ArticlePDF AvailableLiterature Review

Abstract and Figures

Nucleotide oligomerization domain (Nod)-like Receptors (NLRs) are cytosolic sensors that mediate the activation of Caspase-1 and the subsequent processing and secretion of the pro-inflammatory cytokines IL-1β and IL-18, as well as an inflammatory cell death termed pyroptosis. While a multitude of bacteria have been shown to activate one or more NLRs under in vitro conditions, the exact impact of NLR activation during the course of colonization, both of pathogenic and commensal nature, is less understood. In this review, we will focus on the role of intestinal NLRs during the various stages of infection with common gastrointestinal bacterial pathogens, as well as NLR function in controlling and shaping the microbiota.
Content may be subject to copyright.
REVIEW ARTICLE
published: 17 December 2013
doi: 10.3389/fimmu.2013.00462
Interactions between Nod-like receptors and intestinal
bacteria
Marcel R. de Zoete1and Richard A. Flavell1,2*
1Department of Immunobiology,Yale University School of Medicine, New Haven, CT, USA
2Howard Hughes Medical Institute,Yale University, New Haven, CT, USA
Edited by:
Jorg Hermann Fritz, McGill University,
Canada
Reviewed by:
Axel Lorentz, University of
Hohenheim, Germany
Juan J. Garcia-Vallejo,VU University
Medical Center, Netherlands
*Correspondence:
Richard A. Flavell, Department of
Immunobiology,Yale University
School of Medicine, 300 Cedar
Street, TAC S-569, New Haven, CT
06520, USA
e-mail: richard.flavell@yale.edu
Nucleotide oligomerization domain (Nod)-like Receptors (NLRs) are cytosolic sensors that
mediate the activation of Caspase-1 and the subsequent processing and secretion of the
pro-inflammatory cytokines IL-1βand IL-18, as well as an inflammatory cell death termed
pyroptosis. While a multitude of bacteria have been shown to activate one or more NLRs
under in vitro conditions, the exact impact of NLR activation during the course of coloniza-
tion, both of pathogenic and commensal nature, is less understood. In this review, we will
focus on the role of intestinal NLRs during the various stages of infection with common
gastrointestinal bacterial pathogens, as well as NLR function in controlling and shaping the
microbiota.
Keywords: nod-like receptors, microbiota, inflammasome, intestine, pathogen
INTRODUCTION
The human body lives in symbiosis with trillions of microbial cells,
collectively called the microbiota, with the vast majority of these
microbes being bacteria that inhabit the gastrointestinal tract (1).
This symbiosis begins with colonization of the gastrointestinal
tract at birth and then is sustained throughout life by environ-
mental exposures (2). Occasionally this microbial symbiosis is
challenged by invading bacterial pathogens, which perturb the
microbial ecosystem and cause disease.
Our ability to harbor trillions of bacteria within our intestines
relies on the maintenance of a safe distance between these bacteria
and the single layer of intestinal epithelial cells. Crucial protective
mechanisms have evolved to help ensure host-bacteria mutualism.
A major barrier bacteria encounter in the intestine is formed by
the mucus layer, a dense network of glycoproteins that most bacte-
ria are unable to breach (3). To further aid the barrier function of
the mucus layer, intestinal cells also secrete an array of antimicro-
bial proteins, like antimicrobial peptides, lectins, and lysozymes.
Furthermore, secreted IgA specifically targets bacteria for immune
exclusion (4).
At the cellular level, sensing systems continuously scan for bac-
teria that are able to actively surpass the mucus layer and attach
to and/or invade the epithelium. Two major receptor families
that detect microbes are the Toll-like Receptors (TLRs), which
control the extracellular compartment, and Nod-like Receptors
(NLRs), which sense the presence of intracellular microbes (5).
NLRs are crucial for fighting and resolving infections as many
pathogenic bacteria (and under certain conditions also mem-
bers of the commensal microbiota) attempt to exploit and enter
the cytosol for nutrients and to escape extracellular threats (6).
Here, we provide an overview of the role of NLRs in protection
against intestinal pathogenic bacteria and control of the intestinal
microbiota.
INTESTINAL NLRs
Nod-like receptors generally consist of a ligand-sensing domain in
the form of a Leucine Rich Repeat (LRR) domain, a central ATP
binding domain, and a signaling domain (often in the form of
a CARD or Pyrin domain) and are categorized by their domain
structure. While NLRs are expressed widely in a variety of tissues
in humans and mice, we will focus in this review on those that were
shown to function in the defense against bacteria in the intestine.
While Absent In Melanoma 2 (AIM2) theoretically is not part of
the NLR family, we have included it here for completeness.
NOD1 AND NOD2 (NLRC1, NLRC2)
The pattern recognition receptors NOD1 and NOD2 are amongst
the best-studied NLRs, and their ligands are well defined. Both
NOD1 and NOD2 sense cytosolic bacterial peptidoglycan frag-
ments with high specificity: NOD1 is activated by d-glutamyl-
meso-diaminopimelic acid (DAP) containing peptidoglycan frag-
ments, which are mainly found in Gram-negative bacteria (7),
whereas NOD2 was shown to bind and responds to muramyl
dipeptide (MDP), found in all bacteria (8). Despite the pres-
ence of N-terminal CARD domains, NOD1 and NOD2 are non-
inflammasome forming NLRs and do not seem to directly activate
Caspase-1. Instead, after ligand binding the CARD domain of
NOD1 and NOD2 interacts with the signaling kinase RIP2 (RIPK2,
RICK) that initiates a signaling cascade resulting in NF-κB activa-
tion, as well as the activation of ERK, p38 and mitogen-activated
protein kinases (MAPKs) (9,10). These signaling pathways result
in the expression of a variety of pro-inflammatory cytokines
and chemokines, as well as the production of reactive oxygen
species. While NOD2 expression is more restricted, both NODs
are expressed in macrophages, dendritic cells, Paneth cells, and
intestinal epithelial cells, making them highly suited to sense infec-
tions throughout the intestinal tract (10). In recent years, several
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 1
de Zoete and Flavell NLRs and intestinal bacteria
layers of complexity were added onto the basic mechanism of
NOD1 and NOD2 sensing and signaling. For instance, NOD2
was shown to interact with NLRP1, NLRP3, and NLRP12 (11),
NOD1 and NOD2 were found to play a role in autophagy (12),
and NOD1 senses the modification of small rho GTPases injected
by Salmonella during infection (13).
NLRC4
NLRC4 is an N-terminal CARD domain containing NLR. The elu-
cidation of the NLRC4 crystal structure has revealed that, under
resting conditions, NLRC4 resides in a closed monomeric form,
kept in place by an ADP-dependent autoinhibitory mechanism
involving multiple domains including the LRR (14). Ligand bind-
ing is proposed to induce“opening of the structure, the exchange
of ADP for ATP, and subsequent NLRC4 oligomerization. Phos-
phorylation of a conserved serine residue proximal to the LRR
was shown to be required for NLRC4 inflammasome activation in
macrophages, although the exact role in this process requires fur-
ther investigation (15). NLRC4 responds to attaching or invading
pathogens by sensing their bacterial secretion systems. So far, two
bacterial ligands are well defined: flagellin, which is co-secreted
with virulence factors either through type III or type IV secre-
tion systems (T3SS and T4SS, respectively) (1618), and PrgJ, a
structural component of the type III secretion system that leaks
or is secreted into the host cytosol (19). Within the cytosol, fla-
gellin and PrgJ bind to the adapter proteins NLR family, apoptosis
inhibitory protein (NAIP) 2 and NAIP5, respectively (20,21),
which subsequently bind NLRC4 to initiate its oligomerization
into a ring-like inflammasome that recruits the adapter protein
apoptosis-associated speck-like protein containing a CARD (ASC)
(containing both a Pyrin and CARD domain) and Caspase-1 (22).
This complex then processes the pro-inflammatory cytokines pro-
IL-1βand pro-IL-18, and induces pyroptosis, an inflammatory
form of cell death. Interestingly, unlike mice, humans have only
one NAIP protein,which is unresponsive to both flagellin or basal
rod protein but instead binds the conserved T3SS needle protein
to activate NLRC4 (20).
NLRP3
While NLRP3 is probably the best studied of the NLRs, the mech-
anism of receptor activation remains relatively unclear. NLRP3,
or cryopyrin, was originally shown to play a key role in a collec-
tion of autoinflammatory disorders collectively termed cryopyrin-
associated periodic syndromes, which all share mutations in
NLRP3 that lead to inappropriate IL-1β-mediated inflammatory
responses (23). NLRP3 was subsequently found to “sense” a long
list of ligands or stimuli, including ATP, pore-forming toxins,
particulates like asbestos and silica, bacteria, viral, and fungal
infections (24). Initially, three main theories of the activation of
NLRP3 were proposed: potassium efflux, lysosomal rupture and
subsequent cleavage by released Cathepsin, and ROS production.
Several second generation” unifying NLRP3 ligands were pro-
posed to combine the three, including oxidized mitochondrial
DNA released into the cytosol following mitochondrial damage
(25); thioredoxin-interacting protein (26), calcium mobilization
(27), mitochondrial cardiolipin (28), and changes in cell volume
(29). While most of these NLRP3 ligands were recently shown to
lead to potassium efflux might, suggesting this to be the common
trigger in the end (30), NLRP3 activation remains enigmatic; struc-
tural studies similar to those done for NLRC4 might eventually
elucidate the elusive NLRP3 ligand.
A new chapter for NLRP3 has been opened through the elu-
cidation of the non-canonical inflammasome pathway. Due to
the (re)discovery of the presence of a mutated, non-functional
Caspase-11 in the original Caspase-1-deficient mouse, a role
for Caspase-11 was found in NLRP3-inflammasome activation
by Gram-negative bacteria (31). After prolonged (~17h) stim-
ulation of bone-marrow macrophages with bacteria, Caspase-
11 was shown to be activated, leading to cell death and
NLRP3/ASC/Caspase-1-dependent IL-1βand IL-18 secretion. It
was subsequently shown that the TLR4-TRIF-Type I Interferon
pathway was required to induce high levels of Caspase-11 tran-
scription needed for non-canonical inflammasome activation
(32). However, it recently was shown that intracellular LPS serves
as a ligand able to activate the non-canonical inflammasome path-
way, independently of increasing levels of Caspase-11 caused by
Type I Interferon (33). Three major questions regarding non-
canonical inflammasome activation remain currently unanswered:
what is the receptor that senses intracellular LPS or poten-
tially other ligand (presumably a CARD-containing NLR), how
does this complex feed into the NLRP3 inflammasome (Caspase-
11-dependent pyroptosis resulting in potassium efflux?), and is
Caspase-11 activated by any additional receptors?
NLRP6
NLRP6 falls within the group of NLRs that was initially found
to induce NF-κB and Caspase-1 activation during overexpres-
sion in transfected tissue culture cells (34). In this system, human
NLRP6 was also shown to form punctate structures in the cyto-
plasm, but only in the presence of ASC, suggesting the ability of
NLRP6 to form inflammasomes or inflammasome-like structures.
Unlike in humans,where NLRP6 is not highly or widely expressed,
mice exhibit high NLRP6 expression throughout the intestine,kid-
neys, and liver (35,36), which can be regulated by stress factors
(37). Mechanistically, NLRP6 was shown to be a negative regula-
tor of NF-κB and MAPK in cultured bone-marrow macrophages
from NLRP6-deficient mice (38), which is the opposite of what
was initially observed in overexpression studies. NLRP6 function
as a negative regulator of NF-κB and MAPK might play a role
in the increased intestinal tissue proliferation and inflammation
observed in NLRP6-deficient mice (39,40). Furthermore, NLRP6
was shown to be involved in the production of Caspase-1 depen-
dent IL-18 (36), again suggesting the ability of NLRP6 to form an
inflammasome.
NLRP12
Since its identification, NLRP12 has been assigned a num-
ber of different functions. Like NLRP6, NLRP12 was originally
described to induce NF-κB and Caspase-1 activation when co-
expressed with ASC (41). In contrast, without ASC co-expression,
NLRP12 overexpression reduced non-canonical NF-κB activa-
tion and enhanced the expression of non-classical and classical
MHC class I genes (4244). NLRP12-deficient mice were also
reported to have defective dendritic cell and neutrophil responses
Frontiers in Immunology | Molecular Innate Immunity December 2013 | Volume 4 | Article 462 | 2
de Zoete and Flavell NLRs and intestinal bacteria
to chemokines and subsequent defective dendritic cell migration
to draining lymph nodes (45). In addition, NLRP12-deficiency led
to enhanced colon inflammation and colorectal cancer develop-
ment due to increased (non-canonical) NF-κB and ERK activation
(46,47), similar to what was observed previously for NLRP6.
AIM2
AIM2 was originally identified in humans as an interferon-
inducible, putative tumor suppressor protein (48), but subse-
quently found to sense cytoplasmic double stranded (ds) DNA,
form an inflammasome complex together with ASC and Caspase-
1, and trigger the processing of pro-IL-1βand pro-IL-18 (4952).
