Man 2018
Man 2018
The immune system has an extraordinary capacity to the AIM2-like receptor (ALR) family; and pyrin from the
recognize and respond to a range of microbial patterns tripartite motif-containing protein (TRIM) family5
and danger signals. In the gastrointestinal tract, tril- (Fig. 1). Several other sensors have been suggested to
lions of microorganisms colonize the mucosal surface activate the inflammasome, including the NLR family
and lumen and constantly release immunomodulatory members NLRP6, NLRP7, NLRP9 (NLRP9b in mice)
molecules that interact with and shape the immune and NLRP12, the DNA sensor IFNγ-inducible protein
system1. Most of these microorganisms are symbionts 16 (IFI16) and the RNA sensor retinoic acid-inducible
or so-called commensals and do not generally evoke a gene I protein (RIG-I; also known as DDX58); however,
detrimental inflammatory response. However, patho- the ability of these sensors to form inflammasome com-
gens have the ability to invade the mucosal barrier and plexes is still uncertain6. On activation, inflammasome
underlying tissue, inducing the production of cytokines, sensors recruit the adaptor protein apoptosis-associated
chemokines and antimicrobial molecules. speck-like protein containing a caspase activation and
A central and rapid mechanism triggering an inflam- recruitment domain (ASC; also known as PYCARD)
matory response occurs through activation of the innate and the cysteine protease caspase 1, and form a single
immune signalling complex called the inflammasome2,3. cytoplasmic aggregate known as the inflammasome
The inflammasome is formed in a variety of immune speck6. Caspase 1 is activated in this complex, inducing
and non-immune cells when a subset of cytosolic proteolytic cleavage of the pro-inflammatory cytokines
Department of Immunology pattern-recognition receptors known as inflammasome pro-IL-1β and pro-IL-18 and the pore-forming protein
and Infectious Disease, The sensors recognizes pathogen-associated molecular gasdermin D, which results in the release of bioactive
John Curtin School of Medical patterns (PAMPs) or danger-associated molecular pat- IL-1β and IL-18 into the extracellular milieu and pyrop-
Research, The Australian terns (DAMPs)4. To date, members from three families tosis, respectively7. Bioactive IL-1β and IL-18 and cer-
National University,
Canberra, Australia.
of pattern-recognition receptors are known to have an tain DAMPs can be secreted through the gasdermin D
established role in the assembly of an inflammasome: pores before rupture of the host cell membrane, cell lysis
e-mail: siming.man@
anu.edu.au NLRP1, NLRP3, NLRC4 and neuronal apoptosis inhibi and cell death8–10.
https://doi.org/10.1038/ tory proteins (NAIPs) from the NOD-like receptor Inflammation and cell death responses triggered by
s41575-018-0054-1 (NLR) family; absent in melanoma 2 (AIM2) from the inflammasome contribute to the pathogenesis of a
%CPQPKECNKPȱCOOCUQOG 0QPECPQPKECNKPȱCOOCUQOG
0.42D 0.42 0#+2U 0.4% #+/ 2[TKP 0.42
0'- 0'-
%CURCUG
#5% %CURCUG
%CURCUG
%CURCUG
#EVKXG #EVKXG
ECURCUG ECURCUG
)CUFGTOKP& #OKPQ
VGTOKPWU 2TQ+.β +.β
2[TQRVQUKU 2TQ+. +.
Fig. 1 | Inflammasome complexes. The inflammasome sensors NLRP1b, NLRP3, NLRC4, AIM2 and pyrin are all capable
of forming a canonical inflammasome complex containing the adaptor protein ASC and the cysteine protease caspase 1.
NLRP1b and NLRC4 also recruit caspase 1 without ASC owing to the presence of a CARD domain in their structure64–66.
Activation of NLRP3 and NLRC4 requires the kinase NEK7 and the NLR family members neuronal apoptosis inhibitory
proteins (NAIPs), respectively. Caspase 1 cleaves the precursor cytokines pro-IL-1β and pro-IL-18 and the pore-forming
protein gasdermin D. The active fragment of gasdermin D oligomerizes and forms pores on the cell membrane, resulting
in pyroptosis73–80. These pores also allow passive release of biologically active IL-1β and IL-18 from the cell. The non-canonical
inflammasome is defined by a requirement for human caspase 4, human caspase 5 or mouse caspase 11 for the activation
of the NLRP3 inflammasome complex16. Activation of these caspases leads to cleavage of gasdermin D and pyroptosis73–75.
The pore-forming fragment of gasdermin D activates the NLRP3 inflammasome and caspase 1-dependent maturation
of IL-1β and IL-18 (refs73,74).
NAIPs and NLRC4 are expressed in both immune bacteria that naturally escape the pathogen-containing
cells and enterocytes (Fig. 2). Infection of intestinal vacuole, such as S. flexneri and Burkholderia thailan-
macrophages with S. Typhimurium, but not with the densis, transport their LPS into the cytoplasm of macro
commensal bacteria Bacteroides fragilis, Enterococcus fae- phages, where the hexa-acylated lipid A portion of the
calis or Lactobacillus plantarum, triggers activation of the LPS binds and activates caspase 4, caspase 5 and caspase 11
NAIP–NLRC4 inflammasome85, suggesting that innate (refs 22,97,98) . Indeed, the naturally cytosolic bacteria
immune detection of flagellin and the T3SS enables Burkholderia pseudomallei, B. thailandensis and a
the host immune cell to, in part, discriminate between genetically engineered strain of S. Typhimurium that
pathogenic and commensal bacteria (Fig. 4b). In addition, are incapable of remaining in the pathogen-containing
intestinal macrophages, unlike bone marrow-derived vacuole of macrophages and thus enter the host cyto-
macrophages, are relatively unresponsive to activators plasm aberrantly, are rapidly cleared from the host in
of the NLRP3 inflammasome owing to rapid protea a caspase 11-dependent manner 99,100. Furthermore,
somal degradation of NLRP3 and pro-IL-1β95, poten- outer membrane vesicles carrying LPS are shed by
tially representing a mechanism to restrain activation E. coli and most Gram-negative bacteria101. These outer
of an inflammasome sensor in a cell type that normally membrane vesicles are phagocytosed by macrophages,
exposes and responds to a plethora of signals. leading to LPS-induced activation of caspase 11 and
In addition to the NAIP–NLRC4 inflammasome, LPS the non-canonical NLRP3 inflammasome even in the
from Gram-negative bacteria including S. Typhimurium, absence of bacterial invasion of the host cell102.
E. coli and C. rodentium have been found to be recog- Emerging evidence indicates that pyroptosis exe-
nized by caspase 4, caspase 5 and caspase 11 on the basis cuted by NAIP–NLRC4 or caspase 11 is an effective
of cell culture and mouse studies16. LPS from these bacte- host-protective mechanism that either drives bacteria
ria is liberated in the cytoplasm through a host-mediated out of infected host cells or removes an infected cell in
process requiring interferon-inducible GTPases96. These its entirety from the host63. In the former scenario, the
interferon-inducible effector proteins rupture the mem- expelled bacteria released from an infected macrophage
brane of the pathogen-containing vacuole and/or the could be free or trapped within a cellular corpse com-
bacterial cell membrane96. Cytosolic Gram-negative posed of pyroptotic debris, both of which are cleared
by neutrophils84,103. In the latter scenario, enterocytes
in the mouse intestine infected with bacteria, such as
S. Typhimurium, can be physically extruded in their
Caspa e 1
IL- as 4
IL- β e 5
sp se
rin 2
s
NL IPs
NLRC
AS 2
NA18
CaC
R
R
a
M
1
AI
NL P3 –NL
IL- de e 11
in
Gaspa e 1
RP b
rin 2
R a
s
s
3
NLRC4
NLRP1
NL P9
Py 1
Caspa
NL s
NLRC
AS 2
P
NA18
R
M
I
IL-
AI
Dendritic cells (bone marrow-derived) gasdermin D10,106 (Fig. 2). This cell-intrinsic resistance to
Enterocytes pyroptosis enables neutrophils to phagocytose extracel-
Hepatocytes lular bacteria or bacteria entrapped in pyroptotic debris,
Gastric epithelium secrete an abundant amount of IL-1β to amplify local
Goblet cells
Intestinal endothelial cells
inflammation and generate ROS to facilitate intracellular
Intestinal myofibroblasts pathogen killing84,106 (Fig. 4d).
Intestinal phagocytes
Lamina propria inflammatory monocytes NLRP3 and pyrin — sensors of physiological aberra-
Liver immune cells tions. Bacteria that either evade or fail to activate the
Macrophages (bone marrow-derived) NAIP–NLRC4 inflammasome or caspase 11 can be rec-
Neutrophils ognized by other inflammasome complexes22,98 (Fig. 4).
Expression reported Of clinical importance in this bacterial category are
Expression not reported or unknown the Gram-negative bacterium Helicobacter pylori and
several toxin-producing foodborne bacteria, such as
Fig. 2 | Expression of inflammasome sensors and related molecules by cell type.
Staphylococcus aureus. H. pylori, the most common
a | Expression in humans. b | Expression in mice. References for expression data are given
in Supplementary table 1. AIM2, absent in melanoma 2; ASC, apoptosis-associated bacterial cause of gastric cancer107, encodes flagella
speck-like protein containing a caspase activation and recruitment domain (CARD); and flagellin A (FlaA) subunits whose structures evade
NAIP, neuronal apoptosis inhibitory protein; NLRC, nucleotide-binding domain, detection by the NAIP–NLRC4 inflammasome 108.
leucine-rich repeat-containing protein (NLR) family CARD domain-containing protein; Instead, H. pylori infection induces ROS production,
NLRP, NACHT, LRR and PYD domains-containing protein. potassium efflux and lysosomal destabilization in the
Activators PAMPs and DAMPs, LPS, lipid A Bacterial flagellin Microbial Toxin-induced
including microbial and host and the T3SS and RHO GTPase
toxins, RNA, oxidized (needle and inner mammalian modification
DNA–RNA hybrids, phospholipids rod proteins) DNA
mitochondrial DNA,
ATP, K+ efflux and ROS
Caspase 4
ASC Caspase 5
Caspase 1 Caspase 11
Fig. 3 | Inflammasomes recognize gastrointestinal bacteria, viruses, protozoa and helminths. Pathogens carry a
plethora of pathogen-associated molecular patterns (PAMPs). These PAMPs either directly bind and activate an
inflammasome sensor or induce a physiological change in the cell that is sensed by an inflammasome sensor5. Pathogens
also cause substantial damage to the host cell, a process that induces the liberation of danger-associated molecular
patterns (DAMPs) that activate the inflammasome15. AIM2, absent in melanoma 2; ASC, apoptosis-associated speck-like
protein containing a caspase activation and recruitment domain (CARD); LPS, lipopolysaccharide; NAIP, neuronal
apoptosis inhibitory protein; NLRC4, nucleotide-binding domain, leucine-rich repeat-containing protein (NLR) family
CARD domain-containing protein 4; NLRP3, NACHT, LRR and PYD domains-containing protein 3; ROS, reactive oxygen
species; T3SS, type 3 secretion system.
host cell, ultimately leading to activation of the canon- inflammasome sensors. The Gram-positive bacterium
ical NLRP3 inflammasome109–113. Further studies have C. difficile is a causative agent of diarrhoea associated
shown that the H. pylori virulence factors cag patho- with the use of antibiotics, toxic megacolon and colonic
genicity island and urease B subunit potentiate acti- perforation117. The toxins toxin A (TcdA) and toxin B
vation of the NLRP3 inflammasome and secretion of (TcdB) secreted by C. difficile can inactivate host trans-
IL-1β109,112,113. Excessive production of IL-1β could be a forming protein RHOA at the GTPase switch I region;
vital link between H. pylori infection and tumorigenesis this RHO-inactivating activity is sensed by pyrin, result-
in the stomach. Indeed, a study found that a transgenic ing in the assembly of the pyrin inflammasome51,52.
mouse strain engineered to overexpress human IL-1β The physiological function of pyrin in the gastro
in the stomach is prone to the development of gastric intestinal tract remains unexplored, and further studies
cancer, either spontaneously or after colonization with are required to clarify its role. However, wild-type mice
Helicobacter felis, a model organism for H. pylori114. treated with the IL-1R antagonist anakinra are protected
The specific inflammasome involved in the develop- from C. difficile-toxin-induced intestinal injury118. This
ment of gastric cancer is not known; however, an asso- finding indicates that activation of the pyrin inflamma
ciation between polymorphisms in the genes encoding some in response to C. difficile infection might lead to
NLRP3 and caspase 1 and patients with gastric cancer excessive IL-1-mediated inflammation and pathology
has been reported115. Furthermore, H. pylori infection that is detrimental to the host. Pharmacological inhibi-
suppresses the expression of the microRNA molecule tors of microtubules are efficacious in blocking the pyrin
miR-22 in gastric epithelial cells, which leads to elevated inflammasome in cell culture systems54,119,120, but their
expression of NLRP3 and cellular proliferation116. These clinical applications against pyrin-mediated pathology
studies suggest that activation of the inflammasome by have not been tested in animal models.
