You are on page 1of 12

Review

Initiation of Infection

Subversion of trafficking, apoptosis,


and innate immunity by type III
secretion system effectors
Benoit Raymond, Joanna C. Young, Mitchell Pallett, Robert G. Endres,
Abigail Clements, and Gad Frankel
Department of Life Sciences, Imperial College, London, UK

Injection of effector proteins by a type III secretion sys- part of the initial infection process and will be the focus
tem (T3SS) is a common infection strategy employed by of this review. Although the actin cytoskeleton is manip-
many important human pathogens, including enteric ulated by T3SS effectors during infection, this has been
Escherichia coli, Salmonella, Yersinia, and Shigella, to extensively reviewed elsewhere and thus will not be
subvert cell signaling and host responses. In recent covered in this review [1].
years, great advances have been made in understanding
how the T3SS effectors function and execute the diverse Disrupting cellular trafficking
infection strategies employed by these pathogens. In Eukaryotic cells have two vesicular transport systems to
this review, we focus on effectors that subvert signaling mobilize proteins and lipids. The secretory pathway trans-
pathways that impact on endosomal trafficking, cell ports material from the endoplasmic reticulum (ER) to the
survival, and innate immunity, particularly phagocyto- plasma membrane (PM), via the Golgi, whereas the endo-
sis, nuclear factor-kB (NF-kB), and mitogen-activated cytic pathway transports PM-derived endosomes to the
protein (MAP) kinase pathways and the inflammasome. Golgi, ER, lysosomes (for degradation), or back to the
PM via recycling pathways. Both intracellular and extra-
Type III secretion and infection cellular pathogens manipulate endocytic pathways using
The intestinal epithelium provides a critical interface T3SS effectors (Figure 1).
between the body and the external environment acting as
a protective barrier against ingested toxins and patho- Secretory pathway
gens and consists of the largest reservoir of microorgan- Transport steps in the secretory pathway involve cargo
isms that live in the body. Many pathogenic bacteria packaging at the donor membrane followed by vesicle
have acquired sophisticated strategies to breach the budding, transport, tethering, and uncoating at the recep-
intestinal integrity and to exploit the mucosa as a niche tor membrane. These processes are regulated by small
to replicate and disseminate into deeper tissues to cause GTPases. Vesicles responsible for anterograde ER–Golgi
disease. Examples include enteropathogenic and enter- transport contain coat protein complex II (COPII), whereas
ohemorrhagic Escherichia coli (EPEC and EHEC), Shi- COPI coated vesicles are responsible for retrograde intra-
gella, Salmonella, and Yersinia species which all use Golgi and Golgi–ER transport [2]. EPEC/EHEC translo-
their type III secretion system (T3SS) to inject bacterial cate several effectors which inhibit the secretory pathway,
effector proteins to subvert the host defenses and pro- including NleA/EspI, NleF, and EspG. NleA/EspI inhibits
mote gut infection. Although these species all rely on the anterograde ER–Golgi transport by binding to Sec24
their T3SS for pathogenesis, their infection strategies paralogs [3,4] of the COPII vesicle coat. NleA/EspI appears
differ considerably. Whereas EPEC/EHEC and Yersinia to stabilize the COPII vesicles at the Golgi, inhibiting the
are predominantly extracellular pathogens, Salmonella uncoating of the vesicle (with reduced hydrolysis of the
and Shigella trigger their own internalization and sur- small GTPase Sar1) and hence reduced recycling of COPII
vive intracellularly. All of these pathogens use T3SS components back to the ER, resulting in reduced secretion.
effectors to regulate actin dynamics to facilitate their The consequence of this reduced secretion during infection
own attachment or invasion, subvert endocytic traffick- is unclear, although NleA/EspI does significantly contrib-
ing, block phagocytosis, modulate apoptotic pathways, ute to virulence of the EPEC/EHEC-like mouse pathogen
and manipulate innate immunity and host responses as Citrobacter rodentium in vivo [5]. In polarized cultured
cells NleA/EspI can alter cellular tight junctions [6], po-
Corresponding author: Frankel, G. (g.frankel@imperial.ac.uk).
tentially by reducing secretion of replacement tight junc-
Keywords: enteropathogens; T3SS; subversion of cellular trafficking; phagocytosis
remodeling; cell survival modulation; inflammatory response manipulation. tion proteins. Recently, the EHEC effector NleF was shown
0966-842X/$ – see front matter
to bind Tmp21, a COPI vesicle receptor involved in trans-
ß 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tim.2013.06.008 Golgi network function, and may slow ER–Golgi and
430 Trends in Microbiology August 2013, Vol. 21, No. 8
Review Trends in Microbiology August 2013, Vol. 21, No. 8

EPEC
EHEC

Salmonella Shigella

IpgD
PIP2 PIP
SopB Exo70
SipC

Lysosome

MPR Hydrolase
Hydrolase
MVB/ Exocyst
LE SKIP
SifA
SopB
MPR Salmonella Sif
SseF/G

IpaJ

ARF1
Golgi ARFs Rabs
EspG
Espl/NleA Tmp21 GM130 PAKs
Sec24 NleF

ER

Nucleus

TRENDS in Microbiology

Figure 1. Intracellular trafficking disruption. The EspI/NleA effectors from EPEC/EHEC (in blue) bind Sec24 to disrupt protein secretion, potentially by reducing recycling of
COPII to the ER. EspG acts as a Rab-GAP, but also interacts with ARFs, PAKs, and GM130 to disrupt protein secretion and NleF binds to Tmp21 to slow ER–golgi trafficking.
Shigella IpaJ (in orange) acts as cysteine protease that cleaves the N-myristoylated glycine from ARF1 to block cargo transport through the Golgi apparatus. SopB (in green)
and IpgD (in orange) from Salmonella and Shigella, respectively, dephosphorylate phosphatidylinositol 4,5 bisphosphate, PI(4,5)P2 to produce PI5P to regulate membrane
trafficking at the invasion site. SopB can also be found on the SCV. Salmonella SipC binds Exo70 to redirect exocysts to the bacterial attachment site. SseG and SseF
facilitate the extension of Sifs along the microtubules towards the cell periphery. Binding of SifA to SKIP sequesters Rab9, inhibiting the trafficking of MPR to the trans-Golgi
network and downstream trafficking of hydrolases to the lysosome and then the SCV. Abbreviations: EPEC/EHEC, enteropathogenic and enterohemorrhagic Escherichia
coli; COPII, coat protein complex II; ER, endoplasmic reticulum; Rab-GAP, Rab GTPase-activating protein; ARFs, ADP ribosylation factors; PAKs, p21 activated kinases; SCV,
Salmonella-containing vacuole; Sifs, Salmonella induced filaments; MPR, mannose phosphate receptor.

Golgi–PM trafficking, although further work is required to VirA, a homolog of EspG found in Shigella also acts as a
clarify this function [7]. An additional EPEC/EHEC effec- Rab-GAP but during Shigella infection it reduces autop-
tor, EspG, was shown by multiple methods to disrupt the hagic recognition (LC3 recruitment) of cytosolic Shigella,
secretory pathway [8,9]. EspG can function as a Rab thus protecting it from degradation [10]. Recently, the
GTPase-activating protein (Rab-GAP) [10], trapping Shigella effector IpaJ, a cysteine protease, has been shown
Rab-GTPases in their inactive GDP bound form, and re- to cleave the myristoyl group from ARF1, which regulates
ducing ER–Golgi transport. EspG can also bind numerous cargo transport through the Golgi apparatus, and is now
other proteins including ADP ribosylation factor (ARF) the second Shigella T3SS effector shown to alter the host
GTPases [8], p21 activated kinases (PAKs) [8,11], and secretory pathway during infection [12].
the Golgi matrix protein GM130 [9]. EspG interacts with In Salmonella, the C terminus of SipC interacts with the
activated, membrane-embedded ARFs, potentially trans- exocyst targeting component Exo70 [13], causing exocysts to
forming ARF activation into Rab inactivation at appropri- tether at the bacterial attachment site, potentially provid-
ate endomembranes. During infection of cultured cells ing extra membrane to facilitate invasion. To date, this is the
EspG expression reduced interleukin-8 (IL-8) secretion only Salmonella effector which specifically regulates the
[10], suggesting it plays a role in immunomodulation. host secretory pathway, although many Salmonella effec-
The ability of NleA/EspI and NleF to reduce cytokine tors are involved in regulating endolysosome trafficking.
expression during infection has not been reported, thus
it remains to be seen whether interrupting the secretory Endolysosome trafficking
pathway is another mechanism utilized by these patho- The endosomal pathway internalizes extracellular materi-
gens to reduce immune recognition. al as well as plasma membrane proteins and lipids which
431
Review Trends in Microbiology August 2013, Vol. 21, No. 8

