence60,61. might be more complex and diverse (FIG. 3). SopE, SopE2
However, it is too sensitive to detect phenotypes for and SopB (also known as SigD) activate the host Rho
T3SS effectors that contribute to long-term persist- GTPases Cdc42, Rac1 and RhoG, which leads to actin
ence, because these mice do not survive long enough cytoskeletal reorganization, membrane ruffling and bac-
to study the proteins that are necessary for systemic terial internalization by macropinocytosis65–70. However,
colonization. The resurgence of interest in the resistant recent evidence suggests that only Rac1 and RhoG are
Nramp1-positive mouse model has enabled studies to indispensable for the actin remodelling events that are
be performed that are more physiologically relevant generated by Salmonella spp. during host-cell entry70.
to long-term systemic infection49,62. These reports SopE, SopE2 and SopB are all essential for the invasion of
confirmed that SPI2 T3SS effectors are important for epithelial cells, as an S. typhimurium mutant that is
persistent infection and colonization, but also validated defective in all three effectors cannot induce actin

nature reviews | microbiology volume 6 | january 2008 | 55


© 2008 Nature Publishing Group
REVIEWS

a modulating actin dynamics directly. SipA (also


called SspA) helps to initiate actin polymerization
at the site of S. typhimurium entry by decreasing the
critical concentration and increasing the stability

Needle
of actin filaments, whereas SipC (also called SspC),
an effector that also functions as a translocon and is
inserted in the host cell’s plasma membrane with a
cytoplasmic domain that may bind to intermediate
filaments, can nucleate and bundle actin 75–77. In
addition, SipA can enhance the activity of SipC

Base
independently of any host cellular protein, which
indicates that there might be a unique collaboration
between the two effectors78. Although SipA and SipC
might act in concert with SopE, SopE2 and SopB to
mediate the cytoskeletal changes that are required
b
Base
for the formation of membrane ruffles, they can-
not induce membrane ruffling and invasion by
themselves 69,76,77,79. Instead, they seem to facilitate
efficient bacterial uptake by directing the spatial
localization of actin foci beneath the invading bac-
teria and, perhaps, preventing disassembly of the
S. typhimurium-induced actin structures.

Needle Intestinal inflammatory responses


The stimulation of Cdc42 by SopE, SopE2 and SopB
also triggers several mitogen-activated protein kinase
(MAPK) pathways, including the Erk, Jnk and p38 path-
ways, which results in the activation of the transcription
factors activator protein‑1 (AP‑1) and nuclear factor-κB
(NF-κB)16,70,80 (FIG. 3). These transcription factors then
direct the production of pro-inflammatory cytokines,
Figure 2 | Structure of the Salmonella type III
Nature Reviews | Microbiology such as IL‑8, which stimulate PMN transmigration and
secretion system (T3SS). a | Electron micrographs of a
the inflammatory response leading to diarrhoea. Boyle
purified Salmonella enterica serovar Typhimurium
(S. typhimurium) needle complex (NC). b | Electron
and colleagues81 have recently reported that disrup-
cryomicroscopy and surface images of the structures of tion of tight junction structure and function — one
the base and NC of the S. typhimurium T3SS. Part a aspect of S. typhimurium-induced enteritis that is
reproduced, with permission, from Ref. 31 (2002) likely to be important — by the effectors SopB, SopE,
Elsevier Science. Part b reproduced, with permission, SopE2 and SipA also occurs by activation of Rho fam-
from Ref. 185  (2004) American Association for the ily GTPases. In addition, SopB also promotes intestinal
Advancement of Science. disease by increasing the intracellular concentration of
d‑myo-inositol 1,4,5,6-tetrakisphosphate, a compound
rearrangements and, therefore, become intracellular69. that stimulates cellular chloride secretion and fluid
Whereas SopE and SopE2 are potent guanine nucleotide flux69,82. SipB (also called SspB), an effector protein that
exchange factors (GEFs) for all three GTPases, SopB is also part of the SPI1 T3SS translocation machinery,
only stimulates Cdc42 and RhoG indirectly through its adds to the inflammatory response by increasing pro-
phosphoinositide phosphatase activity65,67–70. Although duction of the pro-inflammatory cytokines IL‑1β and
the mechanism of action of SopB is not yet understood, IL‑18 through binding and activating caspase‑1 (Ref. 83).
Patel and colleagues70 have recently shown that activa- Caspase‑1 activation can also occur by the recognition of
tion of RhoG by SopB occurs by a cellular exchange flagellin by the Ipaf cytoplasmic signalling pathway84,85.
factor called SGEF. As SGEF has a phosphoinositide- It is plausible that this is the result of the accidental
binding pleckstrin homology domain, it is plausible translocation of flagellin into the host cell’s cytoplasm
that it is activated by the phosphoinositide fluxes that are by the SPI1 T3SS owing to the conserved evolution of
generated by the enzymatic action of SopB71. Activation the flagellar and the SPI1-encoded secretion systems.
of Rho GTPases then results in the activation of the The observation that caspase‑1-deficient mice are more
Wiskott–Aldrich Syndrome protein (WASP) family susceptible to orally administered S. typhimurium than
members N‑WASP and WAVE2, which leads to recruit- wild-type mice indicates that SPI1 T3SS-mediated cas-
ment of the actin-related protein‑2/3 (Arp2/3) complex pase‑1 activation could be essential to diarrhoeal disease
to sites of membrane ruffles and stimulation of actin and intestinal survival86,87. SopA and SopD also contrib-
polymerization67,72–74. ute to enteritis in calves13,88,89. Recent evidence indicates
There are two SPI1 T3SS effectors that promote that SopA has a HECT (homologous to E6-AP carboxyl
bacterial internalization by binding to actin and terminus)-like E3 ubiquitin ligase activity, which is

56 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS

Table 2 | Effectors of the Salmonella SPI1- and SPI2-encoded type III secretion systems (T3SSs)
Effector Cellular function Host-cell target References
SPI1 T3SS
AvrA Inhibits nuclear factor (NF)‑κB signalling and interleukin (IL)‑8 Unknown 99,186
production; also prevents ubiquitination of β‑catenin
SipA or SspA Decreases the critical concentration of G‑actin and increases the F-actin; T‑plastin 76, 81,187,188
stability of F‑actin; also induces PMN transepithelial migration
and disrupts tight junctions
SipB or SspB* Binds and activates caspase‑1 and induces autophagy in Caspase‑1; cholesterol 83,189,190
macrophages
SipC or SspC* Nucleates and bundles actin F-actin; cytokeratin‑8 and 75,77,191
cytokeratin-18
SopA Stimulates PMN transmigration by HECT-like E3 ubiquitin ligase Unknown 89,90
activity
SopB or SigD Activates Cdc42, RhoG, AktA and chloride secretion through its Unknown 69,70,81,82,192
inositol phosphatase activity and disrupts tight junctions
SopD Stimulates fluid accumulation in bovine ligated ileal loops and Unknown 13,82,91
contributes to diarrhoea in calves and systemic disease in mice
SopE Activates Cdc42, Rac1 and RhoG by its GEF activity and disrupts Cdc42, Rac1 and Rab5 65,81,193
tight junctions
SopE2 Activates Cdc42, Rac1 and RhoG by its GEF activity and disrupts Cdc42 and Rac1 67,68,81
tight junctions
SptP Inhibits Cdc42 and Rac1 by its GAP activity and MAPK signalling Rac1 92,94,95
and IL‑8 secretion through its tyrosine phosphatase activity
SPI2 T3SS
GogB Unknown Unknown 194
PipB Unknown Unknown 157
PipB2 Contributes to Sif formation Kinesin‑1 157,165,170
SifA Induces Sif formation, maintains integrity of the SCV and SKIP and Rab7 154,164,166, 181,195
downregulates kinesin recruitment to the SCV
SifB Unknown Unknown 156
SopD2 Contributes to Sif formation Unknown 91
SpiC* Interferes with endosomal trafficking Hook3 150,151–153
SpvB‡ Actin-specific ADP-ribosyltransferase and downregulates Sif Actin 160, 175,176,196
formation
SseF Contributes to Sif formation and microtubule bundling Unknown 162,169
SseG Contributes to Sif formation and microtubule bundling Unknown 162,169
SseI or SrfH Contributes to host-cell dissemination Filamin and TRIP6 160,197
SseJ Maintains integrity of the SCV and has deacylase activity Unknown 155,180
SseK1 Unknown Unknown 198
SseK2 Unknown Unknown 198
SseL Deubiquitinase Ubiquitin 199
SspH2 Inhibits the rate of actin polymerization and contributes to Filamin and profilin 96,160
virulence in calves
SteA Unknown Unknown 200
SteB Unknown Unknown 200
SteC Unknown Unknown 200
SPI1 and SPI2 T3SS
SlrP Contributes to virulence in calves Unknown 201
SspH1 Inhibits NF‑κB signalling and IL‑8 secretion, contributes to PKN1 95–98
virulence in calves and has E3 ubiquitin ligase activity
*Also a component of the secretion apparatus. ‡Has not been definitively shown to be an SPI2 T3SS effector. GAP, GTPase-activating protein; GEF, guanine
nucleotide exchange factor; HECT, homologous to E6-AP carboxyl terminus; MAPK, mitogen-activated protein kinase; PMN, polymorphonuclear leukocyte; SCV,
Salmonella-containing vacuole; Sif, Salmonella-induced filament; SPI, Salmonella pathogenicity island.

