You are on page 1of 10

Microbes and Infection 13 (2011) 992e1001

www.elsevier.com/locate/micinf

Review

Vibrio parahaemolyticus cell biology and pathogenicity determinants


Christopher A. Broberg1, Thomas J. Calder1, Kim Orth*
Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas TX 75390-9148, USA
Received 26 May 2011; accepted 17 June 2011
Available online 7 July 2011

Abstract

Vibrio parahaemolyticus is a significant cause of gastroenteritis worldwide. Characterization of this pathogen has revealed a unique repertoire
of virulence factors that allow for colonization of the human host and disease. The following describes the known pathogenicity determinants
while establishing the need for continued research.
Ó 2011 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.

Keywords: Vibrio parahaemolyticus; Gastroenteritis; Hemolysin; Type III secretion system; Pathogenesis

1. Introduction Twenty of these people ultimately died. While the source of the
illness was believed to be shirasu, a small partially dried sardine,
Vibrio parahaemolyticus is a Gram-negative, halophilic the etiologic agent was unknown. An intense investigation began
bacterium that thrives in warm climates within marine or estuarine with filtered homogenates from shirasu passed through a guinea
environments. It is commonly found free swimming, attached to pig model to eliminate poisoning by chemicals as a possible
underwater surfaces, or commensally associated with different cause of the disease. When the test animals developed peritonitis,
shellfish species [1]. While some strains of V. parahaemolyticus the homogenates were inoculated to various growth media.
are strictly environmental, many strains are pathogenic to humans. Along with other known bacterial organisms, two species of
Virulent strains can cause distinct diseases, including wound unidentified Gram-negative rods were also isolated. Since these
infections, septicemia, or more commonly acute gastroenteritis organisms could not be separated by isolation streaking, they
which is acquired through the consumption of raw or undercooked were inoculated intraperitoneally into mice. When disease
seafood, especially shellfish. Since its discovery in 1950, V. par- symptoms developed several hours later, ascitic fluid was
ahaemolyticus has become a leading cause of seafood-derived collected and streaked onto blood agar. One of the organisms was
food poisoning throughout the world [2,3]. The global dissemi- identified as Proteus morganii. The other organism was previ-
nation of this pathogen underscores the importance of under- ously unclassified, and was named Pasteurella parahaemolytica.
standing its many virulence factors and their effects on the human Further testing showed that this organism alone was pathogenic
host. This review will focus on the current knowledge of different to mice and could be isolated from the stool samples of afflicted
V. parahaemolyticus strains and their wide repertoire of toxins and individuals from the original outbreak [4e7]. P. para-
type 3 secretion system effectors. haemolytica was reclassified as V. parahaemolyticus when it was
found to grow preferentially on high-salt media [4].
2. V. parahaemolyticus V. parahaemolyticus is a Gram-negative, halophilic, meso-
philic, small rod that may have a single curve to its shape [3,8]. It
In the fall of 1950 in the southern suburbs of Osaka, Japan, an exists as either a swimmer cell with a single polar flagellum, or
outbreak of acute gastroenteritis sickened 272 individuals. a swarmer cell covered in lateral flagella (Fig. 1) [1]. Depending
on environmental conditions, V. parahaemolyticus can produce
* Corresponding author. Tel.: þ1 214 648 1685; fax: þ1 214 648 1488. a capsule, with over 70 different K antigens detected in various
E-mail address: kim.orth@utsouthwestern.edu (K. Orth). strains [7].
1
These authors contributed equally to the preparation of this manuscript.

1286-4579/$ - see front matter Ó 2011 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.
doi:10.1016/j.micinf.2011.06.013
C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001 993

V. parahaemolyticus is found free living in brackish and undercooked seafood including fish, crab and other crustaceans,
estuarine waters, and requires salinity for survival [9]. In and mollusks. Since V. parahaemolyticus is sensitive to heat,
winter months when water temperatures are unfavorable, V. cooking generally kills the bacterium rendering contaminated
parahaemolyticus may be undetectable. It has been proposed food safe to eat. In the United States, the first case of gastro-
that the organism survives in marine sediment, and is rein- enteritis caused by V. parahaemolyticus occurred in 1971 in
troduced to the water column when temperatures rise [3]. Maryland and was associated with contaminated crabmeat. V.
Infaunal burrows have been demonstrated to contain high parahaemolyticus is the primary cause of US Vibrio-associated
levels of V. parahaemolyticus, likely acting as a source for foodborne illness. The majority of these infections are a result of
these organisms to be distributed through an estuary envi- consuming Vibrio-contaminated raw oysters [2].
ronment during favorable conditions [10]. An increase in non-cholera Vibrio infections that began in
In locations where water temperatures do not drop below the mid 1990s has been associated with the detection of a new
15  C, V. parahaemolyticus may be detected year round [3] with clonal group that includes three new serotypes: O3:K6,
the number of organisms detected in water, sediment and oysters O4:K68, and O1:K untypeable [6]. Since 1996, the most
increasing as water temperatures rise [11]. Due to filter feeding, common serotype of V. parahaemolyticus has been O3:K6.
oysters may have a concentration of V. parahaemolyticus up to While non-pandemic variants for this serotype were detected
100-fold higher than surrounding waters. Up to 100% of oysters in Japan as early as 1983, the first reported illness caused by
may be contaminated with V. parahaemolyticus and/or Vibrio this strain was first observed in a Japanese traveler returning
vulnificus during summer months, which increases the chances from Indonesia in 1995. An outbreak of diarrheal disease then
of infection [6]. V. parahaemolyticus is disseminated throughout occurred in Calcutta, India in February of 1996 with 50e80%
the world, and recently, due to warming ocean temperatures, has of the isolates confirmed as O3:K6 [7]. V. parahaemolyticus
been detected in coastal waters as far north as the southern coast has now been detected in North and South America, Europe,
of Alaska [8,12,13]. Africa, and Asia with outbreaks of disease occurring world-
In Japan, V. parahaemolyticus accounts for 20e30% of all wide [7]. To date, at least 12 pathogenic serotypes of V. par-
food poisoning cases and is the leading cause of foodborne ahaemolyticus have been identified [14]. Many strains have
illness [3]. The high rate of infection is attributed to the overall been sequenced including the O3:K6 serotype strains RimD
high seafood diet as well as the common practice of eating 2210633 and AQ3810; the O4:K12 strain Vp10329; and the
seafood raw. Gastroenteritis caused by V. parahaemolyticus is O4:K68 serotype strains AN-5034, K5030 and Peru-466
generally associated with the consumption of raw or (http://www.ncbi.nlm.nih.gov/genomes/lproks.cgi) [14e16].