are then sorted in the early endosome for degradation the adaptive immune response. Phagocytes can recognize
through the late endosomes and lysosomes, or for transport bacteria directly [e.g., through scavenger receptor A recog-
to the trans-Golgi, or back to the plasma membrane via nition of lipopolysaccharide (LPS) [28]] or indirectly
recycling endosomes. Once internalized by host cells Sal- through recognition of immune opsonins (IgG and the
monella subverts the endolysosome trafficking pathway to complement cascade component C3bi), which coat the
create a Salmonella-containing vacuole (SCV), which is bacteria and are recognized by the Fcg receptors and
essential for its intracellular survival. The SCV transiently integrin amb2 (complement receptor 3), respectively
acquires proteins from sorting and recycling endosomes [29,30]. Receptor ligation activates complex signaling
[Rab5, EEA1, and the transferrin receptor (TfR)] before pathways involving tyrosine kinases, small Rho GTPases
acquiring vacuolar ATPase, responsible for phagosome and PIs. Successful pathogens display an extensive array
acidification [14], and lysosomal glycoproteins (lgps), such of strategies for subverting phagocytosis (Figure 2). Intra-
as Lamp1, in a Rab7-dependent process [15]. The SCV does cellular Salmonella and Shigella escape phagocytic killing
not however accumulate the lysosomal hydrolases or the through manipulation of endolysosomal trafficking, as
mannose phosphate receptors (MPRs) that traffic the discussed above, which disrupts phagosome maturation.
hydrolases from the Golgi to the lysosomes. This selective Yersinia cross the intestinal epithelial layer by trans-
exclusion has been attributed to the SPI-2 T3SS effector cytosis through antigen sampling M cells and can then
SifA and its binding partner SKIP, which sequester Rab9 invade macrophages and enterocytes mediated by an in-
to inhibit MPR trafficking from late endosomes to the teraction between Invasin and b1 integrins [31]. However,
trans-Golgi network and hence MPR-dependent trafficking Yersinia predominantly survives extracellularly using the
of the hydrolases from the Golgi to the lysosome [16]. As the translocation of effectors to inhibit cytoskeletal rearrange-
SCVs mature they travel to the microtubule organizing ments and phagocytosis. With the exception of EspT-posi-
center (MTOC), a perinuclear region adjacent to the Golgi, tive invasive strains (2% EPEC strains) [32], EPEC/
where they produce Salmonella induced filaments (Sifs) EHEC inhibit both transcytosis through M cells and inter-
dependent on the SPI-2 T3SS effectors SseF and SseG. Sifs nalization by phagocytes [33,34].
extend along microtubules from the SCV to the cell periph- Yersinia inhibits phagocytosis through the concerted
ery where they recruit Rab9 and Rab11 and take up fluid actions of several effector proteins. YopH is a tyrosine
phase endocytic markers, potentially providing nutrients phosphatase, which acts on several cytoskeletal proteins
for the bacteria from outside the cell [17–19]. including p130Cas, Fyn binding protein (Fyb), focal adhe-
By contrast, internalized Shigella avoids the endolyso- sion kinase (FAK), p85, Lyn, and paxillin [35–38]. Dephos-
some system by rapidly lysing its vacuole through the phorylation of p130Cas disrupts its interaction with Crk
action of the T3SS effectors IpaB and C [20]. IpaB in and subsequent Rac activation required for Yersinia inter-
particular has been shown to form membrane-disrupting nalization [39]. Yersinia also subverts the actin polymeri-
pores [20,21] and some studies suggest that IpaH7.8 may zation required for phagocytosis by the actions of several
also be involved [22]. Time-lapse analysis reported the effectors which manipulate Rho GTPases. The effector
recruitment of host factors, including RhoA and Rac1, to YopE acts as a RhoGAP [40] and inhibits Rac-dependent
lysing vacuoles suggesting the involvement of host signal- actin polymerization either directly or through inactiva-
ing [23]; however, further research is required to fully tion of the upstream RhoG [41,42]. The effector YopO/YpkA
understand this key step in the pathogenesis of Shigella. has a carboxy-terminal GDP-dissociation inhibitor (GDI)-
Phosphoinositide (PI) lipids play a significant regulato- like domain which interacts with Rho GTPases [43] and is
ry role in membrane trafficking. Salmonella SopB/SigD suggested to sequester the GDI-free pool of Rac1 localized
and Shigella IpgD are PI phosphatases that dephosphory- at the membrane [44]. YopT, restricted to Yersinia enter-
late phosphatidylinositol 4,5 bisphosphate, PI(4,5)P2, to ocolitica, acts as a cysteine protease cleaving the carboxy-
produce PI5P. Although SopB aids bacterial invasion by terminal prenylyl groups of RhoA, Rac1, and Cdc42 [45].
localizing to the plasma membrane early in infection [24], This cleavage inactivates Rho GTPases by disrupting their
it later relocalizes to the SCV [25] where it alters the PI membrane localization.
composition to reduce the recruitment of Rab8, Rab13, Extracellular EPEC and EHEC also disrupt Rho
Rab23, and Rab35 and prevent phagolysosome formation. GTPases to prevent internalization through the effector
The formation of PI5P by IpgD at the PM recruits epider- EspH, which inactivates endogenous guanine nucleotide
mal growth factor receptor (EGFR) to the PM on early exchange factors (GEFs), preventing Rho GTPase activa-
endosomes to stop maturation and degradation leading to tion and thus inhibiting phagocytosis [46]. EspB, which
prolonged cell survival through Akt activation [26]. Similar also functions in the translocation pore, interacts with
to SopB, IpgD has pleiotropic effects, including cytoskele- myosin family proteins inhibiting their interaction with
tal reorganization at the bacterial invasion site [27] and actin [47]. This disruption of the actomyosin system
cell migration, in addition to its role in cell survival. results in inhibition of phagocytosis. EspF is central in
inhibiting internalization of non-opsonized bacteria and
Phagocytosis remodeling in preventing transcytosis through M cells [33,48]. Al-
Professional phagocytes of the immune system (including though EspF has been shown to inhibit PI 3 kinase
neutrophils, macrophages, and dendritic cells) mediate the (PI3K)-dependent internalization through its N-terminal
ingestion of foreign material and pathogenic organisms, 101 residues, it does not inhibit opsonophagocytosis [49].
which results in both destruction of the organism and Several binding partners for EspF have been identified
generation of peptides for MHC presentation to prime including actin, profilin, sorting nexin 9 (SNX-9), and
432
Review Trends in Microbiology August 2013, Vol. 21, No. 8

Species inhibing internalizaon Species promong internalizaon

IgG
EPEC
EHEC

Yersinia Shigella Salmonella

FcγR EspF EspB


EspJ
Myosin Internalizaon
Internalizaon Internalizaon
YopH
Disrupon of
p85 P p85
Shigella endomembrane
YopO Fyb P Fyb trafficking
Cas P Cas IpaB IpaC
GDP EspH
YopT
Vacuolar lysis SifA

Salmonella
GDP GTP
Acn
polymerizaon Shigella Sifs

YopE

TRENDS in Microbiology

Figure 2. Disruption of phagocytosis and cell invasion. EPEC/EHEC (effectors in blue) and Yersinia (effectors in pink) inhibit internalization by disrupting phagocytic
signaling. In EPEC/EHEC, EspB prevents the interaction between actin and myosin, whereas EspH inhibits mammalian GEFs disrupting GTPase activation and actin
polymerization. EspF inhibits EPEC/EHEC internalization and EspJ inhibits opsonophagocytosis through unknown mechanisms. In Yersinia, YopH dephosphorylates host
proteins including p130Cas, p85, and Fyb to inhibit signal propagation. YopE, YopO, and YopT inhibit RhoGTPases by promoting GTP hydrolysis, sequestering the inactive
form and cleaving prenylyl groups, respectively. Shigella (in orange) and Salmonella (in green) trigger actin polymerization to promote their internalization. Once
internalized, Shigella disrupts the phagosome dependent on IpaB and IpaC to survive extracellularly. Salmonella prevents phagosome maturation through SifA-dependent
disruption of endomembrane signaling (see Figure 1 for further details). Abbreviations: EPEC/EHEC, enteropathogenic and enterohemorrhagic Escherichia coli; GEFs,
guanine nucleotide exchange factors; Fyb, Fyn binding protein; Sif, Salmonella induced filament.