nature reviews | microbiology volume 6 | january 2008 | 57


© 2008 Nature Publishing Group
REVIEWS

a multifunctional protein that is also involved in the


Membrane reversal of the pro-inflammatory signalling cascade
Tight
ruffle
junction that is associated with invasion94. It has tyrosine phos-
Salmonella phatase activity, which has a role in downregulation
spp.
of the S. typhimurium-induced activation of the MAPK
Actin
Erk. In addition, S. typhimurium can downregulate IL‑8
production after invasion of intestinal epithelial cells,
SopE SipA SopA and SptP and SspH1 participate in this process95. SspH1
Epithelial
cell
SopE2 SipC SptP is a leucine-rich-repeat protein that is localized to the
SopB
SopB Rho GTPases mammalian nucleus and inhibits NF‑κB-dependent
gene expression95,96. It has also been shown to have
Rho GTPases MAPK CI– MAPK SspH1
AvrA ubiquitin ligase activity97. As it interacts with a host
NF-κB
CI– NF-κB serine/threonine protein kinase called PKN1, which if
AP-1 activated also inhibits NF‑κB-dependent gene expres-
sion, SspH1 could interfere with inflammatory signal-
ling by binding to and activating PKN1 (Ref. 98). In fact,
IL-1β
IL-8
IL-18 PKN1 is activated in epithelial cells during infection
with S. typhimurium. However, as this also occurs in
Macrophage the absence of SspH1, it is possible that SspH1 is not
SipB required for this process or that there are additional
PMN
effectors or bacterial mechanisms that are capable of
activating PKN1. Another SPI1 T3SS effector, AvrA,
Figure 3 | SPI1 T3SS-induced changes in host cells. On contact with the epithelial has been reported to inhibit NF‑κB activity and pro-
Nature Reviews | Microbiology
cell, salmonellae assemble the Salmonella pathogenicity island 1(SPI1)-encoded type III inflammatory cytokine secretion99. However, unlike
secretion system (T3SS) and translocate effectors (yellow spheres) into the eukaryotic its Yersinia spp. homologue, YopJ, which binds and
cytoplasm. Effectors, such as SopE, SopE2 and SopB, then activate host Rho GTPases, acetylates MAPK kinases and the inhibitor of NF-κB
which results in the rearrangement of the actin cytoskeleton into membrane ruffles, kinase (IKK), thereby preventing their phosphorylation
induction of mitogen-activated protein kinase (MAPK) pathways and destabilization of
and activation, AvrA has not been shown to interact
tight junctions. Changes in the actin cytoskeleton, which are further modulated by the
actin-binding proteins SipA and SipC, lead to bacterial uptake. MAPK signalling with or affect the activity of any host proteins that are
activates the transcription factors activator protein‑1 (AP‑1) and nuclear factor-κB involved in the NF‑κB pathway100,101.
(NF‑κB), which turn on production of the pro-inflammatory polymorphonuclear The fact that salmonellae use multiple effectors to
leukocyte (PMN) chemokine interleukin (IL)‑8. SipB induces caspase‑1 activation in dampen the host’s inflammatory response indicates
macrophages, with the release of IL‑1β and IL‑18, so augmenting the inflammatory that this function is important for their pathogenesis,
response. In addition, SopB stimulates Cl– secretion by its inositol phosphatase activity. perhaps by contributing to colonization of the intestinal
The destabilization of tight junctions allows the transmigration of PMNs from the tract over prolonged periods of time. Furthermore, it is
basolateral to the apical surface, paracellular fluid leakage and access of bacteria to the interesting to speculate that the evolutionary pressure
basolateral surface. However, the transmigration of PMNs also occurs in the absence of on the host–pathogen interaction for non-typhoidal
tight-junction disruption and is further promoted by SopA. The actin cytoskeleton is
salmonellae is for asymptomatic intestinal colonization,
restored and MAPK signalling is turned off by the enzymatic activities of SptP. This also
results in the down-modulation of inflammatory responses, to which SspH1 and AvrA whereas for typhoidal strains, which have poor intesti-
also contribute by inhibiting activation of NF‑κB. Figure reproduced from an original nal colonization, the selective pressure is for replication
kindly provided by A. Haraga, University of Washington, USA. within professional phagocytes, a condition that is also
less inflammatory than extracellular growth. The bacte-
ria might induce a mild state of inflammatory response
involved in the S. typhimurium-induced transepithelial and their goal might be to evolve towards parasitism or
migration of PMNs90. However, how SopD, an effector that commensalism. In fact, the long period of intestinal colo-
is also expressed under SPI2 T3SS-inducing conditions nization greatly exceeds that of the acute disease phase for
and continues to contribute to later stages of infection, non-typhoidal strains102. Also, typhoidal disease in the
exerts its effects remains unknown33,91. In summary, all absence of antibiotic therapy is often characterized by
of these mechanisms strongly implicate the SPI1 T3SS in periods of few or no symptoms, followed by a relapse that
the induction of gastroenteritis and intestinal disease. is more of a chronic, prolonged febrile illness, in which
intracellular replication is the predominant bacterial
Reversal of cytoskeletal and signalling responses lifestyle1. Thus, it is likely that a dynamic induction of
Interestingly, shortly after bacterial invasion the actin inflammatory responses, followed by their dampening, is
Transepithelial migration
cytoskeleton regains its normal architecture7. SptP has important to the diseases caused by salmonellae.
The movement of cells, such as
neutrophils and invading been shown to participate in this reversal process by
bacteria, from the basolateral its GTPase-activating protein activity on Cdc42 and The SCV as a niche for replication
(bottom) to the apical (top) Rac1 (Ref. 92) (FIG. 3). The opposing activities of SopE Salmonellae are capable of infecting and replicating in
surface, or the reverse, of an or SopE2 and SptP are mediated by the temporal regula- many cell types, but are thought to primarily replicate
epithelial cell layer. Migration
can also occur between two
tion of these proteins93. SopE has a shorter half-life in in macrophages, as they are found in the lymphatic tis-
adjacent cells through tight eukaryotic cells than SptP owing to its ubiquitination sues and organs of the RES during systemic infection1,103.
junctions. and rapid proteasome-dependent degradation. SptP is Salmonella strains that are defective for macrophage

58 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS
Salmonella spp.

intracellularly from hours to days, making it a unique


phagosome with respect to the normal progression of
phagolysosomal maturation and recycling. Although
there has been controversy within the field, several
SspH2 SifA reports have shown that salmonellae can survive within
SpvB PipB2 s macrophages in which the lysosomal compartments
Spacious Ssel tor
phagosome le mo have fused with the SCV107–110. Consequently, the avoid-
u
tub ance of phagolysosomal fusion is unlikely to be a major
M icro Sif
SCV pathogenic strategy of salmonellae. Studies in various
cell types have also demonstrated that the vacuole acidi-
Actin SseJ fies; however, depending on the mechanism of host‑cell
Epithelial cell entry, vacuolar acidification may be delayed in both
macrophages and epithelial cells19,110–112. The SCV has
Microtubules
been shown to interact transiently with the early endo-
cytic pathway and quickly gain and lose early endocytic
markers, such as EEA1 (early endosomal antigen 1), TfR
Nucleus
(transferrin receptor) and the early endocytic traffick-
ing GTPases Rab5 and Rab11 (Refs 113,114). Several late
SseF
SseG endosomal markers are commonly associated with the
SCV at later time points, including the GTPase Rab7,
Golgi Secretory vesicles LAMP1 (lysosomal associated membrane protein 1),
LAMP2, LAMP3 and the vacuolar ATPase110,113,115,116.
There is conflicting data concerning the occurrence of
Figure 4 | Formation of the SCV and induction of the SPI2 Nature
T3SSReviews Microbiology
within| the host cell. M6PR (mannose‑6-phosphate receptor), LBPA (lysobi-
Shortly after internalization by macropinocytosis, salmonellae are enclosed in a sphosphatidic acid) and the lysosomal hydrolase cathep­
spacious phagosome that is formed by membrane ruffles. Later, the phagosome fuses sin D on the SCV115,117–120. Furthermore, cholesterol has
with lysosomes, acidifies and shrinks to become adherent around the bacterium. This is
been reported to accumulate on the SCV118,121. The pres-
called the Salmonella-containing vacuole (SCV), which contains the endocytic marker
lysosomal associated membrane protein 1 (LAMP‑1; purple). The Salmonella
ence or absence of different markers on the persistent
pathogenicity island 2 (SPI2) T3SS (type III secretion system) is induced within the SCV SCV may simply indicate that they are variably detected
and translocates effector proteins (yellow spheres) across the phagosomal membrane rather than reflect whether or not the SCV has matured
several hours after phagocytosis. The SPI2 T3SS effectors SifA and PipB2 contribute to through a normal endocytic pathway. As it has been
Salmonella-induced filament (Sif) formation along microtubules (green) and regulate shown that the SCV can fuse with lysosomes and acidify,
microtubule-motor (yellow star shape) accumulation on the Sif and the SCV. SseJ is a the ability of salmonellae to survive exposure to lyso-
deacylase that is active on the phagosome membrane. SseF and SseG cause somal contents reveals that resistance to antimicrobial
microtubule bundling adjacent to the SCV and direct Golgi-derived vesicle traffic peptides, nitric oxide and oxidative killing are important
toward the SCV. Actin accumulates around the SCV in a SPI2 T3SS-dependent manner, to its survival within macrophages and to virulence122–129.
in which SspH2, SpvB and SseI are thought to have a role.
This is supported by the observations that Salmonella spp.
mutants that are sensitive to these chemicals are attenu-
ated for mouse virulence, whereas mice that are deficient
replication are avirulent in mouse models of infection, for the production of these compounds have increased
which underscores the importance of bacterial survival susceptibility to Salmonella spp.23,104,129–131
and replication in macrophages to disease104. In addi-
tion, the observation that many attenuated strains of Sensing and response to the vacuole
salmonellae are auxotrophic, particularly for purines, Salmonellae sense the acidic environment of the SCV,
pyrimidines, amino acids and other metabolites that resulting in the induction of various regulatory systems
are required for their replication but that are not readily that promote intracellular survival, for example, by
available within mammalian cells, supports the concept surface remodelling of the protein, carbohydrate and
that intracellular replication is essential to salmonellae membrane components of the bacterial envelope19. Such
virulence105. Unlike non-phagocytic cells, salmonellae regulatory systems include OmpR/EnvZ, PhoP/PhoQ,
can enter macrophages by several endocytic processes, RpoS/RpoE, PmrA/PmrB, Cya/Cyp and cyclic diGMP,
including SPI1 and non-SPI1 T3SS-induced macropino- all of which confer resistance to antimicrobial peptides
cytosis20. Following SPI1 T3SS-induced macropinocyto- and oxidative stress18,124,127,129,132–135. The phagosomal
sis, a few SPI1 T3SS effectors, such as SipA, SopB, SopD environment is acidic, with a pH range of <5 to 5.5, has
Auxotrophic
An organism that cannot
and SopE2, persist within the cell and have recently been a concentration of magnesium and calcium in the 1 mM
synthesize certain organic implicated in contributing to the intracellular stages of range and contains antimicrobial peptides and oxygen
compounds, such as amino the infection process47,48,50,51,91,106. Once intracellular, and nitrogen radicals that can damage the bacterial
acids, that are necessary for its salmonellae remain inside a vacuolar compartment, cell19,111,112. Various studies indicate that both pH and
metabolism. For growth,
which has been named the spacious phagosome (SP)20 antimicrobial peptides are important signatures of the
auxotrophic organisms must
be able to take up the lacking (FIG. 4). The SP shrinks over minutes to hours to form phagosomal environment and such conditions activate
compound from the an adherent membrane around one or more bacteria, many of the regulators that are implicated in salmonellae
surrounding environment. which is then referred to as the SCV. The SCV can persist virulence24,25,125,136. It is likely that several sensory systems