Fig. 1. Virulence Associated Factors of V. parahaemolyticus. A single flagellum at one pole of the bacterium is sheathed to extend the bacterial membrane and is
required for swimming motility in liquid environments. During growth on semi-solid surfaces, flagella are produced along the lateral side of the bacterium, which
aid in swarming motility. MAM7 is a multivalent adhesion protein that binds to fibronectin and phosphatidic acid, and it is required for the initial attachment to host
cells. As shown simplistically, V. parahaemolyticus can utilize the siderophores vibrioferrin, ferrichrome, and aerobactin, along with heme, to scavenge iron from
the environment. These iron transporters are internalized by different membrane receptors on the outer membrane of the bacteria and transported to the cytoplasm
by different ABC complexes.
994 C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001

Numerous clinical and environmental strains without occurring on chromosomes 1 and 2, respectively) have been
a sequence have been utilized to understand the biology of V. identified in RimD 2210633 [15,16]. Type VI secretion is
parahaemolyticus [2,17,18]. The RimD 2210633 strain was present in many Gram-negative pathogens, and may be
the first fully sequenced and annotated genome [15] and is involved in the modulation of eukaryotic signaling [22], but
therefore the focus of most studies. currently it has not been investigated in V. parahaemolyticus.
Additionally, activation of these genes is not altered under
3. Disease T3SS-inducing conditions meant to mimic the environment
encountered within a human host [23]. As such, these systems
Infection with V. parahaemolyticus can cause three distinct in V. parahaemolyticus will not be discussed further here.
medical conditions: gastroenteritis, wound infections, and
septicemia. Acute gastroenteritis presents with abdominal 4.1. Flagella, capsule, and biofilm regulation
cramping, diarrhea, nausea, vomiting, low-grade fever, head-
ache, and occasional bloody diarrhea different than that seen in A common cellular factor associated with many intestinal
other enteric infections. Infection occurs 4e96 h after pathogens is the presence of one or more flagella. V. para-
consumption of contaminated food, and lasts up to three days. haemolyticus possesses two different types of flagella with
The illness is self-resolving in immunocompetent individuals distinct functions. A polar flagellum is constitutively
and can be sufficiently treated with oral rehydration alone [2,7]. expressed and is used for swimming. The whip of the
Wound infection is common among fishermen and is flagellum is made of six different flagellin proteins and is
generally acquired when small wounds occur in or around sheathed, which may aid in attachment. The energy to rotate
seawater [19]. This form of V. parahaemolyticus infection is this flagellum is provided by a sodium motive force, which is
sometimes limited to cellulitis, but may progress to necro- advantageous in salt water with an average pH of 8. V. para-
tizing fasciitis, an uncommon infection of soft tissues char- haemolyticus is capable of swimming at speeds up to 60 mm/s
acterized by a rapid spread of bacteria with associated when expressing this single flagellum [1].
inflammation and necrosis of tissues. A decrease in flagellar rotation speed as a consequence of
Septicemia occurs when V. parahaemolyticus enters the blood increased viscosity of the growth environment, or growth
stream of the patient and is disseminated throughout the body. under iron-limiting conditions, induces a switch to a swarmer
Systemic immune activation leads to inflammation and increased cell type. This entails the production of a number of non-
vascular permeability. This can result in hypovolemic shock, sheathed peritrichous flagella which allows the bacteria to
multisystem organ failure and death. The subpopulation of swarm over solid or semi-solid substrates. These flagella are
patients most at risk for septicemia includes those with underlying different than the single polar flagellum in that they are
medical conditions including liver disease, diabetes, cancer, and unsheathed, made from a single flagellin protein, and are
recent gastric surgery [2]. Immunocompromised individuals and powered by the proton motive force [1].
those with liver failure due to liver cirrhosis or hepatitis virus The switch from a swimmer to a swarmer cell is highly
infection seem to be at greatest risk of septicemia [19,20] regulated. The activation of OpaR, a Vibrio harveyii LuxR
homolog and the regulator of capsule production, blocks
4. Cellular factors associated with V. parahaemolyticus production of the lateral flagella (laf ) genes, which are neces-
pathogenicity sary for lateral flagella production [24]. Inactivation of OpaR
switches the cell from opaque (OP) to the translucent (TR) form
Genomic analysis has demonstrated that a common Vibrio that lacks a capsule and can swarm effectively over surfaces or
progenitor gave rise to V. parahaemolyticus, Vibrio cholerae, through viscous liquids. ScrG and ScrC, both GGDEF and EAL-
and the other Vibrio species. The acquisition of a Type III domain containing proteins, regulate c-di-GMP levels in the
Secretion System (T3SS) similar to that found in Yersinia cell. A decrease in c-di-GMP levels leads to lateral flagella
species and herein referred to as T3SS1, was the basis of a V. synthesis and swarming. ScrG can also block CpsA synthesis,
parahaemolyticus ancestor. The acquisition by some strains of which is necessary for capsule synthesis, as well as decrease
a second Type III Secretion System (T3SS2), and thermostable biofilm formation and adherence to surfaces [25,26]. A decrease
direct hemolysin (TDH) and TDH related hemolysin (TRH) in c-di-GMP levels may also enhance expression of other viru-
genes has led to a number of V. parahaemolyticus species with lence factors, including T3SS proteins [27].
varying degrees of pathogenicity. This evolution is separate
from that of V. cholerae, which also acquired T3SS2 as well as 4.2. Adhesion to host cells
the phage-encoded cholera toxin (CTX) in some strains, but
does not possess T3SS1 [21]. In addition to Type III Secretion During infection, a variety of bacterial adhesion factors are
and TDH genes, V. parahaemolyticus possesses flagella for expressed to provide the contact with the host cell necessary for
swimming and swarming, as well as the ability to produce secretion of effector and toxin proteins. In a recent study, an outer
a capsule. These are both factors that likely aid in environ- membrane protein from V. parahaemolyticus was described that
mental survival as well as colonization of the human host. was necessary and sufficient for initial contact of this pathogen
Gene loci for two separate Type VI Secretion Systems with multiple cultured host cell lines. This multivalent adhesion
(VP1386-VP1420, T6SS1 and VPA1030-VPA1043, T6SS2, molecule (MAM) consists of six (MAM6) or seven (MAM7)
C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001 995