Like NLRP receptors, AIM2 contains an N-terminal signaling
Pyrin domain; however, the C-terminal consists of a DNA-binding
HIN200 domain. AIM2 is able to sense the presence of cytosolic
non-sequence-specific dsDNA of both viral and bacterial origin.
Bacterial DNA enters the cytosol mainly by a passive process
following bacterial lysis, for instance during rapid cytosolic bac-
terial replication or after intracellular bacteria-containing vesicles
are compromised, but appears to be always preceded by bacte-
rial invasion into the host cell, making AIM2 a specific sensor
for intracellular bacteria and viruses (5355). In a mechanism
similar to what was observed for NLRC4, the HIN200 domain
functions as a negative regulator of the signaling Pyrin domain.
Non-sequence-specific binding of dsDNA releases this inhibition,
liberating the Pyrin domain to recruit ASC and Caspase-1, and
form an inflammasome surrounding the released bacterial or viral
DNA (56,57).
ENTERIC PATHOGENS AND THEIR INTERACTIONS WITH
NLRs
Foodborne gastrointestinal pathogens are a major cause of bacte-
rial infections in humans (58). Studies on these pathogens, both
in the human host and in various murine models, have provided
great insights into microbial virulence mechanisms as well as the
immunological defense strategies of the host. For NLRs, pathogen-
host interactions have been of great value for the elucidation of the
different functions of this family of innate sensors, both in in vitro
and in vivo model systems. Below (and summarized in Table 1),
we provide an overview of the role of NLRs during infections with
the most commonly studied bacterial enteric pathogens.
SALMONELLA
In humans, infections with the Gram-negative bacterium Salmo-
nella enterica (S.enterica) generally result in one of two distinct
clinical phenotypes. S. enterica serovars Typhi and Paratyphi are
Table 1 | Role of NLRs in intestinal bacterial infections.
Bacteria Model NLR Mechanism of action Reference
S. typhimurium Systemic NLRC4 Flagellin/T3SS-induced pyroptosis, IL-1β, and IL-18 production (7981)
NLRP3 Caspase-1 activation (79)
Systemic,T3SS-1-independent NOD1 Nitric oxide production in dendritic cells (69)
Systemic and colitis in Balb/c NLRC4 IL-1β-mediated neutrophil recruitment (84)
Colitis NLRC4 IL-1βand IL-18 production (80)
NOD1 NOD1-mediated detection of SipA (70)
Colitis, T3SS-1-independent NOD1/2 Innate CD4+T helper type 17 cell responses in the cecum (74,75)
Systemic (intraperitoneal) NLRP6 NLRP6-mediated negative regulation of NF-κB and MAPK activation (38)
C. rodentium Colitis NOD2 NOD2-activation in stromal cells, CCL2/CCR2-dependent recruitment
of inflammatory monocytes, IL-12-mediated bacterial clearance
(92)
NOD1/2 IL-6-dependent IL-17 production in the cecum (75)
NLRC4 IL-1βand IL-18 production (94)
NLRP3 IL-1βand IL-18 production (94)
H. pylori Gastritis NOD1 T4SS-mediated delivery of peptidoglycan, NF-κB-mediated
inflammatory responses
(108)
Unknown IL-18-dependent IL-17 production, T-cell-mediated antibacterial
responses
(110,112)
Unknown IL-1β-dependent impaired bacterial clearance (111,113)
Microbiota Colitis (DSS) NLRP6 IL-18/CCL5 production, increased intestinal epithelial proliferation and
tissue repair
(36,40)
NLRP3 Both increased and decreased susceptibility; microbiota-dependent? (124,125,
127,128)
NOD1/2 Induction of E-cadherin and RegIII-γexpression (120)
Colorectal cancer (DSS-AOM) NLRP6 IL-18/CCL5/IL-6 mediated increased intestinal epithelial proliferation
and tissue repair
(39,40,116)
NLRP3 Caspase-1 activation (126)
Non-alcoholic fatty liver disease NLRP3/6 IL-18-mediated control of microbiota (117)
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 3
de Zoete and Flavell NLRs and intestinal bacteria
the causative agents of Typhoid fever, a life-threatening disease
characterized by systemic spread of ingested bacteria, high fever,
and intestinal bleeding that results in 200,000–600,000 deaths
worldwide each year (59). The more common non-typhoidal S.
enterica serovars, like S.typhimurium and S.enteritidis, cause a self-
limiting gastroenteritis characterized by (bloody) diarrhea, fever,
abdominal cramps, and vomiting that usually lasts 4–7days and
affects over 90 million people worldwide each year (60).
Two different disease models in mice represent the two clini-
cal manifestations of Salmonella infections. The classical murine
model of systemic S. typhimurium infection induces a disease simi-
lar to Typhoid fever. In this model,orally administered salmonellae
reach the distal ileum within hours of ingestion and, aided by
flagella-mediated motility and chemotaxis (61), cross the mucus
layer toward the epithelium. Here, the bacteria target the follicular-
associated epithelium overlying the Peyer’s Patches, with a strong
preference for the M-cells, as a main port of entry. To gain access
into the host cell, Salmonella employs the first of two type III secre-
tion systems (T3SS-1), which is only expressed during this initial
phase of infection, and injects an array of effector proteins into the
host’s cytosol that induce cytoskeletal rearrangements leading to
bacterial invasion (62). Other effector proteins and the activation
of pattern recognition receptors initiate inflammatory responses
that attract neutrophils, monocytes, and macrophages. Salmo-
nella replicates within the Peyer’s Patches and disseminates to
mesenteric lymph nodes (MLN), liver, and spleen within infected
monocytes and dendritic cells. In a T3SS-1-independent manner,
a small proportion of the bacteria is also taken up passively from
the lumen by CD11c+CX3CR1+dendritic cells and transported
directly to the MLN, bypassing the Peyer’s Patches (63). To survive
and replicate in the host’s cells, Salmonella resides within a vac-
uole, the Salmonella-containing Vacuole (SCV), whose integrity
is maintained by the effector proteins secreted into the cytosol
through a second T3SS (T3SS-2). During the later stages of dis-
ease, disseminated bacteria in the liver and spleen are found mostly
within macrophages, in which they rapidly replicate. Continuous
cycles of bacterial replication and dissemination eventually lead to
bacteremia from which mice succumb after a week.
In the murine colitis model, infection with Salmonella is pre-
ceded by a single dose of streptomycin, which is believed to briefly
reduce “colonization resistance” provided by the microbiota that
occupies the more distal intestinal tract and strongly competes
for nutrients. In this short window, Salmonella is able to gain a
foothold in the cecum and colon, and replicates to high numbers
within hours of infection, limiting the need for systemic spread
(64). Colonization is accompanied by mucosal penetration of bac-
teria and the development of colitis, similar to the pathology seen
in human infection with non-typhoidal serovars. For efficient col-
onization salmonellae still require T3SSs, which mediate invasion
of enterocytes and induction of (local) inflammation. A reason
for this was presented in an elegant study by Winter et al. which
revealed that T3SS-induced inflammatory responses are actively
exploited by Salmonella through the ability to utilize tetrathionate,
formed in the intestine under inflammatory conditions, to success-
fully compete with the microbiota (65). In the colitis mouse model,
Salmonella also exploits a T3SS-1-independent,“passive” route for
uptake through CD11c+CX3CR1+dendritic cells. Similar to what
is seen in the systemic model, these two invasion pathways act in
concert, although T3SS-1-mediated invasion seems much more
dominant. While bacteria are able to grow extensively in the dis-
tal intestine in the colitis model, substantial numbers of bacteria
still continue to disseminate to liver and spleen,and infected mice
usually die after 5–6 days.
As several phases of infection largely rely on cellular invasion,
NLRs appear to be the ideal sensing mechanism for Salmonella.
In vitro,Salmonella is sensed by NOD2 in cultured intestinal
epithelial cells, which enables the control of intracellular bacteria
though the induction of antimicrobial responses and autophagy
(6668), and by NOD1 in bone-marrow-derived dendritic cells,
resulting in nitric oxide production (69). Interestingly, while Sal-
monella peptidoglycan may be involved in the activation of NOD1
and NOD2, it was recently shown that the modification of small
Rho GTPases by the T3SS-1 effector SopE, which enables bacter-
ial invasion, is a danger signal sensed by NOD1 (13). Presumably
through a similar mechanism, another T3SS-1 effector SipA acti-
vated NOD1/NOD2-dependent NF-κB responses both in vitro
and in vivo (70). In addition to NOD1 and NOD2, Salmonella
is efficiently sensed by NLRC4, both via flagellin and the basal
rod protein PrgJ, which leads to rapid pyroptosis and secretion
of IL-1βand IL-18 by cultured macrophages, dendritic cells and
B-cells (16,17,19,71,72). These responses are solely dependent
on T3SS-1, which is expressed only at the early logarithmic phase
of bacterial cultures and is believed to represent the early phase in
infection when Salmonella needs to invade host cells. The T3SS-2,
expressed only at the late logarithmic phase, does not contribute to
NLRC4 activation as flagellin expression is now repressed and the
T3SS-2 apparatus is not recognized (19). Finally, Salmonella has
been shown to activate the non-canonical NLRP3 inflammasome
through Caspase-11 in macrophages (73).
In vivo, the role of NLRs during Salmonella infection has
been rather difficult to define, mainly because of differences in
experimental models (systemic versus colitis murine models),
variations in growth phase of Salmonella at time of infection
(T3SS-1-expressing versus T3SS-2-expressing conditions), differ-
ent mouse intestinal microbiotas, and because of the redundancy
in innate receptors. These issues are clearly demonstrated when
studying NOD1 and NOD2. Both of these NLRs were shown to
be dispensable during systemic infection (69). However, under
T3SS-1-independent conditions, during which the bacteria “pas-
sively” cross the epithelial barrier through uptake and trans-
port by dendritic cells, NOD1 deficiency led to higher bacter-
ial loads and mortality. The authors show that NOD1-deficient
CD11b+CD11c+dendritic cells contain higher numbers of Salmo-
nella, likely because of a diminished NOD1-mediated nitric oxide
response. Similarly, during Salmonella colitis, NOD1/NOD2 dou-
ble knockouts or RIP2 deficient mice exhibited reduced inflamma-
tion accompanied by increased mucosal colonization and reduced
early IL-17 responses of innate CD4+T helper type 17 cells in
the lamina propria of the cecum, but again only when Salmonella
was grown under T3SS-2-expressing conditions (74,75); when
T3SS-1 was expressed at the time of oral infection in this model,
no differences as compared to wild-type mice were observed (76).
These data suggest that only one of the two major entry pathways
exploited by Salmonella is controlled by NOD1/NOD2 signaling.
Frontiers in Immunology | Molecular Innate Immunity December 2013 | Volume 4 | Article 462 | 4
de Zoete and Flavell NLRs and intestinal bacteria
During “normal” infections, however, the T3SS-1-mediated inva-
sion seems to outweigh the alternative invasion route, leaving
NOD1 and NOD2 to play a non-significant role. Interestingly,
NOD1/NOD2-activation by the T3SS-1 effector SipA was shown
to lead to a higher gut inflammation score in the colitis model
as compared to mice lacking the receptors (70). Combined, the
above-mentioned studies suggest that NOD1 and NOD2 medi-
ated detection of Salmonella plays a specific but minor role during
salmonellosis. As a further complication, differences in micro-
biota from wild-type and knockout mice dramatically impact
Salmonella susceptibility, as demonstrated by Kaiser et al.; when
”microbiota-matched” littermate controls were used (instead of
independently bred or purchased wild-type mice) to test the role
of RIP2 during Salmonella colitis, the initially observed difference
in Salmonella-induced pathology was completely lost (64).
Caspase-1 has been shown in several publications to pro-
vide moderate protection against Salmonella infection. Without
Caspase-1, mice succumb to bacteremia sooner and have higher
bacterial loads in the MLN, liver and spleen in the typhoid model
of infection, and more colitis accompanied by increased bacte-
rial mucosal infiltration (77,78). While NLRC4 appeared to be
the main upstream candidate for caspase-1 activation, NLRC4-
deficient mice showed only minor or no defects in bacterial control
during infections (77,79,80). Two critical findings explained this
“lack” of NLRC4 function in vivo; first, Salmonella actively evades
recognition by NLRC4 by downregulating both T3SS-1 and fla-
gellin as soon as the bacteria have invaded the host cell,and T3SS-2
is not recognized by NLRC4 (19,81). The in vivo consequence
of NLRC4-evasion, and thereby the role of NLRC4 in protection
against invading pathogens, was elegantly shown by Miao et al.:
when Salmonella was forced to continuously express flagellin,100
times less bacteria were found in the spleen after 48 h during sys-
temic infection. The increased control of bacterial spreading was
attributed to NLRC4-mediated macrophage pyroptosis at periph-
eral sites, which resulted in release of the intracellular bacteria and
subsequent clearance by infiltrating neutrophils (81).