Helicobacter spp. infection contributes to the molecular The haemolysin of the Enterobacteriaceae mem-
basis of gastritis and gastric cancer. ber Proteus mirabilis can activate NLRP3 and drive
Toxins secreted by bacteria can induce a state of inflammation in the mouse intestine 121. Similarly,
intracellular physiological aberration that is sensed by α-haemolysins, β-haemolysins and γ-haemolysins of
the Gram-positive foodborne bacterium S. aureus medi- the human, but not the mouse, genome) is a sensor of
ate activation of the NLRP3 inflammasome in mouse bacterial lipopeptides; its activation in macrophages
macrophages and mice122,123, and the pore-forming impairs intracellular replication of L. monocytogenes
cytolysin listeriolysin O of the Gram-positive food- or S. aureus129. Further studies have shown that, in
borne bacterium L. monocytogenes potentiates activation macrophages or embryonic fibroblasts, inflammasome
of the NLRP3, NAIP–NLRC4 and AIM2 inflamma components accumulate on the phagosome and pro-
somes90,124–128. Infection with either L. monocytogenes mote phagosome–lysosome fusion and phagosomal
or S. aureus activates caspase 1 in human macrophages acidification27,105,130–132. The functional diversity of the
via NLRP3 and NLRP7 (ref.129). NLRP7 (encoded in inflammasome in both immune and non-immune cells
Autophagy
NLRP6
NLRP6 dsRNA ssRNA
NF-κB Pyroptosis
NLRP12
Pyroptosis DHX9 NLRP6
NAIP–NLRC4 and NLRP9b DHX15
caspase 11–NLRP3 ASC MAVS
inflammasome Caspase 1
Goblet cell
Enterocyte Mitochondrion
ROS
Pyroptotic
corpse
Pyroptosis
Expelled
Pyroptotic bacterium
IL-1β and IL-18 macrophage Neutrophil
Fig. 4 | Inflammasomes and related molecules contribute to the killing and clearance of gastrointestinal pathogens
in intestinal cells and immune cells. a | Inflammasomes mediate host protection against Gram-negative bacteria by
inducing the secretion of IL-1β and IL-18 and pyroptosis55,59,84,85,91,92,254,255. NLRP6 and NLRP12 negatively regulate
inflammation133,134,256,257. NLRP6 responds to Toll-like receptor (TLR)-induced autophagy in goblet cells and mediates
secretion of mucus137. RNA-bound DEAH box protein 9 (DHX9) interacts with NLRP9b, inducing the assembly of an
inflammasome complex161. The NLRP6–DHX15 complex binds to viral RNA and induces the production of type I and type III
interferons165. b | Intestinal macrophages can discriminate pathogens from commensals85. c | Activation of caspase 1,
caspase 8 or caspase 11 leads to cell death, which removes and extrudes the infected enterocyte from the epithelium93,94,104.
d | The inflammasome can reduce bacterial load by inhibiting bacterial uptake, which limits macrophage movement and
stiffness, and can promote the production of reactive oxygen species (ROS)105. Pyroptotic macrophages liberate either
whole bacteria or bacteria entrapped within pore-induced intracellular traps103. These entities are phagocytosed by
neutrophils84,106. ASC, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain
(CARD); dsRNA, double-stranded RNA; EMCV, encephalomyocarditis virus; KC, keratinocyte chemoattractant (also known
as CXCL1); MAVS, mitochondrial antiviral-signalling protein; NAIP, neuronal apoptosis inhibitory protein; NF-κB, nuclear
factor-κB; NLRC4, nucleotide-binding domain, leucine-rich repeat-containing protein (NLR) family CARD domain-containing
protein 4; ssRNA, single-stranded RNA.
ensures host protection against a range of bacteria with and increase susceptibility to sepsis150, whereas certain
preferential tropisms and virulence strategies. strains of E. coli, such as strain O21:H+, engage activation
of the NAIP–NLRC4 inflammasome to inhibit the body
NLRP6 and NLRP12 — enigmatic sensors. In line wasting induced by S. Typhimurium infection151. Thus, the
with the functional versatility of other inflammasome inflammasome influences the magnitude of inflammation
sensors, NLRP6 and NLRP12 have multiple functions towards bacterial infection and, in part, determines either
during bacterial infection. The functions of these sen- a protective or detrimental outcome in the host.
sors are enigmatic owing to the lack of expert con-
sensus on their biological functions. Both NLRP6 Viruses
and NLRP12 can negatively regulate activation of the Norwalk and Norwalk-like viruses, rotavirus, astro
nuclear factor-κB (NF-κB) and mitogen-activated pro- viruses, adenoviruses, caliciviruses and coronaviruses
tein kinase (MAPK) pathways in macrophages infected are among the leading causes of acute gastroenteritis
with L. monocytogenes, S. Typhimurium, E. coli or other worldwide152. Evidence has emerged that inflammas-
non-gastrointestinal bacteria (Fig. 4a), resulting in a state omes respond to some of these gastrointestinal viruses.
of reduced inflammation that can be readily exploited Earlier studies had demonstrated that the DNA virus
by bacterial pathogens during infection of a cell or a adenovirus induces the production of IL-1β in circu-
mouse133–136. Others have proposed that NLRP6 and lation and in hepatocytes and Kupffer cells of mice153.
NLRP12 mediate activation of caspase 1 in response to Further functional studies have shown that adenovirus,
infection by C. rodentium and Yersinia pestis, respec- specifically its virion component, triggers activation
tively137,138, and that NLRP12 functions in dampening of the NLRP3 inflammasome and secretion of IL-1β
the cell-intrinsic migratory capacity of neutrophils139. in human THP-1 macrophages and primary mouse
A further study suggests that the C57Bl/6J mice that are macrophages 154. Adenovirus-induced secretion of
used widely in research harbour a missense mutation IL-1β in human monocyte-derived macrophages and
in the Nlrp12 gene that impairs the ability of NLRP12- peripheral blood mononuclear cells partially requires
expressing macrophages to draw neutrophils to the site Toll-like receptor 9 (TLR9)155,156, presumably owing
of inflammation in response to bacterial infection140. to TLR9-mediated sensing of viral DNA as the virus
These conflicting studies highlight that the biological penetrates the cell membrane and enters the endo-
functions of NLRP6 and NLRP12 should be revisited some. Injection of an adenovirus vector into mice trig-
and should take into account the genetic background gers NLRP3-dependent production of IL-1β and other
of the mouse strains, cell types, bacterial agents and the pro-inflammatory cytokines, highlighting a physio-
experimental approaches used. logical role for the NLRP3 inflammasome in licensing
an innate immune response to adenovirus in vivo154.
Detrimental roles of the inflammasome. Overt activation This finding is also consistent with the observation that
of the inflammasome is detrimental, especially during IL-1R, which is activated downstream of inflamma
systemic bacterial dissemination, which might arise from somes, incites an acute and deleterious inflammatory
intestinal perforation. Mice lacking caspase 11 or the signalling cascade in mice infected with adenovirus153.
pyroptosis-inducing effector gasdermin D are remark- The magnitude of the inflammatory response is reduced
ably resistant to LPS-induced or E. coli-induced endo- in mice lacking IL-1R or in wild-type mice treated with
toxaemia compared with wild-type mice16,73,97,98,141–144, an anti-IL-1 antibody153, suggesting that blockade of
suggesting that caspase 11-mediated pyroptosis drives IL-1 signalling prevents overt inflammation that might
lethality in endotoxaemia. Deletion of the Nlrp3 gene in otherwise be damaging to the host.
mice offers protection against polymicrobial endotoxae- Similar to adenovirus, enterovirus EV71, a spor
mia following caecal ligation and puncture145,146. In this adic cause of diarrhoea and a common cause of hand,
model of systemic inflammation, the lack of inflammas- foot and mouth disease in humans157, is sensed by the
ome activity impairs the production of pro-inflammatory NLRP3 inflammasome158,159. Inflammasome-mediated
and endogenous lipid mediators but increases the production of IL-18 is crucial for the protection against
levels of pro-resolving lipid mediators145. Moreover, enterovirus EV71 in mice158,159. Administration of IL-18
LPS-induced endotoxaemia promotes secretion of bile to infected mice reduces viral loads and the production
acids from hepatocytes in mice; bile acids are a form of of pro-inflammatory cytokines159. The precise viral
DAMP that either synergize with ATP to activate the components of enterovirus EV71 that activate NLRP3
NLRP3 inflammasome147 or signal through the mem- are unknown; it is possible that the single-stranded
brane receptor G protein-coupled bile acid receptor 1 RNA (ssRNA) viral genome and/or the structural
(GBAR1) to inhibit the NLRP3 inflammasome148. proteins of the capsid might trigger activation of the
In other models of systemic inflammation, activa- NLRP3 inflammasome.
tion of the NAIP–NLRC4 inflammasome by systemic The double-stranded RNA (dsRNA) virus rotavirus
delivery of cytosolic flagellin to mice results in the pro- is one of the most common causes of gastroenteritis
duction of inflammatory lipid mediators called eicosa- in children160. The role of inflammasomes in the host
noids149. Synthesis of eicosanoids initiates fluid loss into defence against this virus was revealed when the poorly
the mouse intestine and peritoneal cavity and ultimately characterized NLR member NLRP9b was found to
causes rapid host death within 30 min149. The presence of mediate sensing of rotavirus and drive the production
a multidrug-resistant E. coli pathobiont in the gut can also of IL-18 and pyroptosis in organoids generated from
trigger activation of the NAIP–NLRC4 inflammasome the small intestine of mice161. Furthermore, mice lacking
NLRP9b are substantially more susceptible to rotavi- E. histolytica170. Further studies indicated that the para
rus infection, harbour a higher viral load and develop site integrin-binding cysteine protease E. histolytica
more severe intestinal pathology than wild-type mice161. cysteine protease gene 5 (CP5) mediates activation of
Mouse NLRP9b and its human counterpart, NLRP9, the host α5β1 integrin during parasite–macrophage
are strongly and specifically expressed in ileal epithelial contact, leading to the release of ATP and subsequent
cells and primary intestinal epithelial cells, respectively161 activation of the canonical NLRP3 inflammasome171.
(Fig. 2). Both mouse NLRP9b and human NLRP9 coop- The importance of the inflammasome in E. histolytica
erate with the RNA sensor DEAH box protein 9 (DHX9) infection is partially inferred by the observation
to sense dsRNA161 (Fig. 4a), implicating the existence of that recombinant IL-1β and IL-18 are cleaved and
an RNA-sensing NLRP9 inflammasome complex in the inactivated by E. histolytica CP5 (refs172,173).
host defence against rotavirus infection. A protective Similar to pathogenic protozoa, the commensal gut
role of inflammasomes was also observed in a previous protozoan Tritrichomonas musculis in mice also triggers
study showing that administration of bacterial flagel- activation of the inflammasome in intestinal epithelial
lin to mice — causing activation of NLRC4 and the cells in mice174. In this context, T. musculis–induced
membrane-bound flagellin sensor TLR5, which induced activation of the inflammasome promotes IL-18 secre-
the production of IL-18 and IL-22, respectively — tion, T helper 1 (TH1) cell and TH17 cell immunity and
mediates clearance of rotavirus within 24 h (ref.162). protection against S. Typhimurium infection174, high-
Recapitulating the beneficial effects of flagellin, coadmin lighting a favourable outcome of this host–protozoan
istration of IL-18 and IL-22 similarly drives anti- engagement. However, intestinal colonization by
rotavirus immunity in infected mice, supporting the view T. musculis also drives sustained inflammation that ele-
that inflammasome signalling can mediate protection vates the likelihood of developing T cell-driven colitis
against rotavirus infection. and sporadic colorectal tumours in mice174. The pre-
Similar to NLRP9, NLRP6 is strongly expressed in cise inflammasome complex triggered by T. musculis
the intestinal tract of humans and mice163,164 (Fig. 2). In is unknown; however, it is likely that protozoa-induced
concert with the RNA sensor DHX15, NLRP6 interacts activation of the inflammasome in the gastrointestinal
with viral RNA and mediates the recognition of the tract is a double-edged sword that contributes to both
ssRNA viruses encephalomyocarditis virus and murine health and disease.
norovirus165 (Fig. 4a). In this case, NLRP6 does not
assemble an inflammasome. Instead, the RNA-bound Intestinal inflammation, IBD and cancer
NLRP6–DHX15 complex migrates to the mitochondria, The ability of inflammasomes to respond to danger
where it interacts with the signalling adaptor mitochon- signals broadens their clinical relevance to diseases
drial antiviral-signalling protein (MAVS) to induce the other than infectious diseases175,176. Genetic analyses in
production of type I and type III interferons and viral humans have pinpointed an association between muta-
clearance165 (Fig. 4a). These studies underscore emerg- tions in genes encoding NLRs and intestinal inflam-
ing roles of the inflammasome and associated signalling mation, autoinflammatory conditions and cancer14,177.
molecules in antiviral defence of the gastrointestinal The contribution of individual inflammasome sensors
tract. The relative contribution of IL-1β and IL-18 secre- and related molecules in the pathogenesis of intestinal
tion versus pyroptosis in driving immunity to viruses inflammation, IBD and cancer is discussed herein.
remains largely unknown. The current availability of
mice lacking gasdermin D will enable exploration of this NLRP3
emerging research area. NLRP3 is a global sensor of PAMPs and DAMPs and
is expressed in multiple cell types (Fig. 2), contribut-
Protozoa ing to its links to a plethora of clinical manifestations.