neural Wiskott–Aldrich syndrome protein (N-WASP), but and tunicamycin [53]. Treatment with brefeldin A and
the mechanism of action required to inhibit internaliza- tunicamycin induces the intrinsic apoptotic pathway lead-
tion remains unclear [50,51]. Additionally, EspJ is impli- ing to the conformational change and translocation of Bax to
cated in the inhibition of opsonophagocytosis pathways, the mitochondria, mitochondrial permeabilization, and cy-
but it does not prevent internalization of uncoated bacte- tochrome C release. Through direct interaction with Bax
ria [48]. These pathogens therefore use an extensive array inhibitor I (BI-I), NleH blocks the intrinsic pathway [53].
of strategies to manipulate phagocytosis either by impair- Similarly, the Shigella effector IpgD blocks staurosporine-
ing signaling to evade uptake or by facilitating their own induced apoptosis [54] by phosphorylating and stabilizing
entry while blocking phagosomal maturation to establish the double minute 2 protein (MDM2). MDM2 represses p53
an intracellular niche. through binding its trans-activation domain [55]. Inhibition
of p53 blocks NF-kB–p53 proapoptotic signaling, inhibiting
Manipulation of cell survival apoptosis. Furthermore, the Shigella effector VirA promotes
Programmed cell death through apoptosis is a tightly further degradation of p53 via activation of calpain. Al-
regulated process activated by either intrinsic (intracellu- though VirA has an antiapoptotic role, it also drives epithe-
lar: ER and mitochondria-mediated) or extrinsic (extracel- lial cell necrosis and cell detachment in Shigella-infected
lular: receptor-mediated) pathways. Both result in the cells [55]. More recently, the EPEC/EHEC effector NleF has
activation of the caspase cascade, which ultimately leads been shown to specifically bind and inhibit the activity of
to cell shrinkage, chromatin condensation, membrane caspase-4, -8, and -9, inhibiting staurosporine- and TRAIL-
blebbing, and formation of vesicular buds known as apo- induced cell death, when transiently expressed [56].
ptotic bodies [52]. Pathogenic bacteria have developed the Salmonella inhibits apoptosis and activates pro-surviv-
ability to advantageously manipulate and avert host cell al signals, dependent on the effectors AvrA and SopB,
death defense mechanisms to promote colonization and respectively. SopB sustains Akt activation and signaling
infection by injecting specific T3SS effectors (Figure 3). through the pro-survival PI3K–Akt pathway. Infection
with Salmonella enterica serovar Typhimurium (S. Typhi-
Inhibition of apoptosis murium) sopB deletion mutant increases caspase-3 cleav-
The EPEC/EHEC NleH family, homologs of Shigella OspG, age and apoptosis [57]. The antiapoptotic role of AvrA is
blocks apoptosis stimulated with staurosporine, brefeldin A, dependent on its acetyltransferase activity and inhibition
433
Review Trends in Microbiology August 2013, Vol. 21, No. 8

EPEC
EHEC
Survival
signal
Salmonella Shigella Extrinsic
TRAIL, FasL
SopB NIeH
PI3K
Proinflammatory Caspase-8
signal BI-1
SipB
AKT

P
t-Bid
Bad Bax
MKK4/7
Caspase-2 NleF

AvrA
P Mitochondria
JNK EspF

IpgD Cyt C
NleF NleF
APAF-1
P EspH Caspase-9
MDM2
Caspase-4
EspM2 Caspase-3
p65/RelA SopE EspT Cif
Stress
p50 Abcf-2
Apoptosis
p53
EspF
Nucleus Calpain IpgD ER

TRENDS in Microbiology

Figure 3. Manipulation of cell survival. Intrinsic, extrinsic, and proinflammatory pathways involved in apoptosis are targeted by an extensive array of T3SS effectors. The EPEC/
EHEC (blue) NleH family blocks the intrinsic apoptotic pathway through direct interaction with BI-I. NleF binds caspase-4, -8, and -9, inhibiting both intrinsic and extrinsic
pathways. EspF induces cytochrome C (Cyt C) release from the mitochondria and degrades Abcf2 leading to the activation of caspase-9 and -3. Cif and EspH induce caspase-3
cleavage. Caspase-3 cleavage and cell death are recovered by the RhoGEF mimics EspM2, EspT, and SopE (from Salmonella, in green). Salmonella inhibits apoptosis through
SopB and AvrA. SopB sustains Akt activation and AvrA inhibits JNK proapoptotic signaling at the level of MKK4/7. In macrophages, SipB induces caspase-2 activation leading to
cell death. The Shigella effector IpgD blocks apoptosis by phosphorylating and stabilizing the MDM2. Furthermore, VirA promotes further degradation of p53 via activation of
calpain. Abbreviations: EPEC/EHEC, enteropathogenic and enterohemorrhagic Escherichia coli; T3SS, type III secretion system; BI-I, Bax inhibitor I; GEF, guanine nucleotide
exchange factor; JNK, c-Jun NH2-terminal kinase; MMK4/7, mitogen-activated protein kinase kinase 4/7; MDM2, double minute 2 protein.

of c-Jun NH2-terminal kinase (JNK) proapoptotic signal- caspase-3 cleavage and cell death, which is recovered by
ing at the level of mitogen-activated protein kinase kinase coexpression of the RhoGEF mimics EspM2, EspT (EPEC/
4 (MKK4) and MKK7 [58]. EHEC), and SopE (Salmonella) [61].
Furthermore, infection of macrophages with Salmonel-
Induction of cell death la induces caspase-2 cleavage in a SipB-dependent man-
Although Salmonella, Shigella, Yersinia, EPEC, and ner, leading to apoptosis and cell death independent of
EHEC secrete T3SS effectors to block apoptosis, cell death caspase-1 [62].
is apparent in in vivo models and in some in vitro studies.
EspF targets the mitochondria during EPEC infection of Inflammatory response
HeLa cells inducing a loss of mitochondrial outer mem- Extracellular and intracellular pattern recognition recep-
brane potential, cytochrome C release, and activation of tors (PRRs) recognize pathogen-associated molecular pat-
intrinsic apoptosis [59]. EspF binds Abcf2, a protein be- terns (PAMPs), which stimulate inflammatory signaling
longing to the ABC transporter family. During infection, cascades such as the NF-kB and MAPK pathways. Bacte-
Abcf2 is degraded in an EspF-dependent manner leading to rial pathogens have developed multiple mechanisms to
an increase in caspase-9 and caspase-3 cleavage [59]. The disrupt these pathways as outlined below.
EPEC/EHEC effector Cif blocks cell cycle progression and
was recently identified as a late inducer (48 h) of EPEC- Altering signaling to NF-kB
induced apoptosis inducing caspase-3 cleavage and lactate EPEC/EHEC, NleE, NleB, NleC, NleH, and Tir are immu-
dehydrogenase (LDH) release [60]. The role of Cif in apo- nosuppressor T3SS effectors acting cooperatively to target
ptosis may be linked to its function in the inhibition of different proteins in the NF-kB signaling pathway
the ubiquitin/proteasome pathway. EspH also induces (Figure 4). NleE has methyltransferase activity and
434
Review Trends in Microbiology August 2013, Vol. 21, No. 8

EPEC
EHEC TNF-α IL-1β

Shigella
Salmonella Yersinia TNF-α R IL-1β R

Ospl Ubc13
TRAF2 TRAF6
SipA GAPDH ub Tir SH1 ub
EspT IpGB2 NOD1/2 NleB
SopE SopE2 NleE
SopB
TAB2/3
ABIN-1
AvrA
GDP GTP IpaH9.8
SspH1 SseL TAK1

NEMO YopP/J
IKKγ
P
E2–E3 OspG
P complex P Proteosomal
IκBα IκBα degradaon
p65/RelA ub
p50 NleC
RPS3
OspZ
YopE