NLR redundancy is a second reason why NLRC4-deficient
mice do not phenocopy Caspase-1-deficient mice. Late logarith-
mic, non-T3SS-1 expressing salmonellae are able to activate the
non-canonical Caspase-11/NLRP3 inflammasome (73). Similar
to NLRC4, deficiency in only NLRP3 does not lead to differences
in Salmonella infection. However, deletion of both NLRC4 and
NLRP3 recapitulates the Caspase-1 phenotype completely, con-
firming a role for both NLRC4 and NLRP3 during Salmonella
infection (79). This also demonstrates that, as was predicted by
in vitro studies, NLRC4-evasion is not perfect. While pyroptosis
has a clear impact on infection, the cytokines IL-1βand IL-18
appear to play minor roles in the control of the bacteria, since
IL-1βand IL-18-deficent mice show little delay in bacteremia at
72 h (81,82).
With the realization that the Caspase-1 KO was in fact a
Caspase-1/Caspase-11 double knockout, and the elucidation of
the role of Caspase-11 in non-canonical inflammasome activa-
tion, Caspase-11-deficient mice were predicted to result in more
bacterial spread due to diminished control of infection. However,
Caspase-11-deficient mice were indistinguishable from WT mice
during Salmonella infection (79). Surprisingly, Caspase-1 single
deficient mice had even higher numbers of bacteria in liver and
spleen than the Caspase-1/Caspase-11-deficient mice, suggesting
a protective role for Caspase-11 deficiency, but only in the con-
text of Caspase-1-deficiency. A potential explanation for this may
be that, while rapid Caspase-1-mediated pyroptosis clears bacte-
ria, Caspase-11-mediated pyroptosis at later time points is actively
used by Salmonella to escape the “full”macrophage after extensive
replication. Indeed, Caspase-11 senses bacteria escaping from or
leaking out of vacuoles into the cytoplasm (83). In the absence of
Caspase-1, NLRC4 “evasion by Salmonella is complete, resulting
in uncontrolled replication until Caspase-11 is utilized to break out
of the macrophage and invade new host cells. Why non-canonical
Caspase-11-mediated pyroptosis, like NLRC4-activation accom-
panied by IL-1βand IL-18 secretion that induces local inflam-
mation and attracts neutrophils, is less potent than Caspase-1-
mediated pyroptosis in controlling Salmonella infection remains
thus far unclear.
Unlike in C57BL/6 mice,in Balb/c mice NLRC4 appears to have
a more prominent function in controlling Salmonella infection.
In these mice, NLRC4-deficiency leads to more systemic bacter-
ial dissemination and mortality, while less inflammation-induced
pathology in the cecum was observed (84). It was subsequently
shown that Salmonella specifically activates intestinal phagocytes
that respond by producing IL-1βwhich triggered the upregulation
of endothelial adhesion molecules. The basis of the interesting dif-
ferential function of NLRC4 between C57BL/6 and Balb/c remains
to be determined.
ATTACHING AND EFFACING ENTERIC PATHOGENS: CITROBACTER,
EPEC AND EHEC
Citrobacter rodentium (C.rodentium), EnteropathogenicEscherichia
coli (EPEC), and Enterohemorrhagic Escherichia coli (EHEC) are
Gram-negative extracellular enteric pathogens that share a sim-
ilar virulence strategy, termed attaching and effacing (A/E) (85,
86). EPEC and EHEC are human pathogens; EPEC is a major
cause of diarrhea in young children, generally without major com-
plications, while EHEC infections can vary greatly in severity,
ranging from mild gastroenteritis to severe hemorrhagic colitis
and hemolytic uremic syndrome. C.rodentium is a natural mouse
pathogen resulting in self-limiting enteritis. While very little is
known about activation of NLRs by EPEC and EHEC in humans,
several studies have elucidated the role of such responses during
infection of their murine counterpart, C.rodentium.
Within a couple of hours after oral infection of mice, C.roden-
tium reaches its initial site of infection, the lymphoid tissue in the
cecum termed the cecal patch, where it reaches high density over
the following 3 days (87). The cecal patch is structurally similar
to the Peyer’s Patch and, because of their nature as antigen sam-
pling hotspots with decreased mucus layer thickness and absence
of microvilli, provide “easy access/entrance” for several intestinal
pathogens (including Salmonella, as described above). From day
3 to 4, C. rodentium starts to spread throughout the distal colon
(87). Mouse-adapted strains of C.rodentium largely skip the cecal
patch phase and colonize the colon readily, suggesting that colo-
nization of the cecal patch also serves as an adaptation phase to the
mouse intestinal environment (88). Depending on the strain of C.
rodentium used, bacterial numbers peak between day 5 and 14 with
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 5
de Zoete and Flavell NLRs and intestinal bacteria
limited systemic spread to the MLN, liver, and spleen. The colo-
nization then slowly diminishes until bacterial clearance from the
cecum and subsequently the colon after 3–4weeks post infection.
Upon reaching the cell surface of the cecum and colon, C.
rodentium employs a T3SS which injects an array of virulence
factors into the host’s cytosol that result in the attachment of the
bacteria to the enterocytes and the accompanying local destruc-
tion of the brush border microvilli of the epithelium forming
pedestal-like structures termed A/E lesions (86). Two of these vir-
ulence proteins are central for this virulence strategy: the adhesin
Intimin expressed on the bacterial surface and the T3SS-injected
Translocated Intimin Receptor (TIR), which provides a docking
ligand for Intimin on the host epithelial surface (89,90). The
attachment of C. rodentium, in combination with the secretion of
many additional virulence proteins, leads to colonic hyperplasia,
observed readily during the peak of infection as larger intestinal
crypt length and increased colon weight.
Several reports have shown that NOD1 and NOD2 are able
to sense C. rodentium both in vitro and in vivo (75,91,92). In
the absence of NOD2, C. rodentium reaches a higher intestinal
abundance as compared to wild-type mice. At the early stages of
infection, NOD2 signaling was shown to activate the CCL2/CCR2
axis that resulted in the recruitment of inflammatory monocytes
to the site of infection, which initiated IL-12-mediated bacterial
clearance. Interestingly, NOD2-activation took place in intestinal
stromal cells and not immune cells. NOD2-deficiency led to lower
inflammation at the early stages of infection, but more severe coli-
tis later,as a result of reduced clearance and higher bacterial abun-
dance in the intestine (92). In a different study, NOD1 and NOD2
had redundant roles in the protection against C. rodentium infec-
tion and mediated IL-6-dependent IL-17 production in the cecum
at early time point (1–4 days after infection). The observed effect
on infection was similar; lower initial inflammatory responses but
increased levels of bacterial dissemination to the spleen in the sec-
ond week of infection (75). NOD1/NOD2 signaling was shown to
occur mostly in the radio-resistant compartment, but a role for
stromal cells was not further investigated. Although it is expected
that peptidoglycan is the major C. rodentium-derived ligand of
NOD1 and NOD2, T3SS-injected effector proteins may play a role
too, as was shown previously for the Salmonella effector protein
SopE (13). Indeed EspT, which targets small GTPases to induce
membrane rearrangement in a similar way as SopE, was shown
to induce NF-κB, ERK1/2 and JNK activation, common signaling
pathways activated after NOD1/2 signaling (93). Future studies
will determine to what extent effector-mediated NLR activation
contributes to colonization and bacterial clearance.
Caspase-1/Caspase-11-deficient mice were found to be hyper-
susceptible for C. rodentium infection, as determined by increased
intestinal bacterial loads, colitis,and hyp erplasia (94). Both NLRP3
and NLRC4-deficient mice,as well as mice lacking IL-1βand IL-18,
showed similar phenotypes, suggesting an important role for the
NLRP3/NLRC4/IL-1β/IL-18 axis in the control of C. rodentium. In
a different study, a similar but stronger phenotype was observed in
IL-1R-deficient mice, which mostly succumb to infection within
2 weeks (95). In contrast to what was seen in IL-18-deficient mice,
neutralizing this cytokine with antibodies had limited to no effect,
implicating IL-1βor IL-1αas the critical cytokines that mediated
protection against C. rodentium. IL-1R signaling during C. roden-
tium infection led to IFN-γand IL-6 production in the colon,
which mediated epithelial repair and maintained barrier function.
While bacterial loads remained the same, more bacteria dissemi-
nated to the liver in the absence of these cytokines. Like Salmonella
and most other Gram-negative bacteria, C. rodentium is able to
activate the non-canonical Caspase-11/NLRP3 inflammasome in
cultured bone-marrow macrophages, which occurred in a T3SS-
independent (31,94). The activation of the Caspase-11/NLRP3
non-canonical inflammasome during infection was evident when
examining Caspase-11- and TRIF-deficient mice, which were both
more susceptible for C. rodentium infection (96). Interestingly,
while NLRC4 seems to be activated by C. rodentium in vivo, bone-
marrow macrophages did not sense the T3SS of C. rodentium
during in vitro studies. Whether this is due to tightly regulated
T3SS expression or host cell tropism/specificity remains to be
determined.
HELICOBACTER PYLORI
The Gram-negative bacterium Helicobacter pylori (H.pylori ) col-
onizes the gastric mucosa of ~50% of the world’s population,
although substantial variation exists between countries (97). The
majority of people infected by H.pylori do not show any symp-
toms, despite local chronic inflammatory responses induced by
the bacterium. However, in a subset of patients, this inflammatory
response drives the formation of gastric or duodenal ulcers that
can lead to the development of mucosa-associated lymphoid tissue
lymphomas and gastric adenocarcinomas (98100).
In order to survive in the challenging gastric niche and enable
persistent colonization, H.pylori is highly optimized to evade
host antimicrobial strategies. For instance, after ingestion H.pylori
secretes urease, which increases the gastric pH and reduces mucus
viscosity, enabling rapid penetration of the gastric mucus layer and
colonization in close proximity to the pH neutral epithelial cells
(99,101). Also, H. pylori expresses a modified LPS and flagellin to
evade the recognition of TLR4 and TLR5, and has adopted several
mechanisms to counteract the effects of host-produced reactive
oxygen species (102,103). Finally, the secreted pore-forming toxin
VacA induces epithelial cell apoptosis and inhibits T-cell activation
and proliferation (104). In contrast to immune evasion, a subset
of H. pylori strains also actively induces inflammatory responses
by means of the T4SS-mediated delivery of the effector protein
CagA. CagA modifies multiple intracellular signaling pathways of
host cells and is linked to the development of gastric cancer.
While immune evasion appears to be an important part of
the H. pylori life cycle, genetic association studies revealed that
mutations in NOD1, NOD2, and IL-1βmay be associated with
increased risk for the development of gastric cancer, suggesting
that NLRs play a role in controlling H. pylori during human infec-
tion (105107). In addition, several NLR family members have
been shown to sense the bacterium and impact on infection or
colonization, both in in vitro cell culture and in vivo murine mod-
els. In a manner analogous to the “leakage” of flagellin through
the T3SS in Salmonella, peptidoglycan fragments were found to
enter the host cytosol through the T4SS, where they were subse-
quently sensed by NOD1 and initiated NF-κB-mediated inflam-
matory responses (108). NOD1 activation was also observed by
Frontiers in Immunology | Molecular Innate Immunity December 2013 | Volume 4 | Article 462 | 6
de Zoete and Flavell NLRs and intestinal bacteria
peptidoglycan present in secreted bacterial outer membrane vesi-
cles that were taken up by host cells (109). H. pylori was shown to
induce the secretion of IL-1βand IL-18 both in vitro and in vivo
(110,111). As compared to wild-type mice, Caspase-1/Caspase-
11-deficient mice showed decreased numbers of Helicobacter in
the stomach, higher expression of IL-17, and aggravated gastric
immunopathology, which was phenocopied by IL-18 and IL-18R,
but not IL-1R deficient mice. Loss of IL-18 signaling in den-
dritic cells was subsequently shown to result in reduced levels
of regulatory T-cells and stronger T-cell-mediated antibacterial
responses (110,112). In contrast, different groups reported that
Caspase-1/Caspase-11, ASC, IL-1β, and IL-1R-deficient mice were
impaired in the clearance of H. pylori from the stomach, dis-
played decreased gastritis and lower levels of IL-1βand IL-18
(111,113). While the cause of the discrepancies between these
different reports is currently unknown, it appears that H. pylori
strives for the ideal level of inflammasome activation: enough IL-
18 and IL-1βto induce regulatory T-cells and decrease gastric acid
production, respectively (114), but not so much IL-1βas to lead
to T-cell mediated clearance. The nature of the inflammasome
NLR that is activated by Helicobacter remains unclear. While in
cultured dendritic cells NLRP3 was crucial for IL-1βsecretion in a
T4SS-dependent/CagA-VacA-independent manner, this NLR did
not play a role during murine infection.
