You are on page 1of 24

DOMAIN 8 PATHOGENESIS

Adaptive Immune Responses


during Salmonella Infection
LISA A. CUMMINGS,1 BROOKE L. DEATHERAGE,2 AND
BRAD T. COOKSON1,2
1
Department of Laboratory Medicine, University of Washington, Seattle, WA 98195
2
Department of Microbiology, University of Washington, Seattle, WA 98195

ABSTRACT The interaction between Salmonella and its host is complex and dynamic: the
host mounts an immune defense against the pathogen, which in turn acts to reduce,
evade, or exploit these responses to successfully colonize the host. Although the exact
mechanisms mediating protective immunity are poorly understood, it is known that T cells
are a critical component of immunity to Salmonella infection, and a robust T-cell response
is required for both clearance of primary infection and resistance to subsequent challenge.
B-cell functions, including but not limited to antibody production, are also required for
generation of protective immunity. Additionally, interactions among host cells are
essential. For example, antigen-presenting cells (including B cells) express cytokines that
Received: 21 August 2008 participate in CD4+ T cell activation and differentiation. Differentiated CD4+ T cells secrete
Accepted: 17 November 2008 cytokines that have both autocrine and paracrine functions, including recruitment and
Posted: 17 September 2009 activation of phagocytes, and stimulation of B cell isotype class switching and affinity
Supercedes previous posting at EcoSal.org. maturation. Multiple bacterium-directed mechanisms, including altered antigen expression
Editor: Michael S. Donnenberg, University of and bioavailability and interference with antigen-presenting cell activation and function,
Maryland, School of Medicine, Baltimore, MD combine to modify Salmonella’s “pathogenic signature” in order to minimize its susceptibility
Citation: EcoSal Plus 2013; doi:10.1128/ to host immune surveillance. Therefore, a more complete understanding of adaptive
ecosalplus.8.8.11. immune responses may provide insights into pathogenic bacterial functions. Continued
Correspondence: Brad T. Cookson: cookson@u. identification of adaptive immune targets will guide rational vaccine development, provide
washington.edu
insights into host functions required to resist Salmonella infection, and correspondingly
Copyright: © 2013 American Society for
Microbiology. All rights reserved. provide valuable reagents for defining the critical pathogenic capabilities of Salmonella
doi:10.1128/ecosalplus.8.8.11 that contribute to their success in causing acute and chronic infections.

INTRODUCTION
The significance of salmonellosis as a medical problem is readily appreci-
ated by noting that vaccination efforts to protect against the disease began
in the latter part of the 19th century (1). Six decades later, murine models
of Salmonella infection, in which Salmonella enterica serovar Typhimurium
infection in mice served as a model for Salmonella enterica serovar Typhi
infection in humans, began to provide important new insights. Greater
protection was afforded by sublethal infection with viable bacteria than by
administration of heat-killed vaccines (2). Protection generated by live
organisms did not correlate with anti-lipopolysaccharide (anti-LPS)

ASMScience.org/EcoSalPlus 1
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

Figure 1 Disease outcome is the cumulative result of Salmonella interactions with host adaptive immune functions. During Salmonella
infection, there are several points at which pathogen and host immune cells interact. Multiple outcomes for each interaction are possible; the
predominant outcome is determined not only by the virulence of the bacterium, but also the innate resistance or susceptibility of the host, and
previous exposure of the host to the pathogen (host immune status). Uptake of bacteria by APCs (A) results in either APC elimination by
pyroptosis (B) or APC survival (C). Pyroptosis leads to release of the inflammatory cytokines IL-1β and IL-18, and possibly releases bacteria or
bacterial Ags for uptake by bystander APCs. Some APCs, such as macrophages, are capable of destroying intracellular bacteria (D). If APCs
survive, they may be able to process and present Ag in the context of MHC (E). Salmonella interferes with this process via multiple mechanisms
including repression of Ag expression, bacterial surface modifications that reduce Ag bioavailability and APC stimulation/maturation, and other
SPI-2 -dependent processes that mediate bacterial survival within the phagosome (F). Protected from antibody detection, intracellular Salmonella
can utilize APCs as vehicles for systemic dissemination and replication (G). If APCs are able to overcome bacterial interference to process and
present Ag to T cells (E), Salmonella may still inhibit T-cell activation via stimulation of nitric oxide (NO) production and other direct,
suppressive effects (H). Recognition of peptide-MHC on APCs by TCR-expressing naïve T cells leads to activation and expansion of Ag-specific
effector T cells (I). Effector CD4+ T cells provide help for the activation of CD8+ CTLs (leading to cytokine production and lysis of infected host
cells [J]), and B cells (K). B cells and T cells work synergistically: T cells provide help for antibody production, isotype class switching, and cytokine
production by B cells, while B-cell cytokine production supports Th-1 T-cell differentiation, and Ig on B-cell surfaces mediate Ag capture for
processing and presentation to T cells (K). Cytokines such as IFN-γ are produced by effector T cells to further enhance APC function and activate

2 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

antibody responses (2), and passive administration of The generation of protective immunity in the naïve host
antibody prolonged survival but did not afford com- relies on the reactive milieu of innate immune responses
plete protection. Subsequent studies indicated that T elicited at the initiation of bacterial infection by DCs, and
cells contributed to protective immunity (3, 4, 5). Thus, to a lesser extent, macrophages. The inflammatory re-
the principle that both cellular and humoral responses sponse critically benefits the host by helping to generate
were required for protection was established (6). Im- B- and T-cell responses, but recruitment of phagocytic
portantly, this concept was confirmed in 1993 by the cells to the site of initial bacterial invasion and replication
demonstration that adoptive transfer of immune serum may facilitate systemic bacterial spread (15). In addition,
and T cells provided protection to naïve, susceptible Salmonella are capable of destroying both macrophages
hosts (7). and DCs by an inflammatory process called pyroptosis
(see Chapter Host Cell Death) (16, 17, 18), which
Immunity requiring both T-cell responses and immu- abrogates their important APC function(s) and thereby
noglobulins (Igs) from B cells is consistent with our antagonizes initiation of the adaptive immune response
understanding of Salmonella as a facultatively intracel- (Fig. 1B and 2C). Thus, Salmonella infection readily kills
lular pathogen. Bacteria encounter phagocytes either the naïve susceptible murine host. In contrast, the ob-
within the gastrointestinal tract (8), or during penetra- servation that Salmonella also causes chronic coloniza-
tion of the mucosal epithelium (9). Salmonella-specific Ig tion (19, 20), made famous by Typhoid Mary in the last
produced by B cells can opsonize extracellular bacteria century, indicates that Salmonella have evolved mecha-
for phagocytic uptake. Salmonella then spread systemi- nisms to finely balance these seemingly opposing cellular
cally within phagocytes (9) to colonize target organs reactions.
such as liver and spleen (10, 11). Salmonella is able to
survive and replicate within macrophages and dendritic Despite recent advances, many aspects of salmonellosis
cells (DCs) in vivo. Inside these professional antigen- and immunity to Salmonella infection remain enigmatic.
presenting cells (APCs), major histocompatibility com- The mechanisms by which virulent Salmonella evades
plexes (MHCs) serve as receptors surveying the host primary immune responses to mount lethal infections in
cellular cytosol and vacuolar compartments for the pre- naïve hosts are incompletely understood, as are the
sence of foreign peptides. When bound by MHC, these means by which live attenuated bacteria generate pro-
peptides are presented on the APC surface for recogni- tective immunity. The identity of Ags recognized by the
tion by T-cell antigen receptors (TCRs). In addition to adaptive immune response, and the precise roles of T
macrophages and DCs, B cells can also act as APCs. cells and B cells in providing immunity are likewise
These cells also express MHC, and surface-expressed Ig poorly defined. However, our view is that this system
promotes high-efficiency antigen (Ag) capture for Ag provides a rich model for defining significant and fun-
presentation. TCR recognition of Ag presented by APCs damental aspects of the immunopathogenesis of host-
leads to activation and expansion of CD4+ T cells. In pathogen interactions. Both genetic (21, 22) and acquired
addition to Ig production, B cells express cytokines that (23) immune defects lead to increased susceptibility to
appear to participate in the differentiation and function salmonellosis, which is responsible for the annual loss of
of effector CD4+ T cells (12). Activated CD4+ T cells in hundreds of thousands of lives (24). Salmonella infec-
turn provide critical stimulation for B cells to undergo tions therefore remain a contemporary scientific problem
expansion and Ig class switching, as well as for activation of substantial medical and economic significance mea-
and growth of cytotoxic CD8+ T cells and phagocytic sured annually in the billions of dollars worldwide (25,
cells such as macrophages (13). Thus, a pathogen whose 26). Here, we highlight aspects of adaptive immune re-
virulence requires survival inside host phagocytic cells sponses to Salmonella that shed critical light on our
(14) provides a contextual framework for understanding current state of understanding and provide a foundation
adaptive immunity (Fig. 1). to help address important questions in the future.

bacterial degradation by macrophages (L). In an immune host, previously primed Ag-specific memory T cells (M) may be activated by APCs that
process and present Ag (N); activation of these cells and their effector functions (O) is much more rapid than for naïve T cells. In addition,
circulating antibodies primed by previous immunization facilitate bacterial uptake via opsonization (P), accelerating the efficiency of Ag
presentation up to 1,000-fold. Thus, the ultimate outcome of infection is the cumulative result of complex interactions between pathogen and
host.

ASMScience.org/EcoSalPlus 3
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

Figure 2 Development of adaptive immune responses during Salmonella infection. Interactions with Salmonella and its host are dynamic and
complex. Flagellated and nonflagellated Salmonella are present in the gut lumen (A), where they must overcome initial barriers including the
glycocalyx layer, antimicrobial peptides, and sIgA. Salmonella cross the epithelium (B) via M cells, by inducing endocytosis in epithelial cells, or
following uptake by CX3CR1+ DCs. Bacteria in the gut lumen (A), within epithelial cells (B) and in the PP (C) express FliC protein; interaction of
FliC with TLR5 initiates an inflammatory response characterized by production of IL-8 and CCL20 by epithelial cells. Release of inflammatory
mediators triggers infiltration of macrophages, neutrophils, and CCR6+ DC. Interaction of these cells with Salmonella results in at least three
possible outcomes: (1) Phagocytosis of bacteria by DC or macrophages, ultimately resulting in inflammatory cell death (flagellin-dependent
pyroptosis). Pyroptosis eliminates potential APCs, leads to release of the inflammatory cytokines IL-1β and IL-18, and possibly releases Ags for
uptake by bystander APCs (note that flagellin-negative bacteria have a reduced ability to trigger inflammation via TLR5 or pyroptosis). (2) Uptake
of bacteria, which persist within the phagocyte. This interaction can lead to production of NO by APCs (inhibitory for T-cell activation) and
upregulation of MHC and costimulatory molecules on DC. In addition, these cells could provide a means of transport to systemic sites such as the
liver and spleen. (3) Bacteria are phagocytosed by neutrophils or macrophages and degraded. Mature CCR6+ DC in the PP (C) process and
present Ags to naïve T cells; Ags acquired for processing and presentation are restricted to those expressed by bacteria in the gut lumen (A) or PP
(C), or Ags that are present in gut lumen, and disassociated from the bacterial soma ([A] MVs, flagellin). Mature DCs that have processed and
presented Ag on surface MHC enter into an “activation feedback” loop with naïve T cells: TNF-α and IL-12 produced by DCs enhance activation
and expansion of Ag-specific T cells, while IFN-γ secretion by activated T cells further stimulates DC function. Memory T cells primed in the PP
(C) express the α4β7-homing receptor as well as CCR6+, predisposing these cells to traffic to the inflamed gut during a secondary infection.

4 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

ANTIGEN-PRESENTING CELLS subpopulations (32, 33, 34). Macrophages are also capa-
APCs Are Required for the Development of ble of priming naïve Salmonella-specific T-cell responses
Ag-Specific T-Cell Responses in vivo (35). Nevertheless, these two cell types may play
different roles in the development of adaptive immunity
The detection of bacterial Ags by T cells and B cells via
(29, 36).
their variable receptor molecules (TCR and BCR, re-
spectively) is the cornerstone of adaptive immune rec-
DCs are phagocytic cells, but bacterial uptake induces a
ognition. Unlike B cells, T cells do not recognize bacterial
morphological and functional transition from immature
proteins in their native state. Instead, bacterial Ags must
Ag-capturing cells to mature APCs (31). Intracellular
be processed into peptides and presented by professional
Salmonella persist but do not replicate within bone
APCs on specialized surface display structures called
marrow-derived DCs (37), and DCs express low levels of
MHCs. In general, peptides derived from extracellular
lysosomal proteases, degrading bacterial proteins more
pathogens internalized by the host cell or pathogens
slowly than macrophages. (38). Therefore, the environ-
replicating in intracellular vacuoles are displayed on
ment within the mature DC is optimized for Ag
MHC-II molecules. Correspondingly, peptides derived
processing and presentation rather than killing bacteria.
from pathogens replicating in the host cell cytosol are
DCs reside in the subepithelial area of the Peyer’s patches
displayed on MHC-I molecules. Presentation on MHC-II
(PPs), ideally situated for an initial encounter with in-
is required for activation of CD4+ T cells, while CD8+ T
vading Salmonella. In contrast, relatively few macro-
cells respond to peptides presented in the context of
phages reside in the PP (39). After oral infection,
MHC-I molecules. Thus, the mammalian host has
colocalization of Salmonella with DCs in the PP and
evolved mechanisms for immune surveillance of both
mesenteric lymph nodes (MLNs) occurs rapidly (39, 40).
extracellular and intracellular pathogens. The importance
In addition, CCR6+ DCs colocalize with Salmonella-
of APC function in the development of immunity to
specific T cells in the PP suggesting that DCs are the
Salmonella infection, and the mechanisms by which Sal-
primary APCs at early stages of infection (41). DCs in-
monella interfere with this function are discussed below.
fected in vitro are capable of priming Salmonella-specific
CD4+ and CD8+ T-cell responses after adoptive transfer
DCs and Macrophages as Professional APCs to naïve mice (30, 42), demonstrating the potential of
during Salmonella Infection DCs to prime naïve T cells in vivo. Indeed, DCs are
Because of their ability to process and present Ag to T absolutely required for activation and expansion of Sal-
cells, professional APCs are critical for immunity to monella-specific T cells in the PP (41). Taken together
Salmonella infection. Macrophages and DCs are classified these findings confirm that DCs play a critical role in the
as professional APCs, although B cells may also serve this initiation of adaptive immunity to Salmonella.
function (reference 27, and see “B-cell functions in ad-
dition to antibody production,” below). Upon Salmonella Although macrophages are classified as professional
encounter in vitro, both macrophages and DCs are APCs and are capable of presenting Salmonella-derived
stimulated to upregulate the expression of molecules re- Ags to T cells (28, 43, 44, 45), it is unclear whether this is
quired for T-cell activation: costimulatory surface mole- their primary function during Salmonella infection. Hu-
cules CD40, CD80, and CD86 and immunomodulatory man macrophages appear to be more efficient in the
cytokines gamma interferon (IFN-γ), tumor necrosis production of proinflammatory chemokines (CCl5,
factor alpha (TNF-α), interleukin 12 (IL-12), and IL-18 CCL20, and CXCL10) and cytokines (IL-18, TNF-α) than
(28, 29, 30). DCs are well established activators of naïve T DCs (29), whereas DCs are more efficient than macro-
cells (31), and recent advances have begun to reveal a phages in stimulating IFN-γ production by CD4+ T cells
complex and diverse array of functionally distinct (28). This suggests that macrophages may be more

Bacteria within APCs disseminate to MLN (D), where Ag presentation to T cells can also occur. Bacteria in the MLN have undergone complete
adaptations to the intracellular environment: they no longer express FliC, actively reduce Ag bioavailability, and interfere with APC function (see
Fig. 1). Dissemination to systemic sites such as liver and spleen (E) follows MLN colonization. Bacteria replicate within APCs at systemic sites.
However, in naïve hosts, T-cell responses to Ags expressed by intracellular phase bacteria are generally not sufficient in magnitude or quality, or
fail to develop rapidly enough, to combat infection. Further, T cells primed at early stages in the PP (C) will not recognize bacteria growing
intracellularly at systemic sites (E), or that fail to express FliC in the PP (C, left).

