Abstract
Inflammatory bowel disease (IBD) remains a persistent health problem with a global burden surging over 6.8 million cases currently. Clinical pathology of IBD is complicated; however, hyperactive inflammatory and immune responses in the gut is shown to be one of the persistent causes of the disease. Human gut inflammasome, the activator of innate immune system is believed to be a primary underlying cause for the pathology and is largely associated with the progression of IBD. To manage IBD, there is a need to fully understand the role of inflammasome activation in IBD. Since inflammasome potentially play a significant role in IBD, systemic modulation of inflammasome may provide an effective therapeutic and clinical approach to control IBD symptoms. In this review we have focused on this association between IBD and gut inflammasome, and recent advances in the research and therapeutic strategies for IBD. We have discussed inflammasomes and their components, outcomes from the experimental animals and human studies, inflammasome inhibitors, and developments in the inflammasome targeted therapies for IBD.
Keywords: Inflammatory bowel disease, Crohn’s disease, ulcerative colitis, inflammasome, NLRP3
INTRODUCTION
The human gut microbiome is stringently regulated by the intestinal immune system. However, under certain conditions, disruption in gut homeostasis may cause an over-reacted inflammatory immune response. These unproportioned inflammatory immune responses may lead to Crohn’s disease (CD) or Ulcerative colitis (UC) depending on the area they are affecting (1). Both conditions are cumulatively referred to as inflammatory bowel disease (IBD). The pathophysiology and etiology of IBD are complicated. This involves genetic, environmental, and several other factors. Likewise, gut inflammasome, which has been associated with several other inflammatory and autoimmune disorders can also be one of the factors in the development of IBD. Abolition or remission of mucosal inflammation is an ultimate therapeutic aim for IBD treatment. Studies have shown that targeting inflammasome can reduce inflammation in IBD (2). In this review, we have focused on this association between IBD and gut inflammasome, and recent advances in the research and therapeutic strategies for IBD.
INFLAMMATORY BOWEL DISEASE
IBD is a group of Ulcerative colitis and Crohn’s disease, both affecting gastrointestinal system. In 1875, two English physicians Samuel Wilks and Walter Moxon first described possible condition of ulcerative colitis in a woman who died with recurrent diarrhea and fever (3, 4). Whereas, in 1932 American physicians Burrill Crohn, Leon Ginzberg, and Gordon Oppenheimer described the CD (5). Since then, physicians and researchers are working to improve their understanding on the pathophysiology and immunology of these debilitating disorders. Symptoms of both CD and UC include intermittent abdominal pain, weight loss, diarrhea, rectal bleeding, fever, and fatigue. Those who suffer, have increased mortality with a risk of cancer, blood clots, and primary sclerosing cholangitis (6). IBD may occur at any age through adolescents to adults irrespective of the sex. Genetic, environmental, and immunological factors contribute in unison towards compromising the intestinal epithelial barrier function increasing the risk of disease (7).
Ulcerative colitis involves rectum and extending proximally to the large intestine. UC patients develop intestinal lesions which are mostly superficial and extend proximally, but in severe disease condition deep intestinal ulcerations are observed. Complications with UC include anemia, toxic colitis due to the damage to entire thickness of colon wall that can lead to ileus, toxic megacolon and finally colon cancer (1 out of every 200 patients with UC develops colon cancer) (8). Crohn’s disease can involve the entire gastrointestinal tract, however mostly affecting the distal end of small intestine (terminal ileum) and colon (9). Most patients with CD presents chronic inflammation, strictures, intestinal stenosis and fistulas (3). CD patients also encounter malnutrition as damage to small intestine significantly affect the absorption of nutrients (10).
Epidemiology
There is a worldwide increase in the incidence of IBD. IBD, which is incurable reduces the quality of life, increasing disability and thus places a heavy burden on the population and health system. The incidence of IBD varies geographically and a considerable difference between east and west exists. However, this difference between eastern and western countries is shrinking rapidly (11). According to a recent study there were 6.8 million cases of IBD globally in 2017 (1). The age standardized prevalence of IBD between 1990 to 2017 was 464.5 in United States of America, 449.6 in United Kingdom, 520 in Canada, 136.2 in China, and 16.2 in India per 100,000 populations (1). The highest prevalence of UC was 505 per 100,000 population in Northern Europe followed by prevalence of 286 and 44 per 100,000 population in North America and South Asia respectively (12). The highest incidences of UC were 57 in Northern Europe, 23.14 in North America, and 6 in South Asia per 100,000 populations (12). For CD, the highest prevalence was observed in Western Europe (322 per 100,000) followed by North America (318 per 100,000), North Europe (262 per 100,000) and East Europe (200 per 100,000) (12). The highest incidence was in North America (23 per 100,000), Southern (15 per 100,000) and Eastern (14 per 100,000) Europe. Asian countries are also showing increase in both prevalence and incidence of CD. The average prevalence of CD in Asia was 18 and incidence was 4 per 100,000 population (12). The reported numbers can be far more than expected as only partial data is available from most of the underdeveloped countries. Overall, the available data suggest a continuous surge in IBD incidences.