THE INTESTINAL MICROBIOTA AND NLR-MEDIATED
DISORDERS
The intestinal microbiota is predicted to consist of ~100 different
bacterial species per person, and displays great variability between
individuals (115). Alterations in the composition of the microbiota
have been shown to dramatically impact disease susceptibility and
progression. Therefore, controlling and (re)shaping the “healthy”
microbiota is a crucial function of the intestinal immune system.
The role of NLRs in this process is only beginning to be unraveled.
Lack of appropriate immunological control may switch a
healthy microbiota into a pathogenic one, as exemplified by mice
lacking NLRP6. NLRP6-deficient mice show increased levels of
intestinal inflammation during DSS-induced colitis and develop
more severe colorectal cancer in a model of colitis-dependent
tumorigenesis (36,39,40,116). A potential mechanism was pro-
vided by the finding that NLRP6 acts as a negative regulator of
NF-κB and MAPK activation, and reduces the levels of cytokines
and chemokines during infections with intestinal pathogens or
epithelial barrier breach as observed during experimental mod-
els of colitis (38). More severe and prolonged inflammation in
NLRP6-deficient mice results in increased levels of intestinal
epithelial proliferation and increased tissue repair, which was
shown to be CCL5 and IL-6 dependent (36,39,40,116). The
actions of NLRP6 do not seem limited to infectious or damag-
ing episodes, as NLRP6-deficient mice already display continuous
low level inflammation in the steady state, suggesting an interac-
tion with the microbiota (36). 16S rRNA sequencing analysis of the
microbiota revealed that NLRP6-deficient mice harbor a dysbiotic,
colitogenic microbiota that showed a high relative abundance of
Prevotellaceae species, that was transmissible to wild-type control
mice. Similarly,lack of NLRP6-mediated control of the microbiota
induced non-alcoholic fatty liver disease and obesity in mice
and increased colorectal cancer, all of which were transmissible
through microbiota transfer to wild-type mice (116,117).
NLRP12 and NLRP6 may play similar roles in the control
of intestinal homeostasis. Like NLRP6, NLRP12-deficiency leads
to uncontrolled NF-κB signaling and subsequent inflammation
and intestinal cell proliferation. Although extensive analysis of
the composition of the intestinal microbiota of NLRP12-deficient
mice has not been reported, the lack of this NLR might have major
effects on the microbiota, either directly through sensing microbial
products, or indirectly through the induction of an inflammatory
environment via NF-κB dysregulation.
While systemic peptidoglycan from the intestinal microbiota
was shown to boost the development of the intestinal immune sys-
tem and prime immune responses via NOD1 in the bone-marrow
in mice (115,118), NOD1, like NOD2, does not dramatically
influence the composition of the microbiota under homeosta-
tic conditions (119). However, during DSS-induced colitis, the
murine model of inflammatory bowel disease (IBD) that is driven
by the microbiota, NOD1/NOD2-deficiency led to greater suscep-
tibility to colitis (120). Similarly, mutations in NOD2 and NOD1
in humans are associated with susceptibility to Crohn’s disease
and IBD, respectively (121123). The role of NLRP3 in control-
ling the microbiota has been rather controversial. Initially, NLRP3
was reported to have a key role in protecting intestinal home-
ostasis, as NLRP3-deficient mice were shown to have an altered
microbiota and displayed increased susceptibility to DSS-colitis
(124,125) and tumorigenesis (126). However, NLRP3-deficiency
led to resistance to DSS-colitis in a different study (127). As the
DSS-colitis model is highly dependent on the microbiota, differ-
ential compositions of the microbiota may explain the varying
outcomes in these studies. Indeed, co-housing NLRP3-deficient
mice with wild-type mice, which equalized the intestinal micro-
biota, also equalized the inflammatory responses and disease in
both mice (128). In humans, the role of NLRP3 in Crohn’s is
equally confusing; polymorphisms associated with NLRP3 were
shown to contribute to susceptibility to Crohn’s disease (129,130),
but did not replicate in a separate study (131). More detailed
investigation of the interactions between specific members of the
microbiota and NLRs may provide deeper insights in the func-
tion of NLRs in controlling and shaping the microbiota in health
and disease.
CONCLUDING REMARKS
Nod-like receptors are crucial components of the intestinal innate
immune system, controlling both the commensal microbiota as
well as enteropathogenic bacterial infections. While a growing
body of scientific evidence now provides clear insight into the
role of NLRs in controlling intestinal bacteria, several conflicting
reports highlight the importance of precisely controlling experi-
mental conditions like bacterial growth phase and the intestinal
microbiota between wild-type and NLR-deficient mice. Several
key questions still remain unanswered, suck as the nature of the
ligands for NLRP6 and NLRP12, the interplay between NLRs and
adaptive immunity in the intestine, the potential role for other
NLRs like NLRP7 (which senses bacterial lipopeptides in human
cells), NLRP10 (which controls adaptive immune responses), and
NLRC3 (which down-regulates NF-κB), and the role of NLRs in
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 7
de Zoete and Flavell NLRs and intestinal bacteria
human diseases. Future research will undoubtedly shed more light
on these interesting new subjects.
ACKNOWLEDGMENTS
We wish to thank members of the Flavell lab for critically reading
the manuscript. This work was supported by the Howard Hughes
Medical Institute (Richard A. Flavell), a Department of Defense
Grant (W81XWH-11-1-0745) (Richard A. Flavell) and a Rubi-
con Fellowship from the Netherlands Organization of Scientific
Research (NWO) (Marcel R. de Zoete).
REFERENCES
1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial
community variation in human body habitats across space and time. Science
(2009) 326(5960):1694–7. doi:10.1126/science.1177486
2. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diver-
sity, stability and resilience of the human gut microbiota. Nature (2012)
489(7415):220–30. doi:10.1038/nature11550
3. Hansson GC. Role of mucus layers in gut infection and inflammation. Curr
Opin Microbiol (2012) 15(1):57–62. doi:10.1016/j.mib.2011.11.002
4. Ashida H, Ogawa M, Kim M, Mimuro H, Sasakawa C. Bacteria and host
interactions in the gut epithelial barrier. Nat Chem Biol (2011) 8(1):36–45.
doi:10.1038/nchembio.741
5. Palm NW, Medzhitov R. Pattern recognition receptors and control of adap-
tive immunity. Immunol Rev (2009) 227(1):221–33. doi:10.1111/j.1600-065X.
2008.00731.x
6. Ray K, Marteyn B, Sansonetti PJ, Tang CM. Life on the inside: the intra-
cellular lifestyle of cytosolic bacteria. Nat Rev Microbiol (2009) 7(5):333–40.
doi:10.1038/nrmicro2112
7. Girardin SE, Boneca IG, Carneiro LA, Antignac A, Jéhanno M, Viala J, et al.
Nod1 detects a unique muropeptide from gram-negative bacterial peptidogly-
can. Science (2003) 300(5625):1584–7. doi:10.1126/science.1084677
8. Girardin SE, Boneca IG, Viala J, Chamaillard M, Labigne A, Thomas G, et al.
Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP)
detection. J Biol Chem (2003) 278(11):8869–72. doi:10.1074/jbc.C200651200
9. Kobayashi K, Inohara N, Hernandez LD, Galán JE, Núñez G, Janeway CA,
et al. RICK/Rip2/CARDIAK mediates signalling for receptors of the innate
and adaptive immune systems. Nature (2002) 416(6877):194–9. doi:10.1038/
416194a
10. Moreira LO, Zamboni DS. NOD1 and NOD2 signaling in infection and inflam-
mation. Front Immunol (2012) 3:328. doi:10.3389/fimmu.2012.00328
11. Wagner RN, Proell M, Kufer TA, Schwarzenbacher R. Evaluation of Nod-like
receptor (NLR) effector domain interactions. PLoS One (2009) 4(4):e4931.
doi:10.1371/journal.pone.0004931
12. Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim Y-G, Magalhães JG,
et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma
membrane at the site of bacterial entry. Nat Immunol (2009) 11(1):55–62.
doi:10.1038/ni.1823
13. Keestra AM, Winter MG, Auburger JJ, Fräßle SP, Xavier MN, Winter SE, et al.
Manipulation of small Rho GTPases is a pathogen-induced process detected
by NOD1. Nature (2013) 496(7444):233–7. doi:10.1038/nature12025
14. Hu Z, Yan C, Liu P, Huang Z, Ma R, Zhang C, et al. Crystal structure of
NLRC4 reveals its autoinhibitionme chanism.Science (2013) 341(6142):172–5.
doi:10.1126/science.1236381
15. QuY, Misaghi S, Izrael-TomasevicA, Newton K, Gilmour LL, Lamkanfi M, et al.
Phosphorylation of NLRC4 is critical for inflammasome activation. Nature
(2012) 490(7421):539–42. doi:10.1038/nature11429
16. Franchi L, Amer A, Body-Malapel M, Kanneganti T-D, Özören N, Jagirdar R,
et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin
1βin Salmonella-infected macrophages. Nat Immunol (2006) 7(6):576–82.
doi:10.1038/ni1346
17. Miao EA, Alpuche-Aranda CM, Dors M, Clark AE, Bader MW, Miller SI, et al.
Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1βvia
Ipaf. Nat Immunol (2006) 7(6):569–75. doi:10.1038/ni1344
18. Molofsky AB, Byrne BG, Whitfield NN, Madigan CA, Fuse ET, Tateda K, et al.
Cytosolic recognition of flagellin by mouse macrophages restricts Legionella
pneumophila infection. J Exp Med (2006) 203(4):1093–104. doi:10.1084/jem.
20051659
19. Miao EA, Mao DP, Yudkovsky N, Bonneau R, Lorang CG, Warren SE, et al.
Innate immune detection of the type III secretion apparatus through the
NLRC4 inflammasome. Proc Natl Acad Sci U S A (2010) 107(7):3076–80.
doi:10.1073/pnas.0913087107
20. Zhao Y,Yang J, Shi J, Gong Y-N, Lu Q, Xu H, et al. The NLRC4 inflammasome
receptors for bacterial flagellin and type III secretion apparatus. Nature (2011)
477(7366):596–600. doi:10.1038/nature10510
21. Kofoed EM, Vance RE. Innate immune recognition of bacterial ligands by
NAIPs determines inflammasome specificity. Nature (2011) 477(7366):592–5.
doi:10.1038/nature10394
22. Halff EF,D iebolder CA,Versteeg M, Schouten A, Brondijk THC, Huizinga EG.
Formation and structure of a NAIP5-NLRC4 inflammasome induced by direct
interactions with conserved N-and C-terminal regions of flagellin. J Biol Chem
(2012) 287(46):38460–72. doi:10.1074/jbc.M112.393512
23. Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Muta-
tion of a new gene encoding a putative pyrin-like protein causes familial cold
autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet (2001)
29(3):301–5. doi:10.1038/ng756
24. Franchi L, Muñoz-Planillo R, Núñez G. Sensing and reacting to microbes
through the inflammasomes. Nat Immunol (2012) 13(4):325–32. doi:10.1038/
ni.2231
25. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized
mitochondrial DNA activates the NLRP3 inflammasome during apoptosis.
Immunity (2012) 36(3):401–14. doi:10.1016/j.immuni.2012.01.009
26. Zhou R, TardivelA, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting pro-
tein links oxidative stress to inflammasome activation. Nat Immunol (2009)
11(2):136–40. doi:10.1038/ni.1831
27. Murakami T, Ockinger J, Yu J, Byles V, McColl A, Hofer AM, et al. Critical role
for calcium mobilization in activation of the NLRP3 inflammasome. Proc Natl
Acad Sci U S A (2012) 109(28):11282–7. doi:10.1073/pnas.1117765109
28. Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, et al. Mitochondrial
cardiolipin is required for Nlrp3 inflammasome activation. Immunity (2013)
39(2):311–23. doi:10.1016/j.immuni.2013.08.001
29. Compan V, Baroja-Mazo A, López-Castejón G, Gomez AI, Martínez CM,
Angosto D,et al.Cell volume regulation modulates NLRP3 inflammasome acti-
vation. Immunity (2012) 37(3):487–500. doi:10.1016/j.immuni.2012.06.013
30. Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM,
Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome
activation by bacterial toxins and particulate matter. Immunity (2013)
38(6):1142–53. doi:10.1016/j.immuni.2013.05.016
31. Kayagaki N, Warming S, Lamkanfi M, Walle LV, Louie S, Dong J, et al.
Non-canonical inflammasome activation targets caspase-11. Nature (2011)
479(7371):117–21. doi:10.1038/nature10558
32. Rathinam VA, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM,
et al. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation
by gram-negative bacteria. Cell (2012) 150(3):606–19. doi:10.1016/j.cell.2012.
07.007
33. Kayagaki N, Wong MT, Stowe IB, Ramani SR, Gonzalez LC, Akashi-
Takamura S, et al. Noncanonical inflammasome activation by intracellular LPS
independent of TLR4. Science (2013) 341(6151):1246–9. doi:10.1126/science.