Protozoal pathogens cause several neglected tropical Genetic analysis identified an association between six
diseases and are public health risks largely affecting polymorphisms located in a region 4.7 kb downstream
the developing world166. Inflammasomes are known of the NLRP3 gene and the development of Crohn’s dis-
to recognize major protozoal pathogens, including ease in some, but not all, European populations178,179.
Leishmania spp., Plasmodium spp. and Toxoplasma Peripheral blood mononuclear cells derived from
spp. 167. However, information concerning the bio patients with Crohn’s disease who were homozygous for
logical importance of inflammasomes in the context any one of these six polymorphisms have an impaired
of gastrointestinal protozoa is limited. The foodborne ability to express NLRP3 and secrete IL-1β in response
protozoan Entamoeba histolytica causes amoebiasis, to LPS stimulation in vitro178. A further study reported
which is characterized by dysentery, amoebic colitis, that a heterozygous NLRP3 polymorphism, resulting in
amoeboma (the formation of fibrotic tissue masses in a mutation at residue Q705K, is associated with poor
the intestinal wall) and invasive disease of the brain, survival in patients with advanced colorectal cancer
lung and liver168. There is evidence to support the idea (CRC)180. Global expression analysis of genes encod-
that E. histolytica infection activates caspase 1 and ing cytosolic innate immune sensors revealed that the
induces secretion of IL-1β and IL-18 in both human expression of genes encoding NLRP3 and other inflam-
and mouse macrophages169. Pharmacological blockade masome sensors is substantially reduced in tumour
of potassium efflux inhibits activation of caspase 1, tissues compared with non-tumour-associated tissues
IL-1β-release and pyroptosis in THP-1 macrophages or in patients with CRC181. Similar observations have
intestinal epithelial cells that have been infected with been made in mice during colitis-associated CRC182.
However, how reduced expression of these genes is initially tumour burden in inflammasome-deficient mice that had
triggered or executed mechanistically is unknown. been administered AOM and DSS; the same treatment
Several groups have found that mice lacking NLRP3, also ameliorated colitis in inflammasome-deficient mice
ASC or caspase 1 and administered a combination of exposed to the chemical colitogen oxazolone183,187,204,211,214.
the DNA-damaging agent azoxymethane (AOM) and the IL-18 functionally exerts healing properties184,185,187,214 and
chemical colitogen dextran sodium sulfate (DSS) are triggers natural killer cell-mediated killing of colonic
highly sensitive to the development of colitis and tumour cells that have metastasized to the liver215.
colitis-associated CRC183–188. DSS-induced activation of Another study challenged the view that IL-18 is beneficial
the inflammasome in mice also causes a loss of enteric in intestinal inflammation and reported that the IL-18
neurons and onset of abnormal gut motility189. The activ- and IL-18 receptor signalling pathway drives depletion
ity of the NLRP3 inflammasome during DSS-induced of mucus-secreting goblet cells, such that a loss of the
colitis is controlled by the microRNA molecule miR- mucous layer augments sensitivity to DSS-induced coli-
223, which binds the miR-223-binding site within the tis in mice216. Similarly, a transgenic mouse strain over
3′ untranslated region of NLRP3 (ref.190). However, the state expressing IL-18 develops aggressive colitis in response to
of hyporesponsiveness of colonic macrophages to NLRP3 DSS owing to increased levels of infiltrating macrophages
activators is lost during DSS-induced colitis95, which in the colon217. One possibility that might reconcile the
might fuel inflammation. Indeed, studies in mice have also diametrically opposing roles of IL-18 is the ability of
reported a deleterious or non-existent role for NLRP3 or this cytokine to downregulate the soluble IL-22 recep-
caspase 1 and caspase 11 in colitis and colitis-associated tor called IL-22-binding protein (IL-22BP)218. IL-22BP
CRC191–194. The biological basis of these discrepancies modulates the bioavailability of IL-22, a cytokine
is unclear; however, differences in gut microbiota, diet capable of suppressing early intestinal damage but also
and/or housing conditions might affect the activity of promoting tumour development over time218 (Fig. 5a).
the inflammasome (the differences in the microbiota Inappropriate activation of the NLRP3 inflam-
are specifically discussed later). Indeed, a ketogenic masome can result in damage to the small intestine214.
diet or caloric restriction that increases the production In mice, enteropathy induced by NSAIDs, such as indo-
of the ketone body β-hydroxybutyrate from the liver methacin, can be attenuated through the use of neutral-
contributes to the inhibition of the NLRP3 inflammas- izing antibodies to IL-1β or inhibitors of the NLRP3
ome195. Similarly, omega-3 fatty acids signal through the inflammasome, including the ATP scavenger apyrase,
metabolite-sensing receptors G protein-coupled receptor the P2X purinoceptor 7 (P2X7) antagonist brilliant blue
40 (GPR40; also known as FFAR1) and GPR120 (also G and the cytoskeletal inhibitor colchicine214. These
known as FFAR4) to inhibit activation of the NLRP3 studies support the view that unwarranted activation of
inflammasome and prevent diet-induced insulin resist- the NLRP3 inflammasome is a catalyst for small intesti-
ance196. By contrast, short-chain fatty acids from dietary nal damage. The availability of more specific inhibitors
fibres bind to the metabolite-sensing receptor GPR43 of the NLRP3 inflammasome, such as MCC950 and
(also known as FFAR2) on enterocytes, stimulating CY-09 (refs219–221), enables clinical trials to explore new
potassium efflux that drives activation of the NLRP3 options for the treatment of human diseases182.
inflammasome and protection against colitis197,198. A diet
rich in cholesterol or saturated fatty acids also promotes NLRC4
activation of the NLRP3 inflammasome in mice199–203, Studies in humans have revealed that gain-of-function
whereby a cholesterol-rich diet increases inflammation mutations in the gene encoding NLRC4 are associated
and tumour burden199, but a high-fat diet reduces glu- with enterocolitis and autoinflammation222–224. For exam-
cose tolerance and insulin sensitivity200. These studies ple, a substitution mutation within the HD1 domain of
provide evidence to indicate that dietary components, the NLRC4 protein is linked to recurrent enterocolitis
in association with the gut microbiota, are critical mod- and autoinflammation in several members of the same
ulators of inflammasome activity and susceptibility to family222. A newborn baby from this family and carrying
the development of intestinal inflammation, cancer and this mutation died 23 days after birth owing to severe
metabolic syndromes. gastrointestinal complications, fever and systemic inflam-
Activation of the NLRP3 inflammasome leads to mation222. A further study identified that a heterozygous
secretion of both IL-1β and IL-18. Although injection of de novo substitution mutation in the nucleotide-binding
recombinant IL-1β into mice has been shown to mediate domain of NLRC4, potentially causing destabiliza-
protection against DSS-induced colitis204, studies have tion and autoactivation of the protein, was linked to
found variable levels of IL-1β in the colon tissue of macrophage activation syndrome, gastrointestinal
inflammasome-deficient mice in response to pathology, splenomegaly and systemic inflammation223.
DSS163,184,185,204–211. These findings would suggest that the Monocytes or macrophages carrying the aforemen-
role of IL-1β is not always coupled with inflammasome- tioned disease-associated mutations undergo spontane-
mediated protection of intestinal inflammation and ous activation of the inflammasome and pyroptosis and
colitis-associated CRC. secrete increased levels of IL-1β and IL-18 (refs222–224),
The protective role of the NLRP3 inflammasome in suggesting that unchecked inflammasome activation
colitis and CRC is attributed in part to the secretion of contributes to the molecular basis of these autoinflam-
IL-18 (Fig. 5a). Mice lacking IL-18 are highly susceptible matory conditions. Of clinical importance is that the
to the development of colitis and CRC183,212,213. Injection of aforementioned patient with the substitution mutation
recombinant IL-18 reduces intestinal inflammation and in NLRC4 responded to treatment with the recombinant
a Anti-tumour Inflammation
b
Oncogenic assault immunity
(AOM and/or
DSS)
Gut microbiota IL-18 IL-22BP IL-22
Fig. 5 | Development of intestinal inflammation and cancer is regulated by the inflammasome–microbiota axis.
a | Oncogenic assaults such as azoxymethane (AOM) and dextran sodium sulfate (DSS) cause damage, leading to the
release of danger-associated molecular patterns (DAMPs). Bacteria can invade enterocytes and introduce pathogen-
associated molecular patterns (PAMPs) into the host cell. DAMPs and PAMPs are sensed by inflammasomes183–185,187,188,204.
IL-18 promotes downregulation of soluble IL-22-binding protein (IL-22BP), which controls the ability of IL-22 to suppress
inflammation or induce tumorigenesis in the gut218. b | Nucleotide-binding domain, leucine-rich repeat-containing
protein (NLR) family CARD domain-containing protein 4 (NLRC4) and neuronal apoptosis inhibitory proteins (NAIPs)
can block cellular proliferation and tumorigenesis192,226. c | DNA-dependent protein kinase (DNA-PK) induces colorectal
tumorigenesis via activation of AKT and the transcription factor MYC205,206. This response is inhibited by absent in
melanoma 2 (AIM2)205,206. AIM2 also triggers the production of antimicrobial peptides (AMPs) in intestinal epithelial
cells to modulate the gut microbiota207,208. A similar negative regulatory role for NLRC3 has been described229.
d | NLRP6 and NLRP12 contribute to the pathogenesis of gastrointestinal infection, acute colitis and colorectal
cancer133,134,163,234–236,244,248,256,257. Question marks denote unknown mediators. GFR, growth factor receptor; mTOR,
mechanistic target of rapamycin; NLRP, NACHT, LRR and PYD domains-containing protein; PI3Ks, phosphoinositide
3-kinases; ROS, reactive oxygen species; STAT3, signal transducer and activator of transcription 3; T3SS, type 3
secretion system; TLR, Toll-like receptor.
IL-1R antagonist anakinra223. A newly identified patient response to AOM and DSS192,226. Mechanistically, NLRC4
with the heterozygous de novo mutation in NLRC4, who inhibits cellular proliferation and promotes apoptosis
also experienced enterocolitis and skin rash, responded via an undefined mechanism192, whereas NAIPs inhibit
to a combined blockade therapy targeting IL-1β and proliferation in enterocytes by blocking phosphoryl-
IL-18 (ref. 225) . These studies exemplify the success ation and activation of the transcription factor signal
of experimental targeted therapies in the treatment of transducer and activator of transcription 3 (STAT3)
inflammasome-mediated disorders. independently of NLRC4 (ref.226) (Fig. 5b). A possible
In animal studies, deletion of the genes encoding scenario is that either spontaneous activation or loss of
NLRC4 or NAIPs heightens the susceptibility of mice to function associated with the NAIP or NLRC4 signalling
the development of colitis and colitis-associated CRC in axis could upset the equilibrium of the gut ecosystem
and drive intestinal inflammation and tumorigenesis. (CPT-11), a topoisomerase I inhibitor used in the
Immunization of mice with tumour cell lines express- treatment of a variety of cancers, elicits the release of
ing flagellin — to trigger activation of the NAIP–NLRC4 self-DNA, subsequent activation of AIM2 and secre-
and TLR5 pathways – induces clearance of tumour cells tion of IL-1β and IL-18 in enterocytes, leading to intes-
by innate immune cells and tumour-specific CD4+ and tinal mucositis, late-onset diarrhoea and microbiota
CD8+ T cells227. Thus, modulation of the NAIP–NLRC4 disturbance in mice232,233. Deletion of Aim2 or phar-
and/or the IL-1–IL-18 pathways might provide a macological inhibition of the AIM2 inflammasome
promising avenue for cancer immunotherapy. in mice reduces the incidence of irinotecan-induced
diarrhoea without dampening the anticancer efficacy
AIM2 and other signalling molecules of irinotecan232.