mammalian cell entry (mce) domains, and has homologs encoded reproducibility of the KP test is dependent on pH, media
in a number of Gram-negative animal pathogens. salinity, and erythrocyte type. As such, identification of
This work demonstrated that MAM7 bound to both fibro- pathogenic serovars by this method is not always accurate.
nectin and phosphatidic acid, and that blocking the binding of Identification of the tdh gene in samples has been shown to
either of these substrates could prevent adhesion of MAM7 to more accurately predict virulence, as it is a genetic test rather
host cells. Heterologous expression of MAM7 was sufficient than a phenotypic test [34]. The tdh gene is encoded and co-
for attachment of a non-pathogenic E. coli strain to host cells, regulated with T3SS2 genes [23]. The identification of TDH
which could in turn block attachment and attenuate cytotox- may actually serve to identify V. parahaemolyticus strains with
icity of V. parahaemolyticus and a number of other MAM7- T3SS2, the expression of which may be a significant factor in
expressing Gram-negative bacterial pathogens. Furthermore, determining if a serovar can cause pandemic outbreaks of
MAM7 was necessary for attachment early in the infection, as disease.
well as for T3SS-mediated cell death in some cell types. The V. parahaemolyticus RimD 2210633 possesses two copies
characterization of MAM7 provides insight into the initial of the TDH toxin, vpa1314 (tdhA) and vpa1378 (tdhS ) [15].
events of bacterial and host cell interactions, and provides TDH is a reversible amyloid toxin [35] that has been shown to
a potential therapeutic target for not only V. parahaemolyticus, associate with cholesterol and sphingolipid-enriched lipid
but many other Gram-negative pathogens as well [28]. rafts. Disruption of these lipid microdomains abrogated cyto-
toxicity in nucleated cells, but not hemolytic activity against
4.3. Iron acquisition erythrocytes, indicating two potential activities for this toxin
[36]. The x-ray crystallographic structure of TDH showed that
Iron is an essential element for all organisms but usable this protein forms a homotetramer with a central pore 23 Å in
forms of iron are scarcely found in most environments. diameter. This relatively large pore size helps explain previ-
Therefore, V. parahaemolyticus, and other bacteria utilize ously observed low ion selectivity, allowing both water and
several methods of iron acquisition to obtain this element from ions to flow through a membrane with little impedance [37].
the environment (Fig. 1). In humans, iron is associated with This alteration in ion flux from affected cells in the intestine
different molecular complexes such as transferrin, lactoferrin, may be the mechanism for the diarrhea observed during
and hemoglobin. During infection, bacteria utilize iron infection [38]. While TDH was considered to be a major
chelators known as siderophores to scavenge iron [29]. V. virulence factor for V. parahaemolyticus pathogenesis, dele-
parahaemolyticus produces a siderophore known as vibrio- tion of both tdhA and tdhS did not affect cytotoxicity towards
ferrin which is believed to be synthesized and secreted by cultured Hela cells and only showed partial fluid accumulation
proteins from the pvsABCDE operon. For vibrioferrin uptake, in a rabbit ileal loop model [39]. This indicates the existence
the bacteria utilize a siderophore membrane receptor, termed of other virulence factors [40e42]. The cell death of cultured
PvuA, which is coupled to an inner membrane ATP-binding cells infected with TDH-deficient strains was later correlated
cassette (ABC). The ABC transporter system is comprised with functional T3SSs [39].
of proteins from the pvuBCDE gene cluster and is required for TRH is predicted to act in a similar manner to lyse cells
transporting the siderophore across the inner membrane [30]. based upon high sequence homology (68%) between the trh
V. parahaemolyticus can also acquire ferric-bound side- and tdh genes. In a recent study, TRH and TDH were shown to
rophores produced by different fungal and bacterial species or cause similar levels of hemolysis in vitro and size exclusion
through the uptake of heme. Ferrichrome and aerobactin are analysis revealed that TRH also exists in a tetramer complex
two siderophores produced by other organisms that can be [43]. Future studies will unravel the similarities and differ-
retrieved by V. parahaemolyticus with the membrane receptors ences of these two toxins. Another toxin, thermolabile
FhuA and IutA, respectively. Both receptors are believed to be hemolysin (TLH), is found in all strains of V. para-
coupled to the FhuCDB ABC complex [31]. While it is known haemolyticus, but very little is known about its function. The
that V. parahaemolyticus can utilize heme as an iron source, tlh gene was found to be strongly upregulated in a genomic
little is known about the proteins involved in this process. V. screen performed under conditions meant to mimic the intes-
parahaemolyticus was found to contain a homologue of the V. tinal environment of the human host [23]. Therefore, this gene
cholerae heme membrane receptor hutA, but its function may be equally important as the tdh and trh genes in the
remains uncharacterized [32]. process of human infection. Table 1 lists the sequenced strains
of V. parahaemolyticus and the presence of tdh and trh genes,
4.4. Toxins as well as T3SSs.

Nearly all V. parahaemolyticus strains isolated from clinical 4.5. The Type III secretion systems of V.
samples possess b-hemolytic activity attributed to TDH or parahaemolyticus
TRH (Fig. 2). These isolates are able to lyse human erythro-
cytes when plated on a high-salt media called Wagatsuma The T3SS is a bacterial organelle evolved to deliver
agar, a process termed the Kanagawa phenomenon (KP) [33]. proteins, termed effectors, directly into the cytoplasm of
The KP test is commonly used to identify pathogenic V. par- a eukaryotic cell [44]. Made up of 20e30 proteins, the
ahaemolyticus in seafood as well as patient samples. The secretion apparatus consists of a basal body that spans both the
996 C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001

Fig. 2. Known toxins and type 3 secreted effectors of V. parahaemolyticus. The TDH and TLH toxins are secreted from the bacteria and form tetrameric pore
complexes in the host membrane. These pores allow ions to flow freely across the host membrane which leads to hemolysis or cytotoxicity (left panel). V.
parahaemolyticus translocates T3SS1 effectors (VopQ, VopR, VopS, and VPA0450) into host cells to cause cytotoxicity in different cell types such as macrophages
and HeLa cells (middle panel). T3SS2 effectors (VopA, VopC, VopL, and VopT) are translocated into host cells to cause cytotoxicity of colon epithelial cells or
enterotoxicity within the host (right panel).

inner and outer bacterial membranes, a needle that acts as only its lifestyle, but also the disease that it causes. There are
a conduit between the bacterial and eukaryotic cells, and over 100 different characterized effector proteins [47]. While
a translocon pore that is inserted into the eukaryotic cell their activities and targets vary, effectors identified thus far
membrane [45]. Structurally, the apparatus bears resemblance tend to manipulate a limited set of eukaryotic systems.
to a flagellar system from which it may have evolved. Some Common targets of T3SS effectors include the actin cyto-
secretion apparatus proteins have homology to flagellar export skeleton, innate immune signaling, and autophagy. These
proteins, with core transmembrane proteins showing the systems can be up- or down-regulated depending on the
highest level of conservation [46]. specific needs of the pathogen [40,48e52].
The T3SS allows bacteria to translocate effectors from their Translocation occurs in an ATP-dependent manner. Effec-
cytoplasm directly into the host cytoplasm, or to the cyto- tors, which are generally bound to chaperones in a quiescent,
plasmic face of the host cell membrane without release of the partially folded state, are unfolded and threaded through the
effectors to the extracellular space [47]. The specific needle. The chaperone remains in the bacterial cytoplasm
complement of effectors within a pathogen determines not [44,53,54]. Upon entering the host cell cytoplasm, the effec-
tors refold into an active state where they manipulate
eukaryotic signaling cascades to facilitate infection and
Table 1
Occurrence of virulence factors in sequenced pandemic strains.
disrupt the host immune response [55].
Genomic sequencing of V. parahaemolyticus RimD
Strains TDH TRH T3SS1 T3SS2 Ref.
2210633 revealed two pathogenicity islands with one on each
O3:K6 of the two chromosomes [15] (Table 1). The first island
RimD 2210633 D e D D [15]
AQ3810 D e D D [16]
encodes T3SS1 and some of its cognate effectors [56,57]. The
O4:K12 second island, termed Vp-PAI and now also referred to as
Vp10329 D D D D [14] VPaI-7, encodes T3SS2, its known effectors, as well as the tdh
O4:K68 genes (Fig. 2). Both islands have a number of uncharacterized
a
AN-5034 D e D D
a hypothetical proteins [15,23,56,58]. Based on G þ C content,
K5030 D e D D
Peru-466 D e D D a T3SS1 was ancestrally acquired, while T3SS2 was obtained
a through a relatively recent lateral gene transfer [15]. The
http://www.ncbi.nlm.nih.gov/genomes/lproks.cgi e accessed 05/23/2011.
presence of two T3SS2 gene clusters (T3SS2a and T3SS2b) in
C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001 997