1240248
34. Grenier JM, Wang L, Manji GA, Huang W-J, Al-Garawi A, Kelly R, et al.
Functional screening of five PYPAF family members identifies PYPAF5 as
a novel regulator of NF-κB and caspase-1. FEBS Lett (2002) 530(1):73–8.
doi:10.1016/S0014-5793(02)03416-6
35. Lech M, Avila-Ferrufino A, Skuginna V, Susanti HE, Anders H-J. Quantita-
tive expression of RIG-like helicase, NOD-like receptor and inflammasome-
related mRNAs in humans and mice. Int Immunol (2010) 22(9):717–28.
doi:10.1093/intimm/dxq058
36. Elinav E, Strowig T,Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, et al. NLRP6
inflammasome regulates colonic microbial ecology and risk for colitis. Cell
(2011) 145(5):745–57. doi:10.1016/j.cell.2011.04.022
37. Sun Y, Zhang M, Chen CC, Gillilland M, III, Sun X, El-Zaatari M, et al.
Stress-induced corticotropin-releasing hormone-mediated NLRP6 inflamma-
some inhibition and transmissible enteritis in mice. Gastroenterology (2013).
144(7):1478-87.e8. doi:10.1053/j.gastro.2013.02.038
Frontiers in Immunology | Molecular Innate Immunity December 2013 | Volume 4 | Article 462 | 8
de Zoete and Flavell NLRs and intestinal bacteria
38. Anand PK, Malireddi RS, Lukens JR, Vogel P, Bertin J, Lamkanfi M, et al.
NLRP6 negatively regulates innate immunity and host defence against bacterial
pathogens. Nature (2012) 488(7411):389–93. doi:10.1038/nature11250
39. Normand S, Delanoye-Crespin A, Bressenot A, Huot L, Grandjean T, Peyrin-
Biroulet L, et al. Nod-like receptor pyrin domain-containing protein 6
(NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon
injury. Proc Natl Acad Sci U S A (2011) 108(23):9601–6. doi:10.1073/pnas.
1100981108
40. Chen GY, Liu M, Wang F, Bertin J, Núñez G. A functional role for
Nlrp6 in intestinal inflammation and tumorigenesis. J Immunol (2011)
186(12):7187–94. doi:10.4049/jimmunol.1100412
41. Wang L, Manji GA, Grenier JM, Al-Garawi A, Merriam S, Lora JM, et al.
PYPAF7,a novel PYRIN-containing Apaf1-like proteinthat regulates activation
of NF-κB and caspase-1-dependent cytokine processing. J Biol Chem (2002)
277(33):29874–80. doi:10.1074/jbc.M203915200
42. Williams KL, Taxman DJ, Linhoff MW, Reed W, Ting JP-Y. Cutting edge:
monarch-1: a pyrin/nucleotide-binding domain/leucine-rich repeat protein
that controls classical and nonclassical MHC class I genes. J Immunol (2003)
170(11):5354–8.
43. Lich JD, Williams KL, Moore CB, Arthur JC, Davis BK, Taxman DJ,
et al. Monarch-1 suppresses non-canonical NF-kappaB activation and
p52-dependent chemokine expression in monocytes. J Immunol (2007)
178(3):1256–60.
44. Williams KL, Lich JD, Duncan JA, Reed W, Rallabhandi P, Moore C, et al.
The CATERPILLER protein monarch-1 is an antagonist of toll-like receptor-,
tumor necrosis factor alpha-, and Mycobacterium tuberculosis-induced pro-
inflammatory signals. J Biol Chem (2005) 280(48):39914–24. doi:10.1074/jbc.
M502820200
45. Arthur JC,Lich JD,Ye Z,Allen IC, Gris D,Wilson JE, et al. Cutting edge: NLRP12
controls dendritic and myeloid cell migration to affect contact hypersensitivity.
J Immunol (2010) 185(8):4515–9. doi:10.4049/jimmunol.1002227
46. Zaki MH,Vogel P, Malireddi R,Body-Malapel M, Anand PK, Bertin J, et al. The
NOD-like receptor NLRP12 attenuatescolon inflammation and tumorigenesis.
Cancer Cell (2011) 20(5):649–60. doi:10.1016/j.ccr.2011.10.022
47. AllenIC, Wilson JE, Schneider M,Lich JD, Roberts RA,Arthur JC, et al.NLRP12
suppresses colon inflammation and tumorigenesis through the negative reg-
ulation of noncanonical NF-κB signaling. Immunity (2012) 36(5):742–54.
doi:10.1016/j.immuni.2012.03.012
48. DeYoung KL, Ray ME, Su YA, Anzick SL, Johnstone RW, Trapani JA, et al.
Cloning a novel member of the human interferon-inducible gene family associ-
ated with control of tumorigenicity in a model of human melanoma. Oncogene
(1997) 15(4):453. doi:10.1038/sj.onc.1201206
49. Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, et al. HIN-
200 proteins regulate caspase activation in response to foreign cytoplasmic
DNA. Science (2009) 323(5917):1057–60. doi:10.1126/science.1169841
50. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caf-
frey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-
activating inflammasome with ASC. Nature (2009) 458(7237):514–8. doi:10.
1038/nature07725
51. Fernandes-Alnemri T, Yu J-W, Datta P, Wu J, Alnemri ES. AIM2 activates the
inflammasome and cell death in response to cytoplasmic DNA. Nature (2009)
458(7237):509–13. doi:10.1038/nature07710
52. Bürckstümmer T, Baumann C, Blüml S, Dixit E, Dürnberger G, Jahn H,
et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplas-
mic DNA sensor for the inflammasome. Nat Immunol (2009) 10(3):266–72.
doi:10.1038/ni.1702
53. Sauer J-D, Witte CE, Zemansky J, Hanson B, Lauer P, Portnoy DA. Liste-
ria monocytogenes triggers AIM2-mediated pyroptosis upon infrequent bac-
teriolysis in the macrophage cytosol. Cell Host Microbe (2010) 7(5):412–9.
doi:10.1016/j.chom.2010.04.004
54. Singh A, Rahman T, Malik M, Hickey AJ, Leifer CA, Hazlett KR, et al. Discor-
dant results obtained with Francisella tularensis during in vitro and in vivo
immunological studies are attributable to compromised bacterial structural
integrity. PLoS One (2013) 8(3):e58513. doi:10.1371/journal.pone.0058513
55. Ge J, Gong Y-N, Xu Y, Shao F. Preventing bacterial DNA release and absent
in melanoma 2 inflammasome activation by a Legionella effector functioning
in membrane trafficking. Proc Natl Acad Sci U S A (2012) 109(16):6193–8.
doi:10.1073/pnas.1117490109
56. Jin T, Perry A, Jiang J, Smith P, Curry JA, Unterholzner L, et al. Structures
of the HIN domain: DNA complexes reveal ligand binding and activation
mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity (2012)
36(4):561–71. doi:10.1016/j.immuni.2012.02.014
57. Jin T, Perry A, Smith P,Jiang J, Xiao TS. Structure of the absent in melanoma 2
(AIM2) Pyrin domain provides insights into the mechanisms of AIM2 autoin-
hibition and inflammasome assembly. J Biol Chem (2013) 288(19):13225–35.
doi:10.1074/jbc.M113.468033
58. Newell DG, Koopmans M, Verhoef L, Duizer E, Aidara-Kane A, Sprong H,
et al. Food-borne diseases the challenges of 20 years ago still persist while
new ones continue to emerge. Int J Food Microbiol (2010) 139(Suppl 1):S3–15.
doi:10.1016/j.ijfoodmicro.2010.01.021
59. Sur D, Ochiai RL, Bhattacharya SK, Ganguly NK, Ali M, Manna B, et al.
A cluster-randomized effectiveness trial of Vi typhoid vaccine in India.
N Engl J Med (2009) 361(4):335–44. doi:10.1056/NEJMoa0807521
60. Majowicz SE,Musto J, Scallan E, AnguloFJ, Kirk M, O’Brien SJ,et al. The global
burden of nontyphoidal Salmonella gastroenteritis. Clin Infect Dis (2010)
50(6):882–9. doi:10.1086/650733
61. Stecher B, Barthel M, Schlumberger MC, Haberli L, Rabsch W, Kremer M,
et al. Motility allows S. typhimurium to benefit from the mucosal defence. Cell
Microbiol (2008) 10(5):1166–80. doi:10.1111/j.1462-5822.2008.01118.x
62. Haraga A, Ohlson MB,Miller SI. Salmonellae interplay with host cells. Nat Rev
Microbiol (2008) 6(1):53–66. doi:10.1038/nrmicro1788
63. Diehl GE, Longman RS, Zhang J-X, Breart B, Galan C, Cuesta A, et al. Micro-
biota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi
cells. Nature (2013) 494(7435):116–20. doi:10.1038/nature11809
64. Kaiser P, Diard M, Stecher B, Hardt WD. The streptomycin mouse model for
Salmonella diarrhea: functional analysis of the microbiota, the pathogen’s vir-
ulence factors, and the host’s mucosal immune response. Immunol Rev (2012)
245(1):56–83. doi:10.1111/j.1600-065X.2011.01070.x
65. Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL, Crawford RW,
et al. Gut inflammation provides a respiratory electron acceptor for Salmonella.
Nature (2010) 467(7314):426–9. doi:10.1038/nature09415
66. Hisamatsu T, Suzuki M, Reinecker HC, Nadeau WJ, McCormick BA, Podol-
sky DK. CARD15/NOD2 functions as an antibacterial factor in human intesti-
nal epithelial cells. Gastroenterology (2003) 124(4):993–1000. doi:10.1053/gast.
2003.50153
67. Homer CR, Kabi A, Marina-Garcia N, Sreekumar A, Nesvizhskii AI, Nick-
erson KP, et al. A dual role for receptor-interacting protein kinase 2 (RIP2)
kinase activity in nucleotide-binding oligomerization domain 2 (NOD2)-
dependent autophagy. J Biol Chem (2012) 287(30):25565–76. doi:10.1074/jbc.
M111.326835
68. Yamamoto-Furusho JK, Barnich N, Hisamatsu T, Podolsky DK. MDP-
NOD2 stimulation induces HNP-1 secretion, which contributes to NOD2
antibacterial function. Inflamm Bowel Dis (2010) 16(5):736–42. doi:10.1002/
ibd.21144
69. Le Bourhis L, Magalhaes JG, Selvanantham T, Travassos LH, Geddes K, Fritz JH,
et al. Role of Nod1 in mucosal dendritic cells during Salmonella pathogenic-
ity island 1-independent Salmonella enterica serovar typhimurium infection.
Infect Immun (2009) 77(10):4480–6. doi:10.1128/IAI.00519-09
70. Keestra AM,Winter MG, Klein-Douwel D, Xavier MN, Winter SE, Kim A, et al.
ASalmonella virulence factor activates the NOD1/NOD2 signaling pathway.
MBio (2011) 2(6). doi:10.1128/mBio.00266-11
71. Perez-Lopez A, Rosales-Reyes R, Alpuche-Aranda CM, Ortiz-Navarrete V.
Salmonella downregulates Nod-like receptor family CARD domain contain-
ing protein 4 expression to promote its survival in B cells by preventing
inflammasome activation and cell death. J Immunol (2013) 190(3):1201–9.
doi:10.4049/jimmunol.1200415
72. Kupz A, Guarda G, Gebhardt T, Sander LE, Short KR, Diavatopoulos DA,
et al. NLRC4 inflammasomes in dendritic cells regulate noncognate effec-
tor function by memory CD8+ T cells. Nat Immunol (2012) 13(2):162–9.
doi:10.1038/ni.2195
73. Broz P, Ruby T, Belhocine K, Bouley DM, Kayagaki N, Dixit VM, et al. Caspase-
11 increases susceptibility to Salmonella infection in the absence of caspase-1.
Nature (2012) 490(7419):288–91. doi:10.1038/nature11419
74. Geddes K, Rubino S, Streutker C, Cho JH, Magalhaes JG, Le Bourhis L, et al.
Nod1 and Nod2 regulation of inflammation in the Salmonella colitis model.