ASMScience.org/EcoSalPlus 5
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

important in mediating proinflammatory responses and serovar Typhimurium display a bistability phenotype in
that DCs contribute more to Th1-type adaptive re- which a significant number of bacteria in the population
sponses. The effective generation of Salmonella-specific are flagellin negative (51). Additional regulation of bio-
CD8+ T-cell responses and protection against subsequent availability occurs during the transition from extracellu-
challenge in macrophage-depleted mice (46) support lar to intracellular environments within the host, as
the idea that APCs other than macrophages elicit serovar Typhimurium first restrict FliC expression to an
T-cell-mediated protection. Importantly, depletion of intracellular bacterial compartment (50) and then repress
macrophages from immune mice prior to challenge sig- FliC expression below the threshold required for T-cell
nificantly impairs protection as measured by bacterial activation (52). Notably, Ag sequestration alone is not
load in systemic organs (47). Thus, macrophages may sufficient to prevent processing and presentation of this
mediate protection primarily via bacterial clearance Ag by DCs; modifications of the bacterial surface are also
rather than by APC function (36). It is also possible that required (50). The global regulator PhoP, which is acti-
Ag presentation by macrophages may be more important vated during intracellular growth (53) and required for
for stimulation of previously activated effector T cells bacterial survival in the phagosome (54), is one mediator
rather than naïve T cells (35). of these modifications. PhoP regulates extensive re-
modeling of the bacterial surface which reduces its in-
flammatory properties, increases resistance to cationic
Bacterial Interference with APC Function antimicrobial peptides, and alters membrane permeabil-
As described above, adaptive immune responses to Sal- ity (55, 56, 57, 58). In the context of these modifications,
monella infection require the function of professional the processing and presentation of bacterial Ags re-
APCs. Surprisingly, Salmonella virulence depends on the stricted to intracellular compartments is significantly
ability of this pathogen to survive and replicate within the reduced (50). Importantly, repression of FliC expression
very cells required to mount immune responses against by intracellular-phase serovar Typhimurium is also PhoP
it. This can be explained by the fact that coordinated dependent (52, 59). Therefore, at a time when Salmonella
bacterial responses have evolved in the context of a host is residing and replicating within APCs, the organism
immune system in which a generalized innate response to coordinately regulates the expression of genes essential
bacteria leads to the development of a highly specific for both intracellular survival and the restriction of Ag
adaptive response and memory. This coevolutionary bioavailability to APCs.
dynamic between pathogen and host has facilitated the
development of multiple mechanisms by which Salmo- In addition to alterations in Ag bioavailability, Salmo-
nella can subvert or evade APC function (references 48 nella is able to directly interfere with APC function. For
and 49; also discussed below). The ability of Salmonella example, Salmonella causes caspase-1-mediated pro-
to simultaneously survive within APCs and interfere with inflammatory death (pyroptosis) of both macrophages
the presentation of Ags to T cells contributes to virulence: (16, 17) and DCs (18, 60) (see Chapter Nitric Oxide in
activation of adaptive immunity is inhibited while APCs Salmonella and Escherichia coli Infections). Pyroptosis
are exploited as protected reservoirs of replication and represents one potential mechanism (destruction of
means for bacterial dissemination. APCs) for avoiding the activation of adaptive immune
responses, but may also facilitate host responses by pro-
In order for bacterial Ags to be acquired, processed, and viding a source of antigenic material for bystander APCs
presented by APCs to T cells, they must be expressed by (61, 62). Salmonella also affects DC maturation: intra-
bacteria in accessible cellular compartments and at bio- cellular-phase Salmonella inefficiently stimulates MHC-
logically significant levels. Together, these characteristics II upregulation, costimulatory CD86 expression, and
have been described as Ag bioavailability (50). Bacterial secretion of proinflammatory cytokines such as TNF-α
regulation of Ag bioavailability has been most extensively and IL-12 (50). Virulent Salmonella may also inhibit Ag
studied for the dominant Ag FliC, a flagellar subunit processing and presentation on MHC-II by delaying
protein that is abundantly expressed on the bacterial phagosome-lysosome fusion (63). This inhibition re-
surface. Serovar Typhimurium actively regulates FliC quires expression of genes encoded in the Salmonella
bioavailability during infection. The first level of this Pathogenicity Island 2 (SPI2) and does not depend on Ag
regulation exists at the level of nongenetic variation; even uptake, MHC-II upregulation, or APC viability (40, 49).
under conditions permissive for flagellar expression, The ability of Salmonella to interfere with APC function

6 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

may at least partially account for host restriction, because express TCRs consisting of one α- and one β-chain. αβ T
only those serovars able to effectively prevent Ag pro- cells can be further grouped according to the expression
cessing and presentation by DCs within a specific host of CD4 or CD8 coreceptors. CD4+ αβ T cells recognize
can limit activation of adaptive immune responses (64). bacterial peptides from endocytic or phagocytic vacuoles
Importantly, it appears that Salmonella actively inhibits presented in the context of MHC-II molecules. They
APC function. SPI-2 mediated inhibition of Ag pro- function as “helper” T cells by producing cytokines that
cessing and presentation requires live infection (40), and stimulate B-cell antibody class switching, activation and
live but not heat-killed PhoPc mutant bacteria (which growth of cytotoxic T cells, and activation of phagocytes
constitutively express PhoP in its activated state) inhibit such as macrophages (13). CD4+ T cells have been tra-
Ag processing by activated macrophages (45). Therefore, ditionally divided into Th1 and Th2 functional subsets
at least some of the mechanisms that reduce Ag bio- based on expression ratios of various cytokines that lead
availability (PhoP-regulated surface modifications and to control of different classes of pathogens. However,
repressed Ag expression) also reduce the ability of APCs additional CD4+ T-cell subsets have recently been
to process and present Ag to T cells. Finally, Salmonella is identified (71, 72, 73, 74). CD8+ αβ T cells recognize
able to directly inhibit T-cell activation independent of bacterial peptides from the cytosol presented by MHC-I
Ag processing and presentation (65), possibly by down- molecules. CD8+ T cells function as cytotoxic or “killer”
regulation of TCR β-chain expression (66). T cells that lyse infected host cells (13), and like CD4+ T
cells, are capable of secreting cytokines. In addition to αβ
Summary: APCs and Salmonella-Specific Immunity T cells, a small percentage of T cells express a TCR
The processing and presentation of Ags to T cells is a consisting of one γ- and one δ-chain and are concen-
critical step in the development of adaptive immunity. trated in epithelial tissues, particularly in the gut mucosa
Importantly, Salmonella bacteria are not simply passive (75). γδ T cells express TCRs of limited diversity com-
collections of Ags, but rather actively modulate its in- pared with αβ T cells, do not appear to require MHC
teractions with host cells. Multiple bacterium-directed presentation of protein Ags for their activation, and may
mechanisms, including altered Ag expression and bio- function in both innate and adaptive immune responses
availability, and interference with APC activation and (75).
function, combine to modify Salmonella’s “pathogenic
signature” in order to minimize susceptibility to immune Because infection of susceptible hosts with virulent Sal-
surveillance (Fig. 1B, F, and H). monella is rapidly fatal, most studies of the dynamics of
T-cell responses are performed with attenuated bacteria
(76). Salmonella-specific CD4+ and CD8+ T cells are
T CELLS generated during infections with attenuated bacteria in
mice (77, 78, 79) and humans (80, 81, 82, 83, 84). T-cell
T Cells Are a Critical Component of Immunity to activation is quickly initiated after oral infection in mu-
Salmonella Infection cosal lymphoid tissues (85, 86). This is followed by the
As a facultatively intracellular pathogen, Salmonella is expansion of a Salmonella-specific effector population in
capable of replicating within host cells (10, 11), protected the lymphoid tissues that is capable of redistribution to
from recognition by circulating antibodies. Therefore, a nonlymphoid sites of infection (Fig. 2C) (77, 79, 80).
robust T-cell response is required for both clearance of IFN-γ-producing CD4+ and CD8+ T cells can persist for
primary infection and resistance to subsequent challenge several months in mice and humans (43, 69, 82). In ad-
(67, 68, 69, 70). This section discusses the relative con- dition to αβ T cells, γδ T cells can also be detected after
tribution of various T-cell subsets to Salmonella-specific Salmonella infection; a subset is activated at early stages
immunity, the potential mechanisms by which they of infection via the MHC-Ib molecule Qa-1 (87, 88).
mediate protection, and the identity of the Ags they
recognize. Many studies have confirmed that functional T cells are
required for the resolution of both primary and sec-
T-Cell Subsets and Salmonella Immunity ondary Salmonella infections. For example, mice lacking
T lymphocytes recognize Ag via heterodimeric receptors mature αβ T cells are severely impaired in their ability to
(TCRs) expressed on their surface. The majority of T cells control primary oral infection with attenuated aroA
circulating throughout the peripheral lymphoid system Salmonella (67, 88, 89, 70). Interestingly, T-cell function

ASMScience.org/EcoSalPlus 7
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

is not absolutely required in early stages of infection and to naïve mice compared with CD8+ replete control cells
it appears that T-cell-independent mechanisms largely (68, 69).
mediate control of primary serovar Typhimurium in-
fection for up to 21 days (67, 90, 70). Immunized mice The contribution of γδ T cells to Salmonella immunity
that have been depleted of T cells are susceptible to remains unclear. In one study, γδ knockout mice were as
secondary virulent challenge despite antibody production capable as wild-type mice of clearing infection with an
(67, 68, 69). Furthermore, transfer of immune T cells aroA-attenuated serovar Typhimurium strain (67); sim-
(with serum) (7, 69) or T-cell lines derived from immune ilar results were observed after infection with Salmonella
mice (without serum) (91, 92) confers protection on enterica serovar Dublin (70). Other groups detected
naïve susceptible hosts. slight defects in control of primary oral infection in γδ T-
cell-deficient mice, and increased numbers of γδ T cells
The pathogenesis of Salmonella infections is reflected in in the intestinal epithelium during infection (88). The
the particular T-cell subsets most critical for host im- latter observation, together with the fact that γδ T cells
munity. Because Salmonella replicates within phagocytic home to the gut mucosa (75), may indicate that γδ T cells
vacuoles of infected cells it is not surprising that there is have some function during early stages of Salmonella
an absolute requirement for CD4+ T cells in the control infection before systemic spread.
of both primary and secondary Salmonella infection (67,
68, 69). The importance of functional CD4+ T cells is
highlighted by the increased frequency and severity of
nontyphoidal Salmonella infections correlating with re- Mechanisms of T-Cell-Mediated Immunity
duced CD4+ T-cell counts in patients suffering from HIV to Salmonella Infection
infection (93, 94, 95, 96, 97, 98). In addition, CD4+ T Although T cells are required for successful resolution of
cells also appear to actively suppress serovar Typhimu- Salmonella infections, the mechanisms whereby they
rium during chronic infection, as demonstrated by in- contribute to immunity are poorly understood. It is well
creased recurrence of serovar Typhimurium bacteremia established that T cells generated in response to primary
in HIV patients (23, 99), or reactivation of infection in Salmonella infection are capable of producing IFN-γ and
CD4+ T-cell-depleted mice (100). TNF-α (79). Studies using IFN-γ or TNF-α gene knockout
mice or antibody depletion of IFN-γ or TNF-α confirm
CD8+ T cells are generated during Salmonella infection of the importance of these cytokines in control and resolu-
mice and humans, and are restricted not only by MHC-Ia tion of Salmonella infection (67, 68, 103, 104, 105, 106),
molecules, but also nonclassical MHC-Ib molecules (81, although the source of cytokines in these studies is not
101, 102). There is evidence to suggest that Salmonella- clearly defined. These data support the idea that cytokine
specific CD8+ T cells participate in protective immune production is the primary means by which T cells con-
responses. During primary infection with attenuated tribute to immunity, and substantial evidence supports a
aroA Salmonella, MHC-I-deficient mice (lacking func- correlation between CD4+ T-cell function and IFN-γ
tional CD8+ T cells) have higher bacterial loads during production. For example, CD4+ T-cell knockout mice,
primary oral infection than wild-type mice (87). Fur- which are unable to clear an infection with an aroA-at-
thermore, T-cell receptor knockout mice (no functional tenuated serovar Typhimurium strain, produce 100-fold
CD4+ or CD8+ T cells) suffer from more severe infection less IFN-γ than control mice (67). CD4+ T cells from mice
than CD4+ T-cell-deficient MHC-II knockouts (67), deficient in the IFN-γ transcription factor Tbet do not
suggesting that MHC-I-restricted CD8+ T cells partici- produce IFN-γ or promote antibody isotype switching in
pate in bacterial clearance. Additional studies indicate B cells; these mice are also susceptible to infection with
that CD8+ T cells may actually be more important in aroA-attenuated Salmonella (107). Interestingly, expo-
recall of immunity during secondary challenge. For ex- sure to lead induces a shift from a Th1 response to a Th2
ample, although naive β2m−/− mice (lacking functional T-cell response (with a corresponding shift from high to
MHC-I molecules) are eventually able to resolve an initial low levels of IFN-γ and TNF-α production), thereby
infection with attenuated bacteria, immune β2m−/− mice rendering resistant C3H/HeN mice susceptible to oral
are more susceptible than wild-type mice to virulent Salmonella infection (108). Others have demonstrated
challenge (78). Moreover, adoptive transfer of CD8+ IFN-γ production by gut-associated lymphoid tissue and
T-cell-depleted immune cells provides limited protection increased numbers of CD4+ and CD8+ T cells in the gut

8 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

during Salmonella infection (103, 109), suggesting that bacteria accessible to phagocytes. In addition, CTL
localized expression of cytokines by T cells during pri- granules contain the protein granulysin that has been
mary infection may be important. Cytokine expression by shown to have direct antimicrobial activity against Sal-
T cells may account for the ability of immune mice to monella and other pathogens (117). Finally, CD4+ T-cell
control a secondary infection with virulent Salmonella: help is required for B-cell functions such as isotype
rapid release of cytokines by T cells primed to preferen- switching and affinity maturation (Fig. 1K). For example,
tially home to the gut mucosa (110) facilitates rapid in athymic mice (lacking mature T cells), there is a dra-
macrophage activation and recruitment to the site of matically reduced LPS-specific antibody response (89).
initial host encounter with the pathogen (111). This idea CD28-deficient mice (with impaired T-cell activation)
is supported by studies in which administration of anti- exhibit diminished production of IgG1, IgG2a, or IgG2b
TNF-α or IFN-γ antibodies exacerbated secondary viru- antibodies, and only low levels of IgM and IgG3 anti-
lent infection in previously immunized mice (68, 104). bodies (118). It may be that a combination of all of these
Finally, depletion of either IFN-γ or CD4+ T cells results functions is required for optimal immunity to Salmonella
in reactivation of latent Salmonella infection (20, 100), infection (Fig. 1J, K, and L), and further work remains to
suggesting that IFN-γ may mediate CD4+ T-cell control define the exact importance of each contribution.
of latent infections.