The inflammasome-associated sensor AIM2 also con- Fur t her studies have shown t hat NLRP6
tributes to the pathogenesis of colitis and CRC205–208. (refs 137,163,234–237 ) , NLRP1b (ref. 204 ) and caspase 11
The levels of AIM2 expression in tumour tissues often (refs209–211) are important innate immune rheostats of
predict survival in patients with CRC228. Patients whose the gut, providing additional targets for experimental
tumour cells lack AIM2 expression are threefold more therapies. NLRP6 controls intestinal inflammation and
likely to die within 5 years of diagnosis than patients tumorigenesis via multiple mechanisms (discussed further
whose tumour cells retain some level of expression228. later), including modulation of IL-18 secretion163,234,235,237,
Reflecting the scenario in humans, the expression of curtailing the colonization of pro-colitogenic gut bac-
the Aim2 gene is markedly reduced in inflamed and teria163 and promoting secretion of mucus in goblet
tumour-associated tissues of mice, and mice lacking cells137,236 (Fig. 5d). NLRP6 expression in enterocytes and
AIM2 are hypersusceptible to both colitis-associated inflammatory monocytes has been reported to mediate
and spontaneous colorectal tumorigenesis205,206. AIM2 protection against DSS-induced colitis163,235,237. NLRP1b
has functions independent of the inflammasome that regulates activation of the inflammasome and secretion
drive protection against both colitis-associated and of IL-1β and IL-18 specifically in non-haematopoietic
spontaneous CRC, the latter of which arises from a cells 204, whereas caspase 11 triggers IL-1β–IL-18-
mutation in the mouse homologue of the human dependent and IL-1β–IL-18-independent func-
APC gene or from aberrant β-catenin activation205,206. tions209–211, with both sensors mediating protection
AIM2 suppresses the ability of the DNA-binding against DSS-induced colitis. A side-by-side comparison
kinase DNA-dependent protein kinase (DNA-PK) to revealed that, under isobiotic conditions with normal-
activate RAC serine/threonine-protein kinase (AKT) ized gut microbiota across genotypes, mice lacking
and transcription factor MYC proto-oncogene pro- caspase 1, but not mice lacking caspase 11, were hyper-
tein, ultimately preventing excessive proliferation of sensitive to DSS-induced colitis188. Evidence from this
stem cells in the colon205,206 (Fig. 5c). A similar negative study would seem to favour a role for canonical inflam-
regulatory effect of the orphan receptor NLRC3 on masome complexes over the non-canonical inflammas-
the AKT–mechanistic target of rapamycin (mTOR) ome in this mouse model of intestinal inflammation.
pathways activated during colitis-associated CRC Although these studies point towards a protective role
or spontaneous CRC has been described 229 (Fig. 5c). of inflammasomes in the context of intestinal inflam-
Pharmacological inhibitors of the AKT–mTOR mation and tumorigenesis, a study found that deletion
pathway are efficacious against the development of of the genes encoding the mucous layer components
colitis-associated or spontaneous CRC in mice lack- core 1-derived and core 3-derived intestinal O-glycans
ing either AIM2 or NLRC3 (refs205,206,229). The precise in mice causes colonic mucous barrier breach, spon-
identity or source of the ligand(s) activating these taneous colitis and the development of colorectal
sensors in the colon is not known. It is possible that tumours, and further deletion of the genes encoding
self-DNA liberated from the damaged intestinal bar- caspase 1 and caspase 11 reduces the propensity of
rier or DNA derived from the gut microbiota might colitis and colorectal tumorigenesis239. These findings
activate AIM2 (ref.230) (Fig. 5c). Consistent with this illustrate that in the presence of pre-existing immuno-
hypothesis, AIM2 can localize to DNA in the nucleus logical dysregulation triggering overt inflammation,
of mouse intestinal epithelial cells and bone marrow blockade of inflammasome signalling might inhibit
cells following dsDNA breaks caused by ionizing radi- an inflammatory circuitry that normally fuels dis-
ation or chemotherapeutic agents231. The generation ease progression. Development of safe and efficacious
of conditional mouse models that enable deletion modulators of AIM2, NLRP1 and NLRP6 and investi-
or re-expression of AIM2 and other related sensors gations into the bilateral interaction between inflam-
in all cells or in certain cell types once tumours are masomes and the gut ecosystem would create new
established would provide additional insights into the avenues for the treatment of intestinal inflammation
molecular and cellular mechanisms of the operation and cancer.
of these sensors. These studies would also aid further
assessment of the translational potential of targeting Inflammasome–gut microbiota axis
these sensors in cancer immunotherapy. The gut microbiota has an important role in the
Indeed, therapeutic inhibition of the AIM2 inflam- development and progression of a range of clini-
masome might, in some cases, reduce the adverse cal manifestations, especially in association with the
gastrointestinal effects associated with chemotherapy232. gastrointestinal tract. Evidence supporting this view
The chemotherapeutic and cytotoxic agent irinotecan includes the ability of postoperative diversion of the
faecal stream to prevent recurrent IBD in humans and of NLRP6 have been further characterized against the
the use of broad spectrum antibiotics in the amelior backdrop of intestinal inflammation, cancer and meta-
ation of chemical-induced colitis in mice177. The lat- bolic syndromes. NLRP6 senses the microbial metabolite
est developments in the field have suggested that taurine and drives the secretion of IL-18 and antimicro-
the inflammasome regulates gut microbiota compo- bial peptides to maintain a healthy gut microbiota and
sition, which would imply that the inflammasome– to promote antitumour immunity in mice163,248 (Fig. 5d).
gut microbiota axis might be a centrepiece in the By contrast, the microbial metabolites histamine and
development of intestinal inflammation, cancer and spermine suppress NLRP6-dependent IL-18 secretion
metabolic syndromes177,238,240,241. in mice, indicating that components of the gut microbiota
can reciprocate and fine-tune the activities of NLRP6
Colitis and cancer (ref.248) (Fig. 5d). The NLRP6–gut microbiota axis mod-
The concept of inflammasomes controlling the bio- ulates the expression of the chemokine CC-chemokine
geography of intestinal bacteria was spearheaded by ligand 5 (CCL5) and the pro-inflammatory cytokines
studies showing that mice lacking NLRP6 harbour a IL-6 and TNF and, therefore, the overall inflamma-
pro-colitogenic gut microbiota that renders these mice tory state of the intestinal tract163,237,242. In the absence
more susceptible to DSS-induced colitis than wild-type of NLRP6, the dysbiotic gut microbiota developed as a
mice163. This pro-colitogenic signature is defined by an result induces the expression of CCL5 in enterocytes;
increased relative abundance of Prevotellaceae and the CCL5, in turn, mediates the secretion of IL-6 and drives
phylum Candidatus Saccharibacteria, reduced relative proliferation of epithelial cells and tumour formation
abundance of Lactobacillus spp. and the transmissi- in mice163,242. Deletion of the gene encoding CCL5 in
ble nature of the pro-colitogenic microbiota through mice prevents colitis and colitis-associated tumorigen
means of cross-fostering and cohousing techniques163,242. esis despite these mice carrying a dysbiotic gut micro
Furthermore, NLRP6 dampens spontaneous colitis biota163,242, placing dysbiosis upstream of CCL5-mediated
in mice lacking the anti-inflammatory cytokine IL-10 immune dysregulation. Furthermore, inflammatory
by reducing levels of the mucus-degrading bacterium monocytes residing in the lamina propria promote
Akkermansia muciniphila243. Carriage of a transmissible NLRP6-dependent and IL-18-dependent secretion of
pro-colitogenic gut microbiota has also been described TNF and feature a protective role in DSS-induced colitis
in mice lacking the inflammasome components AIM2, and colitis-associated tumorigenesis237.
NLRP3, ASC, caspase 1 or caspase 11 or mice lacking the The relationship between NLRP6 and intestinal
related NLR member NLRP12 (refs163,186,205,207–209,244,245). microorganisms is further exemplified by the ability of
Similar to the findings with NLRP6, reciprocal exchange NLRP6 to respond to Toll-like receptor (TLR)-induced
of the gut microbiota between wild-type mice and autophagy in goblet cells137. This NLRP6 response medi-
mutant mice lacking AIM2 or NLRP12 elevates the ates secretion of mucus via granule exocytosis and pre-
susceptibility of wild-type mice, but reduces the sus- vents colonization of the mouse gut by C. rodentium137.
ceptibility of mutant mice, to colitis-associated tumor- Further studies have shown that sentinel goblet cells
igenesis205,244, suggesting that the colitogenic potential located at the colonic crypt entrance execute NLRP6-
and transmissibility of the gut microbiota influence dependent secretion of mucin 2 (MUC2) to expel
disease outcome. intruding bacteria found in the inner mucous layer236
Other studies, however, challenged the idea that (Fig. 5d). In the small intestine of mice, a loss of endo
NLRP6 controls the biogeography of the gut micro genous NLRP6 expression owing to stress is linked to
biota246,247. Phylogenetic analyses of wild-type mice and the development of intestinal pathology and changes
littermate-controlled mice lacking either NLRP6 or ASC to the relative abundance of Bacteroidetes, Clostridiales,
failed to reveal any difference in the composition of the Firmicutes, Lachnospiraceae, Lactobacillaceae, Rumino
gut microbiota246. Lifetime separation of wild-type mice coccaceae and Streptococcaceae249. The peroxisome
and mice lacking NLRP6 also does not seem to promote proliferator-activated receptor-γ (PPARγ) agonist
alterations of their gut microbiota profile or differential rosiglitazone, among other functions, elevates the
susceptibility to DSS-induced colitis246, casting doubt expression of NLRP6 in the small intestine and pre-
over the previously suggested role of NLRP6 and ASC vents stress-related intestinal pathology in mice164,249.
in driving dysbiosis163. This study also argued that any Whether this therapeutic effect is directly or specifically
difference associated with the gut microbiota might dependent on the ability of rosiglitazone to modulate
be entirely dependent on the facilities used to house the expression of NLRP6 remains unclear.
the animals246. Thus, food sources, gut microbiota and An additional function of NLRP6 is its ability to
sterilization techniques used by different mouse vivar- suppress activation of the canonical NF-κB and MAPK
ium across the globe can influence the activation status pathways in response to infection by the gastrointesti-
of inflammasomes. Future experimental approaches nal pathogens L. monocytogenes, S. Typhimurium and
should be more tightly controlled to examine the role E. coli133 (Fig. 5d). This immunomodulatory pathway
of the gut microbiota in any disease context, including operates in the presence or absence of an altered gut
the use of littermate controls and careful evaluation of microbiota composition133, highlighting that certain
cage-to-cage and room-to-room effects and the overall biological functions of NLRP6 can clearly be uncoupled
sterility of the facility. from the gut microbiota and that previous studies iden-
Despite the uncertainty surrounding the nexus between tifying a connection with the gut microbiota should be
NLRP6 and the gut microbiota, the mechanistic functions interpreted with care and revisited.
1. Belkaid, Y. & Hand, T. W. Role of the microbiota in 28. Zhao, Y. et al. The NLRC4 inflammasome receptors for 54. Park, Y. H., Wood, G., Kastner, D. L. & Chae, J. J.
immunity and inflammation. Cell 157, 121–141 bacterial flagellin and type III secretion apparatus. Pyrin inflammasome activation and RhoA signaling
(2014). Nature 477, 596–600 (2011). in the autoinflammatory diseases FMF and HIDS.