Infect Immun (2010) 78(12):5107–15. doi:10.1128/IAI.00759-10
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 9
de Zoete and Flavell NLRs and intestinal bacteria
75. Geddes K, Rubino SJ, Magalhaes JG, Streutker C, Le Bourhis L, Cho JH, et al.
Identification of an innate T helper type 17 response to intestinal bacterial
pathogens. Nat Med (2011) 17(7):837–44. doi:10.1038/nm.2391
76. Bruno VM, Hannemann S, Lara-Tejero M, Flavell RA, Kleinstein SH, Galán JE.
Salmonella typhimurium type III secretion effectors stimulate innate immune
responses in cultured epithelial cells. PLoS Pathog (2009) 5(8):e1000538.
doi:10.1371/journal.ppat.1000538
77. Lara-Tejero M, Sutterwala FS, OguraY, Grant EP,Bertin J, Coyle AJ, et al. Role
of the caspase-1 inflammasome in Salmonella typhimurium pathogenesis.J Exp
Med (2006) 203(6):1407–12. doi:10.1084/jem.20060206
78. Raupach B, Peuschel S-K, Monack DM, Zychlinsky A. Caspase-1-mediated
activation of interleukin-1β(IL-1β) and IL-18 contributes to innate immune
defenses against Salmonella enterica serovar typhimurium infection. Infect
Immun (2006) 74(8):4922–6. doi:10.1128/IAI.00417-06
79. Broz P, Newton K, Lamkanfi M, Mariathasan S, Dixit VM, Monack DM.
Redundant roles for inflammasome receptors NLRP3 and NLRC4 in host
defense against Salmonella.J Exp Med (2010) 207(8):1745–55. doi:10.1084/
jem.20100257
80. Carvalho FA, Nalbantoglu I, Aitken JD, Uchiyama R, Su Y, Doho GH, et al.
Cytosolic flagellin receptor NLRC4 protectsmice against mucosal and systemic
challenges. Mucosal Immunol (2012) 5(3):288–98. doi:10.1038/mi.2012.8
81. Miao EA, Leaf IA, Treuting PM, Mao DP, Dors M, Sarkar A, et al. Caspase-1-
induced pyroptosis is an innate immune effector mechanism against intracel-
lular bacteria. Nat Immunol (2010) 11(12):1136–42. doi:10.1038/ni.1960
82. Vijay Kumar M, Carvalho FA, Aitken JD, Fifadara NH, Gewirtz AT. TLR5 or
NLRC4 is necessary and sufficient for promotion of humoral immunity by
flagellin. Eur J Immunol (2010) 40(12):3528–34. doi:10.1002/eji.201040421
83. Aachoui Y, Leaf IA, Hagar JA, Fontana MF, Campos CG, Zak DE, et al.
Caspase-11 protects against bacteria that escape the vacuole. Science (2013)
339(6122):975–8. doi:10.1126/science.1230751
84. Franchi L, Kamada N, NakamuraY, Burberry A, Kuffa P, Suzuki S, et al. NLRC4-
driven production of IL-1 [beta] discriminates between pathogenic and com-
mensal bacteria and promotes host intestinal defense. Nat Immunol (2012)
13(5):449–56. doi:10.1038/ni.2263
85. Schmidt MA. LEEways: tales of EPEC,ATEC and EHEC. Cell Microbiol (2010)
12(11):1544–52. doi:10.1111/j.1462-5822.2010.01518.x
86. Mundy R, MacDonald TT, Dougan G, Frankel G, Wiles S. Citrobacter roden-
tium of mice and man. Cell Microbiol (2005) 7(12):1697–706. doi:10.1111/j.
1462-5822.2005.00625.x
87. Wiles S,Clare S, Harker J, HuettA, YoungD, Dougan G, et al. Organ specificity,
colonization and clearance dynamics in vivo following oral challenges with the
murine pathogen Citrobacter rodentium.Cell Microbiol (2004) 6(10):963–72.
doi:10.1111/j.1462-5822.2004.00414.x
88. Bishop AL, Wiles S, Dougan G, Frankel G. Cell attachment properties and
infectivity of host-adapted and environmentally adapted Citrobacter roden-
tium.Microbes Infect (2007) 9(11):1316–24. doi:10.1016/j.micinf.2007.06.006
89. Yang J, Tauschek M, Hart E, Hartland EL, Robins Browne RM. Virulence reg-
ulation in Citrobacter rodentium: the art of timing. Microb Biotechnol (2010)
3(3):259–68. doi:10.1111/j.1751-7915.2009.00114.x
90. Celli J, Deng W, Finlay BB. Enteropathogenic Escherichia coli (EPEC) attach-
ment to epithelial cells: exploiting the host cell cytoskeleton from the outside.
Cell Microbiol (2000) 2(1):1–9. doi:10.1046/j.1462-5822.2000.00033.x
91. LeBlanc PM, Yeretssian G, Rutherford N, Doiron K, Nadiri A, Zhu L, et al.
Caspase-12 modulates NOD signaling and regulates antimicrobial peptide
production and mucosal immunity. Cell Host Microbe (2008) 3(3):146–57.
doi:10.1016/j.chom.2008.02.004
92. Kim Y-G, Kamada N, Shaw MH, Warner N, Chen GY, Franchi L, et al. The
Nod2 sensor promotes intestinal pathogen eradication via the chemokine
CCL2-dependent recruitment of inflammatory monocytes. Immunity (2011)
34(5):769–80. doi:10.1016/j.immuni.2011.04.013
93. Raymond B, CrepinVF, Collins JW, Frankel G. The WxxxE effector EspT trig-
gers expression of immune mediators in an Erk/JNK and NF-κB-dependent
manner. Cell Microbiol (2011) 13(12):1881–93. doi:10.1111/j.1462-5822.2011.
01666.x
94. Liu Z, Zaki MH, Vogel P, Gurung P, Finlay BB, Deng W, et al. Role of inflam-
masomes in host defense against Citrobacter rodentium infection. J Biol Chem
(2012) 287(20):16955–64. doi:10.1074/jbc.M112.358705
95. Lebeis SL, Powell KR, Merlin D, Sherman MA, Kalman D. Interleukin-1
receptor signaling protects mice from lethal intestinal damage caused by the
attaching and effacing pathogen Citrobacter rodentium.Infect Immun (2009)
77(2):604–14. doi:10.1128/IAI.00907-08
96. Gurung P, Malireddi RS, Anand PK, Demon D, Walle LV, Liu Z, et al. Toll or
interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-
β(TRIF)-mediated caspase-11 protease production integrates Toll-like recep-
tor 4 (TLR4) protein-and Nlrp3 inflammasome-mediated host defense against
enteropathogens. J Biol Chem (2012) 287(41):34474–83. doi:10.1074/jbc.
M112.401406
97. Calvet X, Ramírez Lázaro MJ, Lehours P, Mégraud F. Diagnosis and epi-
demiology of Helicobacter pylori infection. Helicobacter (2013) 18(s1):5–11.
doi:10.1111/hel.12071
98. Cover TL,Blaser MJ. Helicobacter pylori in health and disease. Gastroenterology
(2009) 136(6):1863–73. doi:10.1053/j.gastro.2009.01.073
99. Salama NR, Hartung ML, Müller A. Life in the human stomach: persistence
strategies of the bacterial pathogen Helicobacter pylori.Nat Rev Microbiol
(2013) 11(6):385–99. doi:10.1038/nrmicro3016
100. Wroblewski LE, Peek RM, Wilson KT. Helicobacter pylori and gastric cancer:
factors that modulate disease risk. Clin Microbiol Rev (2010) 23(4):713–39.
doi:10.1128/CMR.00011-10
101. Celli JP, Turner BS, Afdhal NH,Keates S, Ghiran I, Kelly CP, et al. Helicobacter
pylori moves through mucus by reducing mucin viscoelasticity. Proc Natl Acad
Sci U S A (2009) 106(34):14321–6. doi:10.1073/pnas.0903438106
102. Cullen TW, Giles DK, Wolf LN, Ecobichon C, Boneca IG, Trent MS. Heli-
cobacter pylori versus the host: remodeling of the bacterial outer mem-
brane is required for survival in the gastric mucosa. PLoS Pathog (2011)
7(12):e1002454. doi:10.1371/journal.ppat.1002454
103. Gewirtz AT, Yu Y, Krishna US, Israel DA, Lyons SL, Peek RM. Helicobacter
pylori flagellin evades toll-like receptor 5-mediated innate immunity. J Infect
Dis (2004) 189(10):1914–20. doi:10.1086/386289
104. Palframan SL, Kwok T, Gabriel K. Vacuolating cytotoxin A (VacA), a key toxin
for Helicobacter pylori pathogenesis. Front Cell Infect Microbiol (2012) 2:92.
doi:10.3389/fcimb.2012.00092
105. Rosenstiel P, Hellmig S, Hampe J, Ott S, Till A, Fischbach W, et al. Influ-
ence of polymorphisms in the NOD1/CARD4 and NOD2/CARD15 genes on
the clinical outcome of Helicobacter pylori infection. Cell Microbiol (2006)
8(7):1188–98. doi:10.1111/j.1462-5822.2006.00701.x
106. Hofner P, Gyulai Z, Kiss ZF, Tiszai A, Tiszlavicz L, Toth G, et al. Genetic poly-
morphisms of NOD1 and IL-8, but not polymorphisms of TLR4 genes, are
associated with Helicobacter pylori-induced duodenal ulcer and gastritis. Heli-
cobacter (2007) 12(2):124–31. doi:10.1111/j.1523-5378.2007.00481.x
107. Sugimoto M,Yamaoka Y, FurutaT. Influence of interleukin polymorphisms on
development of gastric cancer and peptic ulcer. World J Gastroenterol (2010)
16(10):1188. doi:10.3748/wjg.v16.i10.1188
108. Viala J, Chaput C, Boneca IG, Cardona A, Girardin SE, Moran AP, et al. Nod1
responds to peptidoglycan delivered by the Helicobacter pylori cag pathogenic-
ity island. Nat Immunol (2004) 5(11):1166–74. doi:10.1038/ni1131
109. Kaparakis M, Turnbull L, Carneiro L, Firth S, Coleman HA, Parkington HC,
et al. Bacterial membrane vesicles deliver peptidoglycan to NOD1 in epithe-
lial cells. Cell Microbiol (2010) 12(3):372–85. doi:10.1111/j.1462-5822.2009.
01404.x
110. Hitzler I, Sayi A, Kohler E, Engler DB, Koch KN, Hardt W-D, et al. Caspase-1
has both proinflammatory and regulatory properties in Helicobacter infections,
which are differentially mediated by its substrates IL-1βand IL-18. J Immunol
(2012) 188(8):3594–602. doi:10.4049/jimmunol.1103212
111. Kim DJ, Park JH, Franchi L, Backert S, Núñez G. The Cag pathogenicity
island and interaction between TLR2/NOD2 and NLRP3 regulate IL-1βpro-
duction in Helicobacter pylori infected dendritic cells. Eur J Immunol (2013)
43(10):2650–8. doi:10.1002/eji.201243281
112. Oertli M, Sundquist M, Hitzler I, Engler DB, Arnold IC, Reuter S, et al. DC-
derived IL-18 drives Treg differentiation, murine Helicobacter pylori-specific
immune tolerance, and asthma protection. J Clin Invest (2012) 122(3):1082.
doi:10.1172/JCI61029
113. Benoit BN, KobayashiM, Kawakubo M, Takeoka M, Sano K, Zou J, et al. Role of
ASC in the mouse model of Helicobacter pylori infection.J Histochem Cytochem
(2009) 57(4):327–38. doi:10.1369/jhc.2008.952366
Frontiers in Immunology | Molecular Innate Immunity December 2013 | Volume 4 | Article 462 | 10
de Zoete and Flavell NLRs and intestinal bacteria
114. Sugimoto M, Furuta T,Yamaoka Y. Influence of inflammatory cytokine poly-
morphisms on eradication rates of Helicobacter pylori.J Gastroenterol Hepatol
(2009) 24(11):1725–32. doi:10.1111/j.1440-1746.2009.06047.x
115. Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman
AL, et al. The long-term stability of the human gut microbiota. Science (2013)
341(6141). doi:10.1126/science.1237439
116. Hu B, Elinav E, Huber S, Strowig T, Hao L, Hafemann A, et al. Microbiota-
induced activation of epithelial IL-6 signaling links inflammasome-driven
inflammation with transmissible cancer. Proc Natl Acad Sci U S A (2013)
110(24):9862–7. doi:10.1073/pnas.1307575110
117. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al.
Inflammasome-mediated dysbiosis regulates progression of NAFLD and obe-
sity. Nature (2012) 482(7384):179–85. doi:10.1038/nature10809
118. Bouskra D, Brézillon C, Bérard M, Werts C, Varona R, Boneca IG, et al. Lym-
phoid tissue genesis induced by commensals through NOD1 regulates intesti-
nal homeostasis. Nature (2008) 456(7221):507–10. doi:10.1038/nature07450
119. Robertson SJ, Zhou JY, Geddes K, Rubino SJ, Cho JH, Philpott DJ,et al. Nod1
and Nod2 signaling does not alter the composition of intestinal bacterial com-
munities at homeostasis. Gut Microbes (2013) 4(3):222–31. doi:10.4161/gmic.
24373
120. Natividad JM, Petit V, Huang X, de Palma G, Jury J, Sanz Y, et al. Commensal
and probiotic bacteria influence intestinal barrier function and susceptibility to
colitis in Nod1/; Nod2/mice.Inflamm Bowel Dis (2012)18(8):1434–46.
doi:10.1002/ibd.22848
121. Hugot J-P, Chamaillard M, Zouali H, Lesage S, Cézard J-P, Belaiche J, et al.
Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s
disease. Nature (2001) 411(6837):599–603. doi:10.1038/35079107
122. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, et al.