Despite this evidence for the importance of cytokine


production by Ag-specific T cells in Salmonella infection, Antigenic Specificities of
other studies suggest the existence of IFN-γ–independent Salmonella-Specific T Cells
T-cell actions. For example, it appears that cytokine Despite the well-established role for T cells in Salmonella
production alone is insufficient to induce clearance in the immunity, surprisingly little is known about the identity
absence of T cells: T-cell-depleted mice are susceptible to or relative importance of the specific Ags recognized.
challenge with attenuated aroA Salmonella despite high Perhaps the most thoroughly characterized Ag recog-
serum levels of IFN-γ and TNF-α (68). Similarly, IL-12- nized by Salmonella-specific CD4+ T cells is the flagellar
null mice fail to control infection with attenuated serovar subunit protein FliC (43). This protein is abundant on
Typhimurium, despite equivalent IFN-γ and TNF-α the cell surface (a single bacterium can have up to 10
production by T cells in mutant and wild-type mice. flagella, each composed of 30,000 or more flagellin
T cells in these mice did not proliferate in response to Ag, molecules [119]) and is therefore an obvious target for
indicating the importance of IFN-γ-independent, pro- host immune responses. Indeed, multiple epitopes have
liferation-dependent T-cell responses in the control of been identified within the FliC protein (120, 121, 122),
Salmonella infection (112). Together with observations and a significant fraction of Salmonella-specific T cells
that NK-T cells and neutrophils have been identified as a generated by sublethal infection recognize FliC (50, 123).
significant source of IFN-γ production in response to Immunization with purified FliC protein is protective
serovar Typhimurium infection (113), and that T-cell (123, 124). Significantly, the monomeric form of this
function is required only in the later stages of primary protein is a ligand for TLR5 (125, 126) and presumably
sublethal infection (69), this finding suggests that T cells the Nod-like receptor Ipaf (127), and FliC is therefore a
are not the primary source of IFN-γ, at least during early target of both innate and adaptive immune responses.
stages of infection (106).
FliC regulation is complex (51, 128) and acts to limit
Although the exact role of cytokine production by T cells adaptive immune responses: FliC is repressed below the T-
remains to be defined, there are other possible mech- cell activation threshold during systemic growth (52). The
anisms by which T cells could mediate immunity to global regulator PhoP mediates this repression, and it may
Salmonella infection. For example, CD4+ T-cell help is therefore seem surprising that PhoPc bacteria (in which
required to establish and maintain a functional CD8+ FliC expression is constitutively suppressed) are strongly
memory response (Fig. 1J) (114, 115). Salmonella- immunogenic and prime protective immunity. However,
specific CD8+ T cells are capable of lysing infected host given the large number of potential proteins encoded in the
cells both in vitro (78) and in vivo (116). Therefore, Salmonella genome, and a host T-cell compartment with
T-cell-mediated lysis of infected host cells could disrupt the capacity of generating at least 1018 different receptors
the protected intracellular niche to render intracellular (129), it is likely that T cells responding to other

ASMScience.org/EcoSalPlus 9
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

specificities are primed during infection and capable of provide insights into host functions required to resist
mediating protection. Indeed, analysis of proliferative re- Salmonella infection.
sponses to sodium dodecyl sulfate-polyacrylamide gel
electrophoresis (SDS-PAGE)-fractionated whole bacteria
reveals that CD4+ T cells recovered from Salmonella-
immune mice respond to multiple non-FliC specificities B CELLS
(52, 130). Despite this evidence, only a few additional The Role of B Cells in Salmonellosis
proteins have been definitively established as immune Natural oral Salmonella infection progresses from pene-
T-cell targets: the Salmonella invasion protein SipC is tration of the gastrointestinal epithelium to systemic
recognized by CD4+ T cells (131), and peptides derived dissemination, generating B-cell and T-cell immune re-
from GroEL are recognized by CD8+ cytotoxic T cells sponses that collectively promote protective immunity
(132) induced after natural infection. against virulent infection in susceptible hosts. While the
cell-mediated and humoral arms of the adaptive immune
Although the exact identity of additional Ags remains to response possess unique individual functions, interaction
be determined, it appears that, like FliC, other surface- between B and T cells is also critical. Antibody isotype
exposed proteins are good candidates for Salmonella- switching depends on T-cell help, and B cells contribute
specific T-cell recognition. For example, bacterial extracts to the priming of T cells and generation of T-cell mem-
enriched for surface Ags can prime protective immunity, ory. Thus, B-cell functions, including but not limited to
presumably mediated by IFN-γ production from im- antibody production, are critical in the generation of
mune splenocytes and the generation of IgG2a antibodies protective immunity against Salmonella infection.
(133). T-cell lines derived from protectively immunized
mice respond to fimbrial proteins (134, 135). Salmonella
infection also primes T cells responding to outer mem- Studies in B-Cell-Deficient Mice
brane proteins such as OmpC, porins, and iron-regulated The importance of B cells in immunity to serovar
outer membrane proteins (IROMPS); immunization with Typhimurium has been explored by using mice devoid of
these proteins is protective and associated with increased B cells (Igh-6−/−, Igμ−/−) (141). Igμ−/− mice possess a ge-
numbers of CD4+ and CD8+ T cells (136, 137, 138, 139). netic defect that disrupts the gene encoding the constant
Finally, it has recently been shown that membrane region of the IgM heavy chain (μ-chain), rendering them
vesicles (MVs) derived from the bacterial surface contain B-cell-deficient due to arrest in the stage of pre-B-cell
Ags recognized by Salmonella-immune T cells, and im- maturation (141). After intravenous (i.v.) or intraperi-
munization with purified MVs primes a protective CD4+ toneal (i.p.) infection, Igμ−/− mice control and clear an
T-cell response (59, 140). attenuated serovar Typhimurium aroA strain from the
liver and spleen (142, 143, 144, 145). However, unlike
wild-type mice, Igμ−/− mice experience sepsis during
T-Cell Contributions to Salmonella Immunity: primary infection and are not protected against subse-
Future Directions quent oral challenge with virulent serovar Typhimurium
Our understanding of T-cell responses to Salmonella (143, 145). Transfer of immune serum from wild-type
infection has dramatically improved in recent years. mice to Igμ−/− mice confers immunity to virulent chal-
Nonetheless, many questions remain to be answered. lenge, underscoring the importance of B cells in the
What is the exact mechanism by which immune T cells generation of a protective immune response (144). Al-
mediate protection? What is the complete antigenic though B cells do not appear to contribute significantly to
profile of T cells generated by protective immunization? bacterial control during primary infection with aroA-
Do T and B cells recognize the same Ags? When and attenuated Salmonella, Igμ−/− mice are highly susceptible
where are protective T-cell responses generated? There is to primary infection with virulent serovar Typhimurium
a complex relationship between Salmonella and its hosts: compared with wild-type mice (145). These data dem-
immune recognition of microbial Ags selects for patho- onstrate that B cells, or the products of B cells, are
gens capable of exploiting, modulating, or evading that necessary for both the initial control of primary viru-
recognition in order to colonize and infect their hosts. lent serovar Typhimurium, and the development of
Therefore, identification of additional Ags recognized by long-lived resistance against oral challenge with virulent
CD4+ T cells responding to infection will continue to Salmonella.

10 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

The Role of Secretory Antibodies in Protection also increases the incidence of transmission to normally
against Serovar Typhimurium (See Also Chapter resistant wild-type mice, suggesting that innate sIgA may
Mucosal Immune Responses to Escherichia coli play a role in “coating” bacteria present in feces, resulting
and Salmonella Infections) in decreased transmissibility of the organism (153).
Secretory IgA (sIgA) is present in mucosal tissues and
has been proposed to influence the pathogenesis of
mucosal pathogens (146). Salmonella initiate infection The Influence of Salmonella-Specific
via penetration of the mucosal epithelium, where colo- Antibodies on Protection
nization of Peyer's patches (PPs) is required for gener- Susceptibility of mice to Salmonella infection depends on
ation of sIgA (9). The role of Salmonella-specific sIgA, several known genetic loci (154, 155, 156), and likely on
however, appears to be minimal in protective immunity. other strain-specific genetic signatures yet to be deter-
Salmonella-specific monoclonal sIgA reduces adherence mined. These unique host factors are as influential as
of serovar Typhimurium to cells in vitro and confers pathogen-specific virulence factors in determining the
protection to naïve mice against oral challenge when outcome of host-pathogen interactions. The role that
secreted by an implanted hybridoma tumor (147, 148). antibodies play in Salmonella immunity has been inves-
However, although the presence of a sIgA response does tigated in many different mouse strains, leading to a di-
increase the dose necessary for initial PP colonization, verse data set. Eisenstein et al. (157) nicely demonstrated
mice lacking the ability to generate sIgA are able to that, even though naturally resistant and naturally sus-
generate a protective immune response equal to wild- ceptible mice are equally able to generate antibodies, the
type mice, suggesting that serovar Typhimurium-specific resulting protective immunity against Salmonella was not
sIgA are not required for protective immunity (149). To equal. This study provides a helpful framework for in-
reconcile these seemingly conflicting data, it is important terpretation of data exploring the role of Salmonella-
to consider that the concentration of sIgA produced by specific antibodies in protective immunity. To examine
the hybridoma in vivo is likely very high, and may sig- the effects of host background on the generation of a
nificantly exceed the levels of Salmonella-specific sIgA protective response against serovar Typhimurium, four
generated during immunization with viable attenuated Salmonella-derived immunogens were administered i.p.
bacteria. to the inbred mouse strains C3H/HeJ, C3H/FeJ, and
C3H/HeNCrlBR. Although each of these strains is de-
Innate sIgA antibodies are produced in response to rived from the C3H lineage, they vary in their natural
commensal microbiota, and their generation does not susceptibility to Salmonella infection: C3H/FeJ mice are
depend on serovar Typhimurium infection. These anti- hypersusceptible, C3H/HeJ are susceptible, and C3H/
bodies are reactive against a wide range of nonspecific HeNCrlBR mice are resistant. Whereas none of the
Ags, some of which likely cross-react with serovar vaccines protected C3H/HeJ mice, and only administra-
Typhimurium Ags (150). Both conventional B2 B cells tion of acetone-killed Salmonella generated significant
and CD5+ B1 B cells secrete innate sIgA antibodies (151). protection in C3H/FeJ mice, all four immunogens con-
While B2 B cells require direct interaction with CD4+ T ferred significant protection on resistant C3H/HeNCrlBR
cells for stimulation of antibody secretion, B1 B cells do mice. Transfer of immune sera from immunized C3H/
not. Secretion of innate antibodies by B1 B cells is thought HeNCrlBR donor mice to naïve C3H/HeNCrlBR re-
to depend on the stimulating effects of bystander cyto- cipients conferred complete protection against intraper-
kines such as IL-5, IL-6, and IL-10 (152). Unlike Salmo- itoneal challenge. However, the same serum transfer to
nella-specific sIgA (149), innate sIgA is protective against naturally susceptible mouse strains C3H/HeJ and C3H/
natural Salmonella infection (153). In mice that lack the FeJ was unable to provide full protection against i.p.
ability to secrete IgA into their mucosa because of a defect challenge. Thus, the ability of antibodies to provide
in the polymeric immunoglobulin receptor (pIgR−/− protective immunity is strong when the host is naturally
mice), sIgA destined for secretion is instead retained in resistant to Salmonella infection, but naturally suscepti-
the serum. pIgR−/− mice are more susceptible to serovar ble hosts appear to require additional immune factors to
Typhimurium infection via the natural oral route, gain full protective immunity (see “T-cell subsets and
resulting in a higher bacterial load and increased fecal Salmonella immunity,” above). Therefore, the role of
shedding of Salmonella compared with wild-type mice. antibodies in resistance to serovar Typhimurium infec-
Fecal shedding of serovar Typhimurium by pIgR−/− mice tion depends on host susceptibility status.