different V. parahaemolyticus strains indicates this acquisition While the regulation of T3SS1 is just now being elucidated,
has occurred at least twice [21]. the characterization of three T3SS1 effectors, VopQ, VopS and
Several V. parahaemolyticus strains were created from VPA0450, support an infection model that involves the
RimD 2210633 to enable the study of each T3SS and the induction of autophagy, followed by membrane blebbing, cell
characterization of these hypothetical proteins. The POR1 rounding, and then cell lysis. Rapid induction of autophagy by
strain maintains both functional T3SSs, but has tdhAS VopQ causes the target cell to digest itself and prevents
deleted. POR2 is constructed from the POR1 background, phagocytosis of the infecting bacteria. Detachment of the
and has a deletion of vcrD1, an inner membrane structural plasma membrane from the actin cytoskeleton by VPA0450
ring for T3SS1, which prevents the formation of the T3SS1 destabilizes the cell. The collapse of the actin cytoskeleton by
needle. As a result, POR2 only secretes effectors from VopS leads to cell rounding and shrinkage (Table 3). Finally,
T3SS2. POR3 is similar to POR2, but has a deletion of vcrD2 cells lyse and release their contents [62,63].
instead of vcrD1, and can only secrete effectors from T3SS1 An in silico screen for chaperone proteins previously
(Table 2) [42]. identified four potential T3SS1 effectors. VP1680, VP1683,
and VP1686, now referred to as VopQ, VopR and VopS,
respectively, are located within the T3SS1 gene locus.
4.5.1. T3SS1 VPA0450 is unlinked to T3SS1, and is located on the second
T3SS1 is present in all V. parahaemolyticus strains and is chromosome [57]. Secretion of VopQ, VopS, and VPA0450
a defining characteristic of this species [21]. This system was confirmed by a secretion assay using POR1 grown under
causes cytotoxicity in cultured human cells, but does not inducing conditions [52,56,62,63]. VopR was not detected as it
appear to contribute to enterotoxicity during infection as has a pI outside the range of the 2-D gel used to separate the
determined by a rabbit ileal loop model [40e42,48]. T3SS1 is secreted proteins prior to mass spectrometer analysis [56]. The
similar to the Yersinia ysc T3SS based on the number of genes, target and mechanism of action for VopR (VP1683) has not
gene identity, and gene order [15,56,59]. T3SS1 can be been determined.
induced by growing liquid cultures at 37  C in low calcium Burdette et al., showed in previous work that VopQ
media, which is a similar condition used to induce the Yersinia (VP1680) induced PI3-kinase independent autophagy upon
secretion system. infection with POR3 or transfection of vopQ into Hela cells
Transcription of the T3SS1 genes is regulated by three [48]. Microinjection of recombinant VopQ into Hela cells
interacting proteins (ExsC, ExsD, and ExsE) that control the induced autophagy in less than 30 min as measured by the
activity of ExsA, a member of the AraC family of transcrip- conversion of LC3-I to LC3eII and the formation of LC3-GFP
tional activators. Under non-inducing conditions, ExsA is punctae [62]. In addition to inducing autophagy, VopQ was
bound to ExsD, an anti-activator, and rendered inactive. ExsE, able to block phagocytosis of V. parahaemolyticus by RAW
a substrate for T3SS1, is bound to its chaperone ExsC, which 264.7 macrophages, possibly through the sequestration of
is an anti-anti-activator of the system. When low calcium necessary membrane components. VopQ was necessary for
conditions are encountered ExsE is secreted and causes the rapid host cell lysis during infection as cells infected with
release of ExsC, which binds to ExsD. By sequestering ExsD, a DvopQ strain of V. parahaemolyticus lysed 3 h slower than
ExsA is released and is able to activate transcription of T3SS1 those infected with a wild type strain [48]. Recent studies have
genes. This regulatory system requires low-level expression of identified VopQ as an activator of the JNK, p38 and ERK
T3SS1 genes to allow for the initial secretion of ExsE [60]. MAPK pathways in human intestinal epithelial cell cultures.
This may be accomplished through leaky regulation of the MAPK activation resulted in the secretion of IL-8 and was
T3SS1 genes, or additional regulatory systems that allow for necessary for full cytotoxicity [64,65]. The target of VopQ and
some expression under inhibitory conditions. Preliminary the mechanism for this activation were not determined, which
evidence indicates the Heat-stable Nucleoid Structuring leaves the question of whether or not MAPK activation is the
protein (HeNS) may negatively regulate T3SS1. HeNS is intended purpose of VopQ or an off-target effect resulting from
commonly found in Gram-negative bacteria and is known for unregulated induction of autophagy. The molecular target and
genome-wide repression of protein expression by binding to mechanism of action for VopQ are still under investigation.
curved DNA normally found at active sites of transcription VopS (VP1686) is responsible for the rounding of cells and
[61]. It is not currently understood how this protein fits into the cytoplasmic dispersion of actin seen during infection of Hela
regulatory cascade to modulate T3SS1 gene expression [60]. cells with POR3. VopS indirectly targets the actin cytoskeleton
by AMPylating Rho-family GTPases [52]. The Fic domain
Table 2 within VopS mediates the direct transfer of adenosine mono-
V. parahaemolyticus strains used to study effectors are derived from the RimD phosphate from ATP to the switch 1 region of these small G-
2210633 clinical isolate. proteins, which prevents their binding to downstream effec-
TDH T3SS1 T3SS2 tors. This blocks the signaling cascade regulating the actin
RimD 2210633 D D D cytoskeleton, leading to its collapse [66,67].
POR1 e D D VPA0450 has now been shown to be a phosphatidylinosi-
POR2 e e D tide phosphatase homologous to the inositol polyphosphate 5-
POR3 e D e
phosphatase (IPP5C) domain of the eukaryotic protein
998 C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001

Table 3
Known T3SS effectors target diverse substrates to drive cytotoxicity and alter immune function.
T3SS Effector Domain Activity Cellular consequence Ref.
T3SS1 VopQ (VP1680) Unique to VopQ Unknown target or mechanism Induction of autophagy [48, 56]
VopR (VP1683) Unknown Unknown target or mechanism Unknown [56]
VopS (VP1686) Fic domain AMPylates Rho-family GTPases Collapse of the actin cytoskeleton [52,56]
VPA0450 Inositol polyphosphate Removes D5 phosphate from PtdIns(4,5)P2 Destabilization of plasma membrane [56,63]
5-phosphatase
T3SS2 VopC (VPA1321) Cytotoxic Necrotizing Deamidation of Rho GTPases, making them Disregulation of actin network, inhibition [71]
Factor-1 homolog constitutively active (hypothesized) of apoptosis (hypothesized)
VopT (VPA1327) ADP-Ribosyltransferase ADP-ribosylation of Ras Induction of cytotoxicity [71]
VopA/P (VPA1346) Acetyltransferase Inhibition of MAPK signaling Lack of innate immune activation and [71e74]
cytokine production
VopL (VPA1370) WH2, and proline Nucleation of actin polymerization Alteration of cell shape, possible loss of [40]
rich region tight junction integrity