A frameshift mutation in NOD2 associated with susceptibility to Crohn’s dis-
ease. Nature (2001) 411(6837):603–6. doi:10.1038/35079114
123. Lu W-G, Zou Y-F, Feng X-L, Yuan F-L, Gu Y-L, Li X, et al. Association of
NOD1 (CARD4) insertion/deletion polymorphism with susceptibility to IBD:
a meta-analysis. World J Gastroenterol (2010) 16(34):4348. doi:10.3748/wjg.
v16.i34.4348
124. Hirota SA,Ng J, Lueng A,Khaj ah M,Parhar K, Li Y, et al.NLRP3 inflammasome
plays a key role in the regulation of intestinal homeostasis. Inflamm Bowel Dis
(2011) 17(6):1359–72. doi:10.1002/ibd.21478
125. Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti T-D. The
NLRP3 inflammasome protects against loss of epithelial integrity and mortal-
ity during experimental colitis. Immunity (2010) 32(3):379–91. doi:10.1016/j.
immuni.2010.03.003
126. Allen IC, TeKippe EM, Woodford R-MT, Uronis JM, Holl EK, Rogers AB,
et al. The NLRP3 inflammasome functions as a negative regulator of tumori-
genesis during colitis-associated cancer. J Exp Med (2010) 207(5):1045–56.
doi:10.1084/jem.20100050
127. Bauer C, Duewell P, Mayer C, Lehr HA, Fitzgerald KA, Dauer M, et al.
Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by
the NLRP3 inflammasome. Gut (2010) 59(9):1192–9. doi:10.1136/gut.2009.
197822
128. Bauer C, Duewell P, Lehr H-A, Endres S, Schnurr M. Protective and aggra-
vating effects of NLRP3 inflammasome activation in IBD models: influence
of genetic and environmental factors. Dig Dis (2012) 30(Suppl 1):82–90.
doi:10.1159/000341681
129. VillaniA-C, Lemire M, Fortin G, Louis E,Silverberg MS, Collette C, et al. Com-
mon variants in the NLRP3 region contribute to Crohn’s disease susceptibility.
Nat Genet (2008) 41(1):71–6. doi:10.1038/ng.285
130. Schoultz I, Verma D, Halfvarsson J, Törkvist L, Fredrikson M, Sjöqvist U, et al.
Combined polymorphisms in genes encoding the inflammasome components
NALP3 and CARD8 confer susceptibility to Crohn’s disease in Swedish men.
Am J Gastroenterol (2009) 104(5):1180–8. doi:10.1038/ajg.2009.29
131. Lewis GJ, Massey DC, Zhang H, Bredin F, Tremelling M, Lee JC, et al.
Genetic association between NLRP3 variants and Crohn’s disease does not
replicate in a large UK panel. Inflamm Bowel Dis (2011) 17(6):1387–91.
doi:10.1002/ibd.21499
Conflict of Interest Statement: The authors declare that the researchwas conducted
in the absence of any commercial or financial relationships that could be construed
as a potential conflict of interest.
Received: 01 October 2013; accepted: 03 December 2013; published online: 17 December
2013.
Citation: de Zoete MR and Flavell RA (2013) Interactions between Nod-like receptors
and intestinal bacteria. Front. Immunol. 4:462. doi: 10.3389/fimmu.2013.00462
This article was submitted to Molecular Innate Immunity, a section of the journal
Frontiers in Immunology.
Copyright © 2013 de Zoete and Flavell .This is an ope n-access article distributed under
the terms of the Creative Commons Attribution License (CC BY). The use, distribution
or reproduction in other forums is permitted, provided the original author(s) or licensor
are credited and that the original publication in this journal is cited, in accordance with
accepted academic practice. No use, distribution or reproduction is permitted which
does not comply with these terms.
www.frontiersin.org December 2013 | Volume 4 | Article 462 | 11
... In this research, we found that LPC regulated jejuna morphology via multiple signaling pathways, such as toll-like receptor (TLR), nod-like receptor (NLR), and necroptosis pathways. Previous studies indicated that TLR, NLR, and RILR were a variety of pathogen pattern recognition receptors involved in maintaining intestinal morphology, homeostasis and health (32)(33)(34)(35)(36)(37)(38)(39)(40)(41). TLR mediated the inflammatory responses of intestinal mucosa macrophages to resist the pathogen invasion (32). ...
... NLRs mediated inflammatory cell pyroptosis, caspase-1 activation. NLRs might be involved in the common gastrointestinal bacterial pathogen infection in the small intestine (37). NLRs also regulate intestinal microbiota. ...
Article
Full-text available
Lysophosphatidylcholine (LPC) plays a vital role in promoting jejuna morphology in broilers. However, the potential mechanism behind LPC improving the chicken jejuna morphology is unclear. Therefore, the present study was designed to reveal the important genes associated with LPC regulation in birds' jejuna. Thus, GSE94622, the gene expression microarray, was obtained from Gene Expression Omnibus (GEO). GSE94622 consists of 15 broiler jejuna samples from two LPC-treated (LPC500 and LPC1000) and the control groups. Totally 98 to 217 DEGs were identified by comparing LPC500 vs. control, LPC1000 vs. control, and LPC1000 vs. LPC500. Gene ontology (GO) analysis suggested that those DEGs were mainly involved in the one-carbon metabolic process, carbon dioxide transport, endodermal cell differentiation, the positive regulation of dipeptide transmembrane transport, cellular pH reduction, and synaptic transmission. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated the DEGs were enriched in NOD-like receptor (NLR), RIG-I-like receptor (RILR), Toll-like receptor (TLR), and necroptosis signaling pathway. Moreover, many genes, such as RSAD2, OASL, EPSTI1, CMPK2, IFIH1, IFIT5, USP18, MX1, and STAT1 might be involved in promoting the jejuna morphology of broilers. In conclusion, this study enhances our understanding of LPC regulation in jejuna morphology.
... NLRP3 acts as the sensor where the carboxyl-terminal LRR domain is involved in stimuli recognition. Upon receiving an appropriate stimulus, different components start assembling and result in the zymogenic conversion of procaspase-1 to caspase-1, which in turn, activates the proinflammatory cytokines IL-1β and IL-18 [109][110][111]. The inflammasome-mediated activation of IL-1β and IL-18 leads to a quick highly inducible proinflammatory response, which is tightly regulated. ...
Article
Full-text available
Ischemic thrombotic disease, characterized by the formation of obstructive blood clots within arteries or veins, is a condition associated with life-threatening events, such as stroke, myocardial infarction, deep vein thrombosis, and pulmonary embolism. The conventional therapeutic strategy relies on treatments with anticoagulants that unfortunately pose an inherent risk of bleeding complications. These anticoagulants primarily target clotting factors, often overlooking upstream events, including the release of neutrophil extracellular traps (NETs). Neutrophils are integral components of the innate immune system, traditionally known for their role in combating pathogens through NET formation. Emerging evidence has now revealed that NETs contribute to a prothrombotic milieu by promoting platelet activation, increasing thrombin generation, and providing a scaffold for clot formation. Additionally, NET components enhance clot stability and resistance to fibrinolysis. Clinical and preclinical studies have underscored the mechanistic involvement of NETs in the pathogenesis of thrombotic complications, since the clots obtained from patients and experimental models consistently exhibit the presence of NETs. Given these insights, the inhibition of NETs or NET formation is emerging as a promising therapeutic approach for ischemic thrombotic diseases. Recent investigations also implicate a role for the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome as a mediator of NETosis and thrombosis, suggesting that NLRP3 inhibition may also hold potential for mitigating thrombotic events. Therefore, future preclinical and clinical studies aimed at identifying and validating NLRP3 inhibition as a novel therapeutic intervention for thrombotic disorders are imperative.
... These are cytosolic receptors that are predominantly located in immune cells, and whose activation is associated with caspases signaling cascades, leading to the release of proinflammatory cytokines such as IL-1β and IL-18 [43,44]. Such cascade is generated once ligands related to intestinal dysbiosis states, such as microbe-associated molecular patterns (MAMPs), bind to NOD1/2 receptors [44,45]. Therefore, by combating intestinal dysbiosis thanks to its prebiotic effects, DA supplementation could reduce the activity of such receptors and, thus, decrease local and systemic inflammation. ...
Article
Full-text available
Durvillaea antarctica is the seaweed that is the most consumed by the Chilean population. It is recognized worldwide for its high nutritional value in protein, vitamins, minerals, and dietary fiber. This is a narrative review in which an extensive search of the literature was performed to establish the immunomodulator, cardiometabolic, and gut microbiota composition modulation effect of Durvillaea antarctica. Several studies have shown the potential of Durvillaea antarctica to function as prebiotics and to positively modulate the gut microbiota, which is related to anti-obesity, anti-inflammatory, anticancer, lipid-lowering, and hypoglycemic effects. The quantity of Bacteroides was negatively correlated with that of inflammatory monocytes and positively correlated with the levels of several gut metabolites. Seaweed-derived polysaccharides modulate the quantity and diversity of beneficial intestinal microbiota, decreasing phenol and p-cresol, which are related to intestinal diseases and the loss of intestinal function. Additionally, a beneficial metabolic effect related to this seaweed was observed, mainly promoting the decrease in the glycemic levels, lower cholesterol levels and cardiovascular risk. Consuming Durvillaea antarctica has a positive impact on the immune system, and its bioactive compounds provide beneficial effects on glycemic control and other metabolic parameters.
... Sensing of various stimuli can set off inflammasome assembly and result in the conversion of zymogenic procaspase-1 to caspase-1, which in turn leads to the release of pro-inflammatory cytokines IL-1β and IL-18 Strowig et al., 2012;Zoete & Flavell, 2013). Subsequently, inflammasome-mediated processing and secretion of IL-1β and IL-18 enable a quick, yet tightly regulated and highly inducible pro-inflammatory response. ...
Article
Full-text available
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Several pathological hallmarks have been identified, including neuroinflammation. A comprehensive insight into the underlying mechanisms that can fuel the development of novel therapeutic approaches is necessary due to the alarmingly rapid increase in the frequency of incidence. Recently, NLRP3 inflammasome was identified as a critical mediator of neuroinflammation. Activation of nucleotide‐binding domain (NOD)‐like receptor protein 3 (NLRP3) inflammasome by amyloid, neurofibrillary tangles, impaired autophagy and endoplasmic reticulum stress, triggers the release of pro‐inflammatory cytokines such as IL‐1β and IL‐18. Subsequently, these cytokines can promote neurodegeneration and cognitive impairment. It is well established that genetic or pharmacological ablation of NLRP3 alleviates AD related pathological features in in vitro and in vivo models. Therefore, several synthetic and natural compounds have been identified that exhibit the potential to inhibit NLRP3 inflammasome and alleviate AD associated pathology. The current review article will highlight the various mechanisms by which activation of NLRP3 inflammation occurs during Alzheimer's disease, and how it influences neuroinflammation, neurodegeneration and cognitive impairment. Moreover, we will summarize the different small molecules that possess the potential to inhibit NLRP3 and can pave the path for developing novel therapeutic interventions for AD.
... It can lead to inflammatory cell death, secretion and processing of inflammatory cytokines, colonization of gastrointestinal bacterial pathogens, and shaping of gut microbiota that plays a critical role in IBD. 67 In addition, a possible role was demonstrated in an experimental study on CARD9-associated disorders of tryptophan microbial metabolism in the pathogenesis of IBD mediated by aryl-hydrocarbon receptors (AhR). 68 ...
Article
Full-text available
Maheeba Abdulla,1 Nafeesa Mohammed2 1Internal Medicine Department, Ibn AlNafees Hospital, Arabian Gulf University, Manama, Bahrain; 2Salmaniya Medical Complex, Manama, BahrainCorrespondence: Maheeba Abdulla, Consultant Gastroenterologist, Internal Medicine Department, Ibn AlNafees Hospital, Arabian Gulf University, Manama, Bahrain, Email amaheeba@hotmail.comAbstract: Inflammatory bowel diseases are considered immune disorders with a complex genetic architecture involving constantly changing endogenous and exogenous factors. The rapid evolution of genomic technologies and the emergence of newly discovered molecular actors are compelling the research community to reevaluate the knowledge and molecular processes. The human intestinal tract contains intestinal human microbiota consisting of commensal, pathogenic, and symbiotic strains leading to immune responses that can contribute and lead to both systemic and intestinal disorders including IBD. In this review, we attempted to highlight some updates of the new IBD features related to genomics, microbiota, new emerging therapies and some major established IBD risk factors.Keywords: inflammatory bowel disease, immune disorders, genomics, ulcerative colitis, Crohn’s disease
... It is plausible that PAMPs derived from translocating intestinal microbes signal through TLR4 to induce adaptive immunity in the absence of MyD88. Another possibility is that microflora derived molecules, including peptidoglycan or muramyl dipeptide, signal through NOD-like receptors to promote MyD88-independent triggering of immunity [67][68][69]. The adjuvant effect of the microbiota is most likely strongest in the intestinal mucosa and we posit that this explains the robust IFN-γ based immune response and parasite control we observed in this tissue. ...