nature reviews | microbiology volume 6 | january 2008 | 59


© 2008 Nature Publishing Group
REVIEWS

respond to the phagosome environment and cooperate endosome–endosome fusion in vitro and binds to pro-
to orchestrate the complex cascade of events that are nec- teins that are implicated in vesicular trafficking, its role
essary to alter the bacterial surface and promote intracel- as a T3SS effector protein remains controversial150,151. As
lular survival137. The best characterized of these is the it has not been universally observed to be translocated
PhoQ sensor, which promotes resistance to antimicro- into eukaryotic cells and it is required for the transloca-
bial peptides18,25. PhoQ contains a novel acidic domain tion of several, if not all, SPI2 T3SS effectors, as well as
that is bridged to the inner membrane by interactions the surface expression of translocon proteins, it is more
with metal ions and binds to and initiates responses to likely that SpiC is part of the SPI2 secretion apparatus
antimicrobial peptides138,139. It also responds to pH by and is not an effector152,153. The most important trans-
structural changes that are determined by regions of the located effectors, by virtue of causing virulence defects
protein separate from the metal ion bridges involved in in mice when mutated, are SifA, SseJ, SseF, SseG, SopD2
antimicrobial peptide sensing136. and PipB2 (Refs 91,154–158) . The observation that
After phagocytosis, salmonellae undergo extensive S. typhimurium that lacks any single SPI2 T3SS effector
bacterial surface remodelling, as has been shown for the protein cannot cause the same virulence attenuation in
lipid A component of lipopolysaccharide (LPS) during mice as a mutant strain that lacks the entire SPI2 T3SS
growth within macrophages137,140. Bacterial molecules suggests that many effectors function cooperatively to
that the host can recognize as indicators of infection, exert their effects on the host cell. Furthermore, the
such as the SPI1 T3SS and flagellin, are repressed and deletion or mutation of many effector genes has no
the LPS structure is altered85,141. Some of the specific sur- virulence phenotype, which implies that their functions
face modifications include: decreasing the length of the might be redundant. Early studies of SPI2 T3SS effectors,
O antigen, which is the repeating carbohydrate polymer which primarily focused on determining their subcel-
of LPS; alterations to the number of acyl chains in the lular localization in mammalian cells, revealed that
structure of the lipid A component of LPS; and changes they might have specific targeting sequences that direct
in the protein content of the outer membrane, the inner localization to endosomal compartments, the Golgi
membrane and the peptidoglycan layer142–145. Synthesis apparatus, the actin cytoskeleton and the microtubule
of enzymes that allow the bacteria to cope with oxida- network33,155–157,159–162. This indicates that components
tive and nitrogenous radicals also occurs146. Microarray of these host-cell structures might be the intracellular
studies have shown that up to 919 S. typhimurium targets of the SPI2 T3SS.
genes are differentially regulated in response to the
phagosomal environment, demonstrating that dra- Salmonella-induced tubular endosomes
matic transcriptional and post-translational changes In epithelial cells and interferon‑γ-primed macrophages,
probably occur when salmonellae make the transition the SPI2 T3SS induces the formation of long filamen-
from a nutrient-rich extracellular environment to the tous membrane structures — Salmonella-induced fila-
intracellular environment147. ments (Sifs) — that originate from the SCV and extend
throughout the cell157,163 (FIG. 4). These structures are
Protein delivery across the vacuolar membrane LAMP1-positive and have a similar membrane composi-
As a result of sensing the phagosomal environment, tion to the SCV118. Sif formation requires microtubules,
the expression and assembly of the SPI2-encoded T3SS but not the actin cytoskeleton, although these filaments
is induced134,148. Although the function of this T3SS in can be decorated with actin159. Possible mechanisms that
pathogenesis remains poorly defined, it has been shown contribute to Sif formation include repetitive initiation
to be essential for virulence in the mouse model of infec- of vesicular budding from the SCV, in which fission
tion22,149. As mutants of the SPI2 T3SS cannot replicate events are incomplete, or continuous fusion of endocytic
in tissue-culture cells and animal models, it is likely vesicles with the SCV, which would result in endosomal
that the role of this T3SS during disease is to promote tubulation or elongation of the SCV. Regardless of the
intracell­ular replication within the intestine during the mechanism of Sif formation, the filaments may func-
acute phase of the infection and in other organs during tion to increase the size of the SCV to accommodate
the chronic state. A reasonable hypothesis for the main bacterial replication during systemic infection. Sif
function of the SPI2 T3SS is that it promotes intracellular formation is dependent on the SPI2 T3SS effector SifA,
replication by altering host vesicular trafficking, so that and to a lesser extent on SseF, SseG, SopD2 and PipB2
useful metabolic molecules, such as amino acids and lip- (Refs 91,164,165). However, it is a dynamic phenotype
ids, are routed to the SCV and the vesicular compartment that may also be modulated by other effectors. SifA was
membrane is expanded. To date, at least 20 Salmonella recently shown to bind to the host cell protein SKIP (SifA
spp. effector proteins are known to be translocated by and kinesin interacting protein)166. The same authors
the SPI2 T3SS across the phagosomal membrane into the also found that, if SKIP was depleted by RNA interfer-
eukaryotic-cell cytoplasm; however, their specific roles ence in S. typhimurium-infected cells, the cells failed to
in promoting intracellular replication or modifying form Sifs, suggesting that Sif formation requires SKIP.
vesicular movement are not yet understood (for a list In addition, Alto and colleagues167 identified SifA as a
of SPI2 T3SS effectors, their functions and binding part- member of the WxxxE (tryptophan (W)-variable (x)-x-
ners, see TABLE 2). In addition, no individual translocated x-glutamate) family of bacterial effectors that function as
effector has definitively been shown to alter vesicular mimics of GTPases. As SifA contains a carboxy-terminal
trafficking. Although it has been reported that SpiC alters Caax (cysteine (C)-aliphatic residue (a)-a-x) motif that

60 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS

is prenylated and S‑acylated by host-cell enzymes, similar membrane, which has led us to propose that its most
to GTPases, it is possible that SifA uses a GTPase-type likely function during S. typhimurium infection is to
mechanism for membrane localization and, perhaps, Sif cleave phospholipids from the SCV membrane (Megha,
formation168. Furthermore, the region of SKIP that inter- M.B.O. and S.I.M., unpublished observations). The
acts with SifA is a pleckstrin homology domain, which cleavage of acyl chains from phospholipids could then
is commonly found in proteins that bind to signalling promote curvature of the membrane or produce discrete
lipids and other regulatory molecules, including those lipid environments that serve as platforms for promot-
that modulate Rho GTPases166. ing vesicle fusion and binding scaffolding proteins and
The roles of PipB2, SopD2, SseF and SseG in Sif for- such activity could modulate Sif formation. However,
mation are not entirely clear. S. typhimurium strains that how these functions, and Sif formation in general, are
lack any of these effectors do not induce Sifs as efficiently important for Salmonella-induced disease is currently
as wild-type S. typhimurium91,165,169. Instead, infection of not understood. It is plausible that Sifs increase the size
cultured cells with these mutants tends to lead to the for- of the SCV in a specific and directional fashion that
mation of ‘ pseudo-Sifs’, which extend from the SCV and promotes bacterial replication and/or redirect nutri-
co-localize with effectors, but do not contain LAMP1. ent-rich organelles to the SCV. In this regard, the SPI2
This suggests that in the absence of these proteins the T3SS could help salmonellae to replicate inside the
ability to form Sifs — which is defined as being entirely phagosome and gain important nutrients for growth,
LAMP1-positive — is impaired and that induction of but yet avoid some of the host-defence mechanisms and
Sifs involves multiple steps that are orchestrated by dif- inflammatory responses that would result from their
ferent effectors. Transient expression of PipB2 in mam- release into the cytoplasm.
malian cells induces the movement of LAMP1-positive
compartments to the cell periphery, which is probably Microtubule motors and movement of the SCV
the result of its interaction with the plus-end-directed As Sif formation requires microtubules — which have
microtubule motor kinesin165,170. This activity might been observed to accumulate and bundle around the
contribute to the outward extension of the SCV, which SCV — there has been an increased interest in under-
would promote Sif formation. SopD2, which has homo­ standing how microtubules and microtubule motors
logy to the SPI1 T3SS translocated effector SopD, has contribute to the intracellular life cycle of salmonellae
also been shown to cooperate with SifA to induce Sifs, and whether targeting vesicular trafficking along micro-
but by an as yet unidentified mechanism33,91. SseF and tubule networks in infected cells is indeed one of their
SseG promote the aggregation of endosomal vesicles pathogenic strategies162 (FIG. 4). SifA has been reported to
and recruit Golgi-derived exocytic vesicles to the SCV, downregulate the recruitment of the plus-end directed
which suggests that salmonellae are able to usurp both microtubule motor kinesin to the SCV, which is medi-
endocytic and exocytic cellular transport processes171–174. ated by PipB2 (Refs 166,170). It does so by interacting
However, it is not known how these activities directly with SKIP, which, if in a complex with SifA, displaces
influence Sif formation. kinesin from the SCV166. This interference is thought to
The deletion of SseJ and SpvB can cause an increase be crucial for maintaining the integrity of the SCV. The
in Sif formation at later time points during the infection inhibitory activity of SifA on kinesin seems to be domi-
of cultured cells, which suggests that these proteins have nant over the kinesin-recruiting activity of PipB2, which
Sif downregulatory functions175. Furthermore, these is suggestive of a potential coordinate or temporal regu-
effectors have virulence defects in the Nramp1-null ani- lation between the two effectors170. In addition, SseF and
mal model, which indicates that their activities contrib- SseG have been found to co-localize with microtubules
ute to systemic infection156,176. Although SpvB has not and cause microtubule bundling in S. typhimurium-
been formally demonstrated to be an SPI2 T3SS effector, infected cells. In particular, SseF has been implicated
some studies suggest that the SPI2 T3SS is required for in recruiting the minus-end directed motor dynein to
its activity in infected cells177,178. SpvB is an actin-specific the SCV162,173. Therefore, it would appear that the SCV
ADP-ribosyltransferase that promotes actin depolym- accumulates dynein, but not kinesin. However, as SifA
erization176. SseJ has homology to glycerophospholipid inhibits the interaction between Rab7 and its effector
cholesterol acyl transferase enzymes, which can remove RILP, a protein that is associated with the dynein motor
acyl chains from phospholipids and transfer them to complex, it is plausible that SifA also prevents dynein
cholesterol in a two-step deacylase-acyltranferase accumulation on the SCV181,182. Although these results
reaction32,179. In fact, purified SseJ has deacylase activ- paint a paradoxical picture of the need to promote or
ity in vitro, which has been shown to be required for inhibit microtubule-motor accumulation on the SCV,
its virulence in mice180. It has been proposed that SseJ salmonellae might employ a carefully choreographed
Prenylated
and SifA have complementary roles in maintaining the combination of both the recruitment and inhibition
The post-translational addition
of lipid chains, such as farnesyl integrity of the SCV membrane, because sifA-mutant of both types of microtubule motors. For example, at
or geranylgeranyl, to cysteine S. typhimurium tends to lose the SCV membrane but specific times during infection, the recruitment of
residues in proteins that does not do so if SseJ is also lacking. This suggests that, dynein could promote minus-end-directed movement
contain a prenylation motif in the absence of SifA, SseJ could cause damage to the of the SCV to stabilize it near the nucleus or the Golgi
called a CaaX box. This
process facilitates membrane
SCV by its enzymatic activity. In addition, our labora- apparatus, where the SCV would have increased access
localization and/or protein– tory has obtained evidence that SseJ has phospholipase to trafficking vesicles that contain nutrients and mem-
protein interactions. activity in vivo that might be localized to the endosomal brane, whereas at other times the recruitment of kinesin