synaptojanin. By hydrolyzing phosphatidylinositide (4,5)- regulated with T3SS2, it is not necessary for the pathogenic
bisphosphate (PtdIns(4,5)P2) in the plasma membrane, effects observed in these model systems [23,41,58,69].
VPA0450 disrupts the association of actin-binding proteins Induction of Vp-PAI gene expression occurs upon contact
with the membrane. Alone, VPA0450 is sufficient to induce with bile acids. Specific components of crude bile, including
membrane blebbing. In conjunction with the other T3SS1 deoxycholate, taurodeoxycholate, and glycodeoxycholate, were
effectors, the catalytic activity of VPA0450 drives the rapid able to induce a number of genes, which were mostly found
lysis of host cells [63]. within Vp-PAI [23]. The transcriptional regulators, and putative
T3SS1 was initially proposed to kill cells by apoptosis environmental sensors of this genomic island, were identified as
based on Annexin V staining of phosphotidylserine (PS) after the ToxR homologs VtrA (VPA1332) and VtrB (VPA1348).
3 h of infection with V. parahaemolyticus POR3 [56]. Later Both of these proteins contain an N-terminal OmpR-like winged
work demonstrated that LDH is released in as little as 2 h, helix-turn-helix DNA binding domain as well as predicted
indicating the Annexin V staining was likely due to cell single transmembrane regions. Epistasis studies demonstrated
permeability rather than the flipping of PS to the outer leaflet that VtrA initates expression of VtrB. In turn, VtrB activates
of the plasma membrane [48]. Additionally, POR3 failed to transcription of tdhAS, T3SS2 structural genes, and effectors
activate caspases or cleave poly ADP-ribose polymerase including VopC, VopT, VopA, and VopL (Table 3) [58]. As seen
(PARP), both indicators of apoptosis activation [62]. An below, some of these effectors may have a redundant activity,
additional study has indicated cell death proceeds by oncosis and therefore, molecular microbial genetics complemented with
based on the uptake of a membrane impermeable dye as well biochemistry may be required to elucidate the effector’s
as protection of cells from cytotoxicity by PEG3350, an biochemical activity and cellular target.
osmoprotectant [18]. These studies were performed with V. VopC (VPA1321) has homology to cytotoxic necrotizing
parahaemolyticus NY-4 which, as an O3:K6 serovar, harbors factor 1 (CNF1), an exotoxin found in some pathogenic E. coli
a gene encoding TDH [2]. The tdh gene would need to be strains. CNF1 has been shown to specifically activate Rho, Rac
deleted and the osmoprotection assay repeated before oncosis and Cdc42 by deamidating a glutamine residue in the switch 2
could be identified as a mechanism of cell death during Vibrio region of each enzyme, preventing hydrolysis of GTP. This
infection. Apoptosis has been shown to occur in Epithelioma causes different effects inside the cell including the induction
papulosum cyprini (EPC) cells from a carp fish during infec- of numerous actin-dependent phenotypes, the modification of
tion with a Vibrio alginolyticus strain that harbors the VopQ the mitochondrial network, and inhibition of apoptosis [70]. A
homolog Va1680 [68]. It is possible that this fish cell line, or functional analysis of VopC to determine if it shares catalytic
fish in general, do not have the target of VopQ, and the cells activity with CNF1 has not been performed. As such the
default to apoptosis when not directed to induce autophagy. activity and phenotypic consequences of VopC translocation
into host cells is undetermined.
VopT (VPA1327) has homology to the ADP-
4.5.2. Vp-PAI and T3SS2 ribosyltransferase domain of the Pseudomonas aeruginosa
T3SS2 is found primarily in clinical isolates, and is asso- effectors ExoS and ExoT. It has been shown to transfer ADP-
ciated with pandemic strains of V. parahaemolyticus and large ribose to Ras, a small monomeric GTPase. This activity is
outbreaks of disease [42]. Strains without T3SS2 are generally partially responsible for the cytotoxicity seen during infection
considered to lack pathogenic potential [69]. T3SS2 is unlike of Caco-2 monolayers with V. parahaemolyticus [71]. Ras has
any other specific T3SS, but has closest homology to the Hrp1 been implicated in controlling a variety of cellular processes
system found in Pseudomonas syringae [42,47]. The activity including cell growth, differentiation, and apoptosis. It is also
of T3SS2 has been associated with enterotoxicity in the rabbit involved in activating the ERK/MAPK pathway [72]. There-
ileal loop model [42], as well as disruption of tight junction fore, inhibition of Ras by VopT is likely to have numerous
integrity in cultured cell monolayers [41,69]. While tdh is co- effects on host cells during infection.
C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001 999

VopA (VPA1346, also referred to as VopP) was character- concert to orchestrate a disease progression that has been
ized as a YopJ homolog that blocks MAPK signaling by difficult to study due to the lack of a relevant in vivo model.
acetylating a conserved serine, threonine, and lysine residue To facilitate the study of the T3SSs as a whole, in
on MAPKKs. This prevents the phosphorylation and activation conjunction with the effects of TDH, multiple animal models
of the MAPK pathway, which prevents the induction of have been developed. Intraperitoneal injection of V. para-
cytokines. VopA/P differs from YopJ in that it only targets the haemolyticus into mice has demonstrated that both T3SS1 and
MAPK pathway, whereas YopJ also blocks NF-kB signaling TDH contribute to lethality during systemic infection, while
[73,74]. Although Ras activation triggers many signaling T3SS2 is necessary for fluid accumulation in the rabbit ligated
pathways, VopA/P may be considered to be partially redun- ileal loop [78]. These findings are supported by a study in
dant with VopT as both block the activation of the ERK/MPK which piglets infected orogastrically with a V. para-
pathway. haemolyticus NY-4 strain expressing only T3SS2 resulted in
VopL (VPA1370) was identified as a protein containing acute, self-limited diarrhea similar to that seen in humans. A
three Wiskott-Aldrich homology 2 (WH2) domains. The WH2 different strain with only T3SS1 failed to produce symptoms.
domains are able to bind actin monomers and are thought to When these same strains were used in a lung-inhalation model
position them for elongation of an actin filament. Transfection in mice, the T3SS1-expressing strain caused 80e100%
of VopL into Hela cells resulted in the formation of stress mortality in 12 h, while the T3SS2-expressing strain did not
fibers independent of Rho-family GTPase activity [40]. A cause death of the test subjects [79]. These results suggest
homolog from V. cholerae, VopF, has similar domain archi- a role for T3SS2 effectors in causing the initial infection and
tecture but induces aberrant actin protrusions on the cell diarrheal disease, while T3SS1 may be required for survival
surface [75]. Biochemical analysis showed that recombinant from immune clearance once V. parahaemolyticus dissemi-
VopL was sufficient to nucleate actin polymerization inde- nates out of the intestine and into deeper tissues. Continued
pendent of other cellular factors [40]. Depending on the development of relevant model systems will help to improve
cellular target for VopC, VopL could be potentially redundant our understanding of how each T3SS contributes to disease.
with VopC.
While the characterization of each T3SS is progressing, the 6. Conclusion
evolutionary purpose of these T3SSs and their effectors is still
unknown. As V. parahaemolyticus is an incidental pathogen of V. parahaemolyticus is a common human pathogen with an
humans, it is unlikely that T3SSs were acquired specifically to arsenal of virulence factors such a toxins and type 3 secreted
induce disease in this host. Rather, T3SSs may provide for effectors that alter the homeostasis and integrity of human
improved fitness in the environment. cells. Other cellular processes such as capsule formation, iron
While a previous study excluded T3SSs in survival of V. acquisition, and flagellar motility are also required for infec-
parahaemolyticus during co-culture with the amoeba Acan- tion of the host. As the mechanism of each virulence factor is
thamoeba castellanii [76], recent work has detailed the critical elucidated, a new question arisesdhow do these factors work
importance of T3SS2 for survival from predation by a number in concert to contribute to an overall infection model? While
of marine protists. V. parahaemolyticus strains lacking the Vp- this question can be partially answered in vitro with tissue
PAI genes, and specifically T3SS2, were unable to survive in culture cells, it will ultimately require a relevant in vivo
the presence of flagellated and ciliated protists, as well as model.
amoeba. The presence of these genes, however, not only
promoted bacterial survival, but resulted in killing of the Acknowledgments
protists, ascribing a function for T3SS2 in the environment
[77]. Survival from protist grazing would allow for enhanced We thank members of the Orth lab for their critical reviews.
invasion and survival within the estuarine environment by V. K.O., C.A.B. and T.J.C are supported by National Institutes of
parahaemolyticus strains bearing both T3SS1 and 2, while Health-AID Grants R01-AI056404 and R01-AI087808, and
leaving open to question the concerted effects of these systems Grant I-1561 from the Welch Research Foundation. C.A.B. is
on a mammalian host. supported by NIGMS training grant 5T32GM008203 in
cellular and molecular biology. K.O. is a Burroughs Wellcome
5. Mammalian model systems to study V. Investigator in Pathogenesis of Infectious Disease and a W.W.
parahaemolyticus pathogenicity Caruth, Jr. Biomedical Scholar.