2. Martinon, F., Burns, K. & Tschopp, J. The 29. Kortmann, J., Brubaker, S. W. & Monack, D. M. Nat. Immunol. 17, 914–921 (2016).
inflammasome: a molecular platform triggering Cutting edge: inflammasome activation in primary 55. Broz, P. et al. Redundant roles for inflammasome
activation of inflammatory caspases and processing human macrophages is dependent on flagellin. receptors NLRP3 and NLRC4 in host defense
of proIL-beta. Mol. Cell 10, 417–426 (2002). J. Immunol. 195, 815–819 (2015). against Salmonella. J. Exp. Med. 207, 1745–1755
3. Schroder, K. & Tschopp, J. The inflammasomes. 30. Reyes Ruiz, V. M. et al. Broad detection of bacterial (2010).
Cell 140, 821–832 (2010). type III secretion system and flagellin proteins by the 56. Broz, P., von Moltke, J., Jones, J. W., Vance, R. E. &
4. Lamkanfi, M. & Dixit, V. M. Mechanisms and functions human NAIP/NLRC4 inflammasome. Proc. Natl Acad. Monack, D. M. Differential requirement for Caspase-1
of inflammasomes. Cell 157, 1013–1022 (2014). Sci. USA 114, 13242–13247 (2017). autoproteolysis in pathogen-induced cell death and
5. Man, S. M. & Kanneganti, T. D. Converging roles of 31. Kofoed, E. M. & Vance, R. E. Innate immune cytokine processing. Cell Host Microbe 8, 471–483
caspases in inflammasome activation, cell death and recognition of bacterial ligands by NAIPs determines (2010).
innate immunity. Nat. Rev. Immunol. 16, 7–21 (2016). inflammasome specificity. Nature 477, 592–595 57. Fernandes-Alnemri, T. et al. The pyroptosome:
6. Rathinam, V. A. & Fitzgerald, K. A. Inflammasome (2011). a supramolecular assembly of ASC dimers mediating
complexes: emerging mechanisms and effector 32. Rayamajhi, M., Zak, D. E., Chavarria-Smith, J., inflammatory cell death via caspase-1 activation.
functions. Cell 165, 792–800 (2016). Vance, R. E. & Miao, E. A. Cutting edge: mouse NAIP1 Cell Death Differ. 14, 1590–1604 (2007).
7. Broz, P. & Dixit, V. M. Inflammasomes: mechanism detects the type III secretion system needle protein. 58. Man, S. M. et al. Salmonella infection induces
of assembly, regulation and signalling. Nat. Rev. J. Immunol. 191, 3986–3989 (2013). recruitment of caspase-8 to the inflammasome to
Immunol. 16, 407–420 (2016). 33. Rauch, I. et al. NAIP proteins are required for cytosolic modulate IL-1beta production. J. Immunol. 191,
8. Conos, S. A., Lawlor, K. E., Vaux, D. L., Vince, J. E. & detection of specific bacterial ligands in vivo. J. Exp. 5239–5246 (2013).
Lindqvist, L. M. Cell death is not essential for caspase- Med. 213, 657–665 (2016). 59. Franklin, B. S. et al. The adaptor ASC has extracellular
1-mediated interleukin-1beta activation and secretion. 34. Zhao, Y. et al. Genetic functions of the NAIP family of and ‘prionoid’ activities that propagate inflammation.
Cell Death Differ. 23, 1827–1838 (2016). inflammasome receptors for bacterial ligands in mice. Nat. Immunol. 15, 727–737 (2014).
9. Evavold, C. L. et al. The pore-forming protein J. Exp. Med. 213, 647–656 (2016). 60. Lu, A. et al. Unified polymerization mechanism
gasdermin D regulates interleukin-1 secretion from 35. Halff, E. F. et al. Formation and structure of a NAIP5- for the assembly of ASC-dependent inflammasomes.
living macrophages. Immunity 48, 35–44 (2018). NLRC4 inflammasome induced by direct interactions Cell 156, 1193–1206 (2014).
10. Heilig, R. et al. The Gasdermin-D pore acts as a with conserved N− and C-terminal regions of flagellin. 61. Cai, X. et al. Prion-like polymerization underlies signal
conduit for IL-1beta secretion in mice. Eur. J. Immunol. J. Biol. Chem. 287, 38460–38472 (2012). transduction in antiviral immune defense and
48, 584–592 (2018). 36. Hu, Z. et al. Structural and biochemical basis for inflammasome activation. Cell 156, 1207–1222
11. Szabo, G. & Petrasek, J. Inflammasome activation induced self-propagation of NLRC4. Science 350, (2014).
and function in liver disease. Nat. Rev. Gastroenterol. 399–404 (2015). 62. Baroja-Mazo, A. et al. The NLRP3 inflammasome is
Hepatol. 12, 387–400 (2015). 37. D’Osualdo, A. et al. CARD8 and NLRP1 undergo released as a particulate danger signal that amplifies
12. Guo, H., Callaway, J. B. & Ting, J. P. Inflammasomes: autoproteolytic processing through a ZU5-like domain. the inflammatory response. Nat. Immunol. 15,
mechanism of action, role in disease, and PloS one 6, e27396 (2011). 738–748 (2014).
therapeutics. Nat. Med. 21, 677–687 (2015). 38. Frew, B. C., Joag, V. R. & Mogridge, J. Proteolytic 63. Man, S. M., Karki, R. & Kanneganti, T. D. Molecular
13. Levy, M., Kolodziejczyk, A. A., Thaiss, C. A. & Elinav, E. processing of Nlrp1b is required for inflammasome mechanisms and functions of pyroptosis, inflammatory
Dysbiosis and the immune system. Nat. Rev. Immunol. activity. PLoS Pathog. 8, e1002659 (2012). caspases and inflammasomes in infectious diseases.
17, 219–232 (2017). 39. Finger, J. N. et al. Autolytic proteolysis within the Immunol. Rev. 277, 61–75 (2017).
14. Man, S. M. & Kanneganti, T. D. Regulation of function to find domain (FIIND) is required for 64. Poyet, J. L. et al. Identification of Ipaf, a human
inflammasome activation. Immunol. Rev. 265, 6–21 NLRP1 inflammasome activity. J. Biol. Chem. 287, caspase-1-activating protein related to Apaf-1. J. Biol.
(2015). 25030–25037 (2012). Chem. 276, 28309–28313 (2001).
15. Latz, E., Xiao, T. S. & Stutz, A. Activation and 40. Chavarria-Smith, J., Mitchell, P. S., Ho, A. M., 65. Van Opdenbosch, N. et al. Activation of the NLRP1b
regulation of the inflammasomes. Nat. Rev. Immunol. Daugherty, M. D. & Vance, R. E. Functional and inflammasome independently of ASC-mediated
13, 397–411 (2013). evolutionary analyses identify proteolysis as a general caspase-1 autoproteolysis and speck formation.
16. Kayagaki, N. et al. Non-canonical inflammasome mechanism for NLRP1 inflammasome activation. Nat. Commun. 5, 3209 (2014).
activation targets caspase-11. Nature 479, 117–121 PLoS Pathog. 12, e1006052 (2016). 66. Guey, B., Bodnar, M., Manie, S. N., Tardivel, A. &
(2011). 41. Levinsohn, J. L. et al. Anthrax lethal factor cleavage of Petrilli, V. Caspase-1 autoproteolysis is differentially
This study describes the existence of a Nlrp1 is required for activation of the inflammasome. required for NLRP1b and NLRP3 inflammasome
non-canonical inflammasome. PLoS Pathog. 8, e1002638 (2012). function. Proc. Natl Acad. Sci. USA 111,
17. Liston, A. & Masters, S. L. Homeostasis-altering 42. Perregaux, D. & Gabel, C. A. Interleukin-1 beta 17254–17259 (2014).
molecular processes as mechanisms of inflammasome maturation and release in response to ATP and nigericin. 67. Kostura, M. J. et al. Identification of a monocyte
activation. Nat. Rev. Immunol. 17, 208–214 (2017). Evidence that potassium depletion mediated by these specific pre-interleukin 1 beta convertase activity.
18. Hornung, V. et al. AIM2 recognizes cytosolic dsDNA agents is a necessary and common feature of their Proc. Natl Acad. Sci. USA 86, 5227–5231 (1989).
and forms a caspase-1-activating inflammasome with activity. J. Biol. Chem. 269, 15195–15203 (1994). 68. Black, R. A., Kronheim, S. R. & Sleath, P. R. Activation
ASC. Nature 458, 514–518 (2009). 43. Muñoz-Planillo, R. et al. K(+) efflux is the common of interleukin-1 beta by a co-induced protease.
19. Fernandes-Alnemri, T., Yu, J. W., Datta, P., Wu, J. & trigger of NLRP3 inflammasome activation by FEBS Lett. 247, 386–390 (1989).
Alnemri, E. S. AIM2 activates the inflammasome and bacterial toxins and particulate matter. Immunity 38, 69. Thornberry, N. A. et al. A novel heterodimeric cysteine
cell death in response to cytoplasmic DNA. Nature 1142–1153 (2013). protease is required for interleukin-1 beta processing
458, 509–513 (2009). 44. Petrilli, V. et al. Activation of the NALP3 in monocytes. Nature 356, 768–774 (1992).
20. Burckstummer, T. et al. An orthogonal inflammasome is triggered by low intracellular 70. Ghayur, T. et al. Caspase-1 processes IFN-gamma-
proteomic-genomic screen identifies AIM2 as a potassium concentration. Cell Death Differ. 14, inducing factor and regulates LPS-induced IFN-gamma
cytoplasmic DNA sensor for the inflammasome. 1583–1589 (2007). production. Nature 386, 619–623 (1997).
Nat. Immunol. 10, 266–272 (2009). 45. Hornung, V. et al. Silica crystals and aluminum salts 71. Vande Walle, L. & Lamkanfi, M. Pyroptosis. Curr. Biol.
21. Roberts, T. L. et al. HIN-200 proteins regulate caspase mediate NALP-3 inflammasome activation via 26, R568–R572 (2016).
activation in response to foreign cytoplasmic DNA. phagosomal destabilization. Nat. Immunol. 9, 72. Aglietti, R. A. & Dueber, E. C. Recent insights into the
Science 323, 1057–1060 (2009). 847–856 (2008). molecular mechanisms underlying pyroptosis and
References 18–21 show that AIM2 is a cytosolic 46. Zhou, R., Yazdi, A. S., Menu, P. & Tschopp, J. A role gasdermin family functions. Trends Immunol. 38,
inflammasome sensor of double-stranded DNA. for mitochondria in NLRP3 inflammasome activation. 261–271 (2017).
22. Shi, J. et al. Inflammatory caspases are innate Nature 469, 221–225 (2011). 73. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D
immune receptors for intracellular LPS. Nature 514, 47. Shimada, K. et al. Oxidized mitochondrial DNA for non-canonical inflammasome signaling. Nature
187–192 (2014). activates the NLRP3 inflammasome during apoptosis. 526, 666–671 (2015).
23. Mariathasan, S. et al. Differential activation of the Immunity 36, 401–414 (2012). 74. Shi, J. et al. Cleavage of GSDMD by inflammatory
inflammasome by caspase-1 adaptors ASC and Ipaf. 48. Iyer, S. S. et al. Mitochondrial cardiolipin is required caspases determines pyroptotic cell death. Nature
Nature 430, 213–218 (2004). for Nlrp3 inflammasome activation. Immunity 39, 526, 660–665 (2015).
This study provides the first genetic evidence 311–323 (2013). References 73 and 74 demonstrate that
for the existence of an NLRC4 inflammasome 49. Groß, C. J. et al. K+ efflux-independent NLRP3 gasdermin D is a substrate of inflammatory
(previously known as the ICE-protease activating Inflammasome activation by small molecules targeting caspases that upon cleavage converts to a
factor (IPAF) inflammasome). mitochondria. Immunity 45, 761–773 (2016). pro-pyroptotic executor mediating cell lysis.
24. Franchi, L. et al. Cytosolic flagellin requires Ipaf for 50. Murakami, T. et al. Critical role for calcium mobilization 75. He, W. T. et al. Gasdermin D is an executor of
activation of caspase-1 and interleukin 1beta in in activation of the NLRP3 inflammasome. Proc. Natl pyroptosis and required for interleukin-1beta
salmonella-infected macrophages. Nat. Immunol. 7, Acad. Sci. USA 109, 11282–11287 (2012). secretion. Cell Res. 25, 1285–1298 (2015).
576–582 (2006). 51. Xu, H. et al. Innate immune sensing of bacterial 76. Ding, J. et al. Pore-forming activity and structural
25. Miao, E. A. et al. Cytoplasmic flagellin activates modifications of Rho GTPases by the Pyrin autoinhibition of the gasdermin family. Nature 535,
caspase-1 and secretion of interleukin 1beta via Ipaf. inflammasome. Nature 513, 237–241 (2014). 111–116 (2016).
Nat. Immunol. 7, 569–575 (2006). 52. Gavrilin, M. A. et al. Activation of the pyrin 77. Aglietti, R. A. et al. GsdmD p30 elicited by
26. Miao, E. A. et al. Innate immune detection of the type III inflammasome by intracellular Burkholderia caspase-11 during pyroptosis forms pores in
secretion apparatus through the NLRC4 inflammasome. cenocepacia. J. Immunol. 188, 3469–3477 (2012). membranes. Proc. Natl Acad. Sci. USA 113,
Proc. Natl Acad. Sci. USA 107, 3076–3080 (2010). 53. Masters, S. L. et al. Familial autoinflammation 7858–7863 (2016).