nature reviews | microbiology volume 6 | january 2008 | 61


© 2008 Nature Publishing Group
REVIEWS

to the SCV could induce plus-end-directed movement of VAP, presumably by ADP-ribosylating actin and
towards the cell periphery to promote the enlargement promoting actin depolymerization160,176. SspH2 has also
of the SCV and/or elongation of Sifs. been reported to inhibit the rate of actin polymerization
Recent studies have also shown that the SCV in vitro160. In addition, SspH2 can interact with newly
in infected cultured epithelial cells is localized in a polymerized actin, probably by binding to the actin-
perinuclear position and that effectors from both the binding proteins filamin and profilin, through its amino
SPI1- and the SPI2-encoded T3SSs are involved in this and carboxy termini, respectively. Another effector, SseI
process50,161,166,170,173,181. In particular, SifA, SseF, SseG (also called SrfH), shares a high degree of identity with
and SipA seem to control this phenomenon, because the amino terminus of SspH2 and has been shown to
the SCV containing S. typhimurium strains lacking any bind to polymerized actin, probably through filamin.
of these effectors does not localize near the nucleus. As Although transiently expressed SspH2 and SseI are
the disruption of perinuclear localization of the SCV is associated with VAP during S. typhimurium infection,
correlated with decreased bacterial replication, it has they are not required for the formation of this structure.
been proposed that salmonellae require juxtanuclear Thus, unlike the clearly defined and temporally control-
positioning for optimal growth. In fact, Salcedo and led roles of the SPI1 T3SS effectors in manipulating the
Holden161 have shown that SCVs in close proximity actin cytoskeleton during invasion of epithelial cells, the
to the nucleus contain more bacteria on average than biological significance of the SPI2 T3SS-mediated actin
SCVs further away from the nucleus. However, it recruitment to the SCV, and the interaction of the effec-
has not been directly demonstrated that non-nuclear tors of this T3SS with the actin network, remains to be
localization of the SCV, per se, is inhibitory to bacterial understood. It might be important for translocon stabil-
replication. Rather, it seems more likely that the lack ity and translocation, thereby allowing the full effector
of these effectors or disruption of vesicle trafficking complement to be delivered consistently, and it could
along the microtubule network correlates with defective explain the importance of this process to intracellular
replication and that non-nuclear SCV positioning is a replication183. Subsequently, as T3SS effectors seem to
by-product of this more direct effect. Furthermore, the decrease or remodel actin polymerization, there might
positioning of the SCV might be different in polarized be a period in which actin removal is important, for
epithelial cells, in which movement of the vacuole in a example, to decrease inflammatory responses or allow
directional fashion, such as during transcytosis, could fusion with, or remodelling of, the endosomal mem-
be more physiologically relevant. As discussed above, it brane. Alternatively, actin removal could be essential for
is possible that positioning the SCV in close proximity the effectors to interact with the membrane surface, as
to the Golgi, or other organelles, has some advantages certain protein and phospholipid domains that are cov-
for salmonellae in terms of acquiring nutrients, inhibit- ered in polymerized actin may inhibit their binding to
ing immune responses, prolonging the maturation of important regions within the phagosome membrane.
intestinal epithelial cells and allowing more intracellular
time before epithelial shedding into the intestinal lumen. Future perspectives
All of these effects could then contribute to intestinal The study of the molecular basis of Salmonella spp.
colonization or the intracellular growth of salmonellae. pathogenesis in mammals has advanced greatly over
the past 15 years owing to the identification of many
Actin polymerization around the SCV of the key molecules and mechanisms in mammalian
SPI2 T3SS-dependent actin condensation around the model systems and the discovery of important innate
SCV has been observed, which suggests that this T3SS immune receptors and responses to the bacteria. For
is also involved in cytoskeletal modifications160,183 (FIG. 4). example, important findings have been made in our
In addition, the treatment of S. typhimurium-infected understanding of the mechanisms of entrance into non-
cells with inhibitors of actin polymerization results in phagocytic cells, the way bacteria sense the intra­cellular
decreased bacterial replication, which indicates that the environment and remodel their surfaces, and the inflam-
manipulation of the actin cytoskeleton is important for matory responses to salmonellae. Concurrently, the
bacterial replication183. Although the effectors that are identification of innate immune receptors for bacterial
responsible for the recruitment of actin to the SCV have products and characterization of ligand binding by these
not been identified, several have been shown to interact receptors has progressed greatly. We anticipate a fur-
with actin-binding proteins or to directly manipulate ther explosion of information in the next decade about
actin polymerization. However, it is also possible that the details of the intracellular lifestyle of salmonellae,
SCV-associated actin polymerization is not mediated the function of effectors that are translocated across the
by effectors, but is due to the insertion of the T3SS phagosome membrane and the response of the host to
translocon into the SCV membrane, which has also the activity of these bacterial proteins. Studying how
been observed with the similar Yersinia spp. T3SSs184. salmonellae manipulate endosomal trafficking through
Indeed, most SPI2 T3SS effectors that have been shown to the endocytic pathway should lead to important obser-
interact with components of the actin cytoskeleton have vations about unknown mechanisms of endocytic traf-
actin-inhibitory activities, which suggests that the most ficking. In addition, how effectors function to bring
likely goal of salmonellae is to reduce or reverse vacuole- together mammalian protein components that may
associated actin polymerization (VAP). For example, not normally be together could offer important find-
SpvB has been shown to be involved in the disassembly ings with relevance for non-infectious diseases. The

62 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS

study of salmonellae in more-resistant mouse models, model systems. In addition, the availability of human and
coupled with sophisticated imaging, should also lead to animal genotyping will lead to an increase in data about
a greater understanding of the pathophysiological role of the diversity of human and animal responses to infec-
identified virulence mechanisms and the development tion with various salmonellae. Model systems in various
of chronic disease. This may have relevance for other dis- animals that have diverse innate immune modifications
eases at mucosal surfaces, such as inflammatory bowel will also need to be developed to further understand the
disease. The future decade may also take the field of natural world in the context of our available tools and
salmonellae pathogenesis and host responses beyond to, ultimately, reach the stage in which the evolution of
model systems to the diversity of bacteria and hosts in microorganisms and the emergence of infectious diseases
nature. Emerging technologies, such as whole-genome can be observed in real time. Therefore, the study of sal-
sequencing, will allow us to investigate the diversity monellae pathogenesis should yield a rich treasure trove
of effectors that are used by the many pathogenic of information for those who are interested in microbial
Salmonella species, contributing to our view of host pathogenesis, innate immunity, cell biology and genom-
specificity. Specific hypotheses will be generated by sta- ics, and should remain an important model system of
tistical association, which will then need to be tested in host–pathogen interactions for many years to come.