Treatment and disease management
There is no specific treatment available to cure IBD. The ultimate goal of IBD treatment is to achieve clinical remission and then after maintaining this achieved remission of disease condition. The primary line of treatment includes 5-aminosalicylic acid (5-ASA; sulfasalazine, mesalamine), steroids (prednisone, budesonide), and immunomodulators (6-mercaptopurine, methotrexate, azathioprine) (13, 14). Depending on the severity of disease condition and using recent algorithms, treatment approach may vary (15). Additional drugs for the management and inducing the remission in IBD patients include use of calcineurin inhibitors or antibiotics (cyclosporine, tacrolimus, ciprofloxacin, metronidazole), and anti-tumor necrosis factor-α (TNF-α) antibodies or other biologicals (infliximab, adalimumab) (13, 14). However, at least 90% of the IBD patients experience relapse of disease condition once in a lifetime (16, 17). To avoid morbidity in such individuals, surgery is the only option. About 15-47% of patients on medical therapy require surgical intervention (16). Depending on the existing prevalence of IBD in USA, total annual direct cost for the treatment of IBD can be USD 35 billion.
Existing medications to treat IBD are associated with the increased risk of side-effects and complications. The side-effects existing drugs can cause are renal toxicity, hemolytic anemia, osteoporosis, insomnia, pancreatitis, bone marrow suppression, non-Hodgkin lymphoma, hepatosplenic T-cell lymphoma, fatigue, diarrhea, nausea and vomiting (18). Individuals on long-term IBD therapy are vulnerable to other opportunistic infections (8), cervical cancer (19), skin cancer (20), or poor compliance to vaccinations (21, 22). Considering these drawbacks of existing drugs, new therapeutic options are being developed for the treatment of IBD (15, 18).
INFLAMMASOME PATHWAY
Inflammasome is a cytoplasmic multiprotein constituent of the innate immune system. They exist across all immune (macrophages, dendritic cells, etc.) and epithelial cells. Inflammasome is a fundamental and often tightly regulated host defense system that specifically involves sensing microbial, fungal, viral, nematode, and other harmful agents (23). Recognition of pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs) by pattern recognition receptors (PRR) triggers inflammasome assembly and subsequent induction of adaptive immune responses (24). Depending on the stimulus, inflammasome can be activated via canonical caspase-1, or noncanonical caspase-4/5 pathways (caspase-11 in mouse) (25). To date, several kinds of inflammasomes including nucleotide-binding domain and leucine-rich repeat receptors (NLR) pyrin domain-containing family (NLRP), NLR family caspase recruitment domain-containing protein-4 (NLRC4), and double-stranded DNA sensors absent in melanoma-2 (AIM2) have been described.
NLRP3 inflammasome
NLRP3 is a tripartite assembly of NLRP3 protein, caspase-1, and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) (Figure 1). NLRP3 protein complex contains an amino-terminal pyrin domain (PYD), N-terminal domains, a central nucleotide-binding and oligomerization (NACHT) domain, and a carboxy-terminal leucine-rich repeat domain (LRR). A cryo-electron microscopy structure of NLRP3 has been recently determined (26). The atomic model building on a 3.8-A resolution map showed earring-shaped structure of NLRs (PYD-deleted) containing a curved LRR domain and NACHT domain. The earring-shaped structural characteristics were similar to NLRC4 (27), NOD2 (28) and NLR apoptosis inhibitory protein 5 (NAIP5) (29). The caspase-1 has amino-terminal caspase activation and recruitment domain (CARD), a central large catalytic domain (p20), and a carboxy-terminal small catalytic subunit domain (p10) (30). Formation of an active NLRP3 assembly is triggered on sensing stimulations like extracellular ATP, potassium (K+) efflux, calcium (Ca2+) signaling, reactive oxygen species (ROS), toxins, and mitochondrial dysfunction. The NLRP3 then polymerizes through interactions with the NACHT domains, initiating a homotypic pyrin-pyrin domain interaction through ASC (31). Multiple ASC filaments further segregate to form an ASC speck (32). Finally, ASC speck interacts with caspase-1 through CARD-CARD interaction, enabling enzymatic cleavage and activation of caspase-1 (30). The activated caspase-1 enzymatically cleaves pro-interleukin (IL)-1β and pro-IL-18 to their mature forms. The release of IL-1β and IL-18 cytokines triggers inflammatory signals through pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and IL-6, nuclear factor-κB (NF-κB) pathway, and gene transcription.