To date, much of the work characterizing how V. para- References


haemolyticus causes disease has been reductionist in its
approach. Many of its toxins and effectors have been identified [1] L. McCarter, The multiple identities of Vibrio parahaemolyticus, J. Mol.
and characterized. While this has yielded significant and Microbiol. Biotechnol. 1 (1999) 51e57.
[2] P.S. Yeung, K.J. Boor, Epidemiology, pathogenesis, and prevention of
valuable data, how these various bacterial factors work
foodborne Vibrio parahaemolyticus infections, Foodborne Pathog. Dis. 1
together is also of importance. Disease mediated by V. para- (2004) 74e88.
haemolyticus does not occur through the isolated actions of [3] Y.C. Su, C. Liu, Vibrio parahaemolyticus: a concern of seafood safety,
individual effectors. Rather, effectors and toxins work in Food Microbiol. 24 (2007) 549e558.
1000 C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001

[4] S.W. Joseph, R.R. Colwell, J.B. Kaper, Vibrio parahaemolyticus and [25] R.B. Ferreira, L.C. Antunes, E.P. Greenberg, L.L. McCarter, Vibrio
related halophilic Vibrios, Crit. Rev. Microbiol. 10 (1982) 77e124. parahaemolyticus ScrC modulates cyclic dimeric GMP regulation of
[5] T. Honda, T. Iida, Y. Akeda, T. Kodama, Sixty years of Vibrio para- gene expression relevant to growth on surfaces, J. Bacteriol. 190 (2008)
haemolyticus, Microbe 3 (2008) 462e466. 851e860.
[6] J.G. Morris Jr., Cholera and other types of vibriosis: a story of human [26] Y.K. Kim, L.L. McCarter, ScrG, a GGDEF-EAL protein, participates in
pandemics and oysters on the half shell, Clin. Infect. Dis. 37 (2003) regulating swarming and sticking in Vibrio parahaemolyticus, J. Bac-
272e280. teriol. 189 (2007) 4094e4107.
[7] G.B. Nair, T. Ramamurthy, S.K. Bhattacharya, B. Dutta, Y. Takeda, D.A. [27] C.J. Gode-Potratz, R.J. Kustusch, P.J. Breheny, D.S. Weiss, L.L.
Sack, Global dissemination of Vibrio parahaemolyticus serotype O3:K6 McCarter, Surface sensing in Vibrio parahaemolyticus triggers a pro-
and its serovariants, Clin. Microbiol. Rev. 20 (2007) 39e48. gramme of gene expression that promotes colonization and virulence,
[8] N.A. Daniels, L. MacKinnon, R. Bishop, S. Altekruse, B. Ray, R.M. Mol. Microbiol. 79 (2011) 240e263.
Hammond, S. Thompson, S. Wilson, N.H. Bean, P.M. Griffin, L. [28] A.M. Krachler, H. Ham, K. Orth, The outer membrane adhesion factor
Slutsker, Vibrio parahaemolyticus infections in the United States, 1973- MAM7 initiates host cell binding during infection by Gram-negative
1998, J. Infect. Dis. 181 (2000) 1661e1666. pathogens, Proc. Natl. Acad. Sci. U S A (2011).
[9] W. Baffone, R. Tarsi, L. Pane, R. Campana, B. Repetto, G.L. Mariottini, [29] R.J. Kustusch, C.J. Kuehl, J.H. Crosa, Power plays: iron transport and
C. Pruzzo, Detection of free-living and plankton-bound vibrios in coastal energy transduction in pathogenic vibrios, Biometals 24 (2011) 559e566.
waters of the Adriatic Sea (Italy) and study of their pathogenicity- [30] T. Tanabe, T. Funahashi, H. Nakao, S. Miyoshi, S. Shinoda, S. Yama-
associated properties, Environ. Microbiol. 8 (2006) 1299e1305. moto, Identification and characterization of genes required for biosyn-
[10] M.D. Gamble, C.R. Lovell, Infaunal burrows are enrichment zones for thesis and transport of the siderophore vibrioferrin in Vibrio
Vibrio parahaemolyticus, Appl. Environ. Microbiol. (2011). parahaemolyticus, J. Bacteriol. 185 (2003) 6938e6949.
[11] C.N. Johnson, A.R. Flowers, N.F. Noriea 3rd, A.M. Zimmerman, J.C. [31] T. Funahashi, T. Tanabe, K. Shiuchi, H. Nakao, S. Yamamoto, Identifi-
Bowers, A. DePaola, D.J. Grimes, Relationships between environmental cation and characterization of genes required for utilization of desferri-
factors and pathogenic vibrios in the Northern Gulf of Mexico, Appl. ferrichrome and aerobactin in Vibrio parahaemolyticus, Biol. Pharm.
Environ. Microbiol. 76 (2010) 7076e7084. Bull. 32 (2009) 359e365.
[12] G.J. Vasconcelos, W.J. Stang, R.H. Laidlaw, Isolation of Vibrio para- [32] S.M. O’Malley, S.L. Mouton, D.A. Occhino, M.T. Deanda, J.R. Rashidi,
haemolyticus and Vibrio alginolyticus from estuarine areas of South- K.L. Fuson, C.E. Rashidi, M.Y. Mora, S.M. Payne, D.P. Henderson,
eastern Alaska, Appl. Microbiol. 29 (1975) 557e559. Comparison of the heme iron utilization systems of pathogenic Vibrios, J.
[13] J.B. McLaughlin, A. DePaola, C.A. Bopp, K.A. Martinek, N.P. Napolilli, Bacteriol. 181 (1999) 3594e3598.
C.G. Allison, S.L. Murray, E.C. Thompson, M.M. Bird, J.P. Middaugh, [33] M. Nishibuchi, J.B. Kaper, Thermostable direct hemolysin gene of Vibrio
Outbreak of Vibrio parahaemolyticus gastroenteritis associated with parahaemolyticus: a virulence gene acquired by a marine bacterium,
Alaskan oysters, N. Engl. J. Med. 353 (2005) 1463e1470. Infect. Immun. 63 (1995) 2093e2099.