27. Amer, A. et al. Regulation of Legionella phagosome with neutrophilic dermatosis reveals a regulatory 78. Liu, X. et al. Inflammasome-activated gasdermin D
maturation and infection through flagellin and host mechanism of pyrin activation. Sci. Transl Med. 8, causes pyroptosis by forming membrane pores.
Ipaf. J. Biol. Chem. 281, 35217–35223 (2006). 332ra345 (2016). Nature 535, 153–158 (2016).
79. Sborgi, L. et al. GSDMD membrane pore formation 104. Knodler, L. A. et al. Noncanonical inflammasome bacteriolysis in the macrophage cytosol. Cell Host
constitutes the mechanism of pyroptotic cell death. activation of caspase-4/caspase-11 mediates epithelial Microbe 7, 412–419 (2010).
EMBO J. 35, 1766–1778 (2016). defenses against enteric bacterial pathogens. 126. Warren, S. E. et al. Cutting edge: Cytosolic bacterial
80. Chen, X. et al. Pyroptosis is driven by non-selective Cell Host Microbe 16, 249–256 (2014). DNA activates the inflammasome via Aim2. J. Immunol.
gasdermin-D pore and its morphology is different from References 93, 94 and 104 highlight an expulsion 185, 818–821 (2010).
MLKL channel-mediated necroptosis. Cell Res. 26, mechanism induced by the inflammasome to 127. Tsuchiya, K. et al. Involvement of absent in melanoma
1007–1020 (2016). remove an entire infected host cell from the 2 in inflammasome activation in macrophages infected
81. Joosten, L. A., Netea, M. G. & Dinarello, C. A. intestinal epithelium. with Listeria monocytogenes. J. Immunol. 185,
Interleukin-1beta in innate inflammation, autophagy 105. Man, S. M. et al. Actin polymerization as a key 1186–1195 (2010).
and immunity. Semin. Immunol. 25, 416–424 (2013). innate immune effector mechanism to control 128. Meixenberger, K. et al. Listeria
82. Dinarello, C. A., Novick, D., Kim, S. & Kaplanski, G. Salmonella infection. Proc. Natl Acad. Sci. USA monocytogenes-infected human peripheral blood
Interleukin-18 and IL-18 binding protein. Front. 111, 17588–17593 (2014). mononuclear cells produce IL-1beta, depending on
Immunol. 4, 289 (2013). 106. Chen, K. W. et al. The neutrophil NLRC4 inflammasome listeriolysin O and NLRP3. J. Immunol. 184,
83. Navaneethan, U. & Giannella, R. A. Mechanisms of selectively promotes IL-1beta maturation without 922–930 (2010).
infectious diarrhea. Nat. Clin. Pract. Gastroenterol. pyroptosis during acute Salmonella challenge. Cell Rep. 129. Khare, S. et al. An NLRP7-containing inflammasome
Hepatol. 5, 637–647 (2008). 8, 570–582 (2014). mediates recognition of microbial lipopeptides in
84. Miao, E. A. et al. Caspase-1-induced pyroptosis is an 107. O’Connor, A., O’Morain, C. A. & Ford, A. C. human macrophages. Immunity 36, 464–476 (2012).
innate immune effector mechanism against intracellular Population screening and treatment of Helicobacter 130. Akhter, A. et al. Caspase-11 promotes the fusion of
bacteria. Nat. Immunol. 11, 1136–1142 (2010). pylori infection. Nat. Rev. Gastroenterol. Hepatol. 14, phagosomes harboring pathogenic bacteria with
85. Franchi, L. et al. NLRC4-driven production of IL-1beta 230–240 (2017). lysosomes by modulating actin polymerization.
discriminates between pathogenic and commensal 108. Matusiak, M. et al. Flagellin-induced NLRC4 Immunity 37, 35–47 (2012).
bacteria and promotes host intestinal defense. phosphorylation primes the inflammasome for 131. Thurston, T. L. et al. Growth inhibition of cytosolic
Nat. Immunol. 13, 449–456 (2012). activation by NAIP5. Proc. Natl Acad. Sci. USA 112, Salmonella by caspase-1 and caspase-11 precedes
86. Kupz, A. et al. NLRC4 inflammasomes in dendritic 1541–1546 (2015). host cell death. Nat. Commun. 7, 13292 (2016).
cells regulate noncognate effector function by 109. Koch, K. N. et al. Helicobacter urease-induced 132. Sokolovska, A. et al. Activation of caspase-1 by the
memory CD8(+) T cells. Nat. Immunol. 13, 162–169 activation of the TLR2/NLRP3/IL-18 axis protects NLRP3 inflammasome regulates the NADPH oxidase
(2012). against asthma. J. Clin. Invest. 125, 3297–3302 NOX2 to control phagosome function. Nat. Immunol.
87. Man S. M. et al. Inflammasome activation causes (2015). 14, 543–553 (2013).
dual recruitment of NLRC4 and NLRP3 to the same 110. Perez-Figueroa, E. et al. Activation of NLRP3 133. Anand, P. K. et al. NLRP6 negatively regulates innate
macromolecular complex. Proc. Natl. Acad. Sci. USA inflammasome in human neutrophils by Helicobacter immunity and host defence against bacterial
111, 7403–7408 (2014). pylori infection. Innate Immun. 22, 103–112 (2016). pathogens. Nature 488, 389–393 (2012).
88. Suzuki, S. et al. Shigella type III secretion protein MxiI 111. Li, X. et al. Helicobacter pylori induces IL-1beta and 134. Zaki, M. H., Man, S. M., Vogel, P., Lamkanfi, M. &
is recognized by Naip2 to induce Nlrc4 inflammasome IL-18 production in human monocytic cell line through Kanneganti, T. D. Salmonella exploits NLRP12-
activation independently of Pkcdelta. PLoS Pathog. activation of NLRP3 inflammasome via ROS signaling dependent innate immune signaling to suppress host
10, e1003926 (2014). pathway. Pathog. Dis. 73, ftu024 (2015). defenses during infection. Proc. Natl Acad. Sci. USA
89. Suzuki, S. et al. Shigella IpaH7.8 E3 ubiquitin ligase 112. Semper, R. P. et al. Helicobacter pylori-induced 111, 385–390 (2014).
targets glomulin and activates inflammasomes to IL-1beta secretion in innate immune cells is regulated 135. Allen, I. C. et al. Characterization of NLRP12 during
demolish macrophages. Proc. Natl Acad. Sci. USA by the NLRP3 inflammasome and requires the cag the in vivo host immune response to Klebsiella
111, E4254–E4263 (2014). pathogenicity island. J. Immunol. 193, 3566–3576 pneumoniae and Mycobacterium tuberculosis.
90. Wu, J., Fernandes-Alnemri, T. & Alnemri, E. S. (2014). PLOS ONE 8, e60842 (2013).
Involvement of the AIM2, NLRC4, and NLRP3 113. Kim, D. J., Park, J. H., Franchi, L., Backert, S. & 136. Silveira, T. N. et al. NLRP12 negatively regulates
inflammasomes in caspase-1 activation by Listeria Nunez, G. The Cag pathogenicity island and proinflammatory cytokine production and host
monocytogenes. J. Clin. Immunol. 30, 693–702 interaction between TLR2/NOD2 and NLRP3 regulate defense against Brucella abortus. Eur. J. Immunol. 47,
(2010). IL-1beta production in Helicobacter pylori infected 51–59 (2017).
91. Liu, Z. et al. Role of inflammasomes in host defense dendritic cells. Eur. J. Immunol. 43, 2650–2658 137. Wlodarska, M. et al. NLRP6 inflammasome
against Citrobacter rodentium infection. J. Biol. Chem. (2013). orchestrates the colonic host-microbial interface by
287, 16955–16964 (2012). 114. Tu, S. et al. Overexpression of interleukin-1beta regulating goblet cell mucus secretion. Cell 156,
92. Nordlander, S., Pott, J. & Maloy, K. J. NLRC4 induces gastric inflammation and cancer and mobilizes 1045–1059 (2014).
expression in intestinal epithelial cells mediates myeloid-derived suppressor cells in mice. Cancer Cell 138. Vladimer, G. I. et al. The NLRP12 inflammasome
protection against an enteric pathogen. Mucosal 14, 408–419 (2008). recognizes Yersinia pestis. Immunity 37, 96–107
Immunol. 7, 775–785 (2014). 115. Castano-Rodriguez, N., Kaakoush, N. O., Goh, K. L., (2012).
93. Sellin, M. E. et al. Epithelium-Intrinsic NAIP/NLRC4 Fock, K. M. & Mitchell, H. M. The NOD-like receptor 139. Zamoshnikova, A. et al. NLRP12 is a neutrophil-specific,
inflammasome drives infected enterocyte expulsion to signalling pathway in Helicobacter pylori infection and negative regulator of in vitro cell migration but does
restrict Salmonella replication in the intestinal related gastric cancer: a case-control study and gene not modulate LPS- or infection-induced NF-kappaB
mucosa. Cell Host Microbe 16, 237–248 (2014). expression analyses. PLOS ONE 9, e98899 (2014). or ERK signalling. Immunobiology 221, 341–346
94. Rauch, I. et al. NAIP-NLRC4 inflammasomes 116. Li, S. et al. MiR-22 sustains NLRP3 expression and (2016).
coordinate intestinal epithelial cell expulsion with attenuates H. pylori-induced gastric carcinogenesis. 140. Ulland, T. K. et al. Nlrp12 mutation causes C57BL/6J
eicosanoid and IL-18 release via activation of Oncogene 37, 884–896 (2018). strain-specific defect in neutrophil recruitment.
caspase-1 and -8. Immunity 46, 649–659 (2017). 117. Martin, J. S., Monaghan, T. M. & Wilcox, M. H. Nat. Commun. 7, 13180 (2016).
95. Filardy, A. A., He, J., Bennink, J., Yewdell, J. & Clostridium difficile infection: epidemiology, diagnosis 141. Sarkar, A. et al. Caspase-1 regulates Escherichia coli
Kelsall, B. L. Posttranscriptional control of NLRP3 and understanding transmission. Nat. Rev. sepsis and splenic B cell apoptosis independently of
inflammasome activation in colonic macrophages. Gastroenterol. Hepatol. 13, 206–216 (2016). interleukin-1beta and interleukin-18. Am. J. Respir.
Mucosal Immunol. 9, 850–858 (2016). 118. Ng, J. et al. Clostridium difficile toxin-induced Crit. Care Med. 174, 1003–1010 (2006).
96. Ngo, C. C. & Man, S. M. Mechanisms and functions inflammation and intestinal injury are mediated by the 142. Wang, S. et al. Murine caspase-11, an ICE-interacting
of guanylate-binding proteins and related inflammasome. Gastroenterology 139, 542–552 protease, is essential for the activation of ICE. Cell 92,
interferon-inducible GTPases: roles in intracellular lysis (2010). 501–509 (1998).
of pathogens. Cell. Microbiol. 19, e12791 (2017). 119. Van Gorp, H. et al. Familial Mediterranean fever 143. Li, P. et al. Mice deficient in IL-1 beta-converting
97. Kayagaki, N. et al. Noncanonical inflammasome mutations lift the obligatory requirement for enzyme are defective in production of mature IL-1
activation by intracellular LPS independent of TLR4. microtubules in Pyrin inflammasome activation. beta and resistant to endotoxic shock. Cell 80,
Science 341, 1246–1249 (2013). Proc. Natl Acad. Sci. USA 113, 14384–14389 (2016). 401–411 (1995).
98. Hagar, J. A., Powell, D. A., Aachoui, Y., Ernst, R. K. & 120. Gao, W., Yang, J., Liu, W., Wang, Y. & Shao, F. 144. Man, S. M. et al. Differential roles of caspase-1 and
Miao, E. A. Cytoplasmic LPS activates caspase-11: Site-specific phosphorylation and microtubule dynamics caspase-11 in infection and inflammation. Sci. Rep. 7,
implications in TLR4-independent endotoxic shock. control Pyrin inflammasome activation. Proc. Natl 45126 (2017).
Science 341, 1250–1253 (2013). Acad. Sci. USA 113, E4857–E4866 (2016). 145. Lee, S. et al. NLRP3 inflammasome deficiency protects
99. Aachoui, Y. et al. Caspase-11 protects against bacteria 121. Seo, S. U. et al. Distinct commensals induce against microbial sepsis via increased lipoxin B4
that escape the vacuole. Science 339, 975–978 interleukin-1beta via NLRP3 inflammasome in synthesis. Am. J. Respir. Crit. Care Med. 196,
(2013). inflammatory monocytes to promote intestinal 713–726 (2017).