1. Pegues, D. A., Ohl, M. E. & Miller, S. I. in Principles pretreated with streptomycin. Infect. Immun. 73, 29. Kubori, T. et al. Supramolecular structure of the
and Practice of Infectious Diseases (eds Mandell, 3228–3241 (2005). Salmonella typhimurium type III protein secretion
G. L., Bennet, J. E. & Dolin, R.) 2636–2654 (Churchill 16. Hobbie, S., Chen, L. M., Davis, R. J. & Galan, J. E. system. Science 280, 602–605 (1998).
Livingstone, New York, 2005). Involvement of mitogen-activated protein kinase This study provided the first electron microscopy
2. Garcia-del Portillo, F., Foster, J. W. & Finlay, B. B. Role pathways in the nuclear responses and cytokine images of the T3SS needle complex.
of acid tolerance response genes in Salmonella production induced by Salmonella typhimurium in 30. Kimbrough, T. G. & Miller, S. I. Contribution of
typhimurium virulence. Infect. Immun. 61, 4489–4492 cultured intestinal epithelial cells. J. Immunol. 159, Salmonella typhimurium type III secretion components
(1993). 5550–5559 (1997). to needle complex formation. Proc. Natl Acad. Sci.
3. Michetti, P., Mahan, M. J., Slauch, J. M., Mekalanos, 17. Galan, J. E. & Curtiss, R. 3rd Cloning and molecular USA 97, 11008–11013 (2000).
J. J. & Neutra, M. R. Monoclonal secretory characterization of genes whose products allow 31. Kimbrough, T. G. & Miller, S. I. Assembly of the type III
immunoglobulin A protects mice against oral Salmonella typhimurium to penetrate tissue culture secretion needle complex of Salmonella typhimurium.
challenge with the invasive pathogen Salmonella cells. Proc. Natl Acad. Sci. USA 86, 6383–6387 Microbes Infect. 4, 75–82 (2002).
typhimurium. Infect. Immun. 60, 1786–1792 (1992). (1989). 32. Miao, E. A. & Miller, S. I. A conserved amino acid
4. Selsted, M. E., Miller, S. I., Henschen, A. H. & 18. Miller, S. I., Kukral, A. M. & Mekalanos, J. J. A two- sequence directing intracellular type III secretion by
Ouellette, A. J. Enteric defensins: antibiotic peptide component regulatory system (phoP phoQ) controls Salmonella typhimurium. Proc. Natl Acad. Sci. USA
components of intestinal host defense. J. Cell Biol. Salmonella typhimurium virulence. Proc. Natl Acad. 97, 7539–7544 (2000).
118, 929–936 (1992). Sci. USA 86, 5054–5058 (1989). 33. Brumell, J. H. et al. SopD2 is a novel type III secreted
5. Prouty, A. M., Brodsky, I. E., Falkow, S. & Gunn, J. S. This reference identified the PhoP/PhoQ two- effector of Salmonella typhimurium that targets late
Bile‑salt‑mediated induction of antimicrobial and bile component system as being important for the endocytic compartments upon delivery into host cells.
resistance in Salmonella typhimurium. Microbiology regulation of virulence. Traffic 4, 36–48 (2003).
150, 775–783 (2004). 19. Alpuche-Aranda, C. M., Swanson, J. A., Loomis, W. P. 34. Lee, S. H. & Galan, J. E. Salmonella type III secretion-
6. Francis, C. L., Starnbach, M. N. & Falkow, S. & Miller, S. I. Salmonella typhimurium activates associated chaperones confer secretion-pathway
Morphological and cytoskeletal changes in epithelial virulence gene transcription within acidified specificity. Mol. Microbiol. 51, 483–495 (2004).
cells occur immediately upon interaction with macrophage phagosomes. Proc. Natl Acad. Sci. USA 35. Karavolos, M. H. et al. Type III secretion of the
Salmonella typhimurium grown under low-oxygen 89, 10079–10083 (1992). Salmonella effector protein SopE is mediated via an
conditions. Mol. Microbiol. 6, 3077–3087 (1992). 20. Alpuche-Aranda, C. M., Racoosin, E. L., Swanson, J. A. N‑terminal amino acid signal and not an mRNA
7. Takeuchi, A. Electron microscope studies of & Miller, S. I. Salmonella stimulate macrophage sequence. J. Bacteriol. 187, 1559–1567
experimental Salmonella infection. I. Penetration into macropinocytosis and persist within spacious (2005).
the intestinal epithelium by Salmonella typhimurium. phagosomes. J. Exp. Med. 179, 601–608 (1994). 36. Fu, Y. & Galan, J. E. Identification of a specific
Am. J. Pathol. 50, 109–136 (1967). 21. Ochman, H., Soncini, F. C., Solomon, F. & Groisman, chaperone for SptP, a substrate of the centisome 63
This reference showed membrane ruffling that is E. A. Identification of a pathogenicity island required type III secretion system of Salmonella typhimurium.
induced by Salmonella spp. in intestinal cells by for Salmonella survival in host cells. Proc. Natl Acad. J. Bacteriol. 180, 3393–3399 (1998).
microscopy for the first time. Sci. USA 93, 7800–7804 (1996). 37. Hong, K. H. & Miller, V. L. Identification of a novel
8. Baumler, A. J., Tsolis, R. M. & Heffron, F. Contribution References 21 and 22 were the first to show that Salmonella invasion locus homologous to Shigella
of fimbrial operons to attachment to and invasion of the T3SS that is encoded on SPI2 is important for ipgDE. J. Bacteriol. 180, 1793–1802 (1998).
epithelial cell lines by Salmonella typhimurium. Infect. virulence. 38. Bronstein, P. A., Miao, E. A. & Miller, S. I. InvB is a
Immun. 64, 1862–1865 (1996). 22. Shea, J. E., Hensel, M., Gleeson, C. & Holden, D. W. type III secretion chaperone specific for SspA.
9. Kohbata, S., Yokoyama, H. & Yabuuchi, E. Identification of a virulence locus encoding a second J. Bacteriol. 182, 6638–6644 (2000).
Cytopathogenic effect of Salmonella typhi GIFU 10007 type III secretion system in Salmonella typhimurium. 39. Tucker, S. C. & Galan, J. E. Complex function for SicA,
on M cells of murine ileal Peyer’s patches in ligated Proc. Natl Acad. Sci. USA 93, 2593–2597 (1996). a Salmonella enterica serovar Typhimurium type III
ileal loops: an ultrastructural study. Microbiol. 23. Vazquez-Torres, A. et al. Salmonella pathogenicity secretion-associated chaperone. J. Bacteriol. 182,
Immunol. 30, 1225–1237 (1986). island 2‑dependent evasion of the phagocyte NADPH 2262–2268 (2000).
10. Jones, B. D., Ghori, N. & Falkow, S. Salmonella oxidase. Science 287, 1655–1658 (2000). 40. Ehrbar, K., Friebel, A., Miller, S. I. & Hardt, W. D.
typhimurium initiates murine infection by penetrating 24. Miao, E. A., Freeman, J. A. & Miller, S. I. Transcription Role of the Salmonella pathogenicity island 1
and destroying the specialized epithelial M cells of the of the SsrAB regulon is repressed by alkaline pH and (SPI‑1) protein InvB in type III secretion of SopE and
Peyer’s patches. J. Exp. Med. 180, 15–23 (1994). is independent of PhoPQ and magnesium SopE2, two Salmonella effector proteins encoded
11. Vazquez-Torres, A. et al. Extraintestinal dissemination concentration. J. Bacteriol. 184, 1493–1497 outside of SPI‑1. J. Bacteriol. 185, 6950–6967
of Salmonella by CD18-expressing phagocytes. Nature (2002). (2003).
401, 804–808 (1999). 25. Bader, M. W. et al. Regulation of Salmonella 41. Higashide, W. & Zhou, D. The first 45 amino acids of
12. Jepson, M. A., Collares-Buzato, C. B., Clark, M. A., typhimurium virulence gene expression by cationic SopA are necessary for InvB binding and SPI‑1
Hirst, B. H. & Simmons, N. L. Rapid disruption of antimicrobial peptides. Mol. Microbiol. 50, 219–230 secretion. J. Bacteriol. 188, 2411–2420 (2006).
epithelial barrier function by Salmonella typhimurium (2003). 42. Akeda, Y. & Galan, J. E. Chaperone release and
is associated with structural modification of 26. McCormick, B. A., Miller, S. I., Carnes, D. & Madara, unfolding of substrates in type III secretion. Nature
intercellular junctions. Infect. Immun. 63, 356–359 J. L. Transepithelial signaling to neutrophils by 437, 911–915 (2005).
(1995). salmonellae: a novel virulence mechanism for 43. Brown, N. F. et al. Salmonella pathogenicity island 2 is
13. Zhang, S. et al. The Salmonella enterica serotype gastroenteritis. Infect. Immun. 63, 2302–2309 expressed prior to penetrating the intestine. PLoS
Typhimurium effector proteins SipA, SopA, SopB, (1995). Pathog. 1, e32 (2005).
SopD, and SopE2 act in concert to induce diarrhea in 27. McCormick, B. A. et al. Surface attachment of 44. Hensel, M. et al. Functional analysis of ssaJ and the
calves. Infect. Immun. 70, 3843–3855 (2002). Salmonella typhimurium to intestinal epithelia ssaK/U operon, 13 genes encoding components of the
14. Raffatellu, M. et al. SipA, SopA, SopB, SopD, and imprints the subepithelial matrix with gradients type III secretion apparatus of Salmonella pathogenicity
SopE2 contribute to Salmonella enterica serotype chemotactic for neutrophils. J. Cell Biol. 131, island 2. Mol. Microbiol. 24, 155–167 (1997).
Typhimurium invasion of epithelial cells. Infect. Immun. 1599–1608 (1995). 45. Deiwick, J. et al. Mutations in Salmonella pathogenicity
73, 146–154 (2005). 28. Hansen-Wester, I. & Hensel, M. Salmonella island 2 (SPI2) genes affecting transcription of SPI1
15. Stecher, B. et al. Comparison of Salmonella enterica pathogenicity islands encoding type III secretion genes and resistance to antimicrobial agents.
serovar Typhimurium colitis in germfree mice and mice systems. Microbes Infect. 3, 549–559 (2001). J. Bacteriol. 180, 4775–4780 (1998).