ASMScience.org/EcoSalPlus 11
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

In susceptible mouse strains, B cells appear to be most Salmonella-Specific Antibody Responses


important for protection during virulent serovar Typhi- It has long been recognized that antibody responses are
murium challenge. In mice with a functional T-cell re- generated against the abundant Salmonella surface Ag
sponse, B cells are dispensable during primary i.v. LPS. However, it has become clear that antibody re-
infection with aroA-attenuated Salmonella but necessary sponses to protein antigens also contribute to protection,
for protection against subsequent i.p. or i.v. virulent although these responses have not been fully character-
challenge (144). Igμ−/− and wild-type mice are equally ized. The complete repertoire of antibody specificities has
capable of clearing primary infection with aroA-attenu- not been defined in the murine response or in humans
ated serovar Typhimurium, and mice of either back- infected with serovar Typhi, although similarities likely
ground eliminate secondary i.v. challenge with attenuated exist (159). In addition, the contributions of antibodies
bacteria. Wild-type mice with functional B and T cells with particular specificities in the generation of protec-
clear secondary infection with attenuated organisms tive immunity have not been established. We focus below
more rapidly than Igμ−/− mice, indicating that optimal on antibody responses generated by infection with live
bacterial clearance requires both memory T cells and B- Salmonella.
cell functions (144). However, Igμ−/− mice are less able to
control virulent Salmonella i.p. challenge, and Igμ−/− mice LPS, exposed on the surface of gram-negative bacteria,
immunized with attenuated bacteria become as resistant elicits a strong antibody response during serovar Typhi-
to virulent challenge as wild-type animals when wild-type murium infection (159, 160, 161). The majority of the
immune serum is administered prior to challenge (144). B-cell response to LPS targets the serotype-specific O-
Thus, in susceptible mouse strains with functional T-cell antigen polysaccharide chains on the outermost part of
responses, B cells are dispensable during primary and the molecule (162). The O-antigen likely obscures the
secondary infection with aroA-attenuated Salmonella more internal lipid A and core structures from antibody
strains, but resistance to virulent serovar Typhimurium access. Conserved O-antigen residues, found across Sal-
infection via the i.p. and i.v. routes depends on the pre- monella serotypes, are less immunogenic than serotype-
sence of B cells and/or immune serum constituents. specific residues, which may account for the inability of
Consistent with these findings, Mastroeni et al. (143) anti-LPS antibodies to protect against challenge with
demonstrated that protection of naturally susceptible different Salmonella serotypes (163).
mice against virulent oral Salmonella infection requires
both antibodies and T-cell responses. Together with the By using two serotypes of Salmonella with unique O-
data showing the necessity of antibody in protection antigens, Hormaeche et al. (163) elegantly demonstrated
against virulent organisms administered i.p. and i.v., that protection against oral Salmonella challenge depends
these observations confirm the importance of antibodies on antibody responses directed toward protein Ags in
in immunity against virulent serovar Typhimurium in addition to those against LPS moieties. Mice that were
naïve susceptible hosts. The mechanism by which anti- i.v. immunized with either viable serovar Typhimurium
bodies provide protection remains unknown. (O-antigen type O4) or Salmonella enterica serovar
Enteritidis (O-antigen type O9) and orally challenged
Naturally resistant mouse strains show a more variable with the immunizing strain were protected. Conversely,
but still important dependence upon antibody response mice challenged with the Salmonella serotype not used
for Salmonella-specific immunity. Administration of for immunization (termed the “heterologous” strain)
immune sera protects naïve Salmonella-resistant C3H/ were not protected. The inability of heterologous strains,
HeNCrlBR mice but not Salmonella-susceptible C3H/ distinguished by O-antigen type, to confer immunity
HeJ and C3H/FeJ mice from infection (157). Immune B against one another indicates that LPS-specific immune
cells and antibodies confer protection on otherwise sus- responses are responsible for protection. These results
ceptible male xid mice (harboring functionally defective suggested that immunization with serovar Enteritidis O4
B cells but functional T cells) when transferred from would be fully protective against challenge with serovar
immunized Salmonella-resistant female littermates (158). Typhimurium O4, and vice versa. However, only partial
Therefore, antibody production as well as other non- protection is observed when mice are challenged with
antibody-dependent B-cell functions (see below) aids in heterologous bacteria expressing the O-antigen type of
protection against serovar Typhimurium, even in natu- the immunizing strain. These data are consistent with the
rally resistant hosts. presence of antibodies targeting non-LPS (protein) Ags,

12 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

as well as the idea that non-B-cell responses, such as prisingly, this epitope is proposed to lie in the periplasm,
those from CD4+ T cells, also contribute to protective indicating that the B-cell response is directed primarily
immunity. Importantly, generation of antibodies against against a region of the protein that is not surface exposed
protein Ags requires interaction with CD4+ T cells (see (168). However, OmpA has two structurally distinct
“Mechanisms of T-cell-mediated immunity to Salmo- membrane conformations (169). The major OmpA spe-
nella infection,” above), and dependence on protein- and cies is characterized by an N-terminal β-barrel in the
non-protein-specific antibodies for complete protection outer membrane and an α-helix rich C terminus in the
against Salmonella has been observed by several groups periplasm, whereas formation of the minor conformation
(164, 165, 166). Taken together, these data indicate that likely results in surface exposure of parts of the C ter-
the Salmonella-specific antibody response generated minus (169). The C-terminal epitope is more immuno-
during protective immunity depends on multiple Sal- dominant than the N terminus during infection (170),
monella Ags, including both LPS and protein(s). The suggesting that surface exposure of the C terminus
relative contribution of these specificities to protection provides the opportunity for B-cell recognition and an-
remains to be determined. tibody production. The generation of anti-OmpA anti-
bodies also occurs in humans following serovar Typhi
Defining the specificity of antibody responses resulting infection, although the dominant epitope(s) have not
from human serovar Typhi infection is likely to be com- been determined (159, 167).
plex, particularly in regions of the world afflicted with
endemic typhoid fever. Anti-LPS antibody titers are al- Infection has also been confirmed to prime antibody re-
ready elevated in “naïve” subjects prior to immunization sponses to outer membrane porins OmpC and OmpF
or infection, potentially obscuring the significance of anti- (165), and the dominant OmpC epitope is located on a
LPS antibody titers (167), and antibodies directed against surface-exposed loop (162). During natural serovar Typhi
LPS in humans appear to be minor compared with anti- infection of humans, antiporin antibody titers increase
bodies recognizing protein Ags. An inhibition enzyme- 1,000-fold over control sera from uninfected subjects
linked immunosorbent assay (ELISA) method, in which (171), a phenomenon that has been confirmed by ad-
immune serum is preincubated with either outer mem- ditional studies by using both ELISA and immuno-
brane proteins or LPS, was used to assess the ability of blot methods (167, 172). Any contribution of antiporin
these Ags to inhibit antibody binding to solid-phase antibodies to protective immunity, however, remains
bound Salmonella Ags. Greater than 60% inhibition of unknown.
antibody binding to Salmonella Ags occurred after incu-
bation with low concentrations of soluble outer mem- In addition to membrane proteins, abundant surface
brane proteins, whereas preincubation with the highest appendages such as flagella and fimbriae are also targets of
concentrations of LPS inhibited binding by less than 40% the B-cell response. As an abundant extracellular Ag, Sal-
(167). These data suggest that human anti-Salmonella monella FliC elicits a strong antibody response in mice and
antibodies may be directed primarily toward protein Ags. humans, in addition to its role as an important T-cell Ag
(see “Antigenic specificities of Salmonella-specific T cells,”
As in human infections, B cells also generate antibody above) (173, 174). The murine antibody response to FliC is
responses against a group of largely unknown protein Ags generated following both oral and i.p. infection (173, 174).
during natural oral infection of mice. Proposed identities Flagellin also induces antibody production in humans in-
of several B-cell antigens, based only on electrophoretic fected with serovar Typhi (159, 171, 175). The antibody
mobility in one-dimensional SDS-PAGE followed by response to fimbrial Ags depends on which fimbrial sub-
Western blot analysis with immune sera, include OmpA, unit proteins are expressed during infection (176). Al-
outer membrane porins, flagellar subunit protein FliC, though serovar Typhimurium grown in laboratory culture
outer membrane lipoprotein Lpp, and a cellular heat prior to infection expresses only the FimA subunit, ex-
shock protein (159). pression of ten different subunits occurs during murine
infection (176). Expression of various combinations of
Antibody responses to the outer membrane protein these ten subunits occurs in vivo, with a corresponding
OmpA resulting from infection have been experimentally antibody response to the expressed subunit(s), but ex-
confirmed, and the predominant response is directed pression of all ten in an individual mouse is not observed.
against the C-terminal third of the protein (168). Sur- These findings reinforce the concept that bacterial regu-

ASMScience.org/EcoSalPlus 13
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

lation of Ag expression influences adaptive immune re- immune response via secretion of cytokines (12, 184, 185,
sponses (52), and that the most relevant bacterial Ags to 186). Unlike splenocytes from immunized wild-type
investigate will not necessarily be obvious from the ex- mice, splenocytes harvested from immune B-cell-defi-
amination of bacteria grown in the laboratory setting. cient mice are unable to generate Th1-type cytokines
(IFN-γ, IL-2) in response to soluble serovar Typhimu-
Human B cells also generate an antibody response rium Ags in vitro, despite the fact that T-cell populations
against the Vi capsular Ag, which is expressed by serovar in both murine backgrounds are capable of cytokine
Typhi and Salmonella enterica serovar Paratyphi C but production (143). In the absence of B cells, the balance of
not serovar Typhimurium or serovar Paratyphi A and B. the adaptive immune response shifts away from the
Interestingly, whereas only 20% of patients with acute characteristic Th1-type cytokine IFN-γ and toward the
typhoid fever have a detectable anti-Vi antibody titer, production of the Th2-type cytokine IL-4 (27). B-cell
approximately 90% of chronic gall bladder carriers secretion of IFN-γ depends on combined stimulation of
demonstrate an antibody response to the Vi Ag (reviewed TLR2, 4, and 9 (184). These observations suggest that B
in reference 177). The dependence of temporal expres- cells support the generation of a Th1-type cytokine re-
sion of Vi Ag in vivo on environmental signals suggests sponse, either directly through cytokine production and
that, like FliC, altered Vi Ag expression may affect as APCs (see below) or indirectly through undefined
adaptive immune responses (see “Antigenic specificities mechanisms, thereby facilitating the downstream am-
of Salmonella-specific T cells,” above). Immunization plification of the immune responses necessary for pro-
with the Vi capsular Ag or live attenuated serovar Typhi tection against virulent serovar Typhimurium.
Ty21a provides protection against typhoid fever caused
by serovar Typhi (reviewed in reference 178). However, B cells also contribute to Ag presentation in vivo (187,
these licensed typhoid fever vaccines do not provide 188, 189, 190). Early in infection, DCs prime CD4+ T
protection against serovar Paratyphi A infections (179, cells through presentation of immunodominant peptides
180), and the coincident increase in incidence of in the context of MHC-II molecules (see “DCs and
Vi-negative serovar Paratyphi A outbreaks in endemic macrophages as professional APCs during Salmonella
typhoid fever regions is consistent with the immuno- infection,” above). However, once CD4+ T cells are
dominance of the Vi Ag (180, 181, 182). primed, they are able to activate Ag-specific B cells, which
internalize Ag 10,000-fold more efficiently than non-
Identification of Salmonella B-cell Ags has proven diffi- specific cells such as DCs do (191). Activated B cells may,
cult. Ags that have been most frequently characterized in turn, prime naïve T cells through presentation of a
are those expressed by bacteria grown in laboratory much wider range of peptides from the original protein
culture conditions, which may not accurately represent Ag (189). Specifically, the ability of B cells to ingest,
Ag expression and/or abundance in vivo. Selection of process, and present Salmonella Ags depends on the
bacterial targets that approximate what may be seen by immune status of the host. For example, B cells harvested
the host during infection is therefore a critical factor in from naïve mice or mice immunized 2 weeks prior to
Ag discovery. Investigating outer membrane proteins harvest are refractory to infection in vitro, but B cells
and surface appendages such as flagella and fimbriae has isolated from mice immunized 3 months prior to harvest
yielded information about B-cell Ags exposed on the can be infected by live serovar Typhimurium (27). B-cell
bacterial cell surface. It is likely that important B-cell Ags populations exhibit upregulation of the costimulatory
will be revealed with further investigation of other surface molecule CD86 and are able to present soluble serovar
organelles such as MVs (59), which are rich in outer Typhimurium Ags, whether they are obtained from na-
membrane and periplasmic proteins and elicit protective ïve, 2-week immune, or 3-month immune mice (192).
responses when administered as an immunogen to both Such Ags are available for capture independent of the
mice and humans (140, 183). bacteria from which they are derived, and may include
surface organelles such as MVs released by Salmonella in
vivo (193, 194). However, only B cells from 3-month
B-Cell Functions in Addition to Antibody Production immune mice are able to present Ags derived from intact
B cells contribute to the generation of protective immu- heat-killed bacteria (27). Additionally, splenocytes har-
nity against serovar Typhimurium independent of anti- vested from wild-type mice are better able to present
body production. For example, B cells influence the Salmonella Ags to Salmonella-specific CD4+ T-cell lines

14 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

than splenocytes of Igμ−/− mice or B-cell-depleted wild- enter the systemic circulation when doses near the oral
type splenocytes, demonstrating that B cells are an im- LD50 are used (197, 198).
portant APC population in the spleen (27). These data
suggest that B cells have unique Ag presentation capa- Flagella (and the flagellar subunit FliC) are abundantly
bilities at different times postinfection, supporting a expressed on the surface of extracellular-phase Salmo-
model in which presentation of Ags to CD4+ T cells is nella (Fig. 2A) and therefore serve as a large reservoir of
shared between DCs and B cells. ligands for both innate (125, 126) and adaptive (59, 43,
123) immune systems. After bacterial invasion or trans-
location of flagellin across the intestinal epithelial cell
B-Cell Contributions to Salmonella Immunity: layer, TLR5 stimulation results in a strong proinflam-
Future Directions matory response that is not induced by normal
It is clear that the generation of protective immunity microbiota (Fig. 2B) (125, 199, 200, 201). Flagellin-
depends on multiple B-cell functions, including antibody stimulated secretion of chemokines such as IL-8 (125,
production, cytokine secretion, and/or Ag presentation 199, 202) and CCL20 (203) results in the recruitment of
(Fig. 1K). B-cell involvement in protection against pri- phagocytes, including DCs, to the small intestine (re-
mary serovar Typhimurium infection and the generation viewed in reference 204). DCs are highly competent
of protective immunity to secondary challenge may be APCs that can directly sample the gut lumen for Ags to
manifest at many stages of pathogenesis: during initial present to immune effector cells (8). In addition, flagellin
migration to the intestinal epithelia, transition from the stimulates the maturation of both DCs and macrophages
intestinal lumen to the PP, uptake by other immune cells (205, 206). Thus, the proinflammatory response to fla-
such as macrophages, and/or progression to systemic gellin, in addition to the proinflammatory response
organs. The exact stage(s) during which B cells and/or evoked by Salmonella-induced cell death (207), results in
their products act, and how interactions occur between B intestinal localization and activation of cells capable of
cells and Salmonella, are largely unknown. In addition, priming adaptive immune responses. APCs in the PP
the identity of Salmonella Ags that drive these responses (Fig. 2C) therefore initially prime CD4+ T cells that re-
will be a key area of interest in future studies, with the spond to Ags expressed by bacteria invading the host
goal of determining the specificity of antibodies that in- intestinal epithelium or residing in the gut lumen.
duce protective immunity.
Small numbers of bacteria escape the gut and disseminate
by intracellular transport (9) to systemic sites such as the
liver and spleen (197, 198). There, Salmonella replicate
TEMPORAL AND ANATOMICAL ASPECTS OF within macrophage phagosomes (10, 11), an intracellular
SALMONELLA-SPECIFIC ADAPTIVE IMMUNITY: niche providing a safe haven from host antibodies. Sal-
FliC AS A MODEL Ag monella have evolved complex regulatory systems to
During natural infection, Salmonella must survive the precisely tailor gene expression to environmental con-
intestinal environment (Fig. 2A), penetrate the mucosa of text. One global regulator of Salmonella gene expression
the small intestine (Fig. 2B), colonize the PP (Fig. 2C), is PhoP, which is activated by intracellular growth con-
and disseminate to systemic sites such as the liver and ditions and mediates virulence gene expression, bacterial
spleen to replicate within host cells (Fig. 2E). The gut surface modifications, and repression of FliC, a dominant
mucosa presents a major initial barrier to Salmonella Ag for Salmonella-specific CD4+ T cells (52, 53, 54, 55,
dissemination after oral infection. In addition to over- 58).
coming physical barriers such as the glycocalyx and
mucus layers, Salmonella must withstand chemical Regulation of FliC expression is complex. During later
attacks from antimicrobial peptides secreted by the in- stages of Salmonella infection, when bacteria are repli-
testinal mucosa (195, 196). Most Salmonella are con- cating intracellularly at systemic sites, FliC expression is
tained by innate host defenses in the small intestine or are repressed below the T-cell activation threshold in a
eliminated and do not progress farther than the intestinal PhoP-dependent manner (52). In addition to reduced
lumen. Although high doses of Salmonella (many times FliC expression, PhoP-mediated modifications of the
the oral LD50) have often been employed in experimental bacterial surface restrict the access of this Ag to APCs for
models of oral infection, only a few bacteria successfully processing and presentation to T cells (50). The natural