NleH1
p65/RelA
RPS3
p50
Nucleus
TRENDS in Microbiology

Figure 4. NF-kB subversion. NleB, NleE, NleH1, Tir, and NleC from EPEC/EHEC (in blue) inhibit NF-kB activation by targeting TRAF2, TAB1/2, RPS3, TRAF6, or p65 directly,
respectively. NleB reduces TRAF2 polyubiquitination by blocking its interaction with GAPDH. NleE blocks TAK2/3 activation. NleH1 binds and blocks the translocation of
RPS3 into the nucleus. Through its ITIM-like sequence that recruits SHP-1 to bind TRAF6, Tir blocks TRAF6 activation. NleC cleaves p65. Shigella affects NF-kB signaling
through OspG, OspI, IpaH9.8, and OspZ (in orange). OspG interferes with the degradation of IkB-a, OspI blocks the polyubiquitination of TRAF6, IpaH9.8 promotes the
degradation of NEMO, and OspZ blocks the translocation of p65 through an unknown mechanism. Yersinia translocates YopE (in pink), which inhibits activation of NF-kB,
and YopP/J that acts as a serine/threonine acetyltransferase targeting TAK1. SseL, SspH1, and AvrA (in green) secreted by Salmonella have been identified to inhibit NF-kB
signaling pathways. However, the RhoGEF mimics SopE, SopE2, EspT, and IpgB2, as well as the PI phosphatase SopB, induce NF-kB activation through their impact on
RhoGTPases. In addition, SipA activates NF-kB through NOD1. Abbreviations: NF-kB, nuclear factor-kB; EPEC/EHEC, enteropathogenic and enterohemorrhagic Escherichia
coli; T3SS, type III secretion system; RPS3, ribosomal protein S3; TRAF, TNF receptor-associated factor; TAB, TAK1 binding protein; TAK, TGFb activated kinase; ITIM,
immunoreceptor tyrosine-based inhibition motif; IkB-a, inhibitor of kB, NEMO, NF-kB essential modulator;GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GEF,
guanine nucleotide exchange factor; PI, phosphoinositide.

specifically methylates TAB2/3 to block TRAF6-induced TNF-a stimulation. Tir has an immunoreceptor tyrosine-
activation of the TAK1–NF-kB pathway in response to based inhibition motif (ITIM)-like sequence that recruits
tumor necrosis factor-a (TNF-a) and interleukin-1b (IL- SHP-1 to bind TRAF6 and inhibits cytokine production in
1b) [63–65]. NleB has N-acetyl-D-glucosamine transferase cultured cells and in vivo [70,71].
activity and modifies glyceraldehyde 3-phosphate dehydro- Although Shigella peptidoglycan stimulates signaling
genase (GAPDH) to disrupt the TRAF2–GAPDH interac- pathways required for an efficient inflammatory response,
tion and reduce TRAF2 polyubiquitination, which is which is thought to be important for the infection strategy
needed for NF-kB activation [66]. Through its consensus of Shigella, studies have also identified Shigella T3SS
zinc metalloprotease motif, NleC has been shown to cleave effectors which affect the NF-kB pathway, including OspZ
the NF-kB p65 subunit (RelA), blocking the production of and OspG, homologs of NleE and NleH, respectively. Simi-
IL-8 following infection [67]. Although both NleH1 and lar to NleE, OspZ blocks the translocation of NF-kB to the
NleH2 bind to the ribosomal protein S3 (RPS3) subunit nucleus [64], but whether OspZ shares the same cysteine
of NF-kB transcriptional complexes, NleH1, but not methylase activity of NleE is still unknown. Through its
NleH2, blocks translocation of NF-kB into the nucleus, impact on the E2–E3 ubiquitination complex, OspG inter-
without affecting IkB-a degradation [68,69]. The effector feres with the ubiquitin-dependent proteasomal degrada-
Tir, most well characterized for its role in pedestal forma- tion of IkB-a [72,73], in contrast to NleH. OspG also
tion, is also able to inhibit NF-kB activation in response to displays kinase activity, inducing its autophosphorylation
435
Review Trends in Microbiology August 2013, Vol. 21, No. 8

which is necessary for its function. The IpaH effectors are the MAPK signaling pathway by an unknown mechanism
found multiple times in Shigella spp. and the crystal [90].
structure suggests that they belong to a new class of E3 Salmonella AvrA blocks JNK activation at the level of
ubiquitin ligases, with a role in suppressing innate im- the MAPK kinases MKK4 and MKK7 in both AvrA trans-
mune responses [74]. IpaH9.8 dampens NF-kB signaling genic Drosophila and murin cells. Consistent with this
by interacting with NEMO/IKK and ABIN-1. ABIN-1 pro- finding, a DavrA mutant induces higher levels of JNK
motes polyubiquitination of NEMO, which then undergoes activation and inflammation in infected murine intestine
proteasome-dependent degradation, ultimately blocking [58]. Interestingly, the ERK pathway, which is activated
NF-kB activation [75]. OspI selectively deamidates a glu- upon S. Typhimurium infection, is required for the phos-
tamine residue in the E2 enzyme UBC13 to abolish its phorylation of AvrA, which then impairs the JNK pathway
ubiquitin-conjugating activity, which is required for acti- through its interaction with MKK7, but not the p38 or NF-
vation of the TRAF6–NF-kB pathway [76]. kB pathways. In addition, SpvC, a member of the OspF
Similarly, YopE, YopP, and YopJ (in Yersinia enteroco- family, has been shown to abrogate the activation of the
litica, Yersinia pestis, and Yersinia pseudotuberculosis re- ERK, JNK, and p38 kinases [87].
spectively) target the NF-kB pathway. YopE inhibits
activation of NF-kB, which in part prevents the production Upregulation of inflammatory responses
of IL-8 [77]. YopJ acts as a cysteine protease and was Bacterial RhoGEF mimics [1] have a well-defined role in
reported to hydrolyze ubiquitin and SUMO [78]. YopP/J cytoskeleton remodeling and also induce innate immune
also functions as a serine/threonine acetyltransferase tar- responses. S. Typhimurium induces inflammation in cul-
geting the activation loop of Drosophila TAK1, which tured epithelial cells through SopE and SopE2 [91]. SopE
blocks its phosphorylation and subsequent kinase activa- and SopE2 are GEFs for Rac1, Cdc42, RhoA, and RhoG [1].
tion [79]. SopB is a PI phosphatase, which activates RhoG and Cdc42
Despite the profound proinflammatory nature of Sal- by stimulating their endogenous exchange factors [25], and
monella, the effectors SseL [80], SspH1 [81], and possibly also induces inflammation [91]. Activation of these
AvrA [82] have been found to inhibit NF-kB signaling GTPases by SopE, SopE2, and SopB leads to stimulation
pathways. of NF-kB, ERK, JNK, and p38 pathways independently of
the intracellular sensors NOD1, NOD2, and RIP2 kinase
Targeting MAPK [91]. However, it has recently been shown that SopE
MAPK, with the extracellular signal-regulated protein triggers the NOD1–RIP2–NF-kB pathway [92]. It has been
kinases (ERKs), the p38 MAP kinases, and the JNKs suggested that the induction of inflammation is crucial for
cooperate with other signaling pathways to regulate the the ability of Salmonella to grow in the intestinal tract,
expression of inflammatory genes [83]. In addition to NF- because essential nutrients do not become available at this
kB, MAPK is also targeted by T3SS effectors to dampen the site unless such responses are stimulated [93]. Moreover,
inflammatory response. The EPEC effector NleD is a zinc- recent reports indicated that Salmonella is able to use
dependent metalloprotease specifically targeting MAP electron acceptors generated during inflammation in the
kinases JNK and p38 but not ERK, thereby blocking gut, thereby gaining a competitive advantage over com-
nuclear translocation of the AP-1 transcription factor, mensal bacteria [94,95]. The EPEC RhoGEF mimic EspT
which regulates multiple cell processes including inflam- induces production of IL-8 and IL-1b in macrophages
mation [84]. NleC affects the p38 pathway and plays a through Rac1 [96], whereas the Shigella RhoGEF mimic
significant role in the virulence of C. rodentium in vivo [85]. IpgB2 activates NF-kB via a GEF-H1, RhoA, and NOD1
Moreover, deletion of Tir enhanced the activation of ERK, pathway [97], further strengthening the link between the
JNK, and p38 3 h post-EPEC infection [71]. small Rho GTPases/RhoGEF mimic superfamily and im-
Although it is not clear how nuclear IpaH9.8 from mune responses. Salmonella SipA, a non-RhoGEF mimic,
Shigella flexneri affects MAPK signaling, its ubiquitin enhances mucosal inflammation in the mouse colitis model
ligase activity targets the yeast MAPK Ste7 to interrupt and NF-kB activation in cultured cells in a NOD1/NOD2-
pheromone response signaling by promoting the protea- dependent manner [98]. Given the central role MAPKs and
some-dependent destruction of Ste7 [86]. OspF has phos- NF-kB play in antimicrobial signaling, it is not surprising
phothreonine lyase activity and translocates to the that bacterial pathogens have evolved many mechanisms
nucleus, mediating irreversible dephosphorylation and to affect these pathways.
hence inactivation of MAPKs [87]. This also inhibits down-
stream phosphorylation of histone H3 at Ser10, altering Targeting inflammasomes
chromatin accessibility of the IL-8 promoter to NF-kB in The inflammasome is a key component of cytosolic surveil-
epithelial cells [88]. lance in macrophages where the activated NOD-like recep-
Infection of HeLa cells with the Yersinia DyopETHJ tor (NLR) protein family (e.g., NLRC4 and NLRP3) recruits
mutant expressing either YopE or YopT indicates that the adaptor protein apoptosis-associated speck-like pro-
YopE can strongly inhibit activation of JNK, ERK, and tein containing a caspase recruitment domain (ASC),
prevent production of IL-8, whereas YopT moderately which in turn recruits and activates caspase-1. The acti-
inhibited these responses [77]. YopH inactivates the vated caspase-1 regulates maturation of the proinflamma-
PI3K–AKT cascade, which in macrophages correlated with tory cytokines IL-1b and IL-18 and the rapid inflammatory
the downregulation of mRNA coding for monocyte che- form of cell death, called pyroptosis, characterized by the
moattractant protein 1 (MCP-1) [89]. YopP/J also represses formation of pores in the plasma membrane and activation
436
Review Trends in Microbiology August 2013, Vol. 21, No. 8