[14] N. Gonzalez-Escalona, E.A. Strain, A.J. De Jesus, J.L. Jones, A. Depaola, [34] W. Hongping, Z. Jilun, J. Ting, B. Yixi, Z. Xiaoming, Insufficiency of the
Genome sequence of a clinical O4:K12 serotype Vibrio parahaemolyticus Kanagawa hemolytic test for detecting pathogenic Vibrio para-
strain 10329, J. Bacteriol. (2011). doi:10.1128/JB.05044-05011. haemolyticus in Shanghai, China, Diagn. Microbiol. Infect. Dis. 69
[15] K. Makino, K. Oshima, K. Kurokawa, K. Yokoyama, T. Uda, K. Tago- (2011) 7e11.
mori, Y. Iijima, M. Najima, M. Nakano, A. Yamashita, Y. Kubota, S. [35] T. Fukui, K. Shiraki, D. Hamada, K. Hara, T. Miyata, S. Fujiwara, K.
Kimura, T. Yasunaga, T. Honda, H. Shinagawa, M. Hattori, T. Iida, Mayanagi, K. Yanagihara, T. Iida, E. Fukusaki, T. Imanaka, T. Honda, I.
Genome sequence of Vibrio parahaemolyticus: a pathogenic mechanism Yanagihara, Thermostable direct hemolysin of Vibrio parahaemolyticus
distinct from that of V cholerae, Lancet 361 (2003) 743e749. is a bacterial reversible amyloid toxin, Biochemistry 44 (2005)
[16] E.F. Boyd, A.L. Cohen, L.M. Naughton, D.W. Ussery, T.T. Binnewies, O. 9825e9832.
C. Stine, M.A. Parent, Molecular analysis of the emergence of pandemic [36] S. Matsuda, T. Kodama, N. Okada, K. Okayama, T. Honda, T. Iida,
Vibrio parahaemolyticus, BMC Microbiol. 8 (2008) 110. Association of Vibrio parahaemolyticus thermostable direct hemolysin
[17] A.K. Bej, D.P. Patterson, C.W. Brasher, M.C. Vickery, D.D. Jones, C.A. with lipid rafts is essential for cytotoxicity but not hemolytic activity,
Kaysner, Detection of total and hemolysin-producing Vibrio para- Infect. Immun. 78 (2010) 603e610.
haemolyticus in shellfish using multiplex PCR amplification of tlh, tdh [37] I. Yanagihara, K. Nakahira, T. Yamane, S. Kaieda, K. Mayanagi, D.
and trh, J. Microbiol. Methods 36 (1999) 215e225. Hamada, T. Fukui, K. Ohnishi, S. Kajiyama, T. Shimizu, M. Sato, T.
[18] X. Zhou, M.E. Konkel, D.R. Call, Type III secretion system 1 of Vibrio Ikegami, M. Ikeguchi, T. Honda, H. Hashimoto, Structure and functional
parahaemolyticus induces oncosis in both epithelial and monocytic cell characterization of Vibrio parahaemolyticus thermostable direct hemo-
lines, Microbiology 155 (2009) 837e851. lysin, J. Biol. Chem. 285 (2010) 16267e16274.
[19] W.G. Hlady, K.C. Klontz, The epidemiology of Vibrio infections in [38] T. Honda, Y. Ni, T. Miwatani, T. Adachi, J. Kim, The thermostable direct
Florida, 1981-1993, J. Infect. Dis. 173 (1996) 1176e1183. hemolysin of Vibrio parahaemolyticus is a pore-forming toxin, Can. J.
[20] P.A. Blake, M.H. Merson, R.E. Weaver, D.G. Hollis, P.C. Heublein, Microbiol. 38 (1992) 1175e1180.
Disease caused by a marine Vibrio. Clinical characteristics and epide- [39] K.S. Park, T. Ono, M. Rokuda, M.H. Jang, T. Iida, T. Honda, Cytotoxicity
miology, N. Engl. J. Med. 300 (1979) 1e5. and enterotoxicity of the thermostable direct hemolysin-deletion mutants
[21] N. Okada, S. Matsuda, J. Matsuyama, K.S. Park, C. de los Reyes, K. of Vibrio parahaemolyticus, Microbiol. Immunol. 48 (2004) 313e318.
Kogure, T. Honda, T. Iida, Presence of genes for type III secretion system [40] A.D. Liverman, H.C. Cheng, J.E. Trosky, D.W. Leung, M.L. Yarbrough,
2 in Vibrio mimicus strains, BMC Microbiol. 10 (2010) 302. D.L. Burdette, M.K. Rosen, K. Orth, Arp2/3-independent assembly of
[22] G. Chao, X. Jiao, X. Zhou, F. Wang, Z. Yang, J. Huang, Z. Pan, L. Zhou, actin by Vibrio type III effector VopL, Proc. Natl. Acad. Sci. U S A 104
X. Qian, Distribution of genes encoding four pathogenicity islands (2007) 17117e17122.
(VPaIs), T6SS, biofilm, and type I pilus in food and clinical strains of [41] T. Lynch, S. Livingstone, E. Buenaventura, E. Lutter, J. Fedwick, A.G.
Vibrio parahaemolyticus in China, Foodborne Pathog. Dis. 7 (2010) Buret, D. Graham, R. DeVinney, Vibrio parahaemolyticus disruption of
649e658. epithelial cell tight junctions occurs independently of toxin production,
[23] K. Gotoh, T. Kodama, H. Hiyoshi, K. Izutsu, K.S. Park, R. Dryselius, Y. Infect. Immun. 73 (2005) 1275e1283.
Akeda, T. Honda, T. Iida, Bile acid-induced virulence gene expression of [42] K.S. Park, T. Ono, M. Rokuda, M.H. Jang, K. Okada, T. Iida, T. Honda,
Vibrio parahaemolyticus reveals a novel therapeutic potential for bile Functional characterization of two type III secretion systems of Vibrio
acid sequestrants, PLoS One 5 (2010) e13365. parahaemolyticus, Infect. Immun. 72 (2004) 6659e6665.
[24] S. Jaques, L.L. McCarter, Three new regulators of swarming in Vibrio [43] K. Ohnishi, K. Nakahira, S. Unzai, K. Mayanagi, H. Hashimoto, K. Shiraki,
parahaemolyticus, J. Bacteriol. 188 (2006) 2625e2635. T. Honda, I. Yanagihara, Relationship between heat-induced fibrillogenicity
C.A. Broberg et al. / Microbes and Infection 13 (2011) 992e1001 1001