ASMScience.org/EcoSalPlus 15
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

consequence of this repression is that CD4+ T cells directed toward Ags expressed by extracellular bacteria at
primed to respond to Ags (such as FliC) that are ex- early stages of infection (when the risk of systemic dis-
pressed early in infection will fail to recognize intracel- semination is greatest) appears to be an effective strategy.
lular bacteria growing at systemic sites during later stages This idea is supported by the high rate of nontyphoidal
of infection (Fig. 2E). Therefore, it appears that the co- Salmonella bacteremia in AIDS patients compared with
ordinated control of Ag expression and accessibility to immunocompetent hosts (95, 96, 97). Studies using a
APCs (bioavailability) directly imposes temporal and model peptide Ag indicate that systemic Salmonella-
anatomical limits on the corresponding T-cell response. specific CD8+ memory responses appear to be restricted
This idea is supported by the findings that FliC-specific to Ags encountered in the gut mucosa (136). CD4+ and
CD4+ T-cell activation is restricted to the gut, despite CD8+ T-cell levels in the gut increase threefold during
systemic bacterial colonization (86), and that bacteria secondary challenge of protectively immunized mice
present in the PP but not MLN or spleen express FliC (103). This is likely because DCs in the PP (the most
protein. Importantly, Salmonella in the PP do not likely site of T-cell priming during primary infection) are
homogenously express FliC; up to 40% of the bacteria are specifically able to imprint gut-homing specificity on T
FliC negative (51). Heterogeneous gene expression cells (Fig. 2C) (110). Indeed, after oral immunization in
provides a mechanism whereby one bacterial population humans with serovar Typhi, most of the effector (IFN-
provides abundant Ags for T-cell priming, while a sub- γ-producing) CD4+ and CD8+ T cells express the α4β7
population already in “stealth mode” is poised to evade gut-homing receptor (80). Finally, memory T cells ex-
the developing T-cell responses (Fig. 2C left). press CCR6, the only known receptor for CCL20, a
chemokine secreted by intestinal epithelial cells after
Repression of protective Ags like FliC, in concert with stimulation by flagellin (212). Therefore, in an immu-
other genetically co-coordinated modifications of the nized host, it appears that memory T cells primed to
bacterial surface as described above, could facilitate respond to early-phase Ags expressed by extracellular
bacterial growth in vivo by affording Salmonella the bacteria are recruited to the intestine where their
opportunity for additional rounds of replication before resulting activation could facilitate the containment of
the onset of effective adaptive immune recognition. This bacterial replication and dissemination. In addition, it
is a reasonable explanation for the different outcomes seems likely that opsonic Ig would beneficially augment
resulting from infection of naïve, susceptible mice with the host response by facilitating Ag uptake, processing,
virulent versus attenuated bacteria. Attenuated aroA- and presentation (Fig. 1P).
mutant Salmonella grows more slowly in vitro and in
vivo (208, 209) and is cleared by immunocompetent
hosts, while virulent Salmonella rapidly overwhelms host CONCLUDING REMARKS
defenses. The growth advantage of virulent bacteria is As a facultatively intracellular pathogen, Salmonella is an
eliminated by prior immunization, as rapid activation of effective model organism for the study of adaptive im-
localized host T- (and B-) cell functions provides pro- mune responses. Antibodies, the products of B cells,
tection (Fig. 1 right) (210, 211). In this context, it is opsonize extracellular bacteria for phagocytic uptake
important to note that T cells from immune mice can (Fig. 1P), and Ig on the B-cell surface mediates high-
detect intracellular (and therefore FliC) bacteria (52). efficiency Ag capture for Ag presentation (Fig. 1K).
However, it is not known when or where T cells CD4+ and CD8+ T cells generated in response to Sal-
responding to Ags expressed by intracellular phase (FliC) monella infection detect intracellular bacteria and are
bacteria are primed, or whether these T cells are capable required for bacterial clearance. Accordingly, full im-
of mediating protection. Interestingly, it has been shown munity to Salmonella infection in naïve, susceptible hosts
that systemic (spleen and liver) macrophages, although requires both T- and B-cell functions.
capable of processing and presenting Ags to T cells, do
not affect the development of an adaptive T-cell response The requirement of both sera and T cells for immunity
(46). This suggests that APCs at systemic sites (Fig. 2E) suggests that, in addition to their individual functions,
may not be capable of priming a T-cell response of suf- the interaction between T and B cells is important. Al-
ficient speed or magnitude to protect a host from sub- though there has traditionally been a differentiation be-
sequent challenge with virulent Salmonella. From a host tween “cellular” and “humoral” immunity, in reality
perspective, a T-cell response localized to the gut and these responses are tightly interconnected. T cells are

16 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

required to activate B-cell isotype class switching and identification of adaptive immune targets will guide
affinity maturation. B cells may present Ags to T cells and rational vaccine development, provide insights into the
produce cytokines that support the development of a host functions required to resist Salmonella infection,
Th1-type T-cell response (Fig. 1K). CD4+ T cells are and correspondingly provide valuable reagents for de-
required to initiate activation of cytotoxic CD8+ T cells fining the critical pathogenic capabilities of Salmonella
(Fig. 1J). In addition, cytokines released by T cells may that contribute to its ability to cause acute and chronic
activate APCs to more efficiently process and present infections.
Ags, while cytokines released by activated APCs facilitate
T-cell activation and proliferation (Fig. 1E and L). ACKNOWLEDGMENTS
No potential conflicts of interest relevant to this review were reported.
The interaction between Salmonella and its host is
complex and dynamic. The host mounts an immune
REFERENCES
defense against the pathogen, which in turn acts to re- 1. Wright A, Semple D. 1897. Remarks on vacinnation against ty-
duce, evade, or exploit these responses to successfully phoid fever. BMJ 1:256.
colonize the host. Correspondingly, a greater under- 2. Hobson D. 1957. Resistance to reinfection in experimental mouse
standing of adaptive immune responses provides tools typhoid. J Hyg 55:334–343.
and reagents for gaining additional insights into mech- 3. Collins FM, Mackaness GB. 1968. Delayed hypersensitivity and
arthus reactivity in relation to host resistance in Salmonella-infected
anisms of bacterial pathogenesis. Salmonella bacteria are mice. J Immunol 101:830–845.
not just passive collections of Ags, but rather actively 4. Killar LM, Eisenstein TK. 1985. Immunity to Salmonella typhi-
participate in their encounters with host cells. Multiple murium infection in C3H/HeJ and C3H/HeNCrlBR mice: studies with
bacteria-directed mechanisms, including altered Ag ex- an aromatic-dependent live S. typhimurium strain as a vaccine. Infect
Immun 47:605–612.
pression and bioavailability, interference with APC acti-
5. Kotlarski I, Pope M, Doherty K, Attridge SR. 1989. The in vitro
vation and function, and direct inhibition of T-cell proliferative response of lymphoid cells of mice infected with Sal-
activation (65), combine to modify Salmonella’s “patho- monella enteritidis 11RX. Immunol Cell Biol 67(Pt 1):19–29.
genic signature” to minimize susceptibility to immune 6. Ushiba D. 1965. Two types of immunity in experimental ty-
surveillance (Fig. 1B, F, and H). phoid: “cellular immunity” and “humoral immunity.” Keio J Med 14:
45–61.
7. Mastroeni P, Villarreal-Ramos B, Hormaeche CE. 1993. Adoptive
Although our understanding of Salmonella-specific im- transfer of immunity to oral challenge with virulent Salmonellae in
munity has dramatically improved in recent years, many innately susceptible BALB/c mice requires both immune serum and T
unanswered questions remain. For example, the details of cells. Infect Immun 61:3981–3984.
8. Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G,
intestinal DC function, T-cell priming, T-cell migration, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P.
and the generation of T-cell memory in human serovar 2001. Dendritic cells express tight junction proteins and penetrate gut
Typhi infection remain incompletely understood. The epithelial monolayers to sample bacteria. Nat Immunol 2:361–367.
exact mechanisms by which T and B cells act to clear 9. Vazquez-Torres A, Jones-Carson J, Baumler AJ, Falkow S,
Valdivia R, Brown W, Le M, Berggren R, Parks WT, Fang FC. 1999.
Salmonella are unknown. Surprisingly little is known Extraintestinal dissemination of Salmonella by CD18-expressing
about the identity, regulation, and relative contributions phagocytes. Nature 401:804–808.
to immunity of the individual bacterial Ags recognized by 10. Richter-Dahlfors A, Buchan AM, Finlay BB. 1997. Murine sal-
Salmonella-specific B and T cells (the antigenisome). As monellosis studied by confocal microscopy: Salmonella typhimurium
resides intracellularly inside macrophages and exerts a cytotoxic effect
discussed above, emerging data suggest that bacterial on phagocytes in vivo. J Exp Med 186:569–580.
regulation of Ag expression and localization is critically 11. Salcedo SP, Noursadeghi M, Cohen J, Holden DW. 2001. In-
important: temporal and anatomical restrictions imposed tracellular replication of Salmonella typhimurium strains in specific
by bacterial regulation of Ag expression affect the de- subsets of splenic macrophages in vivo. Cell Microbiol 3:587–597.
velopment of FliC-specific T-cell responses (50, 52). MVs 12. Harris DP, Haynes L, Sayles PC, Duso DK, Eaton SM, Lepak
NM, Johnson LL, Swain SL, Lund FE. 2000. Reciprocal regulation of
have recently been identified as bacterial organelles ca- polarized cytokine production by effector B and T cells. Nat Immunol
pable of priming protective immune responses in the 1:475–482.
absence of infection (140). As nonreplicating structures 13. Janeway CA, Travers P, Walport M, Shlomchik M. 2001.
derived from the surface of genetically malleable orga- Immunobiology: the Immune System in Health and Disease, 5th ed.
Garland Publishing, New York, NY.
nisms such as Salmonella, MVs could provide a powerful
14. Fields PI, Swanson RV, Haidaris CG, Heffron F. 1986. Mutants
new tool for both the analysis and delivery of immune of Salmonella typhimurium that cannot survive within the macro-
targets. Collectively, these data suggest that continued phage are avirulent. Proc Natl Acad Sci USA 83:5189–5193.

ASMScience.org/EcoSalPlus 17
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