100. Aachoui, Y. et al. Canonical inflammasomes drive inflammation in response to injury. Immunity 42, 146. Jin, L., Batra, S. & Jeyaseelan, S. Deletion of Nlrp3
IFN-gamma to prime caspase-11 in defense against a 744–755 (2015). augments survival during polymicrobial sepsis by
cytosol-invasive bacterium. Cell Host Microbe 18, 122. Mariathasan, S. et al. Cryopyrin activates the decreasing autophagy and enhancing phagocytosis.
320–332 (2015). inflammasome in response to toxins and ATP. Nature J. Immunol. 198, 1253–1262 (2017).
101. Kaparakis-Liaskos, M. & Ferrero, R. L. Immune 440, 228–232 (2006). 147. Hao, H. et al. Farnesoid X receptor regulation of the
modulation by bacterial outer membrane vesicles. 123. Munoz-Planillo, R., Franchi, L., Miller, L. S. & NLRP3 Inflammasome underlies cholestasis-associated
Nat. Rev. Immunol. 15, 375–387 (2015). Nunez, G. A critical role for hemolysins and bacterial sepsis. Cell. Metab. 25, 856–867 (2017).
102. Vanaja, S. K. et al. Bacterial outer membrane vesicles lipoproteins in Staphylococcus aureus-induced 148. Guo, C. et al. Bile acids control inflammation and
mediate cytosolic localization of LPS and Caspase-11 activation of the Nlrp3 inflammasome. J. Immunol. metabolic disorder through inhibition of NLRP3
activation. Cell 165, 1106–1119 (2016). 183, 3942–3948 (2009). inflammasome. Immunity 45, 802–816 (2016).
103. Jorgensen, I., Zhang, Y., Krantz, B. A. & Miao, E. A. 124. Kim, S. et al. Listeria monocytogenes is sensed by the 149. von Moltke, J. et al. Rapid induction of inflammatory
Pyroptosis triggers pore-induced intracellular traps NLRP3 and AIM2 inflammasome. Eur. J. Immunol. 40, lipid mediators by the inflammasome in vivo.
(PITs) that capture bacteria and lead to their clearance 1545–1551 (2010). Nature 490, 107–111 (2012).
by efferocytosis. J. Exp. Med. 213, 2113–2128 125. Sauer, J. D. et al. Listeria monocytogenes triggers 150. Ayres, J. S., Trinidad, N. J. & Vance, R. E. Lethal
(2016). AIM2-mediated pyroptosis upon infrequent inflammasome activation by a multidrug-resistant
pathobiont upon antibiotic disruption of the 176. Zmora, N., Levy, M., Pevsner-Fishcer, M. & Elinav, E. 200. Wen, H. et al. Fatty acid-induced NLRP3-ASC
microbiota. Nat. Med. 18, 799–806 (2012). Inflammasomes and intestinal inflammation. inflammasome activation interferes with insulin
151. Schieber, A. M. et al. Disease tolerance mediated by Mucosal Immunol. 10, 865–883 (2017). signaling. Nat. Immunol. 12, 408–415 (2011).
microbiome E. coli involves inflammasome and IGF-1 177. Man, S. M., Kaakoush, N. O. & Mitchell, H. M. 201. Robblee, M. M. et al. Saturated fatty acids engage
signaling. Science 350, 558–563 (2015). The role of bacteria and pattern-recognition receptors an IRE1alpha-dependent pathway to activate the
152. Christensen, M. L. Human viral gastroenteritis. in Crohn’s disease. Nat. Rev. Gastroenterol. Hepatol. NLRP3 inflammasome in myeloid cells. Cell Rep. 14,
Clin. Microbiol. Rev. 2, 51–89 (1989). 8, 152–168 (2011). 2611–2623 (2016).
153. Shayakhmetov, D. M., Li, Z. Y., Ni, S. & Lieber, A. 178. Villani, A. C. et al. Common variants in the NLRP3 202. Progatzky, F. et al. Dietary cholesterol directly induces
Interference with the IL-1-signaling pathway improves region contribute to Crohn’s disease susceptibility. acute inflammasome-dependent intestinal inflammation.
the toxicity profile of systemically applied adenovirus Nat. Genet. 41, 71–76 (2009). Nat. Commun. 5, 5864 (2014).
vectors. J. Immunol. 174, 7310–7319 (2005). 179. Lewis, G. J. et al. Genetic association between NLRP3 203. Zhao, S. et al. Deoxycholic acid triggers NLRP3
154. Muruve, D. A. et al. The inflammasome recognizes variants and Crohn’s disease does not replicate in a inflammasome activation and aggravates DSS-induced
cytosolic microbial and host DNA and triggers an large UK panel. Inflamm. Bowel Dis. 17, 1387–1391 colitis in mice. Front. Immunol. 7, 536 (2016).
innate immune response. Nature 452, 103–107 (2011). 204. Williams, T. M. et al. The NLRP1 inflammasome
(2008). 180. Ungerback, J. et al. Genetic variation and alterations attenuates colitis and colitis-associated tumorigenesis.
155. Barlan, A. U., Griffin, T. M., McGuire, K. A. & of genes involved in NFkappaB/TNFAIP3- and NLRP3- J. Immunol. 194, 3369–3380 (2015).
Wiethoff, C. M. Adenovirus membrane penetration inflammasome signaling affect susceptibility and 205. Man, S. M. et al. Critical role for the DNA sensor
activates the NLRP3 inflammasome. J. Virol. 85, outcome of colorectal cancer. Carcinogenesis 33, AIM2 in stem cell proliferation and cancer. Cell 162,
146–155 (2011). 2126–2134 (2012). 45–58 (2015).
156. Teigler, J. E., Kagan, J. C. & Barouch, D. H. 181. Liu, R. et al. Expression profile of innate immune 206. Wilson, J. E. et al. Inflammasome-independent role of
Late endosomal trafficking of alternative serotype receptors, NLRs and AIM2, in human colorectal AIM2 in suppressing colon tumorigenesis via DNA-PK
adenovirus vaccine vectors augments antiviral innate cancer: correlation with cancer stages and and Akt. Nat. Med. 21, 906–913 (2015).
immunity. J. Virol. 88, 10354–10363 (2014). inflammasome components. Oncotarget 6, References 205 and 206 identify a role for the
157. Solomon, T. et al. Virology, epidemiology, pathogenesis, 33456–33469 (2015). DNA sensor AIM2 in preventing colorectal cancer
and control of enterovirus 71. Lancet Infect. Dis. 10, 182. Karki, R., Man, S. M. & Kanneganti, T. D. in mice.
778–790 (2010). Inflammasomes and cancer. Cancer Immunol. Res. 5, 207. Hu, S. et al. The DNA Sensor AIM2 Maintains
158. Wang, H. et al. Reciprocal regulation between 94–99 (2017). Intestinal homeostasis via regulation of epithelial
Enterovirus 71 and the NLRP3 inflammasome. 183. Zaki, M. H., Vogel, P., Body-Malapel, M., Lamkanfi, M. antimicrobial host defense. Cell Rep. 13, 1922–1936
Cell Rep. 12, 42–48 (2015). & Kanneganti, T. D. IL-18 production downstream of (2015).
159. Li, Z. et al. Interleukin-18 protects mice from Enterovirus the Nlrp3 inflammasome confers protection against 208. Ratsimandresy, R. A., Indramohan, M., Dorfleutner, A.
71 infection. Cytokine 96, 132–137 (2017). colorectal tumor formation. J. Immunol. 185, & Stehlik, C. The AIM2 inflammasome is a central
160. Operario, D. J. et al. Etiology of severe acute watery 4912–4920 (2010). regulator of intestinal homeostasis through the
diarrhea in children in the global Rotavirus Surveillance 184. Allen, I. C. et al. The NLRP3 inflammasome functions IL-18/IL-22/STAT3 pathway. Cell. Mol. Immunol. 14,
Network using quantitative polymerase chain reaction. as a negative regulator of tumorigenesis during 127–142 (2017).
J. Infect. Dis. 216, 220–227 (2017). colitis-associated cancer. J. Exp. Med. 207, 209. Demon, D. et al. Caspase-11 is expressed in the
161. Zhu, S. et al. Nlrp9b inflammasome restricts rotavirus 1045–1056 (2010). colonic mucosa and protects against dextran sodium
infection in intestinal epithelial cells. Nature 546, 185. Zaki, M. H. et al. The NLRP3 inflammasome protects sulfate-induced colitis. Mucosal Immunol. 7,
667–670 (2017). against loss of epithelial integrity and mortality 1480–1491 (2014).
This study reports the existence of an NLRP9b during experimental colitis. Immunity 32, 379–391 210. Williams, T. M. et al. Caspase-11 attenuates
inflammasome complex, which is expressed (2010). gastrointestinal inflammation and experimental colitis
specifically in mouse intestinal cells and mediates 186. Hirota, S. A. et al. NLRP3 inflammasome plays pathogenesis. Am. J. Physiol. Gastrointest. Liver
cytosolic recognition of rotavirus. a key role in the regulation of intestinal homeostasis. Physiol. 308, G139–G150 (2014).
162. Zhang, B. et al. Prevention and cure of rotavirus Inflamm. Bowel Dis. 17, 1359–1372 (2011). 211. Oficjalska, K. et al. Protective role for caspase-11
infection via TLR5/NLRC4-mediated production of 187. Dupaul-Chicoine, J. et al. Control of intestinal during acute experimental murine colitis. J. Immunol.
IL-22 and IL-18. Science 346, 861–865 (2014). homeostasis, colitis, and colitis-associated colorectal 194, 1252–1260 (2014).
163. Elinav, E. et al. NLRP6 inflammasome regulates cancer by the inflammatory caspases. Immunity 32, 212. Takagi, H. et al. Contrasting action of IL-12 and IL-18
colonic microbial ecology and risk for colitis. Cell 145, 367–378 (2010). in the development of dextran sodium sulphate colitis
745–757 (2011). 188. Blazejewski, A. J. et al. Microbiota normalization in mice. Scand. J. Gastroenterol. 38, 837–844
164. Kempster, S. L. et al. Developmental control of the reveals that canonical caspase-1 activation exacerbates (2003).
Nlrp6 inflammasome and a substrate, IL-18, in chemically induced intestinal inflammation. Cell Rep. 213. Salcedo, R. et al. MyD88-mediated signaling prevents
mammalian intestine. Am. J. Physiol. Gastrointest. 19, 2319–2330 (2017). development of adenocarcinomas of the colon:
Liver Physiol. 300, G253–G263 (2011). 189. Gulbransen, B. D. et al. Activation of neuronal P2X7 role of interleukin 18. J. Exp. Med. 207, 1625–1636
165. Wang, P. et al. Nlrp6 regulates intestinal antiviral receptor-pannexin-1 mediates death of enteric neurons (2010).
innate immunity. Science 350, 826–830 (2015). during colitis. Nat. Med. 18, 600–604 (2012). 214. Otani, K. et al. Colchicine prevents NSAID-induced
166. Fletcher, S. M., Stark, D., Harkness, J. & Ellis, J. 190. Neudecker, V. et al. Myeloid-derived miR-223 small intestinal injury by inhibiting activation of the
Enteric protozoa in the developed world: a public regulates intestinal inflammation via repression NLRP3 inflammasome. Sci. Rep. 6, 32587 (2016).
health perspective. Clin. Microbiol. Rev. 25, 420–449 of the NLRP3 inflammasome. J. Exp. Med. 214, 215. Dupaul-Chicoine, J. et al. The Nlrp3 Inflammasome
(2012). 1737–1752 (2017). suppresses colorectal cancer metastatic growth in the
167. Zamboni, D. S. & Lima-Junior, D. S. Inflammasomes in 191. Bauer, C. et al. Colitis induced in mice with dextran liver by promoting natural killer cell tumoricidal
host response to protozoan parasites. Immunol. Rev. sulfate sodium (DSS) is mediated by the NLRP3 activity. Immunity 43, 751–763 (2015).
265, 156–171 (2015). inflammasome. Gut 59, 1192–1199 (2010). 216. Nowarski, R. et al. Epithelial IL-18 equilibrium controls
168. Tanyuksel, M. & Petri, W. A. Jr. Laboratory diagnosis of 192. Hu, B. et al. Inflammation-induced tumorigenesis in the barrier function in colitis. Cell 163, 1444–1456
amebiasis. Clin. Microbiol. Rev. 16, 713–729 (2003). colon is regulated by caspase-1 and NLRC4. Proc. Natl (2015).