Table 1. Summary of the activity of the T3SS effectors discussed


T3SS Putative or Putative or known Homolog Associated function Refs
effector known host enzymatic activity
partner
Attaching and NleE TAB2, TAB3 Methyltransferase OspZ Inhibits NF-kB activation [63–65]
effacing (A/E) and inflammatory
pathogens response
NleB GAPDH Glycosyltransferase SseK Inhibits TRAF2–NF-kB [64,66]
activation
EspI Sec24 – Disrupts secretion by [3,4,6]
(NleA) reducing ER–Golgi
trafficking; alters tight
junctions
NleC RelA Zinc-metalloprotease Cleaves RelA; affects the [84,85]
p38 pathway
NleD JNK and p38 Zinc-metalloprotease Degrades JNK and p38 [84]
and blocks the
translocation of AP-1
EspG Rabs, ARFs, PAKs, Rab-GAP VirA Disrupts secretion; [8–11]
GM130, RACK1 inhibits IL-8 secretion
NleH1/ Bax inhibitor I, Serine threonine OspG Blocks the intrinsic [53,68,69]
NleH2 RPS3 kinase apoptotic pathway; NleH1
blocks the translocation of
RPS3 into the nucleus
EspF Abcf2, actin, – TccP/EspFu Disrupts mitochondria; [33,49–51,59]
profilin, SNX-9, inhibits non-opsonized
N-WASP bacteria internalization
and transcytosis through
M cells; inhibition of PI3K;
increases caspase-9 and -3
activation
Tir SHP-1 Has an ITIM-like Blocks TRAF6–NF-kB [70,71]
sequence pathway and inhibits
cytokine production;
blocks MAPK signaling
Cif – – Induces caspase-3 [60]
cleavage; inhibits the
ubiquitin/proteasome
pathway
EspH DH–PH – Inhibits host RhoGEFs; [46]
domain induces caspase-3
RhoGEFs activation; inhibits
phagocytosis
EspB Myosin – Inhibits myosin/actin [47]
interaction; blocks
phagocytosis
EspJ – – Inhibits [48]
opsonophagocytosis
EspT Rac1, cdc42 RhoGEF mimics RhoGEF Invasion; induces IL-8 and [32,96]
mimics IL-1b production in
macrophages
NleF Tmp21, – May affect ER–Golgi and [7,56]
caspase-4/-5 Golgi–PM trafficking;
and -9 inhibits caspase-4/-5/-9
Shigella VirA Rabs, ARFs Rab-GAP EspG Activates calpain; reduces [55]
autophagic recognition
IpgD PI(4,5)P2, PI phosphatase Activates Akt; stabilizes [54]
MDM2 MDM2 and inhibits p53
proapoptotic signaling
IpaC – – Lyses the Shigella vacuole [21]
IpaB Caspase-1 – SipB Activates caspase-1 and [20,21,105]
pyroptosis; causes
Shigella entry into
epithelial cells and escape
from the phagocytic
vacuole
OspZ – Methyltransferase NleE Blocks NF-kB [64]
translocation
OspG Binds various Kinase NleH Blocks the E2–E3 [72,73]
ubiquitin- ubiquitination complex

437
Review Trends in Microbiology August 2013, Vol. 21, No. 8

Table 1 (Continued )
T3SS Putative or Putative or known Homolog Associated function Refs
effector known host enzymatic activity
partner
conjugating enzymes and alters IkB-a
belonging to the degradation
SCFb–TrCP complex
IpaH9.8 NEMO/IKK, ABIN-1 E3 ubiquitin ligases Dampens NF-kB signaling; [74,75,86]
blocks the yeast MAPK
Ste7
OspI UBC13 Glutamine deamidase Deamidates UBC13 and [76]
blocks the TRAF6–NF-kB
pathway
OspF MAPK Phospho-threonine SpvC Dephosphorylates MAPKs [87,88]
lyase and inhibits the
downstream
phosphorylation of
histone H3 at Ser10,
altering the chromatin
accessibility of the IL-8
promoter to NF-kB
IpgB2 RhoA RhoGEF mimics RhoGEF Activates NF-kB [97]
mimics
IpaJ ARF1 Cysteine protease Cleaves the myristoyl [12]
group from ARF1 and
alters the host secretory
pathway
Yersinia spp. YopJ/P IKKb Serine/threonine AvrA Inhibits NF-kB/MAPK pro- [78,79,
acetyltransferase, survival signaling; induces 106,107]
cysteine protease apoptosis, pyroptosis and
IL-1b secretion; hydrolyzes
ubiquitin and SUMO
YopK/ – – Inhibits ASC-dependent [108]
YopQ inflammasome activation
YopH p130Cas, Fyb, FAK, Tyrosine phosphatase Dephosphorylates p130C [35–39,89]
p85, Lyn, paxillin as required for Yersinia
internalization; inhibits
PI3K–AKT in
macrophages, and MCP-1
expression
YopE Rac1 RhoGAP Inhibits NF-kB activation [77,109]
and IL-8 production;
inactivates Rac1; inhibits
JNK and ERK activation;
inhibits caspase-1
activation
YopO/ Rac1 GDI-like domain Inactivates Rac1 and [43,44]
YpkA blocks phagocytosis
YopM Caspase-1 Inhibits inflammasome [110]
assembly
YopT RhoA, Rac1, Cysteine protease Inactivates RhoA, Rac1, [45,77]
and Cdc42 and Cdc42; moderately
inhibits JNK and ERK
Salmonella spp. SipC Exo70 – Provides extra membrane [13]
to facilitate invasion
SifA SKIP, RhoA-GDP – Excludes lysosomal [16]
hydrolases from SCV
SseF – – Produces Sifs [18,19]
SseG – – Produces Sifs [18,19]
SopB PI(4,5)P2 PI phosphatase Facilitates invasion and [25,57,91]
(SigD) prevents phagolysosome
formation; sustains PI3K–
AKT antiapoptotic
signaling; activates RhoG
and Cdc42; induces NF-kB
and MAPKs
AvrA MKK4, MKK7, IkB-a Acetyltransferase; YopJ/P Inhibits JNK proapoptotic [58,82]
deubiquitinase signaling; inhibits NF-kB
activation and host
immune response
SipB Caspase-1 – IpaB [62,104]

