Вы находитесь на странице: 1из 30

Typhoidal Salmonella: distinctive virulence factors and pathogenesis

Rebecca Johnson*, Elli Mylona*, Gad Frankel@

MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial

College London, London, United Kingdom

*These two authors are equal contributors

@
Corresponding author

Gad Frankel, MRC CMBI, Flowers Building, Imperial College, London, SW7 2AZ

g.frankel@imperial.ac.uk

Running title: Pathogenesis of Salmonella Typhi

This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
10.1111/cmi.12939

This article is protected by copyright. All rights reserved.


Summary

While non-typhoidal Salmonella (NTS) (including S. Typhimurium) mainly cause gastroenteritis,

typhoidal serovars (S. Typhi and S. Paratyphi A) cause typhoid fever, the treatment of which is

threatened by increasing drug-resistance. Our understanding of S. Typhi infection in human remains

poorly understood, likely due to the host restriction of typhoidal strains and the subsequent popularity

of the S. Typhimurium mouse typhoid model. However, translating findings with S. Typhimurium

across to S. Typhi has some limitations. Notably, S. Typhi has specific virulence factors, including

typhoid toxin and Vi antigen, involved in symptom development and immune evasion, respectively. In

addition to unique virulence factors, both typhoidal and NTS rely on two pathogenicity-island encoded

type III secretion systems (T3SS), the SPI-1 and SPI-2 T3SS, for invasion and intracellular replication.

Marked differences have been observed in terms of T3SS regulation in response to bile, oxygen and

fever-like temperatures. Moreover, approximately half of effectors found in S. Typhimurium are either

absent or pseudogenes in S. Typhi, with most of the remaining exhibiting sequence variation.

Typhoidal-specific T3SS effectors have also been described. This review discusses what is known about

the pathogenesis of typhoidal Salmonella with emphasis on unique behaviours and key differences

when compared to S. Typhimurium.

This article is protected by copyright. All rights reserved.


Introduction

Salmonella enterica subspecies enterica includes numerous pathogens of warm-blooded animals

including humans. This subspecies is further classified into serovars, based on O (LPS) and H (flagellar)

antigens (Brenner et al., 2000). In humans, infection with Salmonella broadly results in two outcomes

– either self-limiting gastroenteritis, or invasive systemic typhoid fever. The disease outcome

predominantly depends on the infecting serovar, and as such, Salmonella serovars are classified into

two groups: typhoidal and non-typhoidal Salmonella (NTS). The most common NTS, and the most

common cause of gastroenteritis, are the broad host range serovars S. Enteritidis and S. Typhimurium

(CDC, 2015). However, recently NTS variants have been associated with invasive systemic disease and

high mortality rates within immunocompromised patients in sub-Saharan Africa (Feasey et al., 2012).

Typhoidal serovars, which cause typhoid (or enteric) fever, are S. Typhi and S. Paratyphi A (Dougan &

Baker, 2014). Recent studies estimate that there are approximately 10-20 million cases of typhoid per

year, resulting in 100,000-200,000 deaths (Antillón et al., 2017; Mogasale et al., 2014).

Following ingestion, S. Typhi crosses the intestinal epithelium and disseminates to systemic sites,

including the liver, spleen, bone marrow and gallbladder. Symptoms of typhoid typically develop 10-

14 days post-ingestion, and include fever, headache, muscle aches, stomach pain, and constipation or

diarrhoea (Parry et al., 2002). The symptoms of S. Paratyphi A infection are similar, but often milder

(Dobinson et al., 2017). With appropriate antibiotic treatment, typhoid has a mortality rate of

approximately 1% (Parry et al., 2002). Following recovery from acute disease, approximately 3-5% of

infected individuals will continue to shed S. Typhi for several months to years (Gunn et al., 2014).

During chronic carriage, S. Typhi, and S. Paratyphi A can persist asymptomatically within the

gallbladder (Dongol et al., 2012; Gunn et al., 2014). As typhoidal serovars are human restricted, carriers

represent a key reservoir for S. Typhi, which contribute to the transmission and dissemination of typhoid

(Pitzer et al., 2014; Saul et al., 2013).

The global spread of H58 S. Typhi and drug-resistant typhoid

A concerning aspect of S. Typhi epidemiology is the recent expansion and global spread of one

haplotype (H58). In many areas of south and southeast Asia, and sub-Saharan Africa, H58 is the

This article is protected by copyright. All rights reserved.


dominant S. Typhi haplotype, and its introduction and spread within a region is associated with typhoid

outbreaks and epidemics (Baker et al., 2011; Emary et al., 2012; Feasey et al., 2015; Holt et al., 2011;

Yan et al., 2016). The reasons underlying the global success of H58 are unexplained, but H58 strains

are often multi-drug resistant (MDR) (Wong et al., 2015). Alarmingly, the emergence of extensively

drug resistant (XDR) H58 S. Typhi strains, which are resistant to chloramphenicol, ampicillin,

trimethoprim-sulfamethoxazole, fluoroquinolones and third-generation cephalosporins, have recently

been identified in Pakistan (Klemm et al., 2018). H58 strains have previously disseminated rapidly from

the Indian subcontinent to other regions, and therefore the risk of global dissemination of XDR H58

strains is of real concern (Klemm et al., 2018; Wong et al., 2015). The increase in MDR/XDR S. Typhi

isolates poses a real threat to current treatment regimens, and if further resistance develops there is a

risk of typhoid becoming untreatable.

Differences between S. Typhi and S. Typhimurium

Despite the severity of typhoid fever and the concerning rise of antibiotic-resistant S. Typhi, the

molecular pathogenesis of the typhoidal serovars is poorly understood relative to S. Typhimurium. This

is mainly due to the absence of viable in vivo models to study typhoidal strains, as both S. Typhi and S.

Paratyphi are human restricted (Tsolis et al., 2011), and due to biosafety constraints involved in working

with typhoidal serovars (HSE, 2013). In comparison, S. Typhimurium has a broad host-range and

therefore several animal models are available to study infection in vivo (Tsolis et al., 2011). Of particular

importance is the mouse typhoid model, in which susceptible mice (Nramp ), develop a systemic
-

infection somewhat similar to typhoid in humans (Tsolis et al., 2011).

Although research using S. Typhimurium has been invaluable in understanding Salmonella virulence,

there are key differences between S. Typhimurium and S. Typhi (Hiyoshi et al., 2018a), the most

apparent being that in humans, S. Typhimurium predominantly causes gastroenteritis rather than a

systemic disease. At the genomic level, while 89% of genes are shared between the two serovars, almost

500 genes are unique to S. Typhimurium (strain LT2) and over 600 genes are unique to S. Typhi (strain

CT18) (Parkhill et al., 2001). Among the S. Typhi specific genes are those encoding important virulence

factors, including typhoid toxin and the Vi antigen (Sabbagh et al., 2010). Additionally, both S. Typhi

This article is protected by copyright. All rights reserved.


and S. Paratyphi have a high proportion of pseudogenes (approximately 4%) (McClelland et al., 2004;

Parkhill et al., 2001), which are associated with a host restricted lifestyle. In comparison, in S.

Typhimurium approximately 0.9% of genes are pseudogenes (McClelland et al., 2001).

Although S. Typhimurium is a commonly used and long-held model, several studies have observed

marked phenotypic differences between S. Typhimurium and typhoidal serovars (Bishop et al., 2008;

Elhadad et al., 2015, 2016; Eswarappa et al., 2008; Johnson et al., 2017, 2018), in addition to describing

important roles for typhoidal-specific virulence factors (Haghjoo & Galán, 2004; Hiyoshi et al., 2018b;

Raffatellu et al., 2005a; Song et al., 2013; Wilson et al., 2008, 2011; Winter et al., 2015; Yang et al.,

2018). This review will therefore discuss what is known about virulence and pathogenesis of S. Typhi,

and emphasise differences between typhoidal and NTS.

S. Typhi specific virulence factors

The Vi antigen

One of the main characteristics that distinguishes S. Typhi from NTS is the production of a

polysaccharide capsule named the Vi antigen. The Vi capsule inhibits phagocytosis and confers serum

resistance (Hart et al., 2016; Wilson et al., 2011), likely by shielding the O-antigen from antibodies

(Hart et al., 2016). The genes encoding the Vi capsule comprise the viaB locus within Salmonella

pathogenicity island (SPI)-7, which also encodes the type III secretion system (T3SS) effector SopE

and a type IVB pilus (Pickard et al., 2003). The viaB locus encodes genes involved in regulation (tviA),

Vi biosynthesis (tviBCDE), export and retention of the Vi on the bacterial cell surface (vexABCDE)

(Virlogeux et al., 1995). A viaB locus is present in S. Paratyphi C and S. Dublin, but is absent from S.

Paratyphi A, and most gastroenteritis-causing serovars including S. Typhimurium (Bueno et al., 2004;

Parkhill et al., 2001; Pickard et al., 2003; Raffatellu et al., 2005a).

TviA is a positive regulator promoting expression of the viaB locus (Virlogeux et al., 1995), while it

downregulates flagellar and SPI-1 genes under high osmolarity conditions (Winter et al., 2009). Vi

expression is downregulated in the intestine, where flagella and SPI-1 play a role in invasion of

epithelial cells, while it is upregulated in tissues during systemic dissemination, where it prevents

This article is protected by copyright. All rights reserved.


antibody-mediated induction of neutrophil responses (Hiyoshi et al., 2018b; Wangdi et al., 2014; Winter

et al., 2009). TviA-mediated repression of flagellin expression results in limited recognition of flagellin

by NAIP, leading to reduced levels of pyroptosis and IL-1β secretion by macrophages (Winter et al.,

2015) (Fig. 1). Vi has been reported to bind cell surface prohibitin, thus dampening inflammation

through MAPK signalling and IL-8 production (Sharma & Qadri, 2004). Reduced TLR5 and TLR4-

mediated secretion of IL-8 leads to low levels of neutrophil influx (Fig. 1), which is one of the

characteristics of S. Typhi infection that make it distinct from the S. Typhimurium infection (Raffatellu

et al., 2005a; Winter et al., 2008). Strikingly, in contrast to S. Typhimurium, human neutrophils do not

respond to the presence of S. Typhi by forming pseudopods in vitro, as the Vi interferes with

complement activation and complement-dependent chemotactic neutrophil responses (Wangdi et al.,

2014).

S. Paratyphi A is missing the viaB and thus does not express the Vi capsule (Bueno et al., 2004). Instead,

S. Paratyphi A avert antibody binding and antibody-mediated complement activation by elaboration of

long O-chains (Hiyoshi et al., 2018b). Importantly, S. Typhi carries a mutation in the gene responsible

for formation of the long LPS O-chains (fepE) and this mutation is essential for Vi-mediated

inflammation suppression (Crawford et al., 2013). Thus, the ability of the two pathogens to avoid

antibody binding, complement bactericidal effects and the induced phagocyte respiratory burst,

characteristic of (para)typhoid, was acquired through convergent evolution (Hiyoshi et al., 2018b).

The typhoid toxin

Unique to S. Typhi is expression of the typhoid toxin (Haghjoo & Galán, 2004), which is encoded on

SPI-11 (Hodak & Galán, 2013; Spanò et al., 2008). The toxin is expressed exclusively when S. Typhi

is intracellular and localised within the Salmonella containing vacuole (SCV) (Haghjoo & Galán, 2004;

Spanò et al., 2008). The typhoid toxin is an atypical AB toxin, consisting of two enzymatically active

(A) subunits (CdtB and PltA) and a homopentamer of one binding (B) subunit (PltB) (Song et al., 2013).