169. Mortimer, L., Moreau, F., Cornick, S. & Chadee, K. Acad. Sci. USA 107, 21635–21640 (2010). 217. Ishikura, T. et al. Interleukin-18 overproduction
Gal-lectin-dependent contact activates the 193. Siegmund, B., Lehr, H. A., Fantuzzi, G. & Dinarello, C. A. exacerbates the development of colitis with markedly
inflammasome by invasive Entamoeba histolytica. IL-1 beta -converting enzyme (caspase-1) in intestinal infiltrated macrophages in interleukin-18 transgenic
Mucosal Immunol. 7, 829–841 (2014). inflammation. Proc. Natl Acad. Sci. USA 98, mice. J. Gastroenterol. Hepatol. 18, 960–969
170. Marie, C., Verkerke, H. P., Theodorescu, D. & 13249–13254 (2001). (2003).
Petri, W. A. A whole-genome RNAi screen uncovers a 194. Bauer, C., Duewell, P., Lehr, H. A., Endres, S. & 218. Huber, S. et al. IL-22BP is regulated by the
novel role for human potassium channels in cell killing Schnurr, M. Protective and aggravating effects of inflammasome and modulates tumorigenesis in the
by the parasite Entamoeba histolytica. Sci. Rep. 5, Nlrp3 inflammasome activation in IBD models: intestine. Nature 491, 259–263 (2012).
13613 (2015). influence of genetic and environmental factors. 219. Coll, R. C. et al. A small-molecule inhibitor of the
171. Mortimer, L., Moreau, F., Cornick, S. & Chadee, K. Dig. Dis. 30 (Suppl. 1), 82–90 (2012). NLRP3 inflammasome for the treatment of
The NLRP3 Inflammasome is a pathogen sensor for 195. Youm, Y. H. et al. The ketone metabolite inflammatory diseases. Nat. Med. 21, 248–255
invasive Entamoeba histolytica via activation of beta-hydroxybutyrate blocks NLRP3 (2015).
alpha5beta1 integrin at the macrophage-amebae inflammasome-mediated inflammatory disease. This study reports a potent and selective inhibitor
intercellular junction. PLoS Pathog. 11, e1004887 Nat. Med. 21, 263–269 (2015). of the NLRP3 inflammasome and its use in multiple
(2015). 196. Yan, Y. et al. Omega-3 fatty acids prevent inflammation mouse models of inflammatory diseases.
172. Zhang, Z. et al. Entamoeba histolytica cysteine and metabolic disorder through inhibition of NLRP3 220. Guo, W. et al. Small molecule-driven
proteinases with interleukin-1 beta converting enzyme inflammasome activation. Immunity 38, 1154–1163 mitophagy-mediated NLRP3 inflammasome inhibition
(ICE) activity cause intestinal inflammation and tissue (2013). is responsible for the prevention of colitis-associated
damage in amoebiasis. Mol. Microbiol. 37, 542–548 197. Maslowski, K. M. et al. Regulation of inflammatory cancer. Autophagy 10, 972–985 (2014).
(2000). responses by gut microbiota and chemoattractant 221. Jiang, H. et al. Identification of a selective and direct
173. Que, X. et al. A surface amebic cysteine proteinase receptor GPR43. Nature 461, 1282–1286 (2009). NLRP3 inhibitor to treat inflammatory disorders.
inactivates interleukin-18. Infect. Immun. 71, 198. Macia, L. et al. Metabolite-sensing receptors GPR43 J. Exp. Med. 214, 3219 (2017).
1274–1280 (2003). and GPR109A facilitate dietary fibre-induced gut 222. Romberg, N. et al. Mutation of NLRC4 causes a
174. Chudnovskiy, A. et al. Host-protozoan interactions homeostasis through regulation of the inflammasome. syndrome of enterocolitis and autoinflammation.
protect from mucosal infections through activation Nat. Commun. 6, 6734 (2015). Nat. Genet. 46, 1135–1139 (2014).
of the inflammasome. Cell 167, 444–456 (2016). 199. Du, Q. et al. Dietary cholesterol promotes 223. Canna, S. W. et al. An activating NLRC4
175. Mathur, A., Hayward, J. A. & Man, S. M. AOM-induced colorectal cancer through activating the inflammasome mutation causes autoinflammation
Molecular mechanisms of inflammasome signaling. NLRP3 inflammasome. Biochem. Pharmacol. 105, with recurrent macrophage activation syndrome.
J. Leukoc. Biol. 103, 233–257 (2018). 42–54 (2016). Nat. Genet. 46, 1140–1146 (2014).
224. Kitamura, A., Sasaki, Y., Abe, T., Kano, H. & 237. Seregin, S. S. et al. NLRP6 function in inflammatory 252. Rao, S. et al. Pathogen-mediated inhibition of
Yasutomo, K. An inherited mutation in NLRC4 causes monocytes reduces susceptibility to chemically anorexia promotes host survival and transmission.
autoinflammation in human and mice. J. Exp. Med. induced intestinal injury. Mucosal Immunol. 10, Cell 168, 503–516 (2017).
211, 2385–2396 (2014). 434–445 (2017). 253. Wong, M. L. et al. Inflammasome signaling affects
References 222, 223 and 224 collectively report 238. O’Keefe, S. J. Diet, microorganisms and their anxiety- and depressive-like behavior and gut
an association between mutations in NLRC4 and metabolites, and colon cancer. Nat. Rev. microbiome composition. Mol. Psychiatry 21,
inflammatory conditions in humans. They also Gastroenterol. Hepatol. 13, 691–706 (2016). 797–805 (2016).
highlight the therapeutic relevance of inflammasome 239. Bergstrom, K. et al. Defective intestinal mucin-type 254. Lara-Tejero, M. et al. Role of the caspase-1
blockade in the treatment of these clinical O-glycosylation causes spontaneous colitis-associated inflammasome in Salmonella typhimurium
manifestations. cancer in mice. Gastroenterology 151, 152–164 pathogenesis. J. Exp. Med. 203, 1407–1412 (2006).
225. Canna, S. W. et al. Life-threatening NLRC4-associated (2016). 255. Song-Zhao, G. X. et al. Nlrp3 activation in the
hyperinflammation successfully treated with IL-18 240. Powell, N., Walker, M. M. & Talley, N. J. The mucosal intestinal epithelium protects against a mucosal
inhibition. J. Allergy Clin. Immunol. 139, 1698–1701 immune system: master regulator of bidirectional pathogen. Mucosal Immunol. 7, 763–774 (2014).
(2017). gut-brain communications. Nat. Rev. Gastroenterol. 256. Allen, I. C. et al. NLRP12 suppresses colon
226. Allam, R. et al. Epithelial NAIPs protect against Hepatol. 14, 143–159 (2017). inflammation and tumorigenesis through the negative
colonic tumorigenesis. J. Exp. Med. 212, 369–383 241. Man, S. M., Karki, R. & Kanneganti, T. D. DNA-sensing regulation of noncanonical NF-kappaB signaling.
(2015). inflammasomes: regulation of bacterial host defense Immunity 36, 742–754 (2012).
227. Garaude, J., Kent, A., van Rooijen, N. & and the gut microbiota. Pathog. Dis. 74, ftw028 (2016). 257. Zaki, M. H. et al. The NOD-like receptor NLRP12
Blander, J. M. Simultaneous targeting of toll- and 242. Hu, B. et al. Microbiota-induced activation of epithelial attenuates colon inflammation and tumorigenesis.
nod-like receptors induces effective tumor-specific IL-6 signaling links inflammasome-driven inflammation Cancer Cell 20, 649–660 (2011).
immune responses. Sci. Transl Med. 4, 120ra116 with transmissible cancer. Proc. Natl Acad. Sci. USA References 256 and 257 reveal an important role
(2012). 110, 9862–9867 (2013). for NLRP12 in colitis and colitis-associated
228. Dihlmann, S. et al. Lack of Absent in Melanoma 2 243. Seregin, S. S. et al. NLRP6 protects Il10−/− mice from colorectal cancer in mice.
(AIM2) expression in tumor cells is closely associated colitis by limiting colonization of Akkermansia
with poor survival in colorectal cancer patients. muciniphila. Cell Rep. 19, 733–745 (2017). Acknowledgements
Int. J. Cancer 135, 2387–2396 (2014). 244. Chen, L. et al. NLRP12 attenuates colon inflammation S.M.M. is supported by the Australian National University
229. Karki et al. NLRC3 is an inhibitory sensor of by maintaining colonic microbial diversity and Futures Award, The Gretel and Gordon Bootes Medical
PI3K–mTOR pathways in cancer. Nature 540, promoting protective commensal bacterial growth. Research Foundation and the National Health and Medical
583–587 (2016). Nat. Immunol. 18, 541–551 (2017). Research Council of Australia under project grants
230. Man, S. M., Karki, R. & Kanneganti, T. D. 245. Henao-Mejia, J. et al. Inflammasome-mediated (APP1141504 and APP1146864) and the R.G. Menzies Early
AIM2 inflammasome in infection, cancer, and dysbiosis regulates progression of NAFLD and obesity. Career Fellowship (APP1091544). The author apologizes to
autoimmunity: role in DNA sensing, inflammation, Nature 482, 179–185 (2012). researchers whose work was not cited or was cited through
and innate immunity. Eur. J. Immunol. 46, 269–280 246. Mamantopoulos, M. et al. Nlrp6- and ASC-dependent reviews owing to space limitations.
(2016). inflammasomes do not shape the commensal gut
231. Hu, B. et al. The DNA-sensing AIM2 inflammasome microbiota composition. Immunity 47, 339–348 Competing interests
controls radiation-induced cell death and tissue injury. (2017). The author declares no competing interests.
Science 354, 765–768 (2016). 247. Lemire, P. et al. The NLR protein NLRP6 does not
232. Lian, Q. et al. Chemotherapy-induced intestinal impact gut microbiota composition. Cell Rep. 21, Publisher’s note
inflammatory responses are mediated by exosome 3653–3661 (2017). Springer Nature remains neutral with regard to jurisdictional
secretion of double-strand DNA via AIM2 References 246 and 247 demonstrate, using claims in published maps and institutional affiliations.
inflammasome activation. Cell Res. 27, 784–800 littermate-controlled experiments, that certain
(2017). inflammasomes do not influence the composition Reviewer information
233. Alexander, J. L. et al. Gut microbiota modulation of the gut microbiota in mice. Nature Reviews Gastroenterology & Hepatology thanks
of chemotherapy efficacy and toxicity. Nat. Rev. 248. Levy, M. et al. Microbiota-modulated metabolites R. Flavell, T. Monie and the other anonymous reviewer(s) for
Gastroenterol. Hepatol. 14, 356–365 (2017). shape the intestinal microenvironment by regulating their contribution to the peer review of this work.
234. Normand, S. et al. Nod-like receptor pyrin NLRP6 inflammasome signaling. Cell 163,
domain-containing protein 6 (NLRP6) controls 1428–1443 (2015). Review criteria
epithelial self-renewal and colorectal carcinogenesis 249. Sun, Y. et al. Stress-induced corticotropin-releasing A detailed literature review was performed using the PubMed
upon injury. Proc. Natl Acad. Sci. USA 108, hormone-mediated NLRP6 inflammasome inhibition database using a combination of the following search terms:
9601–9606 (2011). and transmissible enteritis in mice. Gastroenterology “inflammasome”, “NLRP1”, “NLRP3”, “NLRC4”, “AIM2”,
235. Chen, G. Y., Liu, M., Wang, F., Bertin, J. & Nunez, G. 144, 1478–1487 (2013). “pyrin”, “caspase-1”, “caspase-4”, “caspase-5”, “caspase-11”,
A functional role for Nlrp6 in intestinal inflammation 250. Pierantonelli, I. et al. Lack of NLRP3-inflammasome “pyroptosis”, “IL-1”, “IL-18”, “infection”, “bacteria”, “viruses”,
and tumorigenesis. J. Immunol. 186, 7187–7194 leads to gut-liver axis derangement, gut dysbiosis and “protozoa”, “colitis”, “IBD”, “cancer” and “microbiota”. Relevant
(2011). a worsened phenotype in a mouse model of NAFLD. English-language papers were evaluated.
236. Birchenough, G. M., Nystrom, E. E., Johansson, M. E. Sci. Rep. 7, 12200 (2017).
& Hansson, G. C. A sentinel goblet cell guards the 251. Lukens, J. R. et al. Dietary modulation of the Supplementary information
colonic crypt by triggering Nlrp6-dependent Muc2 microbiome affects autoinflammatory disease. Supplementary information is available for this paper at
secretion. Science 352, 1535–1542 (2016). Nature 516, 246–249 (2014). https://doi.org/10.1038/s41575-018-0054-1.