438
Review Trends in Microbiology August 2013, Vol. 21, No. 8

Table 1 (Continued )
T3SS Putative or Putative or known Homolog Associated function Refs
effector known host enzymatic activity
partner
Activates caspase-1 and
pyroptosis; activation of
caspase-2-dependent
apoptosis
SseL IkB-a Deubiquitinase Impairs IkB-a degradation [80]
and NF-kB activation
SspH1 PKN1 – Blocks NF-kB activation [81]
SpvC MAPK Phospho-threonine lyase OspF Blocks MAPK signaling [87]
SopE/SopE2 Rho GTPases RhoGEF RhoGEF mimics Activates NF-kB, ERK, [1,91,92,101]
JNK, and p38 pathways;
induces caspase-1
activation
SipA – – Induces NF-kB activation [98]
in a NOD1/NOD2-
dependent manner
PrgJ – – Is recognized by NLRC4 [102]

of an unidentified endonuclease [99]. Because IL-1b tran- bacterial colonization, multiplication, and evasion of in-
scription is highly regulated by NF-kB, all the described nate immunity and host responses. Here, we have
effectors affecting the NF-kB pathway could affect IL-1b reviewed the mechanisms used by EPEC/EHEC, Yersinia,
expression and hence the pool of IL-1b available for cas- Salmonella, and Shigella to disrupt key cellular signaling
pase-1 cleavage. pathways of early host defense processes (Table 1). It is
S. Typhimurium activates caspase-1 in macrophages in a evident that although they have evolved diverse lifestyles
SPI-1 T3SS-dependent manner [100]. In addition to flagellin within the host (i.e., intracellular and extracellular), these
and SopE [101], which induce caspase-1 activation, PrgJ, pathogens share common strategies to counteract host
which forms the periplasmic rod of the T3SS needle complex, defenses and use specific effectors to mitigate cell-specific
is recognized by NLRC4 [102]. During systemic infection, obstacles. For example, whereas inhibition of apoptosis in
Salmonella spp. downregulate expression of the flagellin, epithelial cells is beneficial to promote colonization, induc-
SopE, and PrgJ, while simultaneously upregulating the tion of pyroptosis in macrophages may facilitate dissemi-
SPI-2 T3SS, which is not detected by NLRC4. The Salmo- nation. Another common theme is the existence of a high
nella T3SS also appears to inhibit pyroptosis in B cells level of synergy between different T3SS effectors that
through downregulation of the NLRC4 inflammasome together inhibit or activate specific signaling pathways,
and inhibition of caspase-1 cleavage, although the effector while at the same time other effectors antagonize the
responsible has not been identified [103]. activity of each other. Accordingly, great care should be
The Shigella T3SS effector IpaB and its Salmonella taken when dissecting the function of individual effectors
homolog SipB induce cell death in macrophages with fea- and their contribution to subversion of host cell signaling.
tures of pyroptosis by binding and activating caspase-1
[104,105]. Similarly, although YopP/J was originally de- Acknowledgments
scribed as inducing apoptosis in macrophages through the We are grateful for the financial support from the Biotechnology and
MAPK and NF-kB pathways [106], a recent paper demon- Biological Sciences Research Council (BBSRC) and the Medical Research
council (MRC).
strated that it induces pyroptosis and IL-1b secretion
during infection through caspase-1 [107]. In contrast to
References
this, YopK/YopQ of Yersinia block pyroptosis by preventing 1 Bulgin, R. et al. (2010) Bacterial guanine nucleotide exchange factors
ASC-dependent inflammasome activation in response to SopE-like and WxxxE effectors. Infect. Immun. 78, 1417–1425
the T3SS, promoting bacterial survival and replication. 2 Faini, M. et al. (2013) Vesicle coats: structure, function, and general
Expression of YopK is insufficient to inhibit YopJ-induced principles of assembly. Trends Cell Biol. 23, 279–288
3 Kim, J. et al. (2007) Biogenesis of g-secretase early in the secretory
pyroptosis suggesting that they act on distinct pathways of
pathway. J. Cell Biol. 179, 951–963
caspase-1 activation [108]. YopE and YopT of Y. enteroco- 4 Thanabalasuriar, A. et al. (2012) Sec24 interaction is essential for
litica negatively regulate caspase-1 by targeting Rac1 localization and virulence-associated function of the bacterial effector
[109], and YopM blocks the assembly of the inflammasome protein NleA. Cell. Microbiol. 14, 1206–1218
by directly targeting caspase-1 [110]. Induction of the 5 Mundy, R. et al. (2004) Distribution of espI among clinical
enterohaemorrhagic and enteropathogenic Escherichia coli isolates.
inflammasome and pyroptosis in macrophages may be a
J. Med. Microbiol. 53, 1145–1149
mechanism by which pathogens preferentially trigger im- 6 Thanabalasuriar, A. et al. (2010) The bacterial virulence factor NleA is
mune cell death, resulting in bacterial dissemination and required for the disruption of intestinal tight junctions by
disruption of host innate immune signaling. enteropathogenic Escherichia coli. Cell. Microbiol. 12, 31–41
7 Olsen, R.L. et al. (2013) The enterohemorrhagic Escherichia coli effector
protein NleF binds mammalian Tmp21. Vet. Microbiol. 64, 164–170
Concluding remarks 8 Selyunin, A.S. et al. (2011) The assembly of a GTPase-kinase
Recent advances have enhanced our understanding of how signalling complex by a bacterial catalytic scaffold. Nature 469,
T3SS effectors manipulate host cell processes enabling 107–111