CdtB is homologous to the A subunit of cytolethal distending toxin (CDT), as well as to DNase-I protein

families (Haghjoo & Galán, 2004), while PltA, which has ADP-ribosyl transferase activity, and PltB

share similarities with subunits of pertussis toxin (Spanò et al., 2008). CdtB induces G2/M cell cycle

This article is protected by copyright. All rights reserved.


arrest through its DNase-I-like activity by damaging the host cell DNA, and inducing the DNA-damage

response (Haghjoo & Galán, 2004). Homologues of the genes encoding the typhoid toxin are found in

S. Paratyphi A and several NTS serovars, exhibiting minor sequence differences, but are absent from

S. Typhimurium and S. Enteritidis (Suez et al., 2013).

The typhoid toxin is secreted within vesicles originating from the SCV and released into the

extracellular space (Fig. 1) (Chang et al., 2016; Spanò et al., 2008). Following export, typhoid toxin is

involved in intoxication of infected and uninfected cells through autocrine and paracrine pathways

(Chang et al., 2016; Spanò et al., 2008). Typhoid toxin enters a range of target cells through PltB-

mediated binding to glycans, predominantly those terminated with N-acetylneuraminic acid (Neu5Ac)

(Song et al., 2013). Glycosylated podocalyxin-like protein 1 (PODXL) on human epithelial cells and

CD45 on immune cells, including macrophages, have been identified as typhoid toxin receptors (Song

et al., 2013). Importantly, Neu5Ac is specific to humans, highlighting the host restriction and adaptation

of S. Typhi (Deng et al., 2014). Of note, chronic typhoidal carriage has been linked with gallbladder

cancer (Nath et al., 1997). It would therefore be interesting to investigate whether typhoid toxin is

involved in causing malignancies, through its DNA-damaging activity (Del Bel Belluz et al., 2016).

The role of typhoid toxin in disease and pathogenesis remains elusive. Mice systemically administered

with purified active typhoid toxin developed symptoms including signs of lethargy, reduced circulating

neutrophil numbers, and neurological complications manifested with motor dysfunctions, although not

fever (Song et al., 2013; Yang et al., 2018). The typhoid toxin is suggested to be involved in promoting

chronic S. Typhi infection, although the underlying mechanism warrants further investigation (Song et

al., 2010). These studies suggest that typhoid toxin is responsible for symptom development and

transition from acute to chronic state during typhoid fever and could be a possible target for alleviating

those.

The S. Typhi flagella

Flagella, while contributing to virulence, are also important activators of innate immune responses via

recognition of monomeric flagellin by TLR5 and NAIP receptors (Fig. 1) (Hayashi et al., 2001;

Kortmann et al., 2015). While most NTS exhibit phase variation via alternate expression of two flagellin

This article is protected by copyright. All rights reserved.


genes (fliC and fljB), most S. Typhi strains are monophasic, specifically expressing FliC of the H:d

antigen. Interestingly, some Indonesian S. Typhi strains express H:j, a variation of H:d due to an in-

frame deletion in fliC (Frankel et al., 1989), and/or are biphasic, expressing a plasmid-encoded FljB

analogue of the H:z66 antigen (Baker et al., 2007). H:j and H:z66 antigenic variants are thought to have

recently emerged during S. Typhi evolution (Baker et al., 2008), driven by immune selection in this

high incidence region (Baker et al., 2007). This additional variation seems to play a role in S. Typhi

interactions with host epithelial cells and macrophages and partly in immune evasion (Schreiber et al.,

2015).

Differences in the T3SSs

The SPI-1 and SPI-2 type III secretion systems

Common to both typhoidal and NTS are two pathogenicity-island encoding type III secretion systems

(T3SS): the SPI-1 and SPI-2 T3SS, which are essential for Salmonella virulence. Although there is

increasing evidence of overlapping and cooperative activities (Brawn et al., 2007; Finn et al., 2017), the

SPI-1 T3SS is mainly active when Salmonella are extracellular and permits invasion of non-phagocytic

cells, whilst the SPI-2 T3SS is activated following internalisation, and functions to promote the

development of the SCV (Ramos-Morales, 2012).

In S. Typhimurium, both T3SSs are essential for invasion and intracellular replication in vitro (Galan

& Curtiss III, 1989; Ochman et al., 1996). In S. Typhi, the SPI-1 T3SS is also required for invasion of

non-phagocytic cells (Bishop et al., 2008), but the importance of the SPI-2 T3SS is less clear. Disruption

of the SPI-2 T3SS did not influence the survival of S. Typhi in THP-1 and human monocyte derived

macrophages (Forest et al., 2010), however S. Typhi strains with transposon insertions in the SPI-2

components ssaQ, ssaP or ssaN were negatively selected against during competitive growth in human

macrophages (Sabbagh et al., 2012). The role of SPI-2 during the intracellular lifestyle of typhoidal

serovars therefore warrants further investigation.

This article is protected by copyright. All rights reserved.


Regulation of the T3SSs

The production and activity of T3SSs are tightly regulated, as their synthesis is energetically costly, and

T3SS components can be recognised by the host immune system (Reyes Ruiz et al., 2017; Sturm et al.,

2011). Regulatory pathways governing expression of the T3SSs in Salmonella are complex, reflecting

the myriad of environmental cues to which they are exposed. During disease in humans, typhoidal and

NTS encounter different environments; whilst non-typhoidal serovars are restricted to the small

intestine, typhoidal strains disseminate systemically. Such differences in the site of infections are likely

to contribute to differences in the control of T3SS expression.

A striking difference in the regulation of the SPI-1 T3SS between S. Typhi and S. Typhimurium relates

to their response to bile. Bile is a digestive secretion involved in the digestion and absorption of fats.

Bile serves as an important environmental cue to determine location within the host gastrointestinal

tract, and is also present in the gallbladder at high concentrations prior to release into the intestines.

Several enteric pathogens control production of virulence factors following bile exposure, including

Salmonella (Begley et al., 2005). In S. Typhimurium, expression of the SPI-1 T3SS and subsequently

invasion into non-phagocytic cells is significantly repressed following growth in bile (Prouty & Gunn,

2000). In comparison, in S. Typhi, growth in bile results in significant upregulation of the SPI-1 T3SS

and its associated genes, resulting in a significant increase in epithelial cell invasion (Johnson et al.,

2018). The mechanism(s) underpinning this difference are at present poorly characterised, but the

stability of the dominant SPI-1 regulator HilD is increased approximately three-fold in S. Typhi in bile,

but decreased over three-fold in S. Typhimurium in bile (Eade et al., 2016; Johnson et al., 2018).

However it is unknown if this increase in stability is responsible for driving changes in SPI-1 expression

in bile, or rather reflects an indirect effect on HilD stability, for example via alteration of DNA binding

(as DNA-bound HilD is proposed to be more stable) (Grenz et al., 2018). Differences in regulation of

SPI-1 in S. Typhi may relate to gallbladder carriage, or reflect different infection strategies within the

small intestines where S. Typhi elicits limited inflammatory responses.

SPI-1 expression also appears to be affected differently in response to oxygen availability between

typhoidal and NTS. Comparison of SPI-1 expression between aerobic and microaerobic cultures of S.

This article is protected by copyright. All rights reserved.


Typhimurium revealed expression was broadly similar between the two conditions, but aerobically

grown bacteria were more invasive (Ibarra et al., 2010). For S. Paratyphi A however, SPI-1 expression,

and epithelial cell invasion, decreases significantly following aerobic growth compared to microaerobic

growth (Elhadad et al., 2016). Although equivalent studies have not been performed with S. Typhi,

differences in SopE-mediated invasion have been observed between the two conditions; in S. Typhi,

invasion following aerobic growth is only modestly decreased by the absence of sopE, whilst following

microaerobic growth invasion is almost entirely SopE-dependent (Johnson et al., 2017). An S.

Typhimurium strain does not show these differences, with a ΔsopE strain demonstrating a 40-60%

reduction in invasion relative to WT, following growth under either condition (Johnson et al., 2017).

Oxygen-dependent regulation of SPI-1 expression and activity in typhoidal strains may represent a way

to limit SPI-1 mediated inflammatory responses at the intestinal epithelium, where the oxygen

concentration is higher than in the lumen.

Temperature has also been reported to differentially affect T3SS-activity in typhoidal and non-typhoidal

serovars. Fever-like temperatures of 39-42°C, such as that experienced within an infected host, reduces

invasion and motility of typhoidal but not S. Typhimurium strains (Elhadad et al., 2015). This response

was further characterised in S. Paratyphi A, where a marked decrease in SPI-1 expression, as determined

by RT-qPCR and Western blotting, is observed at growth at 42°C compared to growth at 37°C (Elhadad

et al., 2015). Temperature-dependent effects were also observed on SPI-2 activity; higher temperatures

(42°C) were found to increase SPI-2 expression in both S. Paratyphi A and S. Typhimurium, however

a significant increase in SPI-2 dependent intracellular replication was observed only in S. Paratyphi A

(Elhadad et al., 2015). This response may reflect adaptation to systemic disease – once fever occurs,

typhoidal strains have disseminated, and SPI-1 mediated invasion may no longer be required (as

supported by the SPI-1 T3SS being dispensable for S. Typhimurium infection following intraperitoneal

injection in the mouse typhoid model (Galan & Curtiss III, 1989)), whilst intracellular survival is

important.

This article is protected by copyright. All rights reserved.


Importantly, the mechanisms underpinning the differences in regulation of the T3SS between typhoidal

and NTS have not been well characterised. An important challenge for future studies is therefore to

define the regulatory circuits controlling SPI-1 and SPI-2 expression in typhoidal strains.

The effector proteins of the T3SS

To date, over 40 SPI-1 and SPI-2 effectors have been identified in S. Typhimurium (Table 1) (Ramos-

Morales, 2012). These effectors play diverse roles during S. Typhimurium infection, including

manipulation of the host cytoskeleton, and subversion of immune signalling, intracellular trafficking

and cell survival pathways (Ramos-Morales, 2012). Extensive description of effector functions is

beyond the scope of this review, but are well described in others, and are summarised in Table 1.

Importantly however, of the 42 effectors identified in S. Typhimurium, 21 are absent or pseudogenes

within S. Typhi (Table 1). Several of the effectors that are absent in S. Typhi have previously been

described as required for the full virulence of S. Typhimurium in vivo, including SseJ (Ohlson et al.,

2005), SpvB and SpvC (Matsui et al., 2001), SopD2 (Jiang et al., 2004), and SseI and SseK2 (Lawley

et al., 2006). Interestingly there are also differences in the effector repertoire between S. Typhi and S.

Paratyphi: SopE2 and CigR are present in S. Paratyphi but absent in S. Typhi, whilst SteC, SifB and

SspH2 are absent in S. Paratyphi but present in S. Typhi (Table 1).

The smaller effector repertoire found in typhoidal strains may reflect differences between the human

restricted lifestyle of S. Typhi and S. Paratyphi, and the broad host range lifestyle of NTS. For example,

the absence of one T3SS effector – GtgE – is directly linked with S. Typhi host restriction. Unlike S.

Typhimurium, S. Typhi is unable to survive within murine macrophages (Schwan et al., 2000; Spanò

& Galán, 2012). S. Typhimurium replication is supported by GtgE, which prevents recruitment of

Rab29, Rab32 and Rab38 to the SCV, via proteolytic degradation (Spanò et al., 2011; Spanò & Galán,

2012). However, expression of GtgE or depletion of Rab32 allows S. Typhi to survive in murine

macrophages, indicating that it is the absence of GtgE-mediated Rab32 depletion that prevents S. Typhi

growth in the murine host (Spanò & Galán, 2012).

Pseudogenisation of effectors is also likely to contribute to differences in infection. Introduction of

functional S. Typhimurium effectors into S. Typhi in which they are pseudogenes often has marked

This article is protected by copyright. All rights reserved.


phenotypic consequences. For example, expression of S. Typhimurium SopD2 or SopE2 in S. Typhi

reduces invasion (Trombert et al., 2011; Valenzuela et al., 2015), expression of SopA increases

production of the proinflammatory cytokines IL-8 and IL-18 (Valenzuela et al., 2015), whilst expression

of SseJ increases invasion/intracellular replication within epithelial cells (Trombert et al., 2010).

Aside from effectors which are absent or pseudogenes, additional T3SS-associated genes are reported

to be ‘differentially evolved’ between typhoidal and non-typhoidal serovars, namely SipD, SseD, SseC,

SptP, SseF and SifA (Eswarappa et al., 2008). Of the remaining effectors that are common between S.

Typhimurium and S. Typhi, the protein identity varies: only one effector has 100% identity (SsaB),

whilst the lowest sequence identity is 87% (SipD) (Sabbagh et al., 2010). For most of these effectors it

is unknown if sequence differences translate to functional differences. However, the SPI-1 encoded

effector SptP – which has 94% identity between S. Typhi and S. Typhimurium – has been functionally

characterised in both serovars (Johnson et al., 2017).

SptP has a GTPase activating (GAP) and tyrosine phosphatase domains. In S. Typhimurium SptP is

involved in recovering the cytoskeleton following SopE/SopE2-mediated invasion via inactivation of

Cdc42 and Rac1 (Fu & Galán, 1999), and also in SCV maturation (Humphreys et al., 2009). Despite

the assumption that high protein identity correlates to functional similarity, SptP of S. Typhi contains

multiple point mutations which are clustered within its chaperone binding domain, while additionally,

strains belonging to S. Typhi H58 have a loss-of-function mutation within sptP (Wong et al., 2015).

Replacing the chaperone binding domain in S. Typhimurium with that of S. Typhi results in intracellular

instability and loss of SptP function (Johnson et al., 2017). Consistently, no phenotypic consequences

are observed following deletion of sptP in S. Typhi, with regards to cytoskeletal recovery or invasion

efficiency (Johnson et al., 2017). These results suggest that SptP is non-functional in S. Typhi, which

may have wider ramifications when considering additional effectors with varying sequence identities

between S. Typhi and S. Typhimurium.

Importantly, although there is a bias towards discussing the pseudogenisation and absence of T3SS

effectors in S. Typhi, computational or experimental screens to identify novel effectors have, to our

knowledge, not been performed with S. Typhi. However, based on homology to T3SS effectors from

This article is protected by copyright. All rights reserved.


other bacterial pathogens, putative S. Typhi specific effectors have been identified, including STY1076

(NleG family), STY1423 (EspN) and STY1360 (OspB) (Hannemann & Galán, 2017). Of these,

STY1076 (t1865) can be secreted in a SPI-1 and SPI-2 T3SS-dependent manner, and translocated into

host cells, although a functional role during S. Typhi infection has not yet been described (manuscript

in preparation). As half of the currently identified S. Typhimurium effector repertoire is absent in S.

Typhi, there are potentially numerous, as yet unidentified, typhoidal-specific effectors which may be

functionally important during systemic disease.

Conclusions

Combating S. Typhi infections is a priority with WHO and The Bill & Melinda Gates Foundation, which

are part of the Coalition against Typhoid. The spread of XDR S. Typhi (Klemm et al., 2018) poses a

significant risk to human health, particularly in low and middle-income countries. While there is

concerted international effort to develop a vaccine that provides lasting immunity, in the short-term,

treatment of acutely and chronically infected individuals remains a key strategy to combat and contain

S. Typhi. The development of anti-S. Typhi compounds is dependent on achieving a greater

understanding of S. Typhi biology, gene regulation and virulence factors, particularly within the bile-

rich environment of the gallbladder.

This article is protected by copyright. All rights reserved.


References
Antillón, M., Warren, J. L., Crawford, F. W., Weinberger, D. M., Kürüm, E., Pak, G. D., … Pitzer, V.
E. (2017). The burden of typhoid fever in low- and middle-income countries: A meta-regression
approach. PLOS Neglected Tropical Diseases, 11(2), e0005376.
DOI:10.1371/JOURNAL.PNTD.0005376
Baker, S., Hardy, J., Sanderson, K. E., Quail, M., Goodhead, I., Kingsley, R. A., … Dougan, G.
(2007). A novel linear plasmid mediates flagellar variation in Salmonella Typhi. PLoS
Pathogens, 3(5), 0605–0610. DOI:10.1371/journal.ppat.0030059
Baker, S., Holt, K. E., Clements, A. C. A., Karkey, A., Arjyal, A., Boni, M. F., … Farrar, J. J. (2011).
Combined high-resolution genotyping and geospatial analysis reveals modes of endemic urban
typhoid fever transmission. Open Biology, 1(2), 110008. DOI:10.1098/rsob.110008
Baker, S., Holt, K., Van De Vosse, E., Roumagnac, P., Whitehead, S., King, E., … Dougan, G.
(2008). High-Throughput Genotyping of Salmonella enterica Serovar Typhi Allowing
Geographical Assignment of Haplotypes and Pathotypes within an Urban District of Jakarta,
Indonesia. Journal of Clinical Microbiology, 46(5), 1741–1746. DOI:10.1128/JCM.02249-07
Bakowski, M. A., Cirulis, J. T., Brown, N. F., Finlay, B. B., & Brumell, J. H. (2007). SopD acts
cooperatively with SopB during Salmonella enterica serovar Typhimurium invasion. Cellular
Microbiology, 9(12), 2839–2855. DOI:10.1111/j.1462-5822.2007.01000.x
Bayer-Santos, E., Durkin, C. H., Rigano, L. A., Kupz, A., Alix, E., Cerny, O., … Holden, D. W.
(2016). The Salmonella Effector SteD Mediates MARCH8-Dependent Ubiquitination of MHC
II Molecules and Inhibits T Cell Activation. Cell Host & Microbe, 20(5), 584–595.
DOI:10.1016/j.chom.2016.10.007
Begley, M., Gahan, C., & Hill, C. (2005). The interaction between bacteria and bile. FEMS
Microbiology Reviews, 29, 625–651. DOI:10.1016/j.femsre.2004.09.003
Bernal-Bayard, J., Cardenal-Muñoz, E., & Ramos-Morales, F. (2010). The Salmonella type III
secretion effector, salmonella leucine-rich repeat protein (SlrP), targets the human chaperone
ERdj3. The Journal of Biological Chemistry, 285(21), 16360–16368.
DOI:10.1074/jbc.M110.100669
Bernal-Bayard, J., & Ramos-Morales, F. (2009). Salmonella type III secretion effector SlrP is an E3
ubiquitin ligase for mammalian thioredoxin. The Journal of Biological Chemistry, 284(40),
27587–27595. DOI:10.1074/jbc.M109.010363
Beuzón, C. R., Méresse, S., Unsworth, K. E., Ruíz-Albert, J., Garvis, S., Waterman, S. R., … Holden,
D. W. (2000). Salmonella maintains the integrity of its intracellular vacuole through the action
of SifA. The EMBO Journal, 19(13), 3235–3249. DOI:10.1093/emboj/19.13.3235
Bhavsar, A. P., Brown, N. F., Stoepel, J., Wiermer, M., Martin, D. D. O., Hsu, K. J., … Finlay, B. B.
(2013). The Salmonella Type III Effector SspH2 Specifically Exploits the NLR Co-chaperone
Activity of SGT1 to Subvert Immunity. PLoS Pathogens, 9(7), e1003518.
DOI:10.1371/journal.ppat.1003518
Bishop, A., House, D., Perkins, T., Baker, S., Kingsley, R. A., & Dougan, G. (2008). Interaction of
Salmonella enterica serovar Typhi with cultured epithelial cells: roles of surface structures in
adhesion and invasion. Microbiology, 154(Pt 7), 1914–1926. DOI:10.1099/mic.0.2008/016998-0
Brawn, L. C., Hayward, R. D., & Koronakis, V. (2007). Salmonella SPI1 effector SipA persists after
entry and cooperates with a SPI2 effector to regulate phagosome maturation and intracellular
replication. Cell Host & Microbe, 1(1), 63–75. DOI:10.1016/j.chom.2007.02.001
Brenner, F. W., Villar, R. G., Angulo, F. J., Tauxe, R., & Swaminathan, B. (2000). Salmonella
Nomenclature. Journal of Clinical Microbiology, 38(7), 2465–2467.
Bueno, S. M., Santiviago, C. A., Murillo, A. A., Fuentes, J. A., Trombert, A. N., Rodas, P. I., …
Mora, G. C. (2004). Precise Excision of the Large Pathogenicity Island, SPI7, in Salmonella

This article is protected by copyright. All rights reserved.


enterica Serovar Typhi. Journal of Bacteriology, 186(10), 3202–3213.
DOI:10.1128/JB.186.10.3202
Bulgin, R., Raymond, B., Garnett, J. A., Frankel, G., Crepin, V. F., Berger, C. N., & Arbeloa, A.
(2010). Bacterial Guanine Nucleotide Exchange Factors SopE-Like and WxxxE Effectors.
Infection and Immunity, 78(4), 1417–1425. DOI:10.1128/IAI.01250-09
Carden, S. E., Walker, G. T., Honeycutt, J., Lugo, K., Pham, T., Jacobson, A., … Monack, D. (2017).
Pseudogenization of the Secreted Effector Gene sseI Confers Rapid Systemic Dissemination of
S. Typhimurium ST313 within Migratory Dendritic Cells. Cell Host & Microbe, 21(2), 182–
194. DOI:10.1016/j.chom.2017.01.009
CDC. (2015). National Enteric Disease Surveillance: Salmonella Annual Report, 2015.
Chang, S. J., Song, J., & Galán, J. E. (2016). Receptor-Mediated Sorting of Typhoid Toxin during Its
Export from Salmonella Typhi-Infected Cells. Cell Host and Microbe, 20(5), 682–689.
DOI:10.1016/j.chom.2016.10.005
Crawford, R. W., Wangdi, T., Spees, A. M., Xavier, M. N., Tsolis, R. M., & Bäumler, A. J. (2013).
Loss of very-long O-antigen chains optimizes capsule-mediated immune evasion by Salmonella
enterica serovar Typhi. MBio, 4(4). DOI:10.1128/mBio.00232-13
D’Costa, V. M., Braun, V., Landekic, M., Shi, R., Proteau, A., McDonald, L., … Brumell, J. H.
(2015). Salmonella Disrupts Host Endocytic Trafficking by SopD2-Mediated Inhibition of Rab7.
Cell Reports, 12(9), 1508–1518. DOI:10.1016/j.celrep.2015.07.063
Deiwick, J., Salcedo, S. P., Boucrot, E., Gilliland, S. M., Henry, T., Petermann, N., … Méresse, S.
(2006). The translocated Salmonella effector proteins SseF and SseG interact and are required to
establish an intracellular replication niche. Infection and Immunity, 74(12), 6965–6972.
DOI:10.1128/IAI.00648-06
Del Bel Belluz, L., Guidi, R., Pateras, I. S., Levi, L., Mihaljevic, B., Rouf, S. F., … Frisan, T. (2016).
The Typhoid Toxin Promotes Host Survival and the Establishment of a Persistent Asymptomatic
Infection. PLOS Pathogens, 12(4), e1005528. DOI:10.1371/journal.ppat.1005528
Deng, L., Song, J., Gao, X., Wang, J., Yu, H., Chen, X., … Varki, A. (2014). Host adaptation of a
bacterial toxin from the human pathogen salmonella typhi. Cell, 159(6), 1290–1299.
DOI:10.1016/j.cell.2014.10.057
Dobinson, H. C., Gibani, M. M., Jones, C., Thomaides-Brears, H. B., Voysey, M., Darton, T. C., …
Pollard, A. J. (2017). Evaluation of the Clinical and Microbiological Response to Salmonella
Paratyphi A Infection in the First Paratyphoid Human Challenge Model. Clinical Infectious
Diseases, 64(8), 1066–1073. DOI:10.1093/cid/cix042
Domingues, L., Holden, D. W., & Mota, L. J. (2014). The Salmonella effector SteA contributes to the
control of membrane dynamics of Salmonella-containing vacuoles. Infection and Immunity,
82(7), 2923–2934. DOI:10.1128/IAI.01385-13
Domingues, L., Ismail, A., Charro, N., Rodríguez-Escudero, I., Holden, D. W., Molina, M., … Mota,
L. J. (2016). The Salmonella effector SteA binds phosphatidylinositol 4-phosphate for
subcellular targeting within host cells. Cellular Microbiology, 18(7), 949–969.
DOI:10.1111/cmi.12558
Dongol, S., Thompson, C. N., Clare, S., Nga, T. V. T., Duy, P. T., Karkey, A., … Baker, S. (2012).
The Microbiological and Clinical Characteristics of Invasive Salmonella in Gallbladders from
Cholecystectomy Patients in Kathmandu, Nepal. PLoS ONE, 7(10), e47342.
DOI:10.1371/journal.pone.0047342
Dougan, G., & Baker, S. (2014). Salmonella enterica Serovar Typhi and the Pathogenesis of Typhoid
Fever. Annual Review of Microbiology, 68, 317–336. DOI:10.1146/annurev-micro-091313-
103739
Eade, C. R., Hung, C.-C., Bullard, B., Gonzalez-Escobedo, G., Gunn, J. S., & Altier, C. (2016). Bile

This article is protected by copyright. All rights reserved.


Acids Function Synergistically to Repress Invasion Gene Expression in Salmonella by
Destabilizing the Invasion Regulator HilD. Infection and Immunity, 84(8), 2198–2208.
DOI:10.1128/IAI.00177-16
Elhadad, D., Desai, P., Grassl, G. A., McClelland, M., Rahav, G., & Gal-Mor, O. (2016). Differences
in Host Cell Invasion and Salmonella Pathogenicity Island 1 Expression between Salmonella
enterica Serovar Paratyphi A and Nontyphoidal S. Typhimurium. Infection and Immunity, 84(4),
1150–1165. DOI:10.1128/IAI.01461-15
Elhadad, D., McClelland, M., Rahav, G., & Gal-Mor, O. (2015). Feverlike Temperature is a Virulence
Regulatory Cue Controlling the Motility and Host Cell Entry of Typhoidal Salmonella. The
Journal of Infectious Diseases, 212(1), 147–156. DOI:10.1093/infdis/jiu663
Emary, K., Moore, C. E., Chanpheaktra, N., An, K. P., Chheng, K., Sona, S., … Parry, C. M. (2012).
Enteric fever in Cambodian children is dominated by multidrug-resistant H58 Salmonella
enterica serovar Typhi with intermediate susceptibility to ciprofloxacin. Transactions of the
Royal Society of Tropical Medicine and Hygiene, 106(12), 718–724.
DOI:10.1016/j.trstmh.2012.08.007
Eswarappa, S. M., Janice, J., Nagarajan, A. G., Balasundaram, S. V, Karnam, G., Dixit, N. M., &
Chakravortty, D. (2008). Differentially evolved genes of Salmonella pathogenicity islands:
insights into the mechanism of host specificity in Salmonella. PloS One, 3(12), e3829.
DOI:10.1371/journal.pone.0003829
Feasey, N. A., Dougan, G., Kingsley, R. A., Heyderman, R. S., & Gordon, M. A. (2012). Invasive
non-typhoidal Salmonella disease: an emerging and neglected tropical disease in Africa. The
Lancet, 379(9835), 2489–2499. DOI:10.1016/S0140-6736(11)61752-2
Feasey, N. A., Gaskell, K., Wong, V., Msefula, C., Selemani, G., Kumwenda, S., … Heyderman, R.
S. (2015). Rapid Emergence of Multidrug Resistant, H58-Lineage Salmonella Typhi in Blantyre,
Malawi. PLoS Neglected Tropical Diseases, 9(4), e0003748. DOI:10.1371/journal.pntd.0003748
Figueira, R., Watson, K. G., Holden, D. W., & Helaine, S. (2013). Identification of Salmonella
pathogenicity island-2 type III secretion system effectors involved in intramacrophage
replication of S. enterica serovar typhimurium: implications for rational vaccine design. MBio,
4(2), e00065. DOI:10.1128/mBio.00065-13
Finn, C. E., Chong, A., Cooper, K. G., Starr, T., & Steele-Mortimer, O. (2017). A second wave of
Salmonella T3SS1 activity prolongs the lifespan of infected epithelial cells. PLOS Pathogens,
13(4), e1006354. DOI:10.1371/journal.ppat.1006354
Forest, C. G., Ferraro, E., Sabbagh, S. C., & Daigle, F. (2010). Intracellular survival of Salmonella
enterica serovar Typhi in human macrophages is independent of Salmonella pathogenicity island
(SPI)-2. Microbiology, 156(12), 3689–3698. DOI:10.1099/mic.0.041624-0
Frankel, G., Newton, S. M., Schoolnik, G. K., & Stocker, B. A. (1989). Intragenic recombination in a
flagellin gene: characterization of the H1-j gene of Salmonella typhi. The EMBO Journal, 8(10),
3149–3152.
Freeman, J. A., Ohl, M. E., & Miller, S. I. (2003). The Salmonella enterica serovar typhimurium
translocated effectors SseJ and SifB are targeted to the Salmonella-containing vacuole. Infection
and Immunity, 71(1), 418–427. DOI:10.1128/IAI.71.1.418-427.2003
Freeman, J. A., Rappl, C., Kuhle, V., Hensel, M., & Miller, S. I. (2002). SpiC is required for
translocation of Salmonella pathogenicity island 2 effectors and secretion of translocon proteins
SseB and SseC. Journal of Bacteriology, 184(18), 4971–4980. DOI:10.1128/JB.184.18.4971-
4980.2002
Friebel, A., Ilchmann, H., Aepfelbacher, M., Ehrbar, K., Machleidt, W., & Hardt, W. D. (2001). SopE
and SopE2 from Salmonella typhimurium activate different sets of RhoGTPases of the host cell.
The Journal of Biological Chemistry, 276(36), 34035–34040. DOI:10.1074/jbc.M100609200

This article is protected by copyright. All rights reserved.


Fu, Y., & Galán, J. E. (1999). A salmonella protein antagonizes Rac-1 and Cdc42 to mediate host-cell
recovery after bacterial invasion. Nature, 401(6750), 293–297. DOI:10.1038/45829
Galan, J. E., & Curtiss III, R. (1989). Cloning and molecular characterization of genes whose products
allow Salmonella typhimurium to penetrate tissue culture cells. Microbiology, 86, 6383–6387.
Geddes, K., Worley, M., Niemann, G., & Heffron, F. (2005). Identification of new secreted effectors
in Salmonella enterica serovar Typhimurium. Infection and Immunity, 73(10), 6260–6271.
DOI:10.1128/IAI.73.10.6260-6271.2005
Grabe, G. J., Zhang, Y., Przydacz, M., Rolhion, N., Yang, Y., Pruneda, J. N., … Hare, S. A. (2016).
The Salmonella Effector SpvD Is a Cysteine Hydrolase with a Serovar-specific Polymorphism
Influencing Catalytic Activity, Suppression of Immune Responses, and Bacterial Virulence. The
Journal of Biological Chemistry, 291(50), 25853–25863. DOI:10.1074/jbc.M116.752782
Grenz, J. R., Cott Chubiz, J. E., Thaprawat, P., & Slauch, J. M. (2018). HilE Regulates HilD by
Blocking DNA Binding in Salmonella enterica serovar Typhimurium. Journal of Bacteriology,
JB.00750-17. DOI:10.1128/JB.00750-17
Gunn, J. S., Marshall, J. M., Baker, S., Dongol, S., Charles, R. C., & Ryan, E. T. (2014). Salmonella
chronic carriage: epidemiology, diagnosis, and gallbladder persistence. Trends in Microbiology,
22(11), 648–655. DOI:10.1016/j.tim.2014.06.007
Günster, R. A., Matthews, S. A., Holden, D. W., & Thurston, T. L. M. (2017). SseK1 and SseK3 Type
III Secretion System Effectors Inhibit NF-κB Signaling and Necroptotic Cell Death in
Salmonella-Infected Macrophages. Infection and Immunity, 85(3), e00010-17.
DOI:10.1128/IAI.00010-17
Haghjoo, E., & Galán, J. E. (2004). Salmonella typhi encodes a functional cytolethal distending toxin
that is delivered into host cells by a bacterial-internalization pathway. Proceedings of the
National Academy of Sciences of the United States of America, 101(13), 4614–4619.
DOI:10.1073/pnas.0400932101
Hannemann, S., & Galán, J. E. (2017). Salmonella enterica serovar-specific transcriptional
reprogramming of infected cells. PLOS Pathogens, 13(7), e1006532.
DOI:10.1371/journal.ppat.1006532
Haraga, A., & Miller, S. I. (2003). A Salmonella enterica serovar typhimurium translocated leucine-
rich repeat effector protein inhibits NF-kappa B-dependent gene expression. Infection and
Immunity, 71(7), 4052–4058. DOI:10.1128/IAI.71.7.4052-4058.2003
Haraga, A., & Miller, S. I. (2006). A Salmonella type III secretion effector interacts with the
mammalian serine/threonine protein kinase PKN1. Cellular Microbiology, 8(5), 837–846.
DOI:10.1111/j.1462-5822.2005.00670.x
Hardt, W.-D., Chen, L.-M., Schuebel, K. E., Bustelo, X. R., & Galán, J. E. (1998). S. typhimurium
Encodes an Activator of Rho GTPases that Induces Membrane Ruffling and Nuclear Responses
in Host Cells. Cell, 93(5), 815–826. DOI:10.1016/S0092-8674(00)81442-7
Hart, P. J., O’Shaughnessy, C. M., Siggins, M. K., Bobat, S., Kingsley, R. A., Goulding, D. A., …
Maclennan, C. A. (2016). Differential killing of Salmonella enterica serovar Typhi by antibodies
targeting VI and lipopolysaccharide O:9 antigen. PLoS ONE, 11(1).
DOI:10.1371/journal.pone.0145945
Hayashi, F., Smith, K. D., Ozinsky, A., Hawn, T. R., Yi, E. C., Goodlett, D. R., … Aderem, A.
(2001). The innate immune response to bacterial flagellin is mediated by Toll- like receptor 5.
Nature, 410(6832), 1099–103. DOI:10.1038/35074106
Hayward, R. D., & Koronakis, V. (1999). Direct nucleation and bundling of actin by the SipC protein
of invasive Salmonella. The EMBO Journal, 18(18), 4926–4934.
DOI:10.1093/emboj/18.18.4926
Henry, T., Couillault, C., Rockenfeller, P., Boucrot, E., Dumont, A., Schroeder, N., … Meresse, S.

This article is protected by copyright. All rights reserved.


(2006). The Salmonella effector protein PipB2 is a linker for kinesin-1. Proceedings of the
National Academy of Sciences, 103(36), 13497–13502. DOI:10.1073/pnas.0605443103
Hersh, D., Monack, D. M., Smith, M. R., Ghori, N., Falkow, S., & Zychlinsky, A. (1999). The
Salmonella invasin SipB induces macrophage apoptosis by binding to caspase-1. Proceedings of
the National Academy of Sciences of the United States of America, 96(5), 2396–2401.
DOI:10.1073/PNAS.96.5.2396
Hiyoshi, H., Tiffany, C. R., Bronner, D. N., & Bäumler, A. J. (2018a). Typhoidal Salmonella
serovars: Ecological opportunity and the evolution of a new pathovar. FEMS Microbiology
Reviews. DOI:10.1093/femsre/fuy024
Hiyoshi, H., Wangdi, T., Lock, G., Saechao, C., Raffatellu, M., Cobb, B. A., & Bäumler, A. J.
(2018b). Mechanisms to Evade the Phagocyte Respiratory Burst Arose by Convergent Evolution
in Typhoidal Salmonella Serovars. Cell Reports, 22(7), 1787–1797.
DOI:10.1016/j.celrep.2018.01.016
Ho, T. D., Figueroa-Bossi, N., Wang, M., Uzzau, S., Bossi, L., & Slauch, J. M. (2002). Identification
of GtgE, a novel virulence factor encoded on the Gifsy-2 bacteriophage of Salmonella enterica
serovar Typhimurium. Journal of Bacteriology, 184(19), 5234–5239.
DOI:10.1128/JB.184.19.5234-5239.2002
Hodak, H., & Galán, J. E. (2013). A Salmonella Typhi homologue of bacteriophage muramidases
controls typhoid toxin secretion. EMBO Reports, 14(1), 95–102. DOI:10.1038/embor.2012.186
Holt, K. E., Dolecek, C., Chau, T. T., Duy, P. T., La, T. T. P., Hoang, N. V. M., … Baker, S. (2011).
Temporal fluctuation of multidrug resistant salmonella typhi haplotypes in the mekong river
delta region of Vietnam. PLoS Neglected Tropical Diseases, 5(1), e929.
DOI:10.1371/journal.pntd.0000929
HSE. (2013). The Approved List of biological agents.
Humphreys, D., Hume, P. J., & Koronakis, V. (2009). The Salmonella effector SptP dephosphorylates
host AAA+ ATPase VCP to promote development of its intracellular replicative niche. Cell Host
& Microbe, 5(3), 225–233. DOI:10.1016/j.chom.2009.01.010
Ibarra, J. A., Knodler, L. A., Sturdevant, D. E., Virtaneva, K., Carmody, A. B., Fischer, E. R., …
Steele-Mortimer, O. (2010). Induction of Salmonella pathogenicity island 1 under different
growth conditions can affect Salmonella-host cell interactions in vitro. Microbiology, 156(Pt 4),
1120–1133. DOI:10.1099/mic.0.032896-0
Jackson, L. K., Nawabi, P., Hentea, C., Roark, E. A., & Haldar, K. (2008). The Salmonella virulence
protein SifA is a G protein antagonist. Proceedings of the National Academy of Sciences,
105(37), 14141–14146. DOI:10.1073/pnas.0801872105
Jiang, X., Rossanese, O. W., Brown, N. F., Kujat-Choy, S., Galán, J. E., Finlay, B. B., & Brumell, J.
H. (2004). The related effector proteins SopD and SopD2 from Salmonella enterica serovar
Typhimurium contribute to virulence during systemic infection of mice. Molecular
Microbiology, 54(5), 1186–1198. DOI:10.1111/j.1365-2958.2004.04344.x
Johnson, R., Byrne, A., Berger, C. N., Klemm, E., Crepin, V. F., Dougan, G., & Frankel, G. (2017).
The type III secretion system effector SptP of Salmonella enterica serovar Typhi. Journal of
Bacteriology, 199(4), e00647-16. DOI:10.1128/JB.00647-16
Johnson, R., Ravenhall, M., Pickard, D., Dougan, G., Byrne, A., & Frankel, G. (2018). Comparison of
Salmonella enterica serovars Typhi and Typhimurium reveals typhoidal-specific responses to
bile. Infection and Immunity, 86(3), e00490-17. DOI:10.1128/IAI.00490-17
Jones, R. M., Wu, H., Wentworth, C., Luo, L., Collier-Hyams, L., & Neish, A. S. (2008). Salmonella
AvrA Coordinates Suppression of Host Immune and Apoptotic Defenses via JNK Pathway
Blockade. Cell Host & Microbe, 3(4), 233–244. DOI:10.1016/j.chom.2008.02.016
Kamanova, J., Sun, H., Lara-Tejero, M., & Galán, J. E. (2016). The Salmonella Effector Protein SopA

This article is protected by copyright. All rights reserved.


Modulates Innate Immune Responses by Targeting TRIM E3 Ligase Family Members. PLOS
Pathogens, 12(4), e1005552. DOI:10.1371/journal.ppat.1005552
Kaniga, K., Trollinger, D., & Galán, J. E. (1995). Identification of two targets of the type III protein
secretion system encoded by the inv and spa loci of Salmonella typhimurium that have
homology to the Shigella IpaD and IpaA proteins. Journal of Bacteriology, 177(24), 7078–7085.
Klemm, E. J., Shakoor, S., Page, A. J., Qamar, F. N., Judge, K., Saeed, D. K., … Hasan, R. (2018).
Emergence of an Extensively Drug-Resistant Salmonella enterica Serovar Typhi Clone
Harboring a Promiscuous Plasmid Encoding Resistance to Fluoroquinolones and Third-
Generation Cephalosporins. MBio, 9(1), e00105-18. DOI:10.1128/mBio
Knodler, L. A., Finlay, B. B., & Steele-Mortimer, O. (2005). The Salmonella effector protein SopB
protects epithelial cells from apoptosis by sustained activation of Akt. The Journal of Biological
Chemistry, 280(10), 9058–9064. DOI:10.1074/jbc.M412588200
Knodler, L. A., & Steele-Mortimer, O. (2005). The Salmonella effector PipB2 affects late
endosome/lysosome distribution to mediate Sif extension. Molecular Biology of the Cell, 16(9),
4108–4123. DOI:10.1091/mbc.E05-04-0367
Knodler, L. A., Vallance, B. A., Hensel, M., Jäckel, D., Finlay, B. B., & Steele-Mortimer, O. (2003).
Salmonella type III effectors PipB and PipB2 are targeted to detergent-resistant microdomains
on internal host cell membranes. Molecular Microbiology, 49(3), 685–704.
Kortmann, J., Brubaker, S. W., & Monack, D. M. (2015). Cutting Edge: Inflammasome Activation in
Primary Human Macrophages Is Dependent on Flagellin. The Journal of Immunology, 195(3),
815–819. DOI:10.4049/jimmunol.1403100
Kuhle, V., & Hensel, M. (2002). SseF and SseG are translocated effectors of the type III secretion
system of Salmonella pathogenicity island 2 that modulate aggregation of endosomal
compartments. Cellular Microbiology, 4(12), 813–824.
Kujat Choy, S. L., Boyle, E. C., Gal-Mor, O., Goode, D. L., Valdez, Y., Vallance, B. A., & Finlay, B.
B. (2004). SseK1 and SseK2 Are Novel Translocated Proteins of Salmonella enterica Serovar
Typhimurium. Infection and Immunity, 72(9), 5115–5125. DOI:10.1128/IAI.72.9.5115-
5125.2004
Lawley, T. D., Chan, K., Thompson, L. J., Kim, C. C., Govoni, G. R., & Monack, D. M. (2006).
Genome-wide screen for Salmonella genes required for long-term systemic infection of the
mouse. PLoS Pathogens, 2(2), e11. DOI:10.1371/journal.ppat.0020011
Lesnick, M. L., Reiner, N. E., Fierer, J., & Guiney, D. G. (2001). The Salmonella spvB virulence gene
encodes an enzyme that ADP-ribosylates actin and destabilizes the cytoskeleton of eukaryotic
cells. Molecular Microbiology, 39(6), 1464–1470. DOI:10.1046/j.1365-2958.2001.02360.x
Lilic, M., Galkin, V. E., Orlova, A., VanLoock, M. S., Egelman, E. H., & Stebbins, C. E. (2003).
Salmonella SipA Polymerizes Actin by Stapling Filaments with Nonglobular Protein Arms.
Science, 301(5641), 1918–1921. DOI:10.1126/science.1088433
Lin, S. L., Le, T. X., & Cowen, D. S. (2003). SptP, a Salmonella typhimurium type III-secreted
protein, inhibits the mitogen-activated protein kinase pathway by inhibiting Raf activation.
Cellular Microbiology, 5(4), 267–275.
Lin, Z., Zhang, Y.-G., Xia, Y., Xu, X., Jiao, X., & Sun, J. (2016). Salmonella enteritidis Effector
AvrA Stabilizes Intestinal Tight Junctions via the JNK Pathway. The Journal of Biological
Chemistry, 291(52), 26837–26849. DOI:10.1074/jbc.M116.757393
Lossi, N. S., Rolhion, N., Magee, A. I., Boyle, C., & Holden, D. W. (2008). The Salmonella SPI-2
effector SseJ exhibits eukaryotic activator-dependent phospholipase A and glycerophospholipid :
cholesterol acyltransferase activity. Microbiology, 154(Pt 9), 2680–2688.
DOI:10.1099/mic.0.2008/019075-0
Matsui, H., Bacot, C. M., Garlington, W. A., Doyle, T. J., Roberts, S., & Gulig, P. A. (2001).

This article is protected by copyright. All rights reserved.


Virulence plasmid-borne spvB and spvC genes can replace the 90-kilobase plasmid in conferring
virulence to Salmonella enterica serovar Typhimurium in subcutaneously inoculated mice.
Journal of Bacteriology, 183(15), 4652–4658. DOI:10.1128/JB.183.15.4652-4658.2001
Mazurkiewicz, P., Thomas, J., Thompson, J. A., Liu, M., Arbibe, L., Sansonetti, P., & Holden, D. W.
(2008). SpvC is a Salmonella effector with phosphothreonine lyase activity on host mitogen-
activated protein kinases. Molecular Microbiology, 67(6), 1371–1383. DOI:10.1111/j.1365-
2958.2008.06134.x
McClelland, M., Sanderson, K. E., Clifton, S. W., Latreille, P., Porwollik, S., Sabo, A., … Wilson, R.
K. (2004). Comparison of genome degradation in Paratyphi A and Typhi, human-restricted
serovars of Salmonella enterica that cause typhoid. Nature Genetics, 36(12), 1268–1274.
DOI:10.1038/ng1470
McClelland, M., Sanderson, K. E., Spieth, J., Clifton, S. W., Latreille, P., Courtney, L., … Wilson, R.
K. (2001). Complete genome sequence of Salmonella enterica serovar Typhimurium LT2.
Nature, 413(6858), 852–856. DOI:10.1038/35101614
McLaughlin, L. M., Govoni, G. R., Gerke, C., Gopinath, S., Peng, K., Laidlaw, G., … Monack, D.
(2009). The Salmonella SPI2 Effector SseI Mediates Long-Term Systemic Infection by
Modulating Host Cell Migration. PLoS Pathogens, 5(11), e1000671.
DOI:10.1371/journal.ppat.1000671
Mesquita, F. S., Thomas, M., Sachse, M., Santos, A. J. M., Figueira, R., & Holden, D. W. (2012). The
Salmonella Deubiquitinase SseL Inhibits Selective Autophagy of Cytosolic Aggregates. PLoS
Pathogens, 8(6), e1002743. DOI:10.1371/journal.ppat.1002743
Mogasale, V., Maskery, B., Ochiai, R. L., Lee, J. S., Mogasale, V. V, Ramani, E., … Wierzba, T. F.
(2014). Burden of typhoid fever in low-income and middle-income countries: a systematic,
literature-based update with risk-factor adjustment. The Lancet Global Health, 2(10), e570-80.
DOI:10.1016/S2214-109X(14)70301-8
Myeni, S. K., Wang, L., & Zhou, D. (2013). SipB-SipC Complex Is Essential for Translocon
Formation. PLoS ONE, 8(3), e60499. DOI:10.1371/journal.pone.0060499
Myeni, S. K., & Zhou, D. (2010). The C terminus of SipC binds and bundles F-actin to promote
Salmonella invasion. The Journal of Biological Chemistry, 285(18), 13357–13363.
DOI:10.1074/jbc.M109.094045
Nath, G., Singh, H., & Shukla, V. K. (1997). Chronic typhoid carriage and carcinoma of the
gallbladder. European Journal of Cancer Prevention, 6(6).
Niemann, G. S., Brown, R. N., Gustin, J. K., Stufkens, A., Shaikh-Kidwai, A. S., Li, J., … Heffron, F.
(2011). Discovery of novel secreted virulence factors from Salmonella enterica serovar
Typhimurium by proteomic analysis of culture supernatants. Infection and Immunity, 79(1), 33–
43. DOI:10.1128/IAI.00771-10
Ochman, H., Soncini, F. C., Solomon, F., & Groisman, E. A. (1996). Identification of a pathogenicity
island required for Salmonella survival in host cells. Proceedings of the National Academy of
Sciences of the United States of America, 93, 7800–7804.
Odendall, C., Rolhion, N., Förster, A., Poh, J., Lamont, D. J., Liu, M., … Holden, D. W. (2012). The
Salmonella kinase SteC targets the MAP kinase MEK to regulate the host actin cytoskeleton.
Cell Host & Microbe, 12(5), 657–668. DOI:10.1016/j.chom.2012.09.011
Ohlson, M. B., Fluhr, K., Birmingham, C. L., Brumell, J. H., & Miller, S. I. (2005). SseJ deacylase
activity by Salmonella enterica serovar Typhimurium promotes virulence in mice. Infection and
Immunity, 73(10), 6249–6259. DOI:10.1128/IAI.73.10.6249-6259.2005
Parkhill, J., Dougan, G., James, K. D., Thomson, N. R., Pickard, D., Wain, J., … Barrell, B. G.
(2001). Complete genome sequence of a multiple drug resistant Salmonella enterica serovar
Typhi CT18. Nature, 413(6858), 848–852. DOI:10.1038/35101607

This article is protected by copyright. All rights reserved.


Parry, C., Hien, T. T., Dougan, G., White, N. J., & Farrar, J. J. (2002). Typhoid Fever. The New
England Journal of Medicine, 347(22), 1770–1782. DOI:10.1056/NEJMra020201
Pickard, D., Wain, J., Baker, S., Line, A., Chohan, S., Fookes, M., … Dougan, G. (2003).
Composition, acquisition, and distribution of the Vi exopolysaccharide-encoding Salmonella
enterica pathogenicity island SPI-7. Journal of Bacteriology, 185(17), 5055–5065.
DOI:10.1128/JB.185.17.5055-5065.2003
Pilar, A. V. C., Reid-Yu, S. A., Cooper, C. A., Mulder, D. T., & Coombes, B. K. (2012). GogB Is an
Anti-Inflammatory Effector that Limits Tissue Damage during Salmonella Infection through
Interaction with Human FBXO22 and Skp1. PLoS Pathogens, 8(6), e1002773.
DOI:10.1371/journal.ppat.1002773
Pitzer, V. E., Bowles, C. C., Baker, S., Kang, G., Balaji, V., Farrar, J. J., & Grenfell, B. T. (2014).
Predicting the Impact of Vaccination on the Transmission Dynamics of Typhoid in South Asia:
A Mathematical Modeling Study. PLoS Neglected Tropical Diseases, 8(1), e2642.
DOI:10.1371/journal.pntd.0002642
Poh, J., Odendall, C., Spanos, A., Boyle, C., Liu, M., Freemont, P., & Holden, D. W. (2008). SteC is a
Salmonella kinase required for SPI-2-dependent F-actin remodelling. Cellular Microbiology,
10(1), 20–30. DOI:10.1111/j.1462-5822.2007.01010.x
Prouty, A. M., & Gunn, J. S. (2000). Salmonella enterica Serovar Typhimurium Invasion Is Repressed
in the Presence of Bile. Infection and Immunity, 68(12), 6763–6769.
DOI:10.1128/IAI.68.12.6763-6769.2000
Raffatellu, M., Chessa, D., Wilson, R. P., Dusold, R., Rubino, S., & Bäumler, A. J. (2005a). The Vi
Capsular Antigen of Salmonella enterica Serotype Typhi Reduces Toll-Like Receptor-
Dependent Interleukin-8 Expression in the Intestinal Mucosa. INFECTION AND IMMUNITY,
73(6), 3367–3374. DOI:10.1128/IAI.73.6.3367–3374.2005
Raffatellu, M., Wilson, R. P., Chessa, D., Andrews-Polymenis, H., Tran, Q. T., Lawhon, S., …
Bäumler, A. J. (2005b). SipA, SopA, SopB, SopD, and SopE2 contribute to Salmonella enterica
serotype typhimurium invasion of epithelial cells. Infection and Immunity, 73(1), 146–154.
DOI:10.1128/IAI.73.1.146-154.2005
Ramos-Morales, F. (2012). Impact of Salmonella enterica Type III Secretion System Effectors on the
Eukaryotic Host Cell. ISRN Cell Biology, 2012, 1–36. DOI:10.5402/2012/787934
Reyes Ruiz, V. M., Ramirez, J., Naseer, N., Palacio, N. M., Siddarthan, I. J., Yan, B. M., … Shin, S.
(2017). Broad detection of bacterial type III secretion system and flagellin proteins by the human
NAIP/NLRC4 inflammasome. Proceedings of the National Academy of Sciences of the United
States of America, 114(50), 13242–13247. DOI:10.1073/pnas.1710433114
Rolhion, N., Furniss, R. C. D., Grabe, G., Ryan, A., Liu, M., Matthews, S. A., & Holden, D. W.
(2016). Inhibition of Nuclear Transport of NF-ĸB p65 by the Salmonella Type III Secretion
System Effector SpvD. PLOS Pathogens, 12(5), e1005653. DOI:10.1371/journal.ppat.1005653
Ruiz-Albert, J., Yu, X.-J., Beuzón, C. R., Blakey, A. N., Galyov, E. E., & Holden, D. W. (2002).
Complementary activities of SseJ and SifA regulate dynamics of the Salmonella typhimurium
vacuolar membrane. Molecular Microbiology, 44(3), 645–661. DOI:10.1046/j.1365-
2958.2002.02912.x
Rytkönen, A., Poh, J., Garmendia, J., Boyle, C., Thompson, A., Liu, M., … Holden, D. W. (2007).
SseL, a Salmonella deubiquitinase required for macrophage killing and virulence. Proceedings
of the National Academy of Sciences of the United States of America, 104(9), 3502–3507.
DOI:10.1073/pnas.0610095104
Sabbagh, S. C., Forest, C. G., Lepage, C., Leclerc, J.-M., & Daigle, F. (2010). So similar, yet so
different: uncovering distinctive features in the genomes of Salmonella enterica serovars
Typhimurium and Typhi. FEMS Microbiology Letters, 305(1), 1–13. DOI:10.1111/j.1574-
6968.2010.01904.x

This article is protected by copyright. All rights reserved.


Sabbagh, S. C., Lepage, C., McClelland, M., & Daigle, F. (2012). Selection of Salmonella enterica
Serovar Typhi Genes Involved during Interaction with Human Macrophages by Screening of a
Transposon Mutant Library. PLoS ONE, 7(5), e36643. DOI:10.1371/journal.pone.0036643
Saul, A., Smith, T., & Maire, N. (2013). Stochastic Simulation of Endemic Salmonella enterica
Serovar Typhi: The Importance of Long Lasting Immunity and the Carrier State. PLoS ONE,
8(9), e74097. DOI:10.1371/journal.pone.0074097
Schreiber, F., Kay, S., Frankel, G., Clare, S., Goulding, D., van de Vosse, E., … Baker, S. (2015). The
Hd, Hj, and Hz66 flagella variants of Salmonella enterica serovar Typhi modify host responses
and cellular interactions. Scientific Reports, 5(1), 7947. DOI:10.1038/srep07947
Schwan, W. R., Huang, X.-Z., Hu, L., & Kopecko, D. J. (2000). Differential Bacterial Survival,
Replication, and Apoptosis-Inducing Ability of Salmonella Serovars within Human and Murine
Macrophages. Infection and Immunity, 68(3), 1005–1013. DOI:10.1128/IAI.68.3.1005-
1013.2000
Sharma, A., & Qadri, A. (2004). Vi polysaccharide of Salmonella typhi targets the prohibitin family
of molecules in intestinal epithelial cells and suppresses early inflammatory responses.
Proceedings of the National Academy of Sciences of the United States of America, 101(50),
17492–17497. DOI:10.1073/pnas.0407536101
Song, J., Gao, X., & Galán, J. E. (2013). Structure and function of the Salmonella Typhi chimaeric
A2B5 typhoid toxin. Nature, 499(7458), 350–354. DOI:10.1038/nature12377
Song, J., Willinger, T., Rongvaux, A., Eynon, E. E., Stevens, S., Manz, M. G., … Galán, J. E. (2010).
A mouse model for the human pathogen Salmonella typhi. Cell Host & Microbe, 8(4), 369–376.
DOI:10.1016/j.chom.2010.09.003
Spanò, S., & Galán, J. E. (2012). A Rab32-dependent pathway contributes to Salmonella typhi host
restriction. Science, 338(6109), 960–963. DOI:10.1126/science.1229224
Spanò, S., Liu, X., & Galán, J. E. (2011). Proteolytic targeting of Rab29 by an effector protein
distinguishes the intracellular compartments of human-adapted and broad-host Salmonella.
Proceedings of the National Academy of Sciences, 108(45), 18418–18423.
DOI:10.1073/pnas.1111959108
Spanò, S., Ugalde, J. E., & Galán, J. E. (2008). Delivery of a Salmonella Typhi Exotoxin from a Host
Intracellular Compartment. Cell Host and Microbe, 3(1), 30–38.
DOI:10.1016/j.chom.2007.11.001
Sturm, A., Heinemann, M., Arnoldini, M., Benecke, A., Ackermann, M., Benz, M., … Hardt, W.-D.
(2011). The Cost of Virulence: Retarded Growth of Salmonella Typhimurium Cells Expressing
Type III Secretion System 1. PLoS Pathogens, 7(7), e1002143.
DOI:10.1371/journal.ppat.1002143
Suez, J., Porwollik, S., Dagan, A., Marzel, A., Schorr, Y. I., Desai, P. T., … Gal-Mor, O. (2013).
Virulence Gene Profiling and Pathogenicity Characterization of Non-Typhoidal Salmonella
Accounted for Invasive Disease in Humans. PLoS ONE, 8(3).
DOI:10.1371/journal.pone.0058449
Sun, H., Kamanova, J., Lara-Tejero, M., & Galán, J. E. (2016). A Family of Salmonella Type III
Secretion Effector Proteins Selectively Targets the NF-κB Signaling Pathway to Preserve Host
Homeostasis. PLOS Pathogens, 12(3), e1005484. DOI:10.1371/journal.ppat.1005484
Tobe, T., Beatson, S. A., Taniguchi, H., Abe, H., Bailey, C. M., Fivian, A., … Pallen, M. J. (2006).
An extensive repertoire of type III secretion effectors in Escherichia coli O157 and the role of
lambdoid phages in their dissemination. Proceedings of the National Academy of Sciences,
103(40), 14941–14946. DOI:10.1073/pnas.0604891103
Trombert, A. N., Berrocal, L., Fuentes, J. A., & Mora, G. C. (2010). S. Typhimurium sseJ gene
decreases the S. Typhi cytotoxicity toward cultured epithelial cells. BMC Microbiology, 10, 312.

This article is protected by copyright. All rights reserved.


DOI:10.1186/1471-2180-10-312
Trombert, A. N., Rodas, P. I., & Mora, G. C. (2011). Reduced invasion to human epithelial cell lines
of Salmonella enterica serovar Typhi carrying S. Typhimurium sopD2. FEMS Microbiology
Letters, 322(2), 150–156. DOI:10.1111/j.1574-6968.2011.02347.x
Tsolis, R. M., Townsend, S. M., Miao, E. A., Miller, S. I., Ficht, T. A., Adams, L. G., & Bäumler, A.
J. (1999). Identification of a putative Salmonella enterica serotype typhimurium host range
factor with homology to IpaH and YopM by signature-tagged mutagenesis. Infection and
Immunity, 67(12), 6385–6393.
Tsolis, R. M., Xavier, M. N., Santos, R. L., & Bäumler, A. J. (2011). How to become a top model:
impact of animal experimentation on human Salmonella disease research. Infection and
Immunity, 79(5), 1806–1814. DOI:10.1128/IAI.01369-10
Uchiya, K., Barbieri, M. A., Funato, K., Shah, A. H., Stahl, P. D., & Groisman, E. A. (1999). A
Salmonella virulence protein that inhibits cellular trafficking. The EMBO Journal, 18(14), 3924–
3933. DOI:10.1093/emboj/18.14.3924
Valenzuela, L. M., Hidalgo, A. A., Rodríguez, L., Urrutia, I. M., Ortega, A. P., Villagra, N. A., …
Fuentes, J. A. (2015). Pseudogenization of sopA and sopE2 is functionally linked and
contributes to virulence of Salmonella enterica serovar Typhi. Infection, Genetics and Evolution,
33, 131–142. DOI:10.1016/j.meegid.2015.04.021
Virlogeux, I., Waxin, H., Ecobichon, C., & Popoff, M. Y. (1995). Role of the viaB locus in synthesis,
transport and expression of Salmonella typhi Vi antigen. Microbiology, 141(12), 3039–3047.
DOI:10.1099/13500872-141-12-3039
Wangdi, T., Lee, C.-Y., Spees, A. M., Yu, C., Kingsbury, D. D., Winter, S. E., … Bäumler, A. J.
(2014). The Vi Capsular Polysaccharide Enables Salmonella enterica Serovar Typhi to Evade
Microbe-Guided Neutrophil Chemotaxis. PLoS Pathogens, 10(8), e1004306.
DOI:10.1371/journal.ppat.1004306
Wilson, R. P., Raffatellu, M., Chessa, D., Winter, S. E., Tükel, Ç., & Bäumler, A. J. (2008). The Vi-
capsule prevents Toll-like receptor 4 recognition of Salmonella. Cellular Microbiology, 10(4),
876–890. DOI:10.1111/j.1462-5822.2007.01090.x
Wilson, R. P., Winter, S. E., Spees, A. M., Winter, M. G., Nishimori, J. H., Sanchez, J. F., …
Bäumler, A. J. (2011). The Vi capsular polysaccharide prevents complement receptor 3-
mediated clearance of Salmonella enterica serotype Typhi. Infection and Immunity, 79(2), 830–
837. DOI:10.1128/IAI.00961-10
Winter, S. E., Raffatellu, M., Wilson, P. R., Rüssmann, H., & Bäumler, A. J. (2008). The Salmonella
enterica serotype Typhi regulator TviA reduces interleukin-8 production in intestinal epithelial
cells by repressing flagellin secretion. Cellular Microbiology, 10(1), 247–261.
DOI:10.1111/j.1462-5822.2007.01037.x
Winter, S. E., Winter, M. G., Atluri, V., Poon, V., Romão, E. L., Tsolis, R. M., & Bäumler, A. J.
(2015). The Flagellar Regulator TviA Reduces Pyroptosis by Salmonella enterica Serovar
Typhi. Infection and Immunity, 83(4), 1546–1555. DOI:10.1128/IAI.02803-14
Winter, S. E., Winter, M. G., Thiennimitr, P., Gerriets, V. A., Nuccio, S. P., Rüssmann, H., &
Bäumler, A. J. (2009). The TviA auxiliary protein renders the Salmonella enterica serotype
Typhi RcsB regulon responsive to changes in osmolarity. Molecular Microbiology, 74(1), 175–
193. DOI:10.1111/j.1365-2958.2009.06859.x
Wong, V., Baker, S., Pickard, D., Parkhill, J., Page, A., Feasey, N., … Dougan, G. (2015).
Phylogeographical analysis of the dominant multidrug-resistant H58 clade of Salmonella Typhi
identifies inter- and intracontinental transmission events. Nature Genetics, 47(6), 632–639.
DOI:10.1038/ng.3281
Wood, M. W., Rosqvist, R., Mullan, P. B., Edwards, M. H., & Galyov, E. E. (1996). SopE, a secreted

This article is protected by copyright. All rights reserved.


protein of Salmonella dublin, is translocated into the target eukaryotic cell via a sip-dependent
mechanism and promotes bacterial entry. Molecular Microbiology, 22(2), 327–338.
DOI:10.1046/j.1365-2958.1996.00116.x
Wu, H., Jones, R. M., & Neish, A. S. (2012). The Salmonella effector AvrA mediates bacterial
intracellular survival during infection in vivo. Cellular Microbiology, 14(1), 28–39.
DOI:10.1111/j.1462-5822.2011.01694.x
Yan, M., Li, X., Liao, Q., Li, F., Zhang, J., & Kan, B. (2016). The emergence and outbreak of
multidrug-resistant typhoid fever in China. Emerging Microbes & Infections, 5(6), e62.
DOI:10.1038/emi.2016.62
Yang, Y. A., Lee, S., Zhao, J., Thompson, A. J., McBride, R., Tsogtbaatar, B., … Song, J. (2018). In
vivo tropism of Salmonella Typhi toxin to cells expressing a multiantennal glycan receptor.
Nature Microbiology, 3(2), 155–163. DOI:10.1038/s41564-017-0076-4
Yang, Z., Soderholm, A., Lung, T. W. F., Giogha, C., Hill, M. M., Brown, N. F., … Teasdale, R. D.
(2015). SseK3 Is a Salmonella Effector That Binds TRIM32 and Modulates the Host’s NF-κB
Signalling Activity. PLoS ONE, 10(9), e0138529. DOI:10.1371/journal.pone.0138529
Yu, X.-J., Liu, M., & Holden, D. W. (2016). Salmonella Effectors SseF and SseG Interact with
Mammalian Protein ACBD3 (GCP60) To Anchor Salmonella-Containing Vacuoles at the Golgi
Network. MBio, 7(4), e00474-16. DOI:10.1128/mBio.00474-16
Zhang, Y., Higashide, W. M., McCormick, B. A., Chen, J., & Zhou, D. (2006). The inflammation-
associated Salmonella SopA is a HECT-like E3 ubiquitin ligase. Molecular Microbiology, 62(3),
786–793. DOI:10.1111/j.1365-2958.2006.05407.x

This article is protected by copyright. All rights reserved.


Figure 1. Molecular pathogenesis of S. Typhi. S. Typhi uses an array of virulence factors during

infection, which are distinct from NTS. S. Typhi secretes a pool of effectors into host cells through the

SPI-1 T3SS promoting invasion. However, various effectors present in S. Typhimurium are absent or

pseudogenes in S. Typhi including SptP and GtgE. On the other hand, S. Typhi-specific effectors (e.g.

This article is protected by copyright. All rights reserved.


t1865) are probably associated with S. Typhi pathogenesis. Oxygen availability, feverlike temperatures

(42°C) and the presence of bile results in differential SPI-1 T3SS regulation between typhoidal and

NTS. While the role of SPI-2 T3SS during infection with S. Typhi remains unclear, S. Typhi also lacks

a number of SPI-2 T3SS-dependent effectors found in S. Typhimurium. S. Typhi expresses the Vi

antigen which hinders antibody binding and complement activation, as well as recognition of LPS by

TLR4 and flagellin by TLR5 resulting in reduced downstream TLR-mediated signalling. Vi production

is under the control of TviA which induces expression of the Vi-encoding locus named viaB. Through

the action of TviA, which negatively regulates flagellar and SPI-1-associated genes, the presence of

cytosolic flagellin, as well as SPI-1 T3SS components, is decreased, reducing recognition by NAIP

receptors. This leads to reduced NLRC4 inflammasome and induction of pyroptosis compared to S.

Typhimurium. Another virulence factor associated with typhoidal Salmonella is the typhoid toxin,

which is expressed when S. Typhi is intracellular. It is exported in vesicles originating from the SCV

into the extracellular space, where it can then bind to Neu5Ac-terminated receptors on target cells,

inducing G2/M cell cycle arrest and/or cell death. The action of typhoid toxin results in reduced

circulating neutrophils. Dashed lines depict reduced activation of pathways compared to S.

Typhimurium.

This article is protected by copyright. All rights reserved.


Table 1. The T3SS effector repertoire in S. Typhimurium, S. Typhi and S. Paratyphi
T3SS Effector Genomic S. S. Typhi S. Paratyphi A Function/Comment
location Typhimurium Ty2 (CT18) ATCC9150
LT2
SPI-1 SopB SPI-5 STM1091 t1828 SPA1759 Inositol phosphatase that activates the kinase Akt, inhibiting
(SigD) (STY1121) apoptosis of infected cells both at early and late time points during
infection (Finn et al., 2017; Knodler et al., 2005). Contributes to
Salmonella invasion (Raffatellu et al., 2005b).
SopA STM2066 t0808 P SPA0805P E3 ubiquitin ligase which promotes polymorphonuclear leukocyte
(STY2275)P transmigration and stimulates immune signalling via members of the
TRIM E3 ligases (Kamanova et al., 2016; Zhang et al., 2006).
Pseudogenisation in S. Typhi linked with pseudogenisaton of sopE2
(Valenzuela et al., 2015)
SipA SPI-1 STM2882 t2784 SPA2740 Promotes actin polymerisation near adherent bacteria, contributes to
(STY3005) Salmonella invasion (Lilic et al., 2003; Raffatellu et al., 2005b).
Persists in infected cells to regulate SCV morphology in cooperation
with SifA (Brawn et al., 2007).
SipB SPI-1 STM2885 t2787 SPA2743 SPI-1 T3SS translocon component (Myeni et al., 2013). Induces
(STY3008) apoptotic cell death in macrophages via binding to caspase-1 (Hersh
et al., 1999)
SipC SPI-1 STM2884 t2786 SPA2742 SPI-1 T3SS translocon component (Myeni et al., 2013). Bundles and
(STY3007) nucleates actin promoting Salmonella invasion (Hayward &
Koronakis, 1999; Myeni & Zhou, 2010).
SipD SPI-1 STM2883 t2785 SPA2741 SPI-1 T3SS translocon component. Required for effector
(STY3006) translocation into host cells (Kaniga et al., 1995). Differentially
evolved between typhoidal and non-typhoidal serovars (Eswarappa et
al., 2008)
SptP SPI-1 STM2878 t2780 SPA2736 GTPase-activating protein domain containing effector that
(StpA) (STY3001) antagonises the activity of SopE/SopE2 to promote host cytoskeletal
recovery (Fu & Galán, 1999). Inhibits MAPK signalling (Lin et al.,
2003). Promotes intracellular replication at late stages of infection
(Humphreys et al., 2009). Differentially evolved between typhoidal
and non-typhoidal serovars (Eswarappa et al., 2008), unstable in S.
Typhi (Johnson et al., 2017).
SopD STM2945 t2846 SPA2801 Functions cooperatively with SopB to manipulate host membrane
(STY3073) dynamics (Bakowski et al., 2007). Contributes to Salmonella invasion
(Raffatellu et al., 2005b).

This article is protected by copyright. All rights reserved.


SopE SopEɸ Absent* t4303 SPA2564 Guanine exchange factors that activate Rac1 and Cdc42 to
bacteriophage (SL1344_2674) (STY4609) manipulate the host cytoskeleton and promote invasion (Friebel et al.,
(within SPI-7 2001; Hardt et al., 1998; Wood et al., 1996). sopE2 is not a
in S. Typhi) pseudogene within Paratyphi A (McClelland et al., 2004)
SopE2 Bacteriophage STM1855 t1023 P SPA1018
remnant (STY1987)P
SPI-2 SseF SPI-2 STM1404 t1272 SPA1449 Interacts with SseG to regulate SCV positioning close to the Golgi
(STY1716) network (Deiwick et al., 2006; Kuhle & Hensel, 2002; Yu et al.,
2016). Differentially evolved between typhoidal and non-typhoidal
serovars (Eswarappa et al., 2008)
SseG SPI-2 STM1405 t1273 SPA1448 Interacts with SseF to regulate SCV positioning close to the Golgi
(STY1715) network (Deiwick et al., 2006; Kuhle & Hensel, 2002; Yu et al.,
2016).
SteD STM2139 t0718 SPA0713 Inhibits MHC class II presentation on infected cells, inhibits T cell
(STY2367) activation (Bayer-Santos et al., 2016; Niemann et al., 2011)
GogB Bacteriophage STM2584 Absent Absent E3 ubiquitin ligase that inhibits NF-κB signalling by inhibiting the
(Gifsy-1) activity of host SCF ubiquitin ligase (Pilar et al., 2012).
SseK1 STM4157 Absent Absent Related effectors that inhibit NF-κB signalling (Günster et al., 2017;
SseK2 STM2137 Absent Absent Kujat Choy et al., 2004; Yang et al., 2015).
SseK3 Bacteriophage Absent* Absent Absent
(ST64B) (SL1344_1928)
SteC STM1698 t1612 SPA1178P Kinase the phosphorylates MAP kinases to induce actin formation
(STY1353) around SCVs and regulate intracellular replication (Geddes et al.,
2005; Odendall et al., 2012; Poh et al., 2008)
PipB SPI-5 STM1088 t1830 SPA1762 Localises to SCVs and Sifs (Knodler et al., 2003).
(STY1117)
PipB2 STM2780 t2674 SPA2636 Localises to SCVs and Sifs (Knodler et al., 2003). Recruits kinesin-1
(STY2897) to SCV membranes (Henry et al., 2006). Reorganises host endosomes
and lysosomes promoting the extension of Sifs (Knodler & Steele-
Mortimer, 2005).
SifA STM1224 t1696 SPA1626 Directs recruitment of LAMP1 enriched membranes. Required for the
(STY1264) maintenance and growth of the SCV and Sifs (Beuzón et al., 2000;
Jackson et al., 2008; Ruiz-Albert et al., 2002). Differentially evolved
between typhoidal and non-typhoidal serovars (Eswarappa et al.,
2008)
SifB STM1602 t1511 SPA1266P Member of the WxxxE family of effectors. Localises to the SCV
(STY1462) (Bulgin et al., 2010; Freeman et al., 2003). Pseudogene in Paratyphi
A (McClelland et al., 2004)

This article is protected by copyright. All rights reserved.


SopD2 STM0972 t1962P SPA1826P Interacts with Rab7 to regulate endosomal trafficking and SCV
(STY0971)P membrane dynamics (D’Costa et al., 2015). Expression of S.
Typhimurium sopD2 in S. Typhi reduces invasion (Trombert et al.,
2011)
SpiC SPI-2 STM1393 t1261 SPA1460 Involved in the translocation of SPI-2 proteins. Reported to inhibit
(SsaB) (STY1727) endosomal trafficking (Freeman et al., 2002; Uchiya et al., 1999)
SseI (SrfH) Bacteriophage STM1051 Absent Absent Interacts with IQGAP1 resulting in inhibition of macrophage and
(Gifsy-2) dendritic cell migration (McLaughlin et al., 2009). Pseudogenisation
promotes systemic dissemination (Carden et al., 2017)
SseJ STM1631 t1534 P Absent Regulates SCV membrane dynamics alongside SifA (Lossi et al.,
(STY1439)P 2008; Ruiz-Albert et al., 2002). Expression of S. Typhimurium SseJ
in S. Typhi results in increased intracellular replication (Trombert et
al., 2010)
SseL STM2287 t0576 SPA0576 Deubiquitinase that inhibits autophagic events in infected cells and is
(STY2517) required for macrophage killing (Mesquita et al., 2012; Rytkönen et
al., 2007)
SspH2 Bacteriophage STM2241 t0623 Absent E3 ubiquitin ligase that interacts with SGT1 and ubiquitinates Nod1,
remnant (STY2467) modulating immune signalling (Bhavsar et al., 2013)
SpvB pSLT pSLT039 Absent Absent ADP ribosyltransferase that depolymerises actin filaments (Lesnick et
al., 2001; Matsui et al., 2001)
SpvC pSLT pSLT038 Absent Absent Phosphothreonine lyase that inhibits MAPK signalling (Matsui et al.,
2001; Mazurkiewicz et al., 2008)
CigR SPI-3 STM3762 t3756P SPA3612 Unknown function. Required for replication in bone-marrow derived
(STY4024)P macrophages (Figueira et al., 2013; Niemann et al., 2011). Frameshift
mutation not found in Paratyphi A
GtgA Bacteriophage STM1026 Absent Absent Protease closely related to PipA and GogA. Redundantly inhibits NF-
(Gifsy-2) κB signalling (Niemann et al., 2011; Sun et al., 2016)
SPI-1/ AvrA SPI-1* STM2865 Absent Absent Acetyltransferase that inhibits JNK and NF-κB signalling, and
SPI-2 dampens proapoptotic immune responses. Also stabilises intestinal
tight junctions (Jones et al., 2008; Lin et al., 2016; Wu et al., 2012)
*Absent from SPI-1 in typhoidal strains (Sabbagh et al., 2010)
SlrP STM0800 t2087 P SPA1952P E3 ubiquitin ligase which mediates ubiquitination of mammalian
(STY0833)P thioredoxin and interacts with the chaperone ERdj3, leading to cell
death (Bernal-Bayard et al., 2010; Bernal-Bayard & Ramos-Morales,
2009; Tsolis et al., 1999)
SspH1 Bacteriophage Absent* Absent Absent E3 ubiquitin ligase that interacts with the serine/threonine protein
(Gifsy-3) (STM14_1483) kinase PKN1 and inhibits NF-κB signalling (Haraga & Miller, 2003,
2006)

This article is protected by copyright. All rights reserved.


SteA STM1583 t1493 SPA1285 Binds to phosphatidylinositol 4-phosphate (PI(4)P), localises to SCV
(STY1482) membranes and to Sifs. Implicated in the control of SCV membrane
dynamics (Domingues et al., 2014, 2016; Geddes et al., 2005)
SteB STM1629 Absent Absent Unknown function (Geddes et al., 2005)
SteE Bacteriophage STM2585 Absent Absent Unknown function (Niemann et al., 2011)
(Gifsy-1)
SpvD pSLT pSLT037 Absent Absent Cysteine protease that inhibits NF-κB signalling (Grabe et al., 2016;
Niemann et al., 2011; Rolhion et al., 2016)
GtgE Bacteriophage STM1055 Absent Absent Proteolytically targets Rab29, Rab32, and Rab38, promoting
(Gifsy-2) replication inside murine macrophages. Absence in typhoidal strains
reported to mediate host restriction (Ho et al., 2002; Spanò et al.,
2011; Spanò & Galán, 2012)
Unknown StoD Absent t1865 Absent Homologue of NleG effector family of E3 ubiquitin ligases (Tobe et
(STY1076) al., 2006). Unknown function.
PipA SPI-5 STM1087 t1831 SPA1763 Closely related proteases that are also closely related to GtgA.
(STY1115) Redundantly inhibit NF-κB signalling (Sun et al., 2016).
GogA STM2614 Absent Absent
P
= pseudogene; * = absent from S. Typhimurium LT2, where possible the locus tog of another S. Typhimurium reference (SL1344 or 14028) is given; grey
shading emphasises were differences are apparent between serovars

This article is protected by copyright. All rights reserved.

Вам также может понравиться