Article
Full-text available
Toxoplasma gondii is an orally acquired pathogen that induces strong IFN-γ based immunity conferring protection but that can also be the cause of immunopathology. The response in mice is driven in part by well-characterized MyD88-dependent signaling pathways. Here we focus on induction of less well understood immune responses that do not involve this Toll-like receptor (TLR)/IL-1 family receptor adaptor molecule, in particular as they occur in the intestinal mucosa. Using eYFP-IL-12p40 reporter mice on an MyD88 -/- background, we identified dendritic cells, macrophages, and neutrophils as cellular sources of MyD88-independent IL-12 after peroral T . gondii infection. Infection-induced IL-12 was lower in the absence of MyD88, but was still clearly above noninfected levels. Overall, this carried through to the IFN-γ response, which while generally decreased was still remarkably robust in the absence of MyD88. In the latter mice, IL-12 was strictly required to induce type I immunity. Type 1 and type 3 innate lymphoid cells (ILC), CD4 ⁺ T cells, and CD8 ⁺ T cells each contributed to the IFN-γ pool. We report that ILC3 were expanded in infected MyD88 -/- mice relative to their MyD88 +/+ counterparts, suggesting a compensatory response triggered by loss of MyD88. Furthermore, bacterial flagellin and Toxoplasma specific CD4 ⁺ T cell populations in the lamina propria expanded in response to infection in both WT and KO mice. Finally, we show that My88-independent IL-12 and T cell mediated IFN-γ production require the presence of the intestinal microbiota. Our results identify MyD88-independent intestinal immune pathways induced by T . gondii including myeloid cell derived IL-12 production, downstream type I immunity and IFN-γ production by ILC1, ILC3, and T lymphocytes. Collectively, our data reveal an underlying network of immune responses that do not involve signaling through MyD88.
... NOD-like receptors are another class of PRRs that are expressed by human cells. The important members of the NLRs family are NOD1 andNOD2 which have been known in macrophages, dendritic, Paneth, and intestinal epithelial cells.NLRs are contributed in the activation of caspase 1and mediates pro-inflammatory cytokines IL-1b and IL-18 secretion [12]. Likewise, innate immune responses are induced with TLRs [13]. ...
Preprint
Full-text available
Background: Vibrio cholerae, the causative agent of cholera, as a Gram-negative pathogen tend to colonize the small intestine. The intestinal mucus layer forms mucin physical barrier, consisted from high molecular weight proteins. Regarding the role of toxin–coregulated pilus (TCP) as one of the most important colonization factors of V. cholerae, this experimental study was designed to determine the role of TcpA in induction of mucin production and its regulatory effect on innate immunity molecules including TLRs and NODs using Caco2- PBMC cocultures as an interactive model. Materials and methods: The rTcp protein was expressed in pET-28a-tcpA construct and purified using Ni-column chromatography. The identity of rTcp was confirmed by western immunoblotting analysis using anti-poly-histidine antibody. Nontoxic doses of rTcpA was determined on Caco-2 cell lines. The effects of different concentrations of rTcpA (1, 5, 10 and 50 µg/mL) on the expression of mucin 1,3, 4, toll-like receptors (TLR1, 4), and Nucleotide-binding oligomerization domain-containing proteins (NOD1, 2) genes were evaluated in a co-culture model of human colon carcinoma cell line (Caco-2) and Peripheral Blood Mononuclear cells (PBMCs). Results:The rTcpA protein of V. choleraewas expressed in BL21 E. coli and confirmed by western blotting. The rTcpA showed a statistically significant effect on the expression of muc genes (MUC3 and MUC4) in a dose-dependent manner.This finding is supposed to facilitate physical adhesion and colonization of V. cholerae in intestinal lumen. The rTcpA moderately stimulated the expression of tlr4 and overexpressed tlr1, both of which are supposed to induce a mucosal protective response against bacterial infection and would help a promising protection in prophylaxis applications. No change in NOD2 expression might be attributed to the non-invasive nature of V. cholerae as an intestinal pathogen. Conclusion: In conclusion, the rTcpA protein of V. cholerae showed a statistically significant modulatory effect on the human gut epithelium gene expression which would help promising protection in prophylaxis applications.
Article
Full-text available
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Article
Vibrio cholerae, the causative agent of cholera, tend to colonize the small intestine as a Gram-negative pathogen. The intestinal mucus layer forms mucin physical barrier, consisted of high molecular weight proteins. Regarding the role of toxin–coregulated pilus (TCP) as one of the most important colonization factors of V. cholerae, this experimental study was designed to determine the role of TcpA in induction of mucin production and its regulatory effect on innate immunity molecules including toll like receptors (TLRs) and Nucleotide-binding oligomerization domain-containing proteins (NODs) using Caco2- PBMC co-cultures as an interactive model. The rTcpA protein of V. cholerae was expressed in BL21 Escherichia coli, purified using Ni-column chromatography and confirmed by western blotting. Nontoxic doses of rTcpA was determined on Caco-2 cell lines and different concentrations of rTcpA (1, 5, 10 and 50 μg/mL) showed a statistically significant effect on the expression of muc genes (MUC3 and MUC4) in a dose-dependent manner. This finding is supposed to facilitate physical adhesion and colonization of V. cholerae in intestinal lumen. The rTcpA moderately stimulated the expression of tlr4 and overexpressed tlr1, both of which are supposed to induce a mucosal protective response against bacterial infection. NOD2 was significantly increased which suggests that it may contribute in pro-inflammatory responses observed in cholera disease. No change in NOD1 expression was seen which might be attributed to the non-invasive nature of V. cholerae as an intestinal pathogen. In conclusion, the rTcpA protein of V. cholerae showed a statistically significant modulatory effect on the human gut epithelium gene expression which would help promising protection in prophylaxis applications.
Article
In recent years, a paradigm shift in the bidirectional interactions within the gut-brain axis in normal and pathologic conditions has been evidenced. Although the causal relationship is not completely known, the application of new therapeutic tools such as physical exercise has been described in several studies. However, there are caveats to consider when interpreting the effect of exercise training on the axis. Therefore, an integrative perspective of the gut and the brain's communication pathway is discussed and the role of exercise on influencing this communication highway is explained in this review.
Article
Full-text available
To restrict infection by Legionella pneumophila, mouse macrophages require Naip5, a member of the nucleotide-binding oligomerization domain leucine-rich repeat family of pattern recognition receptors, which detect cytoplasmic microbial products. We report that mouse macrophages restricted L. pneumophila replication and initiated a proinflammatory program of cell death when flagellin contaminated their cytosol. Nuclear condensation, membrane permeability, and interleukin-1β secretion were triggered by type IV secretion-competent bacteria that encode flagellin. The macrophage response to L. pneumophila was independent of Toll-like receptor signaling but correlated with Naip5 function and required caspase 1 activity. The L. pneumophila type IV secretion system provided only pore-forming activity because listeriolysin O of Listeria monocytogenes could substitute for its contribution. Flagellin monomers appeared to trigger the macrophage response from perforated phagosomes: once heated to disassemble filaments, flagellin triggered cell death but native flagellar preparations did not. Flagellin made L. pneumophila vulnerable to innate immune mechanisms because Naip5+ macrophages restricted the growth of virulent microbes, but flagellin mutants replicated freely. Likewise, after intratracheal inoculation of Naip5+ mice, the yield of L. pneumophila in the lungs declined, whereas the burden of flagellin mutants increased. Accordingly, macrophages respond to cytosolic flagellin by a mechanism that requires Naip5 and caspase 1 to restrict bacterial replication and release proinflammatory cytokines that control L. pneumophila infection.
Article
Full-text available
Gram-negative bacteria including Escherichia coli, Citrobacter rodentium, Salmonella typhimurium, and Shigella flexneri are sensed in an ill-defined manner by an intracellular inflammasome complex that activates caspase-11. We show that macrophages loaded with synthetic lipid A, E. coli lipopolysaccharide (LPS), or S. typhimurium LPS activate caspase-11 independently of the LPS receptor Toll-like receptor 4 (TLR4). Consistent with lipid A triggering the noncanonical inflammasome, LPS containing a divergent lipid A structure antagonized caspase-11 activation in response to E. coli LPS or Gram-negative bacteria. Moreover, LPS-mutant E. coli failed to activate caspase-11. Tlr4–/– mice primed with TLR3 agonist polyinosinic:polycytidylic acid [poly(I:C)] to induce pro-caspase-11 expression were as susceptible as wild-type mice were to sepsis induced by E. coli LPS. These data unveil a TLR4-independent mechanism for innate immune recognition of LPS.
Article
Full-text available
Helicobacter pylori colonization of the stomach affects about half of the world population and is associated with the development of gastritis, ulcers and cancer. Polymorphisms in the IL1B gene are linked to an increased risk of H. pylori-associated cancer, but the bacterial and host factors that regulate IL-1β production in response to H. pylori infection remain unknown. Using murine bone marrow-derived DCs, we show that the bacterial virulence factors cagPAI and CagL, but not VacA or CagA, regulate the induction of pro-IL-1β and the production of mature IL-1β in response to H. pylori infection. We further show that the host receptors, TLR2 and NOD2, but not NOD1, are required for induction of pro-IL-1β and NLRP3 in H. pylori-infected DCs. In contrast, NLRP3 and the adaptor ASC were essential for the activation of caspase-1, processing of pro-IL-1β into IL-1β and IL-1β secretion. Finally, we show that mice deficient in caspase-1, IL-1β and IL-1 receptor, but not NLRP3 are impaired in the clearance of CagA-positive H. pylori from the stomach when compared with wild-type mice. These studies identify bacterial cagPAI and the cooperative interaction among host innate receptors TLR2, NOD2 and NLRP3 as important regulators of IL-1β production in H. pylori-infected DCs. This article is protected by copyright. All rights reserved.
Article
Full-text available
Keeping the Inflammasome in Check Nucleotide-binding and oligomerization domain (NOD)–like receptors (NLRs) play an important role in the detection of pathogens by cells of the innate immune system. For several NLR family members, activation results in relief from autoinhibition, oligomerization, and the recruitment of signaling components that together make up the inflammasome, a large multiprotein complex. The inflammasome protects the host by inducing cell death and cytokine secretion. The specific molecular mechanisms that regulate NLR activation and inhibition, however, are not well understood. Hu et al. (p. 172 , published online 13 June) report the crystal structure of autoinhibited NLR family member NLRC4, which reveals the domains that are critical for interaction with adenosine diphosphate to keep NLRC4 in its inactive state and the domains that mediate oligomerization of the protein upon activation.
Article
Nlrp3 inflammasome activation occurs in response to numerous agonists but the specific mechanism by which this takes place remains unclear. All previously evaluated activators of the Nlrp3 inflammasome induce the generation of mitochondrial reactive oxygen species (ROS), suggesting a model in which ROS is a required upstream mediator of Nlrp3 inflammasome activation. Here we have identified the oxazolidinone antibiotic linezolid as a Nlrp3 agonist that activates the Nlrp3 inflammasome independently of ROS. The pathways for ROS-dependent and ROS-independent Nlrp3 activation converged upon mitochondrial dysfunction and specifically the mitochondrial lipid cardiolipin. Cardiolipin bound to Nlrp3 directly and interference with cardiolipin synthesis specifically inhibited Nlrp3 inflammasome activation. Together these data suggest that mitochondria play a critical role in the activation of the Nlrp3 inflammasome through the direct binding of Nlrp3 to cardiolipin.
Article
Inheritance Guts We know little about the stability of the constituent microbiota in the human gut or the extent to which the gut microbiota are a potential target for long-term health interventions. Faith et al. (p. 10.1126/science.1237439 ) analyzed the fecal microbiota of 37 individuals and found that, on average, 60% of bacterial strains remained stable for up to 5 years and many were estimated to remain stable for decades.
Article
The NLRP3 inflammasome is an important component of the innate immune system. However, its mechanism of activation remains largely unknown. We show that NLRP3 activators including bacterial pore-forming toxins, nigericin, ATP, and particulate matter caused mitochondrial perturbation or the opening of a large membrane pore, but this was not required for NLRP3 activation. Furthermore, reactive oxygen species generation or a change in cell volume was not necessary for NLRP3 activation. Instead, the only common activity induced by all NLRP3 agonists was the permeation of the cell membrane to K(+) and Na(+). Notably, reduction of the intracellular K(+) concentration was sufficient to activate NLRP3, whereas an increase in intracellular Na(+) modulated but was not strictly required for inflammasome activation. These results provide a unifying model for the activation of the NLRP3 inflammasome in which a drop in cytosolic K(+) is the common step that is necessary and sufficient for caspase-1 activation.