439
Review Trends in Microbiology August 2013, Vol. 21, No. 8

9 Clements, A. et al. (2011) EspG of enteropathogenic and 34 Goosney, D.L. et al. (1999) Enteropathogenic Escherichia coli inhibits
enterohemorrhagic E. coli binds the Golgi matrix protein GM130 and phagocytosis. Infect. Immun. 67, 490–495
disrupts the Golgi structure and function. Cell. Microbiol. 13, 1429–1439 35 Black, D.S. et al. (1998) Identification of an amino-terminal substrate-
10 Dong, N. et al. (2012) Structurally distinct bacterial TBC-like GAPs binding domain in the Yersinia tyrosine phosphatase that is required
link Arf GTPase to Rab1 inactivation to counteract host defenses. Cell for efficient recognition of focal adhesion targets. Mol. Microbiol. 29,
150, 1029–1041 1263–1274
11 Germane, K.L. and Spiller, B.W. (2011) Structural and functional 36 de la Puerta, M.L. et al. (2009) Characterization of new substrates
studies indicate that the EPEC effector, EspG, directly binds p21- targeted by Yersinia tyrosine phosphatase YopH. PLoS ONE 4, e4431
activated kinase. Biochemistry 50, 917–919 37 Hamid, N. et al. (1999) YopH dephosphorylates Cas and Fyn-binding
12 Burnaevskiy, N. et al. (2013) Proteolytic elimination of N- protein in macrophages. Microb. Pathog. 27, 231–242
myristoylmodifications by the Shigella virulence factor IpaJ. 38 Persson, C. et al. (1997) The PTPase YopH inhibits uptake of Yersinia,
Nature 496, 106–109 tyrosine phosphorylation of p130Cas and FAK, and the associated
13 Nichols, C.D. and Casanova, J.E. (2010) Salmonella-directed accumulation of these proteins in peripheral focal adhesions. EMBO
recruitment of new membrane to invasion foci via the host exocyst J. 16, 2307–2318
complex. Curr. Biol. 20, 1316–1320 39 Weidow, C.L. et al. (2000) CAS/Crk signalling mediates uptake of
14 Meresse, S. et al. (1999) Controlling the maturation of pathogen- Yersinia into human epithelial cells. Cell. Microbiol. 2, 549–560
containing vacuoles: a matter of life and death. Nat. Cell Biol. 1, 40 Von Pawel-Rammingen, U. et al. (2000) GAP activity of the Yersinia
E183–E188 YopE cytotoxin specifically targets the Rho pathway: a mechanism
15 Meresse, S. et al. (1999) The rab7 GTPase controls the maturation of for disruption of actin microfilament structure. Mol. Microbiol. 36,
Salmonella typhimurium-containing vacuoles in HeLa cells. EMBO J. 737–748
18, 4394–4403 41 Andor, A. et al. (2001) YopE of Yersinia, a GAP for Rho GTPases,
16 McGourty, K. et al. (2012) Salmonella inhibits retrograde trafficking selectively modulates Rac-dependent actin structures in endothelial
of mannose-6-phosphate receptors and lysosome function. Science cells. Cell. Microbiol. 3, 301–310
338, 963–967 42 Roppenser, B. et al. (2009) Yersinia enterocolitica differentially
17 Drecktrah, D. et al. (2007) Salmonella trafficking is defined by modulates RhoG activity in host cells. J. Cell Sci. 122, 696–705
continuous dynamic interactions with the endolysosomal system. 43 Prehna, G. et al. (2006) Yersinia virulence depends on mimicry of host
Traffic 8, 212–225 Rho-family nucleotide dissociation inhibitors. Cell 126, 869–880
18 Drecktrah, D. et al. (2008) Dynamic behavior of Salmonella-induced 44 Groves, E. et al. (2010) Sequestering of Rac by the Yersinia effector
membrane tubules in epithelial cells. Traffic 9, 2117–2129 YopO blocks Fcg receptor-mediated phagocytosis. J. Biol. Chem. 285,
19 Rajashekar, R. et al. (2008) Dynamic remodeling of the endosomal 4087–4098
system during formation of Salmonella-induced filaments by 45 Shao, F. et al. (2003) Biochemical characterization of the Yersinia
intracellular Salmonella enterica. Traffic 9, 2100–2116 YopT protease: cleavage site and recognition elements in Rho
20 Blocker, A. et al. (1999) The tripartite type III secreton of Shigella GTPases. Proc. Natl. Acad. Sci. U.S.A. 100, 904–909
flexneri inserts IpaB and IpaC into host membranes. J. Cell Biol. 147, 46 Dong, N. et al. (2010) A bacterial effector targets host DH–PH domain
683–693 RhoGEFs and antagonizes macrophage phagocytosis. EMBO J. 29,
21 High, N. et al. (1992) IpaB of Shigella flexneri causes entry into 1363–1376
epithelial cells and escape from the phagocytic vacuole. EMBO J. 47 Iizumi, Y. et al. (2007) The enteropathogenic E. coli effector EspB
11, 1991–1999 facilitates microvillus effacing and antiphagocytosis by inhibiting
22 Fernandez-Prada, C.M. et al. (2000) Shigella flexneri IpaH(7.8) myosin function. Cell Host Microbe 2, 383–392
facilitates escape of virulent bacteria from the endocytic vacuoles of 48 Marches, O. et al. (2008) EspJ of enteropathogenic and
mouse and human macrophages. Infect. Immun. 68, 3608–3619 enterohaemorrhagic Escherichia coli inhibits opsono-phagocytosis.
23 Mounier, J. et al. (1999) Rho family GTPases control entry of Shigella Cell. Microbiol. 10, 1104–1115
flexneri into epithelial cells but not intracellular motility. J. Cell Sci. 49 Quitard, S. et al. (2006) The enteropathogenic Escherichia coli EspF
112, 2069–2080 effector molecule inhibits PI-3 kinase-mediated uptake independently
24 Zhou, D. and Galan, J. (2001) Salmonella entry into host cells: the of mitochondrial targeting. Cell. Microbiol. 8, 972–981
work in concert of type III secreted effector proteins. Microbes Infect. 50 Peralta-Ramirez, J. et al. (2008) EspF interacts with nucleation-
3, 1293–1298 promoting factors to recruit junctional proteins into pedestals for
25 Marcus, S.L. et al. (2002) Salmonella enterica serovar Typhimurium pedestal maturation and disruption of paracellular permeability.
effector SigD/SopB is membrane-associated and ubiquitinated inside Infect. Immun. 76, 3854–3868
host cells. Cell. Microbiol. 4, 435–446 51 Alto, N.M. et al. (2007) The type III effector EspF coordinates
26 Ramel, D. et al. (2011) Shigella flexneri infection generates the lipid membrane trafficking by the spatiotemporal activation of two
PI5P to alter endocytosis and prevent termination of EGFR signaling. eukaryotic signaling pathways. J. Cell Biol. 178, 1265–1278
Sci. Signal. 4, ra61 52 Taylor, R.C. et al. (2008) Apoptosis: controlled demolition at the
27 Niebuhr, K. et al. (2002) Conversion of PtdIns(4,5)P(2) into PtdIns(5)P cellular level. Nat. Rev. Mol. Cell Biol. 9, 231–241
by the S. flexneri effector IpgD reorganizes host cell morphology. 53 Hemrajani, C. et al. (2010) NleH effectors interact with Bax inhibitor-
EMBO J. 21, 5069–5078 1 to block apoptosis during enteropathogenic Escherichia coli
28 Peiser, L. et al. (2000) Macrophage class A scavenger receptor- infection. Proc. Natl. Acad. Sci. U.S.A. 107, 3129–3134
mediated phagocytosis of Escherichia coli: role of cell 54 Clark, C.S. and Maurelli, A.T. (2007) Shigella flexneri inhibits
heterogeneity, microbial strain, and culture conditions in vitro. staurosporine-induced apoptosis in epithelial cells. Infect. Immun.
Infect. Immun. 68, 1953–1963 75, 2531–2539
29 Anderson, C.L. et al. (1990) Phagocytosis mediated by three distinct Fcg 55 Bergounioux, J. et al. (2012) Calpain activation by the Shigella
receptor classes on human leukocytes. J. Exp. Med. 171, 1333–1345 flexneri effector VirA regulates key steps in the formation and life
30 Ross, G.D. et al. (1992) Macrophage cytoskeleton association with CR3 of the bacterium’s epithelial niche. Cell Host Microbe 11, 240–252
and CR4 regulates receptor mobility and phagocytosis of iC3b- 56 Blasche, S. et al. (2013) The E. coli effector protein NleF is a caspase
opsonized erythrocytes. J. Leukoc. Biol. 51, 109–117 inhibitor. PLoS ONE 8, e58937
31 Isberg, R.R. (1990) Pathways for the penetration of enteroinvasive 57 Knodler, L.A. et al. (2005) The Salmonella effector protein SopB
Yersinia into mammalian cells. Mol. Biol. Med. 7, 73–82 protects epithelial cells from apoptosis by sustained activation of
32 Bulgin, R. et al. (2009) The T3SS effector EspT defines a new category Akt. J. Biol. Chem. 280, 9058–9064
of invasive enteropathogenic E. coli (EPEC) which form intracellular 58 Jones, R.M. et al. (2008) Salmonella AvrA coordinates suppression of
actin pedestals. PLoS Pathog. 5, e1000683 host immune and apoptotic defenses via JNK pathway blockade. Cell
33 Martinez-Argudo, I. et al. (2007) Translocation of enteropathogenic Host Microbe 3, 233–244
Escherichia coli across an in vitro M cell model is regulated by its type 59 Nougayrede, J.P. and Donnenberg, M.S. (2004) Enteropathogenic
III secretion system. Cell. Microbiol. 9, 1538–1546 Escherichia coli EspF is targeted to mitochondria and is required to