nature reviews | microbiology volume 6 | january 2008 | 63


© 2008 Nature Publishing Group
REVIEWS

46. Steele-Mortimer, O. et al. The invasion-associated 67. Stender, S. et al. Identification of SopE2 from 89. Wood, M. W. et al. The secreted effector protein of
type III secretion system of Salmonella enterica Salmonella typhimurium, a conserved guanine Salmonella dublin, SopA, is translocated into
serovar Typhimurium is necessary for intracellular nucleotide exchange factor for Cdc42 of the host cell. eukaryotic cells and influences the induction of
proliferation and vacuole biogenesis in epithelial cells. Mol. Microbiol. 36, 1206–1221 (2000). enteritis. Cell Microbiol. 2, 293–303 (2000).
Cell. Microbiol. 4, 43–54 (2002). 68. Friebel, A. et al. SopE and SopE2 from Salmonella 90. Zhang, Y., Higashide, W. M., McCormick, B. A., Chen, J.
47. Hernandez, L. D., Hueffer, K., Wenk, M. R. & Galan, typhimurium activate different sets of RhoGTPases of & Zhou, D. The inflammation-associated Salmonella
J. E. Salmonella modulates vesicular traffic by the host cell. J. Biol. Chem. 276, 34035–34040 SopA is a HECT-like E3 ubiquitin ligase. Mol. Microbiol.
altering phosphoinositide metabolism. Science 304, (2001). 62, 786–793 (2006).
1805–1807 (2004). 69. Zhou, D., Chen, L. M., Hernandez, L., Shears, S. B. & 91. Jiang, X. et al. The related effector proteins SopD and
48. Drecktrah, D., Knodler, L. A., Galbraith, K. & Steele- Galan, J. E. A Salmonella inositol polyphosphatase SopD2 from Salmonella enterica serovar Typhimurium
Mortimer, O. The Salmonella SPI1 effector SopB acts in conjunction with other bacterial effectors to contribute to virulence during systemic infection of
stimulates nitric oxide production long after invasion. promote host cell actin cytoskeleton rearrangements mice. Mol. Microbiol. 54, 1186–1198 (2004).
Cell. Microbiol. 7, 105–113 (2005). and bacterial internalization. Mol. Microbiol. 39, 92. Fu, Y. & Galan, J. E. A Salmonella protein
49. Lawley, T. D. et al. Genome-wide screen for Salmonella 248–259 (2001). antagonizes Rac‑1 and Cdc42 to mediate host-cell
genes required for long-term systemic infection of the 70. Patel, J. C. & Galan, J. E. Differential activation and recovery after bacterial invasion. Nature 401,
mouse. PLoS Pathog. 2, e11 (2006). function of Rho GTPases during Salmonella-host cell 293–297 (1999).
50. Brawn, L. C., Hayward, R. D. & Koronakis, V. interactions. J. Cell Biol. 175, 453–463 (2006). References 92 and 93 identified the
Salmonella SPI1 effector SipA persists after entry and 71. Ellerbroek, S. M. et al. SGEF, a RhoG guanine GTPase-antagonizing activity of SptP and showed
cooperates with a SPI2 effector to regulate nucleotide exchange factor that stimulates how the activities of SopE and SptP are temporally
phagosome maturation and intracellular replication. macropinocytosis. Mol. Biol. Cell 15, 3309–3319 regulated by proteasome-dependent degradation.
Cell Host Microbe 1, 63–75 (2007). (2004). 93. Kubori, T. & Galan, J. E. Temporal regulation of
51. Giacomodonato, M. N. et al. SipA, SopA, SopB, SopD 72. Criss, A. K. & Casanova, J. E. Coordinate regulation of Salmonella virulence effector function by proteasome-
and SopE2 effector proteins of Salmonella enterica Salmonella enterica serovar Typhimurium invasion of dependent protein degradation. Cell 115, 333–342
serovar Typhimurium are synthesized at late stages of epithelial cells by the Arp2/3 complex and Rho (2003).
infection in mice. Microbiology 153, 1221–1228 GTPases. Infect. Immun. 71, 2885–2891 (2003). 94. Murli, S., Watson, R. O. & Galan, J. E. Role of tyrosine
(2007). 73. Unsworth, K. E., Way, M., McNiven, M., Machesky, L. kinases and the tyrosine phosphatase SptP in the
52. Li, J. et al. Relationship between evolutionary rate and & Holden, D. W. Analysis of the mechanisms of interaction of Salmonella with host cells. Cell.
cellular location among the Inv/Spa invasion proteins Salmonella-induced actin assembly during invasion of Microbiol. 3, 795–810 (2001).
of Salmonella enterica. Proc. Natl Acad. Sci. USA 92, host cells and intracellular replication. Cell. Microbiol. 95. Haraga, A. & Miller, S. I. A Salmonella enterica
7252–7256 (1995). 6, 1041–1055 (2004). serovar Typhimurium translocated leucine-rich repeat
53. Ochman, H. & Groisman, E. A. Distribution of 74. Shi, J., Scita, G. & Casanova, J. E. WAVE2 signaling effector protein inhibits NF‑kappa B‑dependent gene
pathogenicity islands in Salmonella spp. Infect. mediates invasion of polarized epithelial cells by expression. Infect. Immun. 71, 4052–4058 (2003).
Immun. 64, 5410–5412 (1996). Salmonella typhimurium. J. Biol. Chem. 280, 96. Miao, E. A. et al. Salmonella typhimurium leucine-
54. Boyd, E. F., Li, J., Ochman, H. & Selander, R. K. 29849–29855 (2005). rich repeat proteins are targeted to the SPI1 and
Comparative genetics of the inv-spa invasion gene 75. Hayward, R. D. & Koronakis, V. Direct nucleation and SPI2 type III secretion systems. Mol. Microbiol. 34,
complex of Salmonella enterica. J. Bacteriol. 179, bundling of actin by the SipC protein of invasive 850–864 (1999).
1985–1991 (1997). Salmonella. EMBO J. 18, 4926–4934 (1999). 97. Rohde, J. R., Breitkreutz, A., Chenal, A., Sansonetti,
55. Hensel, M. et al. Analysis of the boundaries of References 75 and 76 detected direct P. J. & Parsot, C. Type III secretion effectors of the
Salmonella pathogenicity island 2 and the manipulation of actin by the SPI1 T3SS effectors IpaH family are E3 ubiquitin ligases. Cell Host Microbe
corresponding chromosomal region of Escherichia coli SipA and SipC. 1, 77–83 (2007).
K‑12. J. Bacteriol. 179, 1105–1111 (1997). 76. Zhou, D., Mooseker, M. S. & Galan, J. E. Role of the 98. Haraga, A. & Miller, S. I. A Salmonella type III
56. Hensel, M., Nikolaus, T. & Egelseer, C. Molecular and S. typhimurium actin-binding protein SipA in bacterial secretion effector interacts with the mammalian
functional analysis indicates a mosaic structure of internalization. Science 283, 2092–2095 (1999). serine/threonine protein kinase PKN1. Cell. Microbiol.
Salmonella pathogenicity island 2. Mol. Microbiol. 31, 77. Scherer, C. A., Cooper, E. & Miller, S. I. The Salmonella 8, 837–846 (2006).
489–498 (1999). type III secretion translocon protein SspC is inserted 99. Collier-Hyams, L. S. et al. Cutting edge: Salmonella
57. Porwollik, S., Wong, R. M. & McClelland, M. into the epithelial cell plasma membrane upon AvrA effector inhibits the key proinflammatory, anti-
Evolutionary genomics of Salmonella: gene infection. Mol. Microbiol. 37, 1133–1145 (2000). apoptotic NF‑kappaB pathway. J. Immunol. 169,
acquisitions revealed by microarray analysis. 78. McGhie, E. J., Hayward, R. D. & Koronakis, V. 2846–2850 (2002).
Proc. Natl Acad. Sci. USA 99, 8956–8961 (2002). Cooperation between actin-binding proteins of 100. Mittal, R., Peak-Chew, S. Y. & McMahon, H. T.
58. Hormaeche, C. E. Natural resistance to Salmonella invasive Salmonella: SipA potentiates SipC nucleation Acetylation of MEK2 and I kappa B kinase (IKK)
typhimurium in different inbred mouse strains. and bundling of actin. EMBO J. 20, 2131–2139 activation loop residues by YopJ inhibits signaling.
Immunology 37, 311–318 (1979). (2001). Proc. Natl Acad. Sci. USA 103, 18574–18579
59. Vidal, S. M., Malo, D., Vogan, K., Skamene, E. & 79. Higashide, W., Dai, S., Hombs, V. P. & Zhou, D. (2006).
Gros, P. Natural resistance to infection with Involvement of SipA in modulating actin dynamics 101. Mukherjee, S. et al. Yersinia YopJ acetylates and
intracellular parasites: isolation of a candidate for Bcg. during Salmonella invasion into cultured epithelial inhibits kinase activation by blocking phosphorylation.
Cell 73, 469–485 (1993). cells. Cell. Microbiol. 4, 357–365 (2002). Science 312, 1211–1214 (2006).
60. Hensel, M. et al. Genes encoding putative effector 80. Chen, L. M., Hobbie, S. & Galan, J. E. Requirement 102. Buchwald, D. S. & Blaser, M. J. A review of human
proteins of the type III secretion system of of CDC42 for Salmonella-induced cytoskeletal and salmonellosis: II. Duration of excretion following
Salmonella pathogenicity island 2 are required for nuclear responses. Science 274, 2115–2118 infection with nontyphi Salmonella. Rev. Infect. Dis. 6,
bacterial virulence and proliferation in macrophages. (1996). 345–356 (1984).
Mol. Microbiol. 30, 163–174 (1998). 81. Boyle, E. C., Brown, N. F. & Finlay, B. B. Salmonella 103. Richter-Dahlfors, A., Buchan, A. M. & Finlay, B. B.
61. Beuzon, C. R. & Holden, D. W. Use of mixed enterica serovar Typhimurium effectors SopB, SopE, Murine salmonellosis studied by confocal microscopy:
infections with Salmonella strains to study virulence SopE2 and SipA disrupt tight junction structure and Salmonella typhimurium resides intracellularly inside
genes and their interactions in vivo. Microbes Infect. function. Cell. Microbiol. 8, 1946–1957 (2006). macrophages and exerts a cytotoxic effect on
3, 1345–1352 (2001). 82. Norris, F. A., Wilson, M. P., Wallis, T. S., Galyov, E. E. & phagocytes in vivo. J. Exp. Med. 186, 569–580
62. Monack, D. M., Bouley, D. M. & Falkow, S. Salmonella Majerus, P. W. SopB, a protein required for virulence (1997).
typhimurium persists within macrophages in the of Salmonella dublin, is an inositol phosphate 104. Fields, P. I., Swanson, R. V., Haidaris, C. G. & Heffron, F.
mesenteric lymph nodes of chronically infected phosphatase. Proc. Natl Acad. Sci. USA 95, Mutants of Salmonella typhimurium that cannot survive
Nramp1+/+ mice and can be reactivated by IFNgamma 14057–14059 (1998). within the macrophage are avirulent. Proc. Natl Acad.
neutralization. J. Exp. Med. 199, 231–241 (2004). 83. Hersh, D. et al. The Salmonella invasin SipB induces Sci. USA 83, 5189–5193 (1986).
63. Behlau, I. & Miller, S. I. A PhoP-repressed gene macrophage apoptosis by binding to caspase‑1. This important study demonstrated that survival
promotes Salmonella typhimurium invasion of Proc. Natl. Acad. Sci. USA 96, 2396–2401 (1999). within macrophages is important for S. typhimurium
epithelial cells. J. Bacteriol. 175, 4475–4484 (1993). 84. Franchi, L. et al. Cytosolic flagellin requires Ipaf for virulence.
64. Barthel, M. et al. Pretreatment of mice with activation of caspase‑1 and interleukin 1b in 105. O’Callaghan, D., Maskell, D., Liew, F. Y., Easmon, C. S.
streptomycin provides a Salmonella enterica serovar Salmonella-infected macrophages. Nature Immunol. 7, & Dougan, G. Characterization of aromatic- and
Typhimurium colitis model that allows analysis of both 576–582 (2006). purine-dependent Salmonella typhimurium: attention,
pathogen and host. Infect. Immun. 71, 2839–2858 85. Miao, E. A. et al. Cytoplasmic flagellin activates persistence, and ability to induce protective immunity
(2003). caspase‑1 and secretion of interleukin 1b via Ipaf. in BALB/c mice. Infect. Immun. 56, 419–423 (1988).
65. Hardt, W. D., Chen, L. M., Schuebel, K. E., Bustelo, Nature Immunol. 7, 569–575 (2006). 106. Dukes, J. D. et al. The secreted Salmonella dublin
X. R. & Galan, J. E. S. typhimurium encodes an 86. Lara-Tejero, M. et al. Role of the caspase‑1 phosphoinositide phosphatase, SopB, localizes to
activator of Rho GTPases that induces membrane inflammasome in Salmonella typhimurium PtdIns(3)P-containing endosomes and perturbs
ruffling and nuclear responses in host cells. Cell 93, pathogenesis. J. Exp. Med. 203, 1407–1412 (2006). normal endosome to lysosome trafficking. Biochem. J.
815–826 (1998). 87. Raupach, B., Peuschel, S. K., Monack, D. M. & 395, 239–247 (2006).
This work elegantly demonstrated the ability of Zychlinsky, A. Caspase‑1‑mediated activation of 107. Carrol, M. E., Jackett, P. S., Aber, V. R. & Lowrie,
SopE to activate GTPases and subsequent actin interleukin-1b (IL-1b) and IL‑18 contributes to innate D. B. Phagolysosome formation, cyclic adenosine
rearrangements and transcriptional responses. immune defenses against Salmonella enterica serovar 3′:5′-monophosphate and the fate of Salmonella
66. Bakshi, C. S. et al. Identification of SopE2, a Typhimurium infection. Infect. Immun. 74, 4922–4926 typhimurium within mouse peritoneal macrophages.
Salmonella secreted protein which is highly (2006). J. Gen. Microbiol. 110, 421–429 (1979).
homologous to SopE and involved in bacterial invasion 88. Jones, M. A. et al. Secreted effector proteins of 108. Buchmeier, N. A. & Heffron, F. Inhibition of macrophage
of epithelial cells. J. Bacteriol. 182, 2341–2344 Salmonella dublin act in concert to induce enteritis. phagosome–lysosome fusion by Salmonella
(2000). Infect. Immun. 66, 5799–5804 (1998). typhimurium. Infect. Immun. 59, 2232–2238 (1991).