The canonical activation of NLRP3 is by a two-step process (Figure 2). In the first priming step, PAMPs or DAMPs recognition by toll-like receptors (TLR) initiates the NF-κB-mediated signaling and activation pathway. This downstream events to NF-κB pathway upregulates the transcriptional and translational induction of inflammasome sensors, NLRP3, proIL-1β, and proIL-18. In the second activation step, NLRP3 protein, ASC, and caspase-1 are assembled into an active NLRP3 inflammasome assembly, which converts caspase-1, proIL-18, and proIL-1β to their active forms. The formation of active NLRP3 assembly also induces programmed necrosis or pyroptosis in cells by the formation of gasdermin-D (GSDMD; a pore-forming protein) (25).
GSDMD-mediated pyroptosis is a crucial component in executing inflammasome triggered innate immune responses. Recent developments redefine pyroptosis as GSDMD-mediated programmed necrosis (33). Caspase-1 once activated, cleaves at aspartate-275 site within the GSDMD-C terminal linker loop (34). Recent study uncovers more specific structural details of this engagement (35). The GSDMD-C terminal linker loop binds to the exosite and catalytic groove of caspase-1 via hydrogen bonds forming a dual-site engagement for more specific recognition and efficient cleavage of GSDMD. The linker loop between C-terminal (repressor domain) and N-terminal (pore-forming domain) of GSDMD is responsible for the autoinhibition of GSDMD (36). Whereas the N-terminal domain of GSDMD is in-charge for forming the membrane pores and inducing pyroptosis (34). The GSDMD-N terminal domain is itself sufficient to drive pyroptosis. GSDMD-N domain can form pores on phosphoinositides on the plasma membrane, disrupting the osmotic potential and K+ efflux, causing cell swelling and eventually cell lysis (36). GSDMD-pore probably even allows the release of IL-1β and IL-18 cytokines (37). However, the possibility of releasing these cytokines through pyroptotic membrane rupture or through normal cytokine release mechanism in conjunction with the timing of cell death is still unclear.
The canonical activation and oligomerization of inflammasome was triggered by the efflux of K+ or Cl- ions, flux of Ca2+ ions, lysosomal disruption, mitochondrial dysfunction, and metabolic changes (38). Additional stimuli for canonical NLRP3 activation are: (1) the disassembly of trans-Golgi network can cause aggregation and activation of NLRP3, (2) bacterial lipopolysaccharide (LPS) can initiate NLRP3 activation through TLR4 ligand, (3) priming step that can induce post-translational modifications of NLRP3, and recently discovered (4) the post-transcriptional deubiquitination of NLRP3 (39).
The non-canonical activation of NLRP3 occurs through the activation pathways of human caspases-4/5 and mouse caspase-11 (Figure 2). Activation of GSDMD by the enzymatic cleavage from caspases-4/5 or 11 leads to the K+ efflux and autocrine activation of NLRP3, which subsequently leads to the maturation of IL-1β and IL-18 cytokines. The non-canonical pathway of inflammasome activation is independent of LPS detection by TLR. Until now it was believed that the CARD domain of caspase-4/5/11 recognizes cytosolic LPS which is internalized through phagocytosis, inducing the oligomerization and cleavage of GSDMD (40-43). However, recent studies reported the necessity of interferon-induced guanylate binding proteins (GBP) for detection of cytosolic LPS by caspase-4/5/11 and non-canonical inflammasome activation (44-47). GBP acts as a sensor for cytosolic LPS and their interactions are through electrostatic interactions between Lipid A and inner core of LPS and hydrophobic LPS binding sites CD14 and MD-2 of GBP (44). GBP-LPS complex then recruits and activates caspase-4 for subsequent release of cytokines.
Recently an alternative pathway for NLRP3 activation in human and porcine monocytes has also been discovered (48). This involves TLR4 activation by LPS. The binding of extracellular ATP to ATP-gated ion channel-P2X7 triggers the downstream TLR4 signaling cascade (TLR-4-TRIF-RIPK1-FADD) activating caspase-8 and ultimately formation of NLRP3 inflammasome assembly. The mechanism was independent of pyroptosome formation, pyroptosis and K+ efflux for inflammasome activation. The LPS and ATP co-stimulation of THP-1 macrophages increased the expression of RIPK1 and activation of caspase-8 (49). The alternative pathway of NLRP3 activation has been involved in the development of inflammation and autoimmunity (50-52). However, additional components of this pathway are yet to be explored.