and hemolytic activity of thermostable direct hemolysin and a related [63] C.A. Broberg, L. Zhang, H. Gonzalez, M.A. Laskowski-Arce, K. Orth, A
hemolysin of Vibrio parahaemolyticus, FEMS Microbiol. Lett. 318 (2011) Vibrio effector protein is an inositol phosphatase and disrupts host cell
10e17. membrane integrity, Science 329 (2010) 1660e1662.
[44] J.E. Galan, H. Wolf-Watz, Protein delivery into eukaryotic cells by type [64] K. Matlawska-Wasowska, R. Finn, A. Mustel, C.P. O’Byrne, A.W. Baird,
III secretion machines, Nature 444 (2006) 567e573. E.T. Coffey, A. Boyd, The Vibrio parahaemolyticus Type III Secretion
[45] T. Izore, V. Job, A. Dessen, Biogenesis, regulation, and targeting of the Systems manipulate host cell MAPK for critical steps in pathogenesis,
type III secretion system, Structure 19 (2011) 603e612. BMC Microbiol. 10 (2010) 329.
[46] T.C. Marlovits, C.E. Stebbins, Type III secretion systems shape up as [65] T. Shimohata, M. Nakano, X. Lian, T. Shigeyama, H. Iba, A. Hamamoto,
they ship out, Curr. Opin. Microbiol. 13 (2010) 47e52. M. Yoshida, N. Harada, H. Yamamoto, M. Yamato, K. Mawatari, T.
[47] G.R. Cornelis, The type III secretion injectisome, Nat. Rev. Microbiol. 4 Tamaki, Y. Nakaya, A. Takahashi, Vibrio parahaemolyticus infection
(2006) 811e825. induces modulation of IL-8 secretion through dual pathway via VP1680
[48] D.L. Burdette, J. Seemann, K. Orth, Vibrio VopQ induces PI3-kinase- in Caco-2 cells, J. Infect. Dis. 203 (2011) 537e544.
independent autophagy and antagonizes phagocytosis, Mol. Microbiol. [66] P. Luong, L.N. Kinch, C.A. Brautigam, N.V. Grishin, D.R. Tomchick, K.
73 (2009) 639e649. Orth, Kinetic and structural insights into the mechanism of AMPylation
[49] S. Mukherjee, G. Keitany, Y. Li, Y. Wang, H.L. Ball, E.J. Goldsmith, K. by VopS Fic domain, J. Biol. Chem. 285 (2010) 20155e20163.
Orth, Yersinia YopJ acetylates and inhibits kinase activation by blocking [67] A.R. Woolery, P. Luong, C.A. Broberg, K. Orth, AMPylation: something
phosphorylation, Science 312 (2006) 1211e1214. old is new again, Front. Microbiol. 1 (2010) 1e6.
[50] M. Ogawa, T. Yoshimori, T. Suzuki, H. Sagara, N. Mizushima, C. [68] Z. Zhao, C. Chen, C.Q. Hu, C.H. Ren, J.J. Zhao, L.P. Zhang, X. Jiang, P.
Sasakawa, Escape of intracellular Shigella from autophagy, Science 307 Luo, Q.B. Wang, The type III secretion system of Vibrio alginolyticus
(2005) 727e731. induces rapid apoptosis, cell rounding and osmotic lysis of fish cells,
[51] U. Von Pawel-Rammingen, M.V. Telepnev, G. Schmidt, K. Aktories, Microbiol. 156 (2010) 2864e2872.
H. Wolf-Watz, R. Rosqvist, GAP activity of the Yersinia YopE [69] G. Caburlotto, M.M. Lleo, T. Hilton, A. Huq, R.R. Colwell, J.B. Kaper,
cytotoxin specifically targets the Rho pathway: a mechanism for Effect on human cells of environmental Vibrio parahaemolyticus strains
disruption of actin microfilament structure, Mol. Microbiol. 36 (2000) carrying type III secretion system 2, Infect. Immun. 78 (2010) 3280e3287.
737e748. [70] A.G. Miraglia, S. Travaglione, S. Meschini, L. Falzano, P. Matarrese, M.
[52] M.L. Yarbrough, Y. Li, L.N. Kinch, N.V. Grishin, H.L. Ball, K. Orth, G. Quaranta, M. Viora, C. Fiorentini, A. Fabbri, Cytotoxic necrotizing
AMPylation of Rho GTPases by Vibrio VopS disrupts effector binding factor 1 prevents apoptosis via the Akt/IkappaB kinase pathway: role of
and downstream signaling, Science 323 (2009) 269e272. nuclear factor-kappaB and Bcl-2, Mol. Biol. Cell 18 (2007) 2735e2744.
[53] Y. Akeda, J.E. Galan, Chaperone release and unfolding of substrates in [71] T. Kodama, M. Rokuda, K.S. Park, V.V. Cantarelli, S. Matsuda, T. Iida, T.
type III secretion, Nature 437 (2005) 911e915. Honda, Identification and characterization of VopT, a novel ADP-
[54] R. Arnold, A. Jehl, T. Rattei, Targeting effectors: the molecular recog- ribosyltransferase effector protein secreted via the Vibrio parahaemolyticus
nition of Type III secreted proteins, Microbes Infect. 12 (2010) 346e358. type III secretion system 2, Cell Microbiol. 9 (2007) 2598e2609.
[55] J.E. Galan, Common themes in the design and function of bacterial [72] M.C. Lawrence, A. Jivan, C. Shao, L. Duan, D. Goad, E. Zaganjor, J.
effectors, Cell Host Microbe 5 (2009) 571e579. Osborne, K. McGlynn, S. Stippec, S. Earnest, W. Chen, M. Cobb, The
[56] T. Ono, K.S. Park, M. Ueta, T. Iida, T. Honda, Identification of proteins roles of MAPKs in disease, Cell Res. 18 (2008) 436e442.
secreted via Vibrio parahaemolyticus type III secretion system 1, Infect. [73] J.E. Trosky, Y. Li, S. Mukherjee, G. Keitany, H. Ball, K. Orth, VopA
Immun. 74 (2006) 1032e1042. inhibits ATP binding by acetylating the catalytic loop of MAPK kinases,
[57] E.M. Panina, S. Mattoo, N. Griffith, N.A. Kozak, M.H. Yuk, J.F. Miller, J. Biol. Chem. 282 (2007) 34299e34305.
A genome-wide screen identifies a Bordetella type III secretion effector [74] J.E. Trosky, S. Mukherjee, D.L. Burdette, M. Roberts, L. McCarter, R.
and candidate effectors in other species, Mol. Microbiol. 58 (2005) M. Siegel, K. Orth, Inhibition of MAPK signaling pathways by VopA
267e279. from Vibrio parahaemolyticus, J. Biol. Chem. 279 (2004) 51953e51957.
[58] T. Kodama, K. Gotoh, H. Hiyoshi, M. Morita, K. Izutsu, Y. Akeda, K.S. [75] V.C. Tam, M. Suzuki, M. Coughlin, D. Saslowsky, K. Biswas, W.I.
Park, V.V. Cantarelli, R. Dryselius, T. Iida, T. Honda, Two regulators of Lencer, S.M. Faruque, J.J. Mekalanos, Functional analysis of VopF
Vibrio parahaemolyticus play important roles in enterotoxicity by activity required for colonization in Vibrio cholerae, MBio 1 (2010). doi:
controlling the expression of genes in the Vp-PAI region, PLoS One 5 10.1128/mBio.00289e00210.
(2010) e8678. [76] M.A. Laskowski-Arce, K. Orth, Acanthamoeba castellanii promotes the
[59] D.M. Monack, J. Mecsas, N. Ghori, S. Falkow, Yersinia signals macro- survival of Vibrio parahaemolyticus, Appl. Environ. Microbiol. 74
phages to undergo apoptosis and YopJ is necessary for this cell death, (2008) 7183e7188.
Proc. Natl. Acad. Sci. U S A 94 (1997) 10385e10390. [77] C. Matz, B. Nouri, L. McCarter, J. Martinez-Urtaza, Acquired type III
[60] T. Kodama, C. Yamazaki, K.S. Park, Y. Akeda, T. Iida, T. Honda, secretion system determines environmental fitness of Epidemic Vibrio
Transcription of Vibrio parahaemolyticus T3SS1 genes is regulated by parahaemolyticus in the interaction with Bacterivorous protists, PLoS
a dual regulation system consisting of the ExsACDE regulatory cascade One 6 (2011) e20275.
and H-NS, FEMS Microbiol. Lett. (2010). [78] H. Hiyoshi, T. Kodama, T. Iida, T. Honda, Contribution of Vibrio par-
[61] C.J. Dorman, H-NS: a universal regulator for a dynamic genome, Nat. ahaemolyticus virulence factors to cytotoxicity, enterotoxicity, and
Rev. Microbiol. 2 (2004) 391e400. lethality in mice, Infect. Immun. 78 (2010) 1772e1780.
[62] D.L. Burdette, M.L. Yarbrough, A. Orvedahl, C.J. Gilpin, K. Orth, Vibrio [79] P. Pineyro, X. Zhou, L.H. Orfe, P.J. Friel, K. Lahmers, D.R. Call,
parahaemolyticus orchestrates a multifaceted host cell infection by Development of two animal models to study the function of Vibrio
induction of autophagy, cell rounding, and then cell lysis, Proc. Natl. parahaemolyticus type III secretion systems, Infect. Immun. 78 (2010)
Acad. Sci. U S A 105 (2008) 12497e12502. 4551e4559.

You might also like