440
Review Trends in Microbiology August 2013, Vol. 21, No. 8

initiate the mitochondrial death pathway. Cell. Microbiol. 6, 85 Sham, H.P. et al. (2011) Attaching and effacing bacterial effector NleC
1097–1111 suppresses epithelial inflammatory responses by inhibiting NF-kB
60 Samba-Louaka, A. et al. (2009) The enteropathogenic Escherichia coli and p38 mitogen-activated protein kinase activation. Infect. Immun.
effector Cif induces delayed apoptosis in epithelial cells. Infect. 79, 3552–3562
Immun. 77, 5471–5477 86 Rohde, J.R. et al. (2007) Type III secretion effectors of the IpaH family
61 Wong, A.R. et al. (2012) The interplay between the Escherichia coli are E3 ubiquitin ligases. Cell Host Microbe 1, 77–83
Rho guanine nucleotide exchange factor effectors and the mammalian 87 Li, H. et al. (2007) The phosphothreonine lyase activity of a bacterial
RhoGEF inhibitor EspH. mBio 3, e00250–11, (http://dx.doi.org/ type III effector family. Science 315, 1000–1003
10.1128/mBio.00250-11) 88 Arbibe, L. et al. (2007) An injected bacterial effector targets chromatin
62 Jesenberger, V. et al. (2000) Salmonella-induced caspase-2 activation access for transcription factor NF-kB to alter transcription of host
in macrophages: a novel mechanism in pathogen-mediated apoptosis. genes involved in immune responses. Nat. Immunol. 8, 47–56
J. Exp. Med. 192, 1035–1046 89 Sauvonnet, N. et al. (2002) YopH prevents monocyte chemoattractant
63 Nadler, C. et al. (2010) The type III secretion effector NleE inhibits protein 1 expression in macrophages and T-cell proliferation through
NF-kB activation. PLoS Pathog. 6, e1000743 inactivation of the phosphatidylinositol 3-kinase pathway. Mol.
64 Newton, H.J. et al. (2010) The type III effectors NleE and NleB from Microbiol. 45, 805–815
enteropathogenic E. coli and OspZ from Shigella block nuclear 90 Orth, K. et al. (1999) Inhibition of the mitogen-activated protein kinase
translocation of NF-kB p65. PLoS Pathog. 6, e1000898 kinase superfamily by a Yersinia effector. Science 285, 1920–1923
65 Vossenkamper, A. et al. (2010) Inhibition of NF-kB signaling in 91 Bruno, V.M. et al. (2009) Salmonella Typhimurium type III secretion
human dendritic cells by the enteropathogenic Escherichia coli effectors stimulate innate immune responses in cultured epithelial
effector protein NleE. J. Immunol. 185, 4118–4127 cells. PLoS Pathog. 5, e1000538
66 Gao, X. et al. (2013) NleB, a bacterial effector with glycosyltransferase 92 Keestra, A.M. et al. (2013) Manipulation of small Rho GTPases is a
activity, targets GAPDH function to inhibit NF-kB activation. Cell pathogen-induced process detected by NOD1. Nature 496, 233–237
Host Microbe 13, 87–99 93 Stecher, B. et al. (2007) Salmonella enterica serovar typhimurium
67 Yen, H. et al. (2010) NleC, a type III secretion protease, compromises exploits inflammation to compete with the intestinal microbiota.
NF-kB activation by targeting p65/RelA. PLoS Pathog. 6, e1001231 PLoS Biol. 5, 2177–2189
68 Pham, T.H. et al. (2012) Functional differences and interactions 94 Lopez, C.A. et al. (2012) Phage-mediatedacquisition of a type III secreted
between the Escherichia coli type III secretion system effectors effector protein boosts growth of salmonella by nitrate respiration.
NleH1 and NleH2. Infect. Immun. 80, 2133–2140 MBio 3, e00143–12, (http://dx.doi.org/10.1128/mBio.00143-12)
69 Gao, X. et al. (2009) Bacterial effector binding to ribosomal protein s3 95 Winter, S.E. et al. (2010) Gut inflammation provides a respiratory
subverts NF-kB function. PLoS Pathog. 5, e1000708 electron acceptor for Salmonella. Nature 467, 426–429
70 Ruchaud-Sparagano, M.H. et al. (2011) The enteropathogenic E. coli 96 Raymond, B. et al. (2011) The WxxxE effector EspT triggers
(EPEC) Tir effector inhibits NF-kB activity by targeting TNFa expression of immune mediators in an Erk/JNK and NF-kB-
receptor-associated factors. PLoS Pathog. 7, e1002414 dependent manner. Cell. Microbiol. 13, 1881–1893
71 Yan, D. et al. (2012) Inhibition of TLR signaling by a bacterial protein 97 Fukazawa, A. et al. (2008) GEF-H1 mediated control of NOD1
containing immunoreceptor tyrosine-based inhibitory motifs. Nat. dependent NF-kB activation by Shigella effectors. PLoS Pathog. 4,
Immunol. 13, 1063–1071 e1000228
72 Kim, D.W. et al. (2005) The Shigella flexneri effector OspG interferes 98 Keestra, A.M. et al. (2011) A Salmonella virulence factor activates the
with innate immune responses by targeting ubiquitin-conjugating NOD1/NOD2 signaling pathway. mBio 2, e00266–11, (http://
enzymes. Proc. Natl. Acad. Sci. U.S.A. 102, 14046–14051 dx.doi.org/10.1128/mBio.00266-11)
73 Zhou, Y. et al. (2013) The Shigella type three secretion system effector 99 Bergsbaken, T. et al. (2009) Pyroptosis: host cell death and
OspG directly and specifically binds to host ubiquitin for activation. inflammation. Nat. Rev. Microbiol. 7, 99–109
PLoS ONE 8, e57558 100 Mariathasan, S. et al. (2004) Differential activation of the
74 Singer, A.U. et al. (2008) Structure of the Shigella T3SS effector IpaH inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430,
defines a new class of E3 ubiquitin ligases. Nat. Struct. Mol. Biol. 15, 213–218
1293–1301 101 Muller, A.J. et al. (2009) The S. Typhimurium effector SopE induces
75 Ashida, H. et al. (2010) A bacterial E3 ubiquitin ligase IpaH9.8 targets caspase-1 activation in stromal cells to initiate gut inflammation. Cell
NEMO/IKKg to dampen the host NF-kB-mediated inflammatory Host Microbe 6, 125–136
response. Nat. Cell Biol. 12, 66–73 102 Miao, E.A. et al. (2010) Innate immune detection of the type III
76 Sanada, T. et al. (2012) The Shigella flexneri effector OspI deamidates secretion apparatus through the NLRC4 inflammasome. Proc.
UBC13 to dampen the inflammatory response. Nature 483, 623–626 Natl. Acad. Sci. U.S.A. 107, 3076–3080
77 Viboud, G.I. et al. (2006) Comparison of YopE and YopT activities in 103 Perez-Lopez, A. et al. (2013) Salmonella downregulates Nod-like
counteracting host signalling responses to Yersinia receptor family CARD domain containing protein 4 expression to
pseudotuberculosis infection. Cell. Microbiol. 8, 1504–1515 promote its survival in B cells by preventing inflammasome
78 Orth, K. et al. (2000) Disruption of signaling by Yersinia effector YopJ, activation and cell death. J. Immunol. 190, 1201–1209
a ubiquitin-like protein protease. Science 290, 1594–1597 104 Hersh, D. et al. (1999) The Salmonella invasin SipB induces
79 Paquette, N. et al. (2012) Serine/threonine acetylation of TGFb- macrophage apoptosis by binding to caspase-1. Proc. Natl. Acad.
activated kinase (TAK1) by Yersinia pestis YopJ inhibits innate Sci. U.S.A. 96, 2396–2401
immune signaling. Proc. Natl. Acad. Sci. U.S.A. 109, 12710–12715 105 Hilbi, H. et al. (1998) Shigella-induced apoptosis is dependent on
80 Le Negrate, G. et al. (2008) Salmonella secreted factor L caspase-1 which binds to IpaB. J. Biol. Chem. 273, 32895–32900
deubiquitinase of Salmonella typhimurium inhibits NF-kB, 106 Zhang, Y. et al. (2005) Inhibition of MAPK and NF-kB pathways is
suppresses IkBa ubiquitination and modulates innate immune necessary for rapid apoptosis in macrophages infected with Yersinia.
responses. J. Immunol. 180, 5045–5056 J. Immunol. 174, 7939–7949
81 Haraga, A. and Miller, S.I. (2003) A Salmonella enterica serovar 107 Philip, N.H. and Brodsky, I.E. (2012) Cell death programs in Yersinia
typhimurium translocated leucine-rich repeat effector protein immunity and pathogenesis. Front. Cell. Infect. Microbiol. 2, 149
inhibits NF-kB-dependent gene expression. Infect. Immun. 71, 108 Brodsky, I.E. et al. (2010) A Yersinia effector protein promotes
4052–4058 virulence by preventing inflammasome recognition of the type III
82 Collier-Hyams, L.S. et al. (2002) Cutting edge: Salmonella AvrA secretion system. Cell Host Microbe 7, 376–387
effector inhibits the key proinflammatory, anti-apoptotic NF-kB 109 Schotte, P. et al. (2004) Targeting Rac1 by the Yersinia effector protein
pathway. J. Immunol. 169, 2846–2850 YopE inhibits caspase-1-mediated maturation and release of
83 Zhang, Y.L. and Dong, C. (2005) MAP kinases in immune responses. interleukin-1b. J. Biol. Chem. 279, 25134–25142
Cell. Mol. Immunol. 2, 20–27 110 LaRock, C.N. and Cookson, B.T. (2012) The Yersinia virulence effector
84 Baruch, K. et al. (2011) Metalloprotease type III effectors that YopM binds caspase-1 to arrest inflammasome assembly and
specifically cleave JNK and NF-kB. EMBO J. 30, 221–231 processing. Cell Host Microbe 12, 799–805

441

You might also like