15. Rydstrom A, Wick MJ. 2007. Monocyte recruitment, activation, triggers dendritic cells and macrophages to adopt distinct migration
and function in the gut-associated lymphoid tissue during oral Sal- patterns in vivo. Eur J Immunol 36:2939–2950.
monella infection. J Immunol 178:5789–5801. 31. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ,
16. Brennan MA, Cookson BT. 2000. Salmonella induces macro- Pulendran B, Palucka K. 2000. Immunobiology of dendritic cells.
phage death by caspase-1-dependent necrosis. Mol Microbiol 38:31– Annu Rev Immunol 18:767–811.
40. 32. Geissmann F, Auffray C, Palframan R, Wirrig C, Ciocca A,
17. Fink SL, Cookson BT. 2007. Pyroptosis and host cell death Campisi L, Narni-Mancinelli E, Lauvau G. 2008. Blood monocytes:
responses during Salmonella infection. Cell Microbiol 9:2562–2570. distinct subsets, how they relate to dendritic cells, and their possible
18. van der Velden AW, Velasquez M, Starnbach MN. 2003. Sal- roles in the regulation of T-cell responses. Immunol Cell Biol 86:398–
monella rapidly kill dendritic cells via a caspase-1-dependent mech- 408.
anism. J Immunol 171:6742–6749. 33. Sundquist M, Wick MJ. 2005. TNF-alpha-dependent and
19. Levine MM, Black RE, Lanata C. 1982. Precise estimation of the -independent maturation of dendritic cells and recruited CD11c(int)
numbers of chronic carriers of Salmonella typhi in Santiago, Chile, an CD11b+ cells during oral Salmonella infection. J Immunol 175:3287–
endemic area. J Infect Dis 146:724–726. 3298.
20. Monack DM, Bouley DM, Falkow S. 2004. Salmonella typhi- 34. Wick MJ. 2007. Monocyte and dendritic cell recruitment and
murium persists within macrophages in the mesenteric lymph nodes activation during oral Salmonella infection. Immunol Lett 112:68–
of chronically infected Nramp1+/+ mice and can be reactivated by 74.
IFNgamma neutralization. J Exp Med 199:231–241. 35. Pozzi LA, Maciaszek JW, Rock KL. 2005. Both dendritic cells
21. Fieschi C, Dupuis S, Catherinot E, Feinberg J, Bustamante J, and macrophages can stimulate naive CD8 T cells in vivo to prolif-
Breiman A, Altare F, Baretto R, Le Deist F, Kayal S, Koch H, erate, develop effector function, and differentiate into memory cells.
Richter D, Brezina M, Aksu G, Wood P, Al-Jumaah S, Raspall M, J Immunol 175:2071–2081.
Da Silva Duarte AJ, Tuerlinckx D, Virelizier JL, Fischer A, Enright 36. Niedergang F, Sirard JC, Blanc CT, Kraehenbuhl JP. 2000. Entry
A, Bernhoft J, Cleary AM, Vermylen C, Rodriguez-Gallego C, and survival of Salmonella typhimurium in dendritic cells and pre-
Davies G, Blutters-Sawatzki R, Siegrist CA, Ehlayel MS, Novelli V, sentation of recombinant antigens do not require macrophage-spe-
Haas WH, Levy J, Freihorst J, Al-Hajjar S, Nadal D, De Moraes cific virulence factors. Proc Natl Acad Sci USA 97:14650–14655.
Vasconcelos D, Jeppsson O, Kutukculer N, Frecerova K, Caragol I, 37. Jantsch J, Cheminay C, Chakravortty D, Lindig T, Hein J,
Lammas D, Kumararatne DS, Abel L, Casanova JL. 2003. Low Hensel M. 2003. Intracellular activities of Salmonella enterica in
penetrance, broad resistance, and favorable outcome of interleukin 12 murine dendritic cells. Cell Microbiol 5:933–945.
receptor beta1 deficiency: medical and immunological implications. 38. Delamarre L, Pack M, Chang H, Mellman I, Trombetta ES.
J Exp. Med. 197:527–535. 2005. Differential lysosomal proteolysis in antigen-presenting cells
22. MacLennan C, Fieschi C, Lammas DA, Picard C, Dorman SE, determines antigen fate. Science 307:1630–1634.
Sanal O, MacLennan JM, Holland SM, Ottenhoff TH, Casanova JL, 39. Hopkins SA, Niedergang F, Corthesy-Theulaz IE, Kraehenbuhl
Kumararatne DS. 2004. Interleukin (IL)-12 and IL-23 are key cyto- JP. 2000. A recombinant Salmonella typhimurium vaccine strain is
kines for immunity against Salmonella in humans. J Infect. Dis. 190: taken up and survives within murine Peyer's patch dendritic cells. Cell
1755–1757. Microbiol 2:59–68.
23. Gordon MA, Banda HT, Gondwe M, Gordon SB, Boeree MJ, 40. Cheminay C, Mohlenbrink A, Hensel M. 2005. Intracellular
Walsh AL, Corkill JE, Hart CA, Gilks CF, Molyneux ME. 2002. Salmonella inhibit antigen presentation by dendritic cells. J Immunol
Non-typhoidal Salmonella bacteraemia among HIV-infected 174:2892–2899.
Malawian adults: high mortality and frequent recrudescence. AIDS 16: 41. Salazar-Gonzalez RM, Niess JH, Zammit DJ, Ravindran R,
1633–1641. Srinivasan A, Maxwell JR, Stoklasek T, Yadav R, Williams IR, Gu
24. Pang T, Bhutta ZA, Finlay BB, Altwegg M. 1995. Typhoid fever X, McCormick BA, Pazos MA, Vella AT, Lefrancois L, Reinecker
and other salmonellosis: a continuing challenge. Trends Microbiol 3: HC, McSorley SJ. 2006. CCR6-mediated dendritic cell activation of
253–255. pathogen-specific T cells in Peyer's patches. Immunity 24:623–632.
25. Hayes C, Lyons RA, Warde C. 1991. A large outbreak of sal- 42. Yrlid U, Svensson M, Hakansson A, Chambers BJ, Ljunggren
monellosis and its economic cost. Isr Med J 84:65–66. HG, Wick MJ. 2001. In vivo activation of dendritic cells and T cells
26. Rubino JR. 1997. The economic impact of human Salmonella during Salmonella enterica serovar Typhimurium infection. Infect
infection. Clin Microbiol Newslett 19:25–29. Immun 69:5726–5735.
27. Ugrinovic S, Menager N, Goh N, Mastroeni P. 2003. Charac- 43. Cookson BT, Bevan MJ. 1997. Identification of a natural T cell
terization and development of T-cell immune responses in B-cell- epitope presented by Salmonella-infected macrophages and recog-
deficient [Igh-6(−/−)] mice with Salmonella enterica serovar Typhi- nized by T cells from orally immunized mice. J Immunol 158:4310–
murium infection. Infect Immun 71:6808–6819. 4319.
28. Kalupahana RS, Mastroeni P, Maskell D, Blacklaws BA. 2005. 44. Martin-Orozco N, Isibasi A, Ortiz-Navarrete V. 2001. Macro-
Activation of murine dendritic cells and macrophages induced by phages present exogenous antigens by class I major histocompatibility
Salmonella enterica serovar Typhimurium. Immunology 115:462–472. complex molecules via a secretory pathway as a consequence of in-
29. Pietila TE, Veckman V, Kyllonen P, Lahteenmaki K, terferon-gamma activation. Immunology 103:41–48.
Korhonen TK, Julkunen I. 2005. Activation, cytokine production, 45. Wick MJ, Harding CV, Twesten NJ, Normark SJ, Pfeifer JD.
and intracellular survival of bacteria in Salmonella-infected human 1995. The phoP locus influences processing and presentation of Sal-
monocyte-derived macrophages and dendritic cells. J Leukoc Biol 78: monella typhimurium antigens by activated macrophages. Mol
909–920. Microbiol 16:465–476.
30. Zhao C, Wood MW, Galyov EE, Hopken UE, Lipp M, Bodmer 46. Wijburg OL, Van Rooijen N, Strugnell RA. 2002. Induction of
HC, Tough DF, Carter RW. 2006. Salmonella typhimurium infection CD8+ T lymphocytes by Salmonella typhimurium is independent of

18 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

Salmonella pathogenicity island 1-mediated host cell death. J Immunol 63. Tobar JA, Gonzalez PA, Kalergis AM. 2004. Salmonella
169:3275–3283. escape from antigen presentation can be overcome by targeting bac-
47. Wijburg OL, Simmons CP, van Rooijen N, Strugnell RA. 2000. teria to Fc gamma receptors on dendritic cells. J Immunol 173:4058–
Dual role for macrophages in vivo in pathogenesis and control of 4065.
murine Salmonella enterica var. Typhimurium infections. Eur J 64. Bueno SM, Gonzalez PA, Carreno LJ, Tobar JA, Mora GC,
Immunol 30:944–953. Pereda CJ, Salazar-Onfray F, Kalergis AM. 2008. The capacity of
48. Bueno SM, Gonzalez PA, Schwebach JR, Kalergis AM. 2007. Salmonella to survive inside dendritic cells and prevent antigen pre-
T cell immunity evasion by virulent Salmonella enterica. Immunol sentation to T cells is host specific. Immunology 124:522–533.
Lett 111:14–20. 65. van der Velden AW, Copass MK, Starnbach MN. 2005. Sal-
49. Tobar JA, Carreno LJ, Bueno SM, Gonzalez PA, Mora JE, monella inhibit T cell proliferation by a direct, contact-dependent
Quezada SA, Kalergis AM. 2006. Virulent Salmonella enterica immunosuppressive effect. Proc Natl Acad Sci USA 102:17769–17774.
serovar typhimurium evades adaptive immunity by preventing den- 66. van der Velden AW, Dougherty JT, Starnbach MN. 2008. Down-
dritic cells from activating T cells. Infect Immun 74:6438–6448. modulation of TCR expression by Salmonella enterica serovar
50. Alaniz RC, Cummings LA, Bergman MA, Rassoulian-Barrett Typhimurium. J Immunol 180:5569–5574.
SL, Cookson BT. 2006. Salmonella typhimurium coordinately 67. Hess J, Ladel C, Miko D, Kaufmann SH. 1996. Salmonella
regulates FliC location and reduces dendritic cell activation and an- typhimurium aroA− infection in gene-targeted immunodeficient
tigen presentation to CD4+ T cells. J Immunol 177:3983–3993. mice: major role of CD4+ TCR-alpha beta cells and IFN-gamma in
51. Cummings LA, Wilkerson WD, Bergsbaken T, Cookson BT. bacterial clearance independent of intracellular location. J Immunol
2006. In vivo, fliC expression by Salmonella enterica serovar Typhi- 156:3321–3326.
murium is heterogeneous, regulated by ClpX, and anatomically re- 68. Mastroeni P, Villarreal RB, Hormaeche CE. 1992. Role of T cells,
stricted. Mol Microbiol 61:795–809. TNF alpha and IFN gamma in recall of immunity to oral challenge
52. Cummings LA, Barrett SL, Wilkerson WD, Fellnerova I, with virulent Salmonellae in mice vaccinated with live attenuated aro-
Cookson BT. 2005. FliC-specific CD4+ T cell responses are restricted Salmonella vaccines. Microb Pathog 13:477–491.
by bacterial regulation of antigen expression. J Immunol 174:7929– 69. Nauciel C. 1990. Role of CD4+ T cells and T-independent
7938. mechanisms in acquired resistance to Salmonella typhimurium in-
53. Alpuche Aranda CM, Swanson JA, Loomis WP, Miller SI. 1992. fection. J Immunol 145:1265–1269.
Salmonella typhimurium activates virulence gene transcription within 70. Weintraub BC, Eckmann L, Okamoto S, Hense M, Hedrick SM,
acidified macrophage phagosomes. Proc Natl Acad Sci USA 89:10079– Fierer J. 1997. Role of alphabeta and gammadelta T cells in the host
10083. response to Salmonella infection as demonstrated in T-cell-receptor-
54. Groisman EA. 2001. The pleiotropic two-component regulatory deficient mice of defined Ity genotypes. Infect Immun 65:2306–2312.
system PhoP-PhoQ. J Bacteriol 183:1835–1842. 71. Bettelli E, Korn T, Kuchroo VK. 2007. Th17: the third member of
55. Ernst RK, Guina T, Miller SI. 1999. How intracellular bacteria the effector T cell trilogy. Curr Opin Immunol 19:652–657.
survive: surface modifications that promote resistance to host innate 72. Harrington LE, Mangan PR, Weaver CT. 2006. Expanding the
immune responses. J Infect Dis 179(Suppl. 2):S326–S330. effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol
56. Gunn JS. 2001. Bacterial modification of LPS and resistance to 18:349–356.
antimicrobial peptides. J Endotoxin Res 7:57–62.
73. Korn T, Oukka M, Kuchroo V, Bettelli E. 2007. Th17 cells:
57. Kawasaki K, Ernst RK, Miller SI. 2004. 3-O-deacylation of lipid effector T cells with inflammatory properties. Semin Immunol 19:
A by PagL, a PhoP/PhoQ-regulated deacylase of Salmonella typhi- 362–371.
murium, modulates signaling through Toll-like receptor 4. J Biol
Chem 279:20044–20048. 74. Weaver CT, Hatton RD, Mangan PR, Harrington LE. 2007.
IL-17 family cytokines and the expanding diversity of effector T cell
58. Murata T, Tseng W, Guina T, Miller SI, Nikaido H. 2007. lineages. Annu Rev Immunol 25:821–852.
PhoPQ-mediated regulation produces a more robust permeability
barrier in the outer membrane of Salmonella enterica serovar typhi- 75. Hayday AC. 2000. [gamma][delta] cells: a right time and a right
murium. J Bacteriol 189:7213–7222. place for a conserved third way of protection. Annu Rev Immunol 18:
975–1026.
59. Bergman MA, Cummings LA, Barrett SL, Smith KD, Lara JC,
Aderem A, Cookson BT. 2005. CD4+ T cells and toll-like receptors 76. Ravindran R, McSorley SJ. 2005. Tracking the dynamics of T-cell
recognize Salmonella antigens expressed in bacterial surface activation in response to Salmonella infection. Immunology 114:450–
organelles. Infect Immun 73:1350–1356. 458.
60. Fink SL, Bergsbaken T, Cookson BT. 2008. Anthrax lethal toxin 77. Kirby AC, Sundquist M, Wick MJ. 2004. In vivo compartmen-
and Salmonella elicit the common cell death pathway of caspase- talization of functionally distinct, rapidly responsive antigen-specific
1-dependent pyroptosis via distinct mechanisms. Proc Natl Acad Sci T-cell populations in DNA-immunized or Salmonella enterica serovar
USA 105:4312–4317. Typhimurium-infected mice. Infect Immun 72:6390–6400.
61. Eguchi M, Sekiya Y, Kikuchi Y, Takaya A, Yamamoto T, 78. Lo WF, Ong H, Metcalf ES, Soloski MJ. 1999. T cell responses to
Matsui H. 2007. Expressed Salmonella antigens within macrophages Gram-negative intracellular bacterial pathogens: a role for CD8+
enhance the proliferation of CD4+ and CD8+ T lymphocytes by T cells in immunity to Salmonella infection and the involvement of
means of bystander dendritic cells. FEMS Immunol Med Microbiol 50: MHC class Ib molecules. J Immunol 162:5398–5406.
411–420. 79. Mittrucker HW, Kohler A, Kaufmann SH. 2002. Characteriza-
62. Yrlid U, Wick MJ. 2000. Salmonella-induced apoptosis of in- tion of the murine T-lymphocyte response to Salmonella enterica
fected macrophages results in presentation of a bacteria-encoded serovar Typhimurium infection. Infect Immun 70:199–203.
antigen after uptake by bystander dendritic cells. J Exp Med 191:613– 80. Lundin BS, Johansson C, Svennerholm AM. 2002. Oral immu-
624. nization with a Salmonella enterica serovar typhi vaccine induces