64 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group
REVIEWS

109. Oh, Y. K. et al. Rapid and complete fusion of survival in mice, antioxidant defence and killing of 152. Freeman, J. A., Rappl, C., Kuhle, V., Hensel, M. &
macrophage lysosomes with phagosomes containing macrophages: role of cyclic diGMP. Mol. Microbiol. Miller, S. I. SpiC is required for translocation of
Salmonella typhimurium. Infect. Immun. 64, 56, 1234–1245 (2005). Salmonella pathogenicity island 2 effectors and
3877–3883 (1996). 130. Humphreys, S., Stevenson, A., Bacon, A., Weinhardt, secretion of translocon proteins SseB and SseC.
110. Drecktrah, D., Knodler, L. A., Howe, D. & Steele- A. B. & Roberts, M. The alternative sigma factor, J. Bacteriol. 184, 4971–4980 (2002).
Mortimer, O. Salmonella trafficking is defined by sigmaE, is critically important for the virulence of 153. Yu, X. J. et al. SpiC is required for secretion of
continuous dynamic interactions with the Salmonella typhimurium. Infect. Immun. 67, Salmonella pathogenicity island 2 type III secretion
endolysosomal system. Traffic 8, 212–225 (2007). 1560–1568 (1999). system proteins. Cell. Microbiol. 4, 531–540 (2002).
111. Rathman, M., Sjaastad, M. D. & Falkow, S. 131. Rosenberger, C. M., Gallo, R. L. & Finlay, B. B. 154. Beuzon, C. R. et al. Salmonella maintains the
Acidification of phagosomes containing Salmonella Interplay between antibacterial effectors: a integrity of its intracellular vacuole through the
typhimurium in murine macrophages. Infect. Immun. macrophage antimicrobial peptide impairs action of SifA. EMBO J. 19, 3235–3249 (2000).
64, 2765–2773 (1996). intracellular Salmonella replication. Proc. Natl Acad. 155. Ruiz-Albert, J. et al. Complementary activities of SseJ
112. Martin-Orozco, N. et al. Visualization of vacuolar Sci. USA 101, 2422–2427 (2004). and SifA regulate dynamics of the Salmonella
acidification-induced transcription of genes of 132. Curtiss, R. & Kelly, S. M. Salmonella typhimurium typhimurium vacuolar membrane. Mol. Microbiol. 44,
pathogens inside macrophages. Mol. Biol. Cell 17, deletion mutants lacking adenylate cyclase and cyclic 645–661 (2002).
498–510 (2006). AMP receptor protein are avirulent and immunogenic. 156. Freeman, J. A., Ohl, M. E. & Miller, S. I. The
113. Steele-Mortimer, O., Meresse, S., Gorvel, J. P., Toh, Infect. Immun. 55, 3035–3043 (1987). Salmonella enterica serovar Typhimurium translocated
B. H. & Finlay, B. B. Biogenesis of Salmonella 133. Fang, F. C. et al. The alternative sigma factor katF effectors SseJ and SifsB are targeted to the
typhimurium-containing vacuoles in epithelial cells (rpoS) regulates Salmonella virulence. Proc. Natl Acad. Salmonella-containing vacuole. Infect. Immun. 71,
involves interactions with the early endocytic pathway. Sci. USA 89, 11978–11982 (1992). 418–427 (2003).
Cell. Microbiol. 1, 33–49 (1999). 134. Lee, A. K., Detweiler, C. S. & Falkow, S. OmpR 157. Knodler, L. A. et al. Salmonella type III effectors PipB
114. Smith, A. C., Cirulis, J. T., Casanova, J. E., Scidmore, regulates the two-component system SsrA-SsrB in and PipB2 are targeted to detergent-resistant
M. A. & Brumell, J. H. Interaction of the Salmonella- Salmonella pathogenicity island 2. J. Bacteriol. 182, microdomains on internal host cell membranes. Mol.
containing vacuole with the endocytic recycling 771–781 (2000). Microbiol. 49, 685–704 (2003).
system. J. Biol. Chem. 280, 24634–24641 (2005). 135. Testerman, T. L. et al. The alternative sigma factor sE 158. Deiwick, J. et al. The translocated Salmonella effector
115. Garcia-del Portillo, F. & Finlay, B. B. Targeting of controls antioxidant defences required for Salmonella proteins SseF and SseG interact and are required to
Salmonella typhimurium to vesicles containing virulence and stationary-phase survival. Mol. establish an intracellular replication niche. Infect.
lysosomal membrane glycoproteins bypasses Microbiol. 43, 771–782 (2002). Immun. 74, 6965–6972 (2006).
compartments with mannose 6‑phosphate receptors. 136. Prost, L. R. et al. Activation of the bacterial sensor 159. Brumell, J. H., Goosney, D. L. & Finlay, B. B. SifA, a
J. Cell Biol. 129, 81–97 (1995). kinase PhoQ by acidic pH. Mol. Cell 26, 165–174 type III secreted effector of Salmonella typhimurium,
116. Meresse, S., Steele-Mortimer, O., Finlay, B. B. & (2007). directs Salmonella-induced filament (Sif) formation
Gorvel, J. P. The rab7 GTPase controls the maturation References 136 and 138 show that PhoQ directly along microtubules. Traffic 3, 407–415 (2002).
of Salmonella typhimurium-containing vacuoles in senses antimicrobial peptides and low pH. 160. Miao, E. A. et al. Salmonella effectors translocated
HeLa cells. EMBO J. 18, 4394–4403 (1999). 137. Heithoff, D. M. et al. Coordinate intracellular across the vacuolar membrane interact with the
117. Hashim, S., Mukherjee, K., Raje, M., Basu, S. K. & expression of Salmonella genes induced during actin cytoskeleton. Mol. Microbiol. 48, 401–415
Mukhopadhyay, A. Live Salmonella modulate infection. J. Bacteriol. 181, 799–807 (1999). (2003).
expression of Rab proteins to persist in a specialized 138. Bader, M. W. et al. Recognition of antimicrobial 161. Salcedo, S. P. & Holden, D. W. SseG, a virulence
compartment and escape transport to lysosomes. peptides by a bacterial sensor kinase. Cell 122, protein that targets Salmonella to the Golgi network.
J. Biol. Chem. 275, 16281–16288 (2000). 461–472 (2005). EMBO J. 22, 5003–5014 (2003).
118. Brumell, J. H., Tang, P., Mills, S. D. & Finlay, B. B. 139. Cho, U. S. et al. Metal bridges between the PhoQ 162. Kuhle, V., Jackel, D. & Hensel, M. Effector proteins
Characterization of Salmonella-induced filaments (Sifs) sensor domain and the membrane regulate encoded by Salmonella pathogenicity island 2
reveals a delayed interaction between Salmonella- transmembrane signaling. J. Mol. Biol. 356, interfere with the microtubule cytoskeleton after
containing vacuoles and late endocytic compartments. 1193–1206 (2006). translocation into host cells. Traffic 5, 356–370
Traffic 2, 643–653 (2001). 140. Gibbons, H. S., Kalb, S. R., Cotter, R. J. & Raetz, C. R. (2004).
119. Garvis, S. G., Beuzon, C. R. & Holden, D. W. A role for Role of Mg2+ and pH in the modification of Salmonella 163. Garcia-del Portillo, F., Zwick, M. B., Leung, K. Y. &
the PhoP/Q regulon in inhibition of fusion between lipid A after endocytosis by macrophage tumour cells. Finlay, B. B. Salmonella induces the formation of
lysosomes and Salmonella-containing vacuoles in Mol. Microbiol. 55, 425–440 (2005). filamentous structures containing lysosomal
macrophages. Cell. Microbiol. 3, 731–744 (2001). 141. Ernst, R. K., Guina, T. & Miller, S. I. Salmonella membrane glycoproteins in epithelial cells.
120. Cuellar-Mata, P. et al. Nramp1 modifies the fusion of typhimurium outer membrane remodeling: role in Proc. Natl Acad. Sci. USA 90, 10544–10548 (1993).
Salmonella typhimurium-containing vacuoles with resistance to host innate immunity. Microbes Infect. 3, References 163 and 164 were the first to show
cellular endomembranes in macrophages. J. Biol. 1327–1334 (2001). that S. typhimurium produces Sifs in infected
Chem. 277, 2258–2265 (2002). 142. Guo, L. et al. Regulation of lipid A modifications by cultured cells and that the effector that is
121. Catron, D. M. et al. The Salmonella-containing vacuole Salmonella typhimurium virulence genes phoP-phoQ. responsible for this activity is SifA.
is a major site of intracellular cholesterol accumulation Science 276, 250–253 (1997). 164. Stein, M. A., Leung, K. Y., Zwick, M., Garcia-del
and recruits the GPI-anchored protein CD55. Cell. This study demonstrates that PhoP/PhoQ regulates Portillo, F. & Finlay, B. B. Identification of a Salmonella
Microbiol. 4, 315–328 (2002). the remodelling of the LPS structure in a way that virulence gene required for formation of filamentous
122. Fields, P. I., Groisman, E. A. & Heffron, F. A Salmonella makes it less immunostimulatory by TLR4 signalling. structures containing lysosomal membrane
locus that controls resistance to microbicidal proteins 143. Gunn, J. S. et al. PmrA‑PmrB‑regulated genes glycoproteins within epithelial cells. Mol. Microbiol.
from phagocytic cells. Science 243, 1059–1062 necessary for 4‑aminoarabinose lipid A modification 20, 151–164 (1996).
(1989). and polymyxin resistance. Mol. Microbiol. 27, 165. Knodler, L. A. & Steele-Mortimer, O. The Salmonella
123. Miller, S. I. & Mekalanos, J. J. Constitutive expression 1171–1182 (1998). effector PipB2 affects late endosome/lysosome
of the PhoP regulon attenuates Salmonella virulence 144. Guo, L. et al. Lipid A acylation and bacterial resistance distribution to mediate Sif extension. Mol. Biol. Cell.
and survival within macrophages. J. Bacteriol. 172, against vertebrate antimicrobial peptides. Cell 95, 16, 4108–4123 (2005).
2485–2490 (1990). 189–198 (1998). 166. Boucrot, E., Henry, T., Borg, J. P., Gorvel, J. P. &
124. Nickerson, C. A. & Curtiss, R. Role of sigma factor 145. Hilbert, F., Garcia-del Portillo, F. & Groisman, E. A. Meresse, S. The intracellular fate of Salmonella
RpoS in initial stages of Salmonella typhimurium A periplasmic d‑alanyl‑d-alanine dipeptidase in the depends on the recruitment of kinesin. Science 308,
infection. Infect. Immun. 65, 1814–1823 (1997). Gram-negative bacterium Salmonella enterica. 1174–1178 (2005).
125. Bearson, B. L., Wilson, L. & Foster, J. W. A low pH- J. Bacteriol. 181, 2158–2165 (1999). 167. Alto, N. M. et al. Identification of a bacterial type III
inducible, PhoPQ-dependent acid tolerance response 146. Fang, F. C. et al. Virulent Salmonella typhimurium has effector family with G protein mimicry functions. Cell
protects Salmonella typhimurium against inorganic two periplasmic Cu, Zn-superoxide dismutases. Proc. 124, 133–145 (2006).
acid stress. J. Bacteriol. 180, 2409–2417 (1998). Natl Acad. Sci. USA 96, 7502–7507 (1999). 168. Reinicke, A. T. et al. A Salmonella typhimurium
126. Shiloh, M. U. et al. Phenotype of mice and 147. Eriksson, S., Lucchini, S., Thompson, A., Rhen, M. & effector protein SifA is modified by host cell
macrophages deficient in both phagocyte oxidase and Hinton, J. C. Unravelling the biology of macrophage prenylation and S‑acylation machinery. J. Biol. Chem.
inducible nitric oxide synthase. Immunity 10, 29–38 infection by gene expression profiling of intracellular 280, 14620–14627 (2005).
(1999). Salmonella enterica. Mol. Microbiol. 47, 103–118 169. Guy, R. L., Gonias, L. A. & Stein, M. A. Aggregation of
127. Gunn, J. S., Ryan, S. S., Van Velkinburgh, J. C., (2003). host endosomes by Salmonella requires SPI2
Ernst, R. K. & Miller, S. I. Genetic and functional 148. Cirillo, D. M., Valdivia, R. H., Monack, D. M. & Falkow, translocation of SseFG and involves SpvR and the fms-
analysis of a PmrA–PmrB‑regulated locus necessary S. Macrophage-dependent induction of the Salmonella aroE intragenic region. Mol. Microbiol. 37,
for lipopolysaccharide modification, antimicrobial pathogenicity island 2 type III secretion system and its 1417–1435 (2000).
peptide resistance, and oral virulence of Salmonella role in intracellular survival. Mol. Microbiol. 30, 170. Henry, T. et al. The Salmonella effector protein PipB2
enterica serovar Typhimurium. Infect. Immun. 68, 175–188 (1998). is a linker for kinesin‑1. Proc. Natl Acad. Sci. USA
6139–6146 (2000). 149. Hensel, M. et al. Simultaneous identification of 103, 13497–13502 (2006).
128. Vazquez-Torres, A., Jones-Carson, J., Mastroeni, P., bacterial virulence genes by negative selection. 171. Hansen-Wester, I., Stecher, B. & Hensel, M. Type III
Ischiropoulos, H. & Fang, F. C. Antimicrobial actions of Science 269, 400–403 (1995). secretion of Salmonella enterica serovar Typhimurium
the NADPH phagocyte oxidase and inducible nitric 150. Uchiya, K. et al. A Salmonella virulence protein that translocated effectors and SseFG. Infect. Immun. 70,
oxide synthase in experimental salmonellosis. I. inhibits cellular trafficking. EMBO J. 18, 3924–3933 1403–1409 (2002).
Effects on microbial killing by activated peritoneal (1999). 172. Kuhle, V. & Hensel, M. SseF and SseG are
macrophages in vitro. J. Exp. Med. 192, 227–236 151. Shotland, Y., Kramer, H. & Groisman, E. A. The translocated effectors of the type III secretion system
(2000). Salmonella SpiC protein targets the mammalian of Salmonella pathogenicity island 2 that modulate
129. Hisert, K. B. et al. A glutamate‑alanine‑leucine (EAL) Hook3 protein function to alter cellular trafficking. aggregation of endosomal compartments. Cell.
domain protein of Salmonella controls bacterial Mol. Microbiol. 49, 1565–1576 (2003). Microbiol. 4, 813–824 (2002).