NLRP1 inflammasome
NLRP1 is a first inflammasome discovered and has PYD, NACHT and LRR domains. The interactions between human NLRP1 (hNLRP1) and caspase-1 CARD is through electrostatic interactions (53). NLRP1 oligomerization can directly recruit caspase-1 via the C-terminal CARD domain independent of ASC (54). The hNLRP1 and mouse NLRP1b homolog have a C-terminal extension containing a function-to-find domain (FIIND) which is not commonly observed in other NLRP’s (55). The autolytic proteolysis within the FIIND domain between the “found in ZO-1 and UNC5” and “conserved in UNC5, PIDD, and Ankyrin; (UPA)” sub-domains generates two non-covalent polypeptide chains (N-terminal and C-terminal fragments) (56). FIIND domain is required for the activation of NLRP1 assembly (57), as inhibition of the autoproteolytic activity of FIIND domain blocks the activation of NLRP1 inflammasome (57, 58). Studies have shown that direct proteolytic cleavage of NLRP1 by anthrax lethal toxin (LT) or IpaH7.8, a Shigella flexneri ubiquitin ligase secreted effector is required for the NLRP1 activation, formation of IL-1β and pyroptosis (58, 59). The activation of NLRP1 by IpaH7.8 needed E3 ligase and host proteosome but independent of N-end rule ubiquitin ligase Ubr2 (59, 60). Studies have also shown that selective inhibition of dipeptidyl peptidases 8 and 9 (DPP8/9) activate NLRP1 in human THP-1 and mouse RAW 264.7 macrophage cell lines to induce pyroptosis without efficiently activating IL-1β (60-62). DPP8/9 can directly bind to the FIIND domain mediating the NLRP1 inflammasome activation (55, 63). However, underlying molecular mechanisms of this pathway are still subject to further investigation.
NLRP6 inflammasome
NLRP6 (PYPAF5) is a newly discovered inflammasome involved in the regulation of NF-κB and mitogen-activated protein kinase (MAPK) pathways (64). NLRP6 can be regulated at both transcriptional and post-translational levels by transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ), TNF-α and miRNA-331-3p (65-67). Activation of NLRP6 mediates the formation of an active inflammasome assembly with ASC and caspase-1 or 11 leading to the production of IL-18 and IL-1β. Some of the activators of NLRP6 includes deubiquitinase Cyld (68), carcinogenic bacterial lipoteichoic acid (LTA) (69), and LPS (70). Interestingly, LTA-induced formation of NLRP6 assembly has been shown to follow the non-canonical pathway of inflammasome activation (71).
The structure of NLRP6 resembles NLRP3 with few differences in the PYD component (72, 73). Recent cryo-EM and crystal structure of NLRP6 showed that the N-terminal NLRP6PYD filament is a core element to recruit inflammasome assembly (72). The NLRP6 constructs (full-length NLRP6, NLRP6PYD, and NLRP6PYD+nucleotide-binding domain, NBD) were able to induce ASC-PYD polymerization. Unlike NLPR3, PYD of NLRP6 at higher concentrations is independently capable of nucleating ASC-PYD polymerization. The NLRP6PYD is a hollow cylindrical structure assembly of right-handed helix containing six antiparallel α-helices. The α2 and α3 loop of NLRP6PYD are involved and facilitates confirmational changes and molecular interactions in the NLRP6 inflammasome assembly (72).
NAIP/NLRC4 inflammasome
NAIP/NLRC4 inflammasome is important during lung, spleen and liver infections, sepsis, and intestinal inflammation. The NAIP/NLRC4 complex is formed upon detection of bacterial flagellin and type-3 secretion system components as activating ligands and induce activation of caspase-1 and pyroptosis (74-76). NLRC4 cannot directly recognize activation signals and thus activation of NLRC4 is through NAIP. NAIP as a sensor and NLRC4 as adaptor protein forms a single inflammasome complex (77). A three domain structure of NLRC4 complex comprises of N-terminal homotypic interaction domain, a central NBD domain, and a series of C-terminal LRR (77). Upon activation, a single NAIP protein forms assembly with 10 to 11 NLRC4 molecules leading to a multi-subunit disk-like structure of NAIP/NLRC4 inflammasome (27). NLRC4 can directly bind to caspase-1 through CARD-CARD domain independent of ASC. However, binding to ASC can efficiently activate caspase-1 (78). Other reviews have detailed discussion on NAIP/NLRC4 inflammasome (77, 79