ASMScience.org/EcoSalPlus 19
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

specific circulating mucosa-homing CD4(+) and CD8(+) T cells in 2007. Risk of recurrent nontyphoid Salmonella bacteremia in HIV-
humans. Infect Immun 70:5622–5627. infected patients in the era of highly active antiretroviral therapy and
81. Salerno-Goncalves R, Fernandez-Vina M, Lewinsohn DM, an increasing trend of fluoroquinolone resistance. Clin Infect Dis 45:
Sztein MB. 2004. Identification of a human HLA-E-restricted CD8+ T e60–e67.
cell subset in volunteers immunized with Salmonella enterica serovar 97. Kankwatira AM, Mwafulirwa GA, Gordon MA. 2004. Non-
Typhi strain Ty21a typhoid vaccine. J Immunol 173:5852–5862. typhoidal Salmonella bacteraemia—an under-recognized feature of
82. Salerno-Goncalves R, Pasetti MF, Sztein MB. 2002. Character- AIDS in African adults. Trop Doct 34:198–200.
ization of CD8(+) effector T cell responses in volunteers immunized 98. Nelson MR, Shanson DC, Hawkins DA, Gazzard BG. 1992.
with Salmonella enterica serovar Typhi strain Ty21a typhoid vaccine. Salmonella, Campylobacter and Shigella in HIV-seropositive patients.
J Immunol 169:2196–2203. AIDS 6:1495–1498.
83. Salerno-Goncalves R, Wyant TL, Pasetti MF, Fernandez-Vina 99. Glaser JB, Morton-Kute L, Berger SR, Weber J, Siegal FP, Lopez
M, Tacket CO, Levine MM, Sztein MB. 2003. Concomitant induc- C, Robbins W, Landesman SH. 1985. Recurrent Salmonella typhi-
tion of CD4+ and CD8+ T cell responses in volunteers immunized murium bacteremia associated with the acquired immunodeficiency
with Salmonella enterica serovar typhi strain CVD 908-htrA. J syndrome. Ann Intern Med 102:189–193.
Immunol 170:2734–2741. 100. van Diepen A, Koudijs JSvan de Gevel MM, Ossendorp F,
84. Sztein MB, Tanner MK, Polotsky Y, Orenstein JM, Levine MM. Beekhuizen H, Janssen R, JTvan Dissel. 2005. Gamma irradiation or
1995. Cytotoxic T lymphocytes after oral immunization with atten- CD4+-T-cell depletion causes reactivation of latent Salmonella
uated vaccine strains of Salmonella typhi in humans. J Immunol 155: enterica serovar Typhimurium infection in C3H/HeN mice. Infect.
3987–3993. Immun. 73:2857–2862.
85. Bumann D. 2001. In vivo visualization of bacterial colonization, 101. Soloski MJ, Metcalf ES. 2001. The involvement of class Ib
antigen expression, and specific T-cell induction following oral ad- molecules in the host response to infection with Salmonella and its
ministration of live recombinant Salmonella enterica serovar Typhi- relevance to autoimmunity. Microbes Infect 3:1249–1259.
murium. Infect Immun 69:4618–4626. 102. Ugrinovic S, Brooks CG, Robson J, Blacklaws BA, Hormaeche
86. McSorley SJ, Asch S, Costalonga M, Reinhardt RL, Jenkins MK. CE, Robinson JH. 2005. H2–M3 major histocompatibility complex
2002. Tracking Salmonella-specific CD4 T cells in vivo reveals a local class Ib-restricted CD8 T cells induced by Salmonella enterica serovar
mucosal response to a disseminated infection. Immunity 16:365–377. Typhimurium infection recognize proteins released by Salmonella
87. Davies A, Lopez-Briones S, Ong H, O’Neil-Marshall C, serovar Typhimurium. Infect Immun 73:8002–8008.
Lemonnier FA, Nagaraju K, Metcalf ES, Soloski MJ. 2004. 103. Bao S, Beagley KW, France MP, Shen J, Husband AJ. 2000.
Infection-induced expansion of a MHC Class Ib-dependent intestinal Interferon-gamma plays a critical role in intestinal immunity against
intraepithelial gammadelta T cell subset. J Immunol 172:6828–6837. Salmonella typhimurium infection. Immunology 99:464–472.
88. Mixter PF, Camerini V, Stone BJ, Miller VL, Kronenberg M. 104. Mastroeni P, Villarreal-Ramos B, Hormaeche CE. 1993. Effect
1994. Mouse T lymphocytes that express a gamma delta T-cell antigen of late administration of anti-TNF alpha antibodies on a Salmonella
receptor contribute to resistance to Salmonella infection in vivo. Infect infection in the mouse model. Microb Pathog 14:473–480.
Immun 62:4618–4621. 105. Muotiala A, Makela PH. 1993. Role of gamma interferon in late
89. Sinha K, Mastroeni P, Harrison J, de Hormaeche RD, stages of murine salmonellosis. Infect Immun 61:4248–4253.
Hormaeche CE. 1997. Salmonella typhimurium aroA, htrA, and aroD 106. Nauciel C, Espinasse-Maes F. 1992. Role of gamma interferon
htrA mutants cause progressive infections in athymic (nu/nu) BALB/c and tumor necrosis factor alpha in resistance to Salmonella typhi-
mice. Infect Immun 65:1566–1569. murium infection. Infect Immun 60:450–454.
90. Hormaeche CE, Mastroeni P, Arena A, Uddin J, Joysey HS. 107. Ravindran R, Foley J, Stoklasek T, Glimcher LH, McSorley SJ.
1990. T cells do not mediate the initial suppression of a Salmonella 2005. Expression of T-bet by CD4 T cells is essential for resistance to
infection in the RES. Immunology 70:247–250. Salmonella infection. J Immunol 175:4603–4610.
91. Paul C, Shalala K, Warren R, Smith R. 1985. Adoptive transfer of 108. Fernandez-Cabezudo MJ, Ali SA, Ullah A, Hasan MY,
murine host protection to salmonellosis with T-cell growth factor- Kosanovic M, Fahim MA, Adem A, al-Ramadi BK. 2007. Pro-
dependent, Salmonella-specific T-cell lines. Infect Immun 48:40–43. nounced susceptibility to infection by Salmonella enterica serovar
92. Paul CC, Norris K, Warren R, Smith RA. 1988. Transfer of Typhimurium in mice chronically exposed to lead correlates with a
murine host protection by using interleukin-2-dependent T-lym- shift to Th2-type immune responses. Toxicol Appl Pharmacol 218:
phocyte lines. Infect Immun 56:2189–2192. 215–226.
93. Centers for Disease Control Prevention. 1992. 1993 revised 109. Ramarathinam L, Shaban RA, Niesel DW, Klimpel GR. 1991.
classification system for HIV infection and expanded surveillance case Interferon gamma (IFN-gamma) production by gut-associated lym-
definition for AIDS among adolescents and adults. MMWR Recom- phoid tissue and spleen following oral Salmonella typhimurium
mend Rep 41:1–19. challenge. Microb Pathog 11:347–356.
94. Grant AD, Sidibe K, Domoua K, Bonard D, Sylla-Koko F, 110. Mora JR, Bono MR, Manjunath N, Weninger W, Cavanagh
Dosso M, Yapi A, Maurice C, Whitaker JP, Lucas SB, Hayes RJ, LL, Rosemblatt M, Von Andrian UH. 2003. Selective imprinting of
Wiktor SZ, De Cock KM, Greenberg AE. 1998. Spectrum of gut-homing T cells by Peyer’s patch dendritic cells. Nature 424:88–93.
disease among HIV-infected adults hospitalised in a respiratory 111. Mastroeni P. 2002. Immunity to systemic Salmonella infections.
medicine unit in Abidjan, Cote d’Ivoire. Int. J Tuberc. Lung Dis. 2: Curr Mol Med 2:393–406.
926–934. 112. Price JD, Simpfendorfer KR, Mantena RR, Holden J, Heath
95. Hohmann EL. 2001. Nontyphoidal salmonellosis. Clin Infect Dis WR, van Rooijen N, Strugnell RA, Wijburg OL. 2007. Gamma
32:263–269. interferon-independent effects of interleukin-12 on immunity to
96. Hung CC, Hung MN, Hsueh PR, Chang SY, Chen MY, Hsieh Salmonella enterica serovar Typhimurium. Infect Immun 75:5753–
SM, Sheng WH, Sun HY, Huang YT, Lo YC, Hsiao CF, Chang SC. 5762.

20 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

113. Ramarathinam L, Niesel DW, Klimpel GR. 1993. Salmonella typhimurium for presentation of a CD4 T cell epitope from the Sal-
typhimurium induces IFN-gamma production in murine splenocytes. monella invasion protein C (SipC). Eur J Immunol 32:2664–2671.
Role of natural killer cells and macrophages. J Immunol 150:3973– 132. Lo WF, Woods AS, DeCloux A, Cotter RJ, Metcalf ES,
3981. Soloski MJ. 2000. Molecular mimicry mediated by MHC class Ib
114. Shedlock DJ, Shen H. 2003. Requirement for CD4 T cell help in molecules after infection with gram-negative pathogens. Nat Med 6:
generating functional CD8 T cell memory. Science 300:337–339. 215–218.
115. Sun JC, Williams MA, Bevan MJ. 2004. CD4+ T cells are re- 133. Ochoa-Reparaz J, Garcia B, Solano C, Lasa I, Irache JM,
quired for the maintenance, not programming, of memory CD8+ T Gamazo C. 2005. Protective ability of subcellular extracts from Sal-
cells after acute infection. Nat Immunol 5:927–933. monella Enteritidis and from a rough isogenic mutant against sal-
116. Luu RA, Gurnani K, Dudani R, Kammara R, van Faassen H, monellosis in mice. Vaccine 23:1491–1501.
Sirard JC, Krishnan L, Sad S. 2006. Delayed expansion and con- 134. Ogunniyi AD, Kotlarski I, Morona R, Manning PA. 1997. Role
traction of CD8+ T cell response during infection with virulent Sal- of SefA subunit protein of SEF14 fimbriae in the pathogenesis of
monella typhimurium. J Immunol 177:1516–1525. Salmonella enterica serovar Enteritidis. Infect Immun 65:708–717.
117. Stenger S, Hanson DA, Teitelbaum R, Dewan P, Niazi KR, 135. Ogunniyi AD, Manning PA, Kotlarski I. 1994. A Salmonella
Froelich CJ, Ganz T, Thoma-Uszynski S, Melian A, Bogdan C, enteritidis 11RX pilin induces strong T-lymphocyte responses. Infect
Porcelli SA, Bloom BR, Krensky AM, Modlin RL. 1998. An anti- Immun 62:5376–5383.
microbial activity of cytolytic T cells mediated by granulysin. Science 136. Diaz-Quinonez A, Martin-Orozco N, Isibasi A, Ortiz-
282:121–125. Navarrete V. 2004. Two Salmonella OmpC K(b)-restricted epitopes
118. Mittrucker HW, Kohler A, Mak TW, Kaufmann SH. 1999. for CD8+-T-cell recognition. Infect Immun 72:3059–3062.
Critical role of CD28 in protective immunity against Salmonella 137. Galdiero M, De Martino L, Marcatili A, Nuzzo I, Vitiello M,
typhimurium. J Immunol 163:6769–6776. Cipollaro de l’Ero G. 1998. Th1 and Th2 cell involvement in immune
119. Yonekura K, Maki-Yonekura S, Namba K. 2002. Growth response to Salmonella typhimurium porins. Immunology 94:5–13.
mechanism of the bacterial flagellar filament. Res Microbiol 153:191– 138. Gupta S. 1998. Priming of T-cell responses in mice by porins of
197. Salmonella typhimurium. Scand J Immunol 48:136–143.
120. Bergman MA, Cummings LA, Alaniz RC, Mayeda L, 139. Sood S, Rishi P, Vohra H, Sharma S, Ganguly NK. 2005.
Fellnerova I, Cookson BT. 2005. CD4+-T-cell responses generated Cellular immune response induced by Salmonella enterica serotype
during murine Salmonella enterica serovar Typhimurium infection are Typhi iron-regulated outer-membrane proteins at peripheral and
directed towards multiple epitopes within the natural antigen FliC. mucosal levels. J Med Microbiol 54:815–821.
Infect Immun 73:7226–7235.
140. Alaniz RC, Deatherage BL, Lara JC, Cookson BT. 2007.
121. Joys TM, Schödel F. 1991. Epitope mapping of the d flagellar Membrane vesicles are immunogenic facsimiles of Salmonella typhi-
antigen of Salmonella muenchen. Infect Immun 59:3330–3332. murium that potently activate dendritic cells, prime B and T cell re-
122. Joys TM, Street NE. 1993. Mapping of T-cell epitopes of flagellar sponses, and stimulate protective immunity in vivo. J Immunol 179:
antigen d of Salmonella muenchen. Infect Immun 61:1146–1148. 7692–7701.
123. McSorley SJ, Cookson BT, Jenkins MK. 2000. Characterization 141. Kitamura D, Roes J, Kuhn R, Rajewsky K. 1991. A B cell-
of CD4+ T cell responses during natural infection with Salmonella deficient mouse by targeted disruption of the membrane exon of the
typhimurium. J Immunol 164:986–993. immunoglobulin mu chain gene. Nature 350:423–426.
124. Strindelius L, Filler M, Sjoholm I. 2004. Mucosal immunization 142. Cunningham AF, Gaspal F, Serre K, Mohr E, Henderson IR,
with purified flagellin from Salmonella induces systemic and mucosal Scott-Tucker A, Kenny SM, Khan M, Toellner KM, Lane PJ,
immune responses in C3H/HeJ mice. Vaccine 22:3797–3808. MacLennan IC. 2007. Salmonella induces a switched antibody re-
125. Gewirtz AT, Navas TA, Lyons S, Godowski PJ, Madara JL. sponse without germinal centers that impedes the extracellular spread
2001. Cutting edge: bacterial flagellin activates basolaterally expressed of infection. J Immunol 178:6200–6207.
TLR5 to induce epithelial proinflammatory gene expression. J 143. Mastroeni P, Simmons C, Fowler R, Hormaeche CE, Dougan
Immunol 167:1882–1885. G. 2000. Igh-6(−/−) (B-cell-deficient) mice fail to mount solid ac-
126. Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett quired resistance to oral challenge with virulent Salmonella enterica
DR, Eng JK, Akira S, Underhill DM, Aderem A. 2001. The innate serovar typhimurium and show impaired Th1 T-cell responses to
immune response to bacterial flagellin is mediated by Toll-like re- Salmonella antigens. Infect Immun 68:46–53.
ceptor 5. Nature 410:1099–1103. 144. McSorley SJ, Jenkins MK. 2000. Antibody is required for pro-
127. Miao EA, Andersen-Nissen E, Warren SE, Aderem A. 2007. tection against virulent but not attenuated Salmonella enterica serovar
TLR5 and Ipaf: dual sensors of bacterial flagellin in the innate immune typhimurium. Infect Immun 68:3344–3348.
system. Semin Immunopathol 29:275–288. 145. Mittrucker HW, Raupach B, Kohler A, Kaufmann SH. 2000.
128. Kutsukake K, Ohya Y, Iino T. 1990. Transcriptional analysis of Cutting edge: role of B lymphocytes in protective immunity against
the flagellar regulon of Salmonella typhimurium. J Bacteriol 172:741– Salmonella typhimurium infection. J Immunol 164:1648–1652.
747. 146. Childers NK, Bruce MG, McGhee JR. 1989. Molecular mech-
129. Cookson BT, Cummings LA, Rassoulian Barrett SL. 2001. anisms of immunoglobulin A defense. Annu Rev Microbiol 43:503–
Bacterial antigens elicit T cell responses via adaptive and transitional 536.
immune recognition. Curr. Opin. Microbiol. 4:267–273. 147. Michetti P, Mahan MJ, Slauch JM, Mekalanos JJ, Neutra MR.
130. Vordermeier HM, Kotlarski I. 1990. Identification of antigens 1992. Monoclonal secretory immunoglobulin A protects mice against
which stimulate T lymphocytes of Salmonella enteritidis 11RX im- oral challenge with the invasive pathogen Salmonella typhimurium.
munized mice. Immunol Cell Biol 68(Pt 5):299–305. Infect Immun 60:1786–1792.
131. Musson JA, Hayward RD, Delvig AA, Hormaeche CE, 148. Michetti P, Porta N, Mahan MJ, Slauch JM, Mekalanos JJ,
Koronakis V, Robinson JH. 2002. Processing of viable Salmonella Blum AL, Kraehenbuhl JP, Neutra MR. 1994. Monoclonal immu-

ASMScience.org/EcoSalPlus 21
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Cummings et al.