nature reviews | microbiology volume 6 | january 2008 | 65


© 2008 Nature Publishing Group
REVIEWS

173. Abrahams, G. L., Muller, P. & Hensel, M. Functional 185. Marlovits, T. C. et al. Structural insights into the SpvB. Mol. Microbiol. 39, 606–619 (2001).
dissection of SseF, a type III effector protein involved assembly of the type III secretion needle complex. 197. Worley, M. J., Nieman, G. S., Geddes, K. & Heffron, F.
in positioning the Salmonella-containing vacuole. Science 306, 1040–1042 (2004). Salmonella typhimurium disseminates within its host
Traffic 7, 950–965 (2006). 186. Sun, J., Hobert, M. E., Rao, A. S., Neish, A. S. & by manipulating the motility of infected cells. Proc.
174. Kuhle, V., Abrahams, G. L. & Hensel, M. Intracellular Madara, J. L. Bacterial activation of b-catenin Natl Acad. Sci. USA 103, 17915–17920 (2006).
Salmonella enterica redirect exocytic transport signaling in human epithelia. Am. J. Physiol. 198. Kujat Choy, S. L. et al. SseK1 and SseK2 are novel
processes in a Salmonella pathogenicity island Gastrointest. Liver Physiol. 287, G220–227 (2004). translocated proteins of Salmonella enterica serovar
2‑dependent manner. Traffic 7, 716–730 (2006). 187. Zhou, D., Mooseker, M. S. & Galan, J. E. An invasion- Typhimurium. Infect. Immun. 72, 5115–5125
175. Birmingham, C. L., Jiang, X., Ohlson, M. B., Miller, S. I. associated Salmonella protein modulates the actin- (2004).
& Brumell, J. H. Salmonella-induced filament formation bundling activity of plastin. Proc. Natl Acad. Sci. USA 199. Rytkonen, A. et al. SseL, a Salmonella deubiquitinase
is a dynamic phenotype induced by rapidly replicating 96, 10176–10181 (1999). required for macrophage killing and virulence. Proc.
Salmonella enterica serovar Typhimurium in epithelial 188. Lee, C. A. et al. A secreted Salmonella protein induces Natl Acad. Sci. USA 104, 3502–3507 (2007).
cells. Infect. Immun. 73, 1204–1208 (2005). a proinflammatory response in epithelial cells, which 200. Geddes, K., Worley, M., Niemann, G. & Heffron, F.
176. Lesnick, M. L., Reiner, N. E., Fierer, J. & Guiney, D. G. promotes neutrophil migration. Proc. Natl Acad. Sci. Identification of new secreted effectors in Salmonella
The Salmonella spvB virulence gene encodes an USA 97, 12283–12288 (2000). enterica serovar Typhimurium. Infect. Immun. 73,
enzyme that ADP-ribosylates actin and destabilizes 189. Hernandez, L. D., Pypaert, M., Flavell, R. A. & Galan, 6260–6271 (2005).
the cytoskeleton of eukaryotic cells. Mol. Microbiol. J. E. A Salmonella protein causes macrophage cell 201. Tsolis, R. M., Adams, L. G., Ficht, T. A. & Baumler, A. J.
39, 1464–1470 (2001). death by inducing autophagy. J. Cell Biol. 163, Contribution of Salmonella typhimurium virulence
177. Browne, S. H., Lesnick, M. L. & Guiney, D. G. Genetic 1123–1131 (2003). factors to diarrheal disease in calves. Infect. Immun.
requirements for Salmonella-induced cytopathology in 190. Hayward, R. D. et al. Cholesterol binding by the 67, 4879–4885 (1999).
human monocyte-derived macrophages. Infect. bacterial type III translocon is essential for virulence
Immun. 70, 7126–7135 (2002). effector delivery into mammalian cells. Mol. Microbiol.
178. Gotoh, H. et al. Extracellular secretion of the virulence 56, 590–603 (2005). Acknowledgments
plasmid-encoded ADP-ribosyltransferase SpvB in 191. Carlson, S. A., Omary, M. B. & Jones, B. D. We would like to thank the entire Salmonella pathogenesis
Salmonella. Microb. Pathog. 34, 227–238 (2003). Identification of cytokeratins as accessory mediators of community for its work that made this Review possible. In
179. Brumlik, M. J. & Buckley, J. T. Identification of the Salmonella entry into eukaryotic cells. Life Sci. 70, particular, we are grateful to members of the Miller labora-
catalytic triad of the lipase/acyltransferase from 1415–1426 (2002). tory, past and present, for their contributions to the ideas
Aeromonas hydrophila. J. Bacteriol. 178, 2060–2064 192. Knodler, L. A., Finlay, B. B. & Steele-Mortimer, O. The that are presented here. We would also like to apologize to
(1996). Salmonella effector protein SopB protects epithelial those authors whose work was not cited owing to space limi-
180. Ohlson, M. B., Fluhr, K., Birmingham, C. L., Brumell, cells from apoptosis by sustained activation of Akt. tations. A.H. is supported by a Career Development Award
J. H. & Miller, S. I. SseJ deacylase activity by J. Biol. Chem. 280, 9058–9064 (2005). from the Northwest Regional Center of Excellence for
Salmonella enterica serovar Typhimurium promotes 193. Mukherjee, K., Parashuraman, S., Raje, M. & Biodefense and Emerging Infectious Diseases Research
virulence in mice. Infect. Immun. 73, 6249–6259 Mukhopadhyay, A. SopE acts as an Rab5-specific (National Institute of Allergy and Infectious Diseases (NIAID)
(2005). nucleotide exchange factor and recruits non- grant U54 AI057141). M.B.O. is supported by the
181. Harrison, R. E. et al. Salmonella impairs RILP prenylated Rab5 on Salmonella-containing Comprehensive Training in Inter-Disciplinary Oral Health
recruitment to Rab7 during maturation of invasion phagosomes to promote fusion with early endosomes. Research T32 grant DE07132. S.I.M. is supported by the
vacuoles. Mol. Biol. Cell 15, 3146–3154 (2004). J. Biol. Chem. 276, 23607–23615 (2001). National Institutes of Health, NIAID grants R01 AI30479,
182. Marsman, M., Jordens, I., Kuijl, C., Janssen, L. & 194. Coombes, B. K. et al. Genetic and molecular analysis R01 AI048683 and U54 AI057141 for the Northwest
Neefjes, J. Dynein-mediated vesicle transport controls of GogB, a phage-encoded type III-secreted substrate Regional Center of Excellence for Biodefense and Emerging
intracellular Salmonella replication. Mol. Biol. Cell 15, in Salmonella enterica serovar Typhimurium with Infectious Diseases Research.
2954–2964 (2004). autonomous expression from its associated phage.
183. Meresse, S. et al. Remodelling of the actin cytoskeleton J. Mol. Biol. 348, 817–830 (2005).
is essential for replication of intravacuolar Salmonella. 195. Brumell, J. H., Rosenberger, C. M., Gotto, G. T., DATABASES
Cell. Microbiol. 3, 567–577 (2001). Marcus, S. L. & Finlay, B. B. SifA permits survival Entrez Genome Project: http://www.ncbi.nlm.nih.gov/
184. Viboud, G. I. & Bliska, J. B. A bacterial type III and replication of Salmonella typhimurium in murine entrez/query.fcgi?db=geneomeprj
secretion system inhibits actin polymerization to macrophages. Cell. Microbiol. 3, 75–84 (2001). Salmonella typhi | Salmonella typhimurium
prevent pore formation in host cell membranes. 196. Tezcan-Merdol, D. et al. Actin is ADP-ribosylated by All links are active in the online pdfF
EMBO J. 20, 5373–5382 (2001). the Salmonella enterica virulence-associated protein

66 | january 2008 | volume 6 www.nature.com/reviews/micro


© 2008 Nature Publishing Group

You might also like