noglobulin A prevents adherence and invasion of polarized epithelial lipopolysaccharide of Salmonella typhimurium. Microb Pathog 21:
cell monolayers by Salmonella typhimurium. Gastroenterology 107: 249–263.
915–923. 166. Svenson SB, Nurminen M, Lindberg AA. 1979. Artificial Sal-
149. Uren TK, Wijburg OL, Simmons C, Johansen FE, Brandtzaeg monella vaccines: O-antigenic oligosaccharide-protein conjugates in-
P, Strugnell RA. 2005. Vaccine-induced protection against gastro- duce protection against infection with Salmonella typhimurium. Infect
intestinal bacterial infections in the absence of secretory antibodies. Immun 25:863–872.
Eur J Immunol 35:180–188. 167. Verdugo-Rodriguez A, Gam LH, Devi S, Koh CL, Puthucheary
150. Pecquet SS, Ehrat C, Ernst PB. 1992. Enhancement of mucosal SD, Calva E, Pang T. 1993. Detection of antibodies against Salmonella
antibody responses to Salmonella typhimurium and the microbial typhi outer membrane protein (OMP) preparation in typhoid fever
hapten phosphorylcholine in mice with X-linked immunodeficiency patients. Asian Pac J Allergy Immunol 11:45–52.
by B-cell precursors from the peritoneal cavity. Infect Immun 60:503– 168. Puohiniemi R, Karvonen M, Vuopio-Varkila J, Muotiala A,
509. Helander IM, Sarvas M. 1990. A strong antibody response to the
151. Kantor AB, Herzenberg LA. 1993. Origin of murine B cell periplasmic C-terminal domain of the OmpA protein of Escherichia
lineages. Annu Rev Immunol 11:501–538. coli is produced by immunization with purified OmpA or with whole
152. Whitmore AC, Haughton G, Arnold LW. 1992. Isotype E. coli or Salmonella typhimurium bacteria. Infect Immun 58:1691–
switching in CD5 B cells. Ann N Y Acad Sci 651:143–151. 1696.
153. Wijburg OL, Uren TK, Simpfendorfer K, Johansen FE, 169. Sugawara E, Nikaido H. 1994. OmpA protein of Escherichia coli
Brandtzaeg P, Strugnell RA. 2006. Innate secretory antibodies outer membrane occurs in open and closed channel forms. J Biol
protect against natural Salmonella typhimurium infection. J Exp Med Chem 269:17981–17987.
203:21–26. 170. Singh SP, Williams YU, Miller S, Nikaido H. 2003. The C-
154. Hormaeche CE. 1990. Dead salmonellae or their endotoxin terminal domain of Salmonella enterica serovar typhimurium OmpA is
accelerate the early course of a Salmonella infection in mice. Microb an immunodominant antigen in mice but appears to be only partially
Pathog 9:213–218. exposed on the bacterial cell surface. Infect Immun 71:3937–3946.
155. O’Brien AD, Metcalf ES, Rosenstreich DL. 1982. Defect in 171. Calderon I, Lobos SR, Rojas HA, Palomino C, Rodriguez LH,
macrophage effector function confers Salmonella typhimurium sus- Mora GC. 1986. Antibodies to porin antigens of Salmonella typhi in-
ceptibility on C3H/HeJ mice. Cell Immunol 67:325–333. duced during typhoid infection in humans. Infect Immun 52:209–212.
156. Vidal SM, Malo D, Vogan K, Skamene E, Gros P. 1993. Natural 172. Ortiz V, Isibasi A, Garcia-Ortigoza E, Kumate J. 1989. Im-
resistance to infection with intracellular parasites: isolation of a can- munoblot detection of class-specific humoral immune response to
didate for Bcg. Cell 73:469–485. outer membrane proteins isolated from Salmonella typhi in humans
157. Eisenstein TK, Killar LM, Sultzer BM. 1984. Immunity to in- with typhoid fever. J Clin Microbiol 27:1640–1645.
fection with Salmonella typhimurium: mouse-strain differences in 173. De Almeida ME, Newton SM, Ferreira LC. 1999. Antibody
vaccine- and serum-mediated protection. J Infect Dis 150:425–435. responses against flagellin in mice orally immunized with attenuated
158. O’Brien AD, Scher I, Metcalf ES. 1981. Genetically conferred Salmonella vaccine strains. Arch Microbiol 172:102–108.
defect in anti-Salmonella antibody formation renders CBA/N mice 174. Sbrogio-Almeida ME, Mosca T, Massis LM, Abrahamsohn IA,
innately susceptible to Salmonella typhimurium infection. J Immunol Ferreira LC. 2004. Host and bacterial factors affecting induction of
126:1368–1372. immune responses to flagellin expressed by attenuated Salmonella
159. Brown A, Hormaeche CE. 1989. The antibody response to vaccine strains. Infect Immun 72:2546–2555.
salmonellae in mice and humans studied by immunoblots and ELISA. 175. Tacket CO, Sztein MB, Losonsky GA, Wasserman SS, Nataro
Microb Pathog 6:445–454. JP, Edelman R, Pickard D, Dougan G, Chatfield SN, Levine MM.
160. Matsiota-Bernard P, Mahana W, Avrameas S, Nauciel C. 1993. 1997. Safety of live oral Salmonella typhi vaccine strains with deletions
Specific and natural antibody production during Salmonella typhi- in htrA and aroC aroD and immune response in humans. Infect
murium infection in genetically susceptible and resistant mice. Im- Immun 65:452–456.
munology 79:375–380. 176. Humphries A, Deridder S, Baumler AJ. 2005. Salmonella
161. Xu HR, Tan YY, Hsu HS, Moncure CW, Wang XM. 1993. enterica serotype Typhimurium fimbrial proteins serve as antigens
Comparative antibody response to Salmonella antigens in genetically during infection of mice. Infect Immun 73:5329–5338.
resistant and susceptible mice. Clin Exp Immunol 91:73–77. 177. Levine MM, Tacket CO, Sztein MB. 2001. Host-Salmonella
162. Singh SP, Williams YU, Klebba PE, Macchia P, Miller S. 2000. interaction: human trials. Microbes Infect 3:1271–1279.
Immune recognition of porin and lipopolysaccharide epitopes of 178. Bhan MK, Bahl R, Bhatnagar S. 2005. Typhoid and paratyphoid
Salmonella typhimurium in mice. Microb Pathog 28:157–167. fever. Lancet 366:749–762.
163. Hormaeche CE, Mastroeni P, Harrison JA, Demarco de 179. Arya SC, Sharma KB. 1995. Urgent need for effective vaccine
Hormaeche R, Svenson S, Stocker BA. 1996. Protection against oral against Salmonella paratyphi A, B and C. Vaccine 13:1727–1728.
challenge three months after i.v. immunization of BALB/c mice with 180. Ochiai RL, Wang X, von Seidlein L, Yang J, Bhutta ZA,
live Aro Salmonella typhimurium and Salmonella enteritidis vaccines Bhattacharya SK, Agtini M, Deen JL, Wain J, Kim DR, Ali M,
is serotype (species)-dependent and only partially determined by the Acosta CJ, Jodar L, Clemens JD. 2005. Salmonella paratyphi A rates,
main LPS O antigen. Vaccine 14:251–259. Asia. Emerg. Infect. Dis. 11:1764–1766.
164. Lindberg AA, Segall T, Weintraub A, Stocker BA. 1993. An- 181. Sood S, Kapil A, Dash N, Das BK, Goel V, Seth P. 1999.
tibody response and protection against challenge in mice vaccinated Paratyphoid fever in India: an emerging problem. Emerg Infect Dis 5:
intraperitoneally with a live aroA O4–O9 hybrid Salmonella dublin 483–484.
strain. Infect Immun 61:1211–1221. 182. Vollaard AM, Ali S, Widjaja HAvan Asten S, Visser LG,
165. Singh SP, Williams YU, Benjamin WH, Klebba PE, Boyd D. Surjadi C, JTvan Dissel. 2004. Risk factors for typhoid and paraty-
1996. Immunoprotection by monoclonal antibodies to the porins and phoid fever in Jakarta, Indonesia. JAMA 291:2607–2615.

22 ASMScience.org/EcoSalPlus
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Adaptive Immune Responses during Salmonella Infection

183. Sexton K, Lennon D, Oster P, Crengle S, Martin D, 199. Gewirtz AT, Simon PO Jr, Schmitt CK, Taylor LJ, Hagedorn
Mulholland K, Percival T, Reid S, Stewart J, O’Hallahan J. 2004. CH, O’Brien AD, Neish AS, Madara JL. 2001. Salmonella typhi-
The New Zealand Meningococcal Vaccine Strategy: a tailor-made murium translocates flagellin across intestinal epithelia, inducing a
vaccine to combat a devastating epidemic. N Z Med J 117:U1015. proinflammatory response. J Clin Invest 107:99–109.
184. Barr TA, Brown S, Ryan G, Zhao J, Gray D. 2007. TLR- 200. Tallant T, Deb A, Kar N, Lupica J, DiDonato MJde Veer JA.
mediated stimulation of APC: distinct cytokine responses of B cells 2004. Flagellin acting via TLR5 is the major activator of key signaling
and dendritic cells. Eur J Immunol 37:3040–3053. pathways leading to NF-kappa B and proinflammatory gene program
185. Harris DP, Goodrich S, Mohrs K, Mohrs M, Lund FE. 2005. activation in intestinal epithelial cells. BMC Microbiol 4:33.
Cutting edge: the development of IL-4-producing B cells (B effector 2 201. Zeng H, Carlson AQ, Guo Y, Yu Y, Collier-Hyams LS, Madara
cells) is controlled by IL-4, IL-4 receptor alpha, and Th2 cells. JL, Gewirtz AT, Neish AS. 2003. Flagellin is the major proinflam-
J Immunol 175:7103–7107. matory determinant of enteropathogenic Salmonella. J Immunol 171:
186. Skok J, Poudrier J, Gray D. 1999. Dendritic cell-derived IL-12 3668–3674.
promotes B cell induction of Th2 differentiation: a feedback regulation 202. Huang FC, Werne A, Li Q, Galyov EE, Walker WA, Cherayil
of Th1 development. J Immunol 163:4284–4291. BJ. 2004. Cooperative interactions between flagellin and SopE2 in the
187. Chesnut RW, Grey HM. 1981. Studies on the capacity of B cells epithelial interleukin-8 response to Salmonella enterica serovar
to serve as antigen-presenting cells. J Immunol 126:1075–1079. typhimurium infection. Infect Immun 72:5052–5062.
203. Sierro F, Dubois B, Coste A, Kaiserlian D, Kraehenbuhl JP,
188. Davidson HW, Watts C. 1989. Epitope-directed processing of
specific antigen by B lymphocytes. J Cell Biol 109:85–92. Sirard JC. 2001. Flagellin stimulation of intestinal epithelial cells
triggers CCL20-mediated migration of dendritic cells. Proc Natl Acad
189. Mamula MJ, Janeway CA Jr. 1993. Do B cells drive the diver- Sci USA 98:13722–13727.
sification of immune responses? Immunol. Today 14:151–154.
204. Ramos HC, Rumbo M, Sirard JC. 2004. Bacterial flagellins:
190. Rock KL, Benacerraf B, Abbas AK. 1984. Antigen presentation mediators of pathogenicity and host immune responses in mucosa.
by hapten-specific B lymphocytes. I Role of surface immunoglobulin Trends Microbiol 12:509–517.
receptors J Exp Med 160:1102–1113.
205. McDermott PF, Ciacci-Woolwine F, Snipes JA, Mizel SB. 2000.
191. Lanzavecchia A. 1985. Antigen-specific interaction between T High-affinity interaction between gram-negative flagellin and a cell
and B cells. Nature 314:537–539. surface polypeptide results in human monocyte activation. Infect
192. Yrlid U, Wick MJ. 2002. Antigen presentation capacity and Immun 68:5525–5529.
cytokine production by murine splenic dendritic cell subsets upon 206. Means TK, Hayashi F, Smith KD, Aderem A, Luster AD. 2003.
Salmonella encounter. J Immunol 169:108–116. The Toll-like receptor 5 stimulus bacterial flagellin induces matura-
193. Garcia-del Portillo F, Stein MA, Finlay BB. 1997. Release of tion and chemokine production in human dendritic cells. J Immunol
lipopolysaccharide from intracellular compartments containing Sal- 170:5165–5175.
monella typhimurium to vesicles of the host epithelial cell. Infect 207. Fink SL, Cookson BT. 2005. Apoptosis, pyroptosis, and necrosis:
Immun 65:24–34. mechanistic description of dead and dying eukaryotic cells. Infect
194. Rosenberger CM, Gallo RL, Finlay BB. 2004. Interplay between Immun 73:1907–1916.
antibacterial effectors: a macrophage antimicrobial peptide impairs 208. Hoiseth SK, Stocker BA. 1981. Aromatic-dependent Salmonella
intracellular Salmonella replication. Proc Natl Acad Sci USA 101: typhimurium are non-virulent and effective as live vaccines. Nature
2422–2427. 291:238–239.
195. Ayabe T, Ashida T, Kohgo Y, Kono T. 2004. The role of Paneth 209. O’Callaghan D, Maskell D, Liew FY, Easmon CS, Dougan G.
cells and their antimicrobial peptides in innate host defense. Trends 1988. Characterization of aromatic- and purine-dependent Salmonella
Microbiol 12:394–398. typhimurium: attention, persistence, and ability to induce protective
196. Ganz T. 2003. Defensins: antimicrobial peptides of innate im- immunity in BALB/c mice. Infect Immun 56:419–423.
munity. Nat Rev Immunol 3:710–720. 210. Mastroeni P, Menager N. 2003. Development of acquired im-
197. Meynell GG. 1957. The applicability of the hypothesis of inde- munity to Salmonella. J Med Microbiol 52:453–459.
pendent action to fatal infections in mice given Salmonella typhimu- 211. Mittrucker HW, Kaufmann SH. 2000. Immune response to
rium by mouth. J Gen Microbiol 16:396–404. infection with Salmonella typhimurium in mice. J Leukoc Biol 67:457–
198. Meynell GG, Stocker BA. 1957. Some hypotheses on the 463.
aetiology of fatal infections in partially resistant hosts and their ap- 212. Liao F, Rabin RL, Smith CS, Sharma G, Nutman TB, Farber
plication to mice challenged with Salmonella paratyphi-B or Sal- JM. 1999. CC-chemokine receptor 6 is expressed on diverse memory
monella typhimurium by intraperitoneal injection. J Gen Microbiol subsets of T cells and determines responsiveness to macrophage in-
16:38–58. flammatory protein 3 alpha. J Immunol 162:186–194.

ASMScience.org/EcoSalPlus 23
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28
Downloaded from www.asmscience.org by
IP: 137.111.226.20
On: Mon, 28 Nov 2016 16:04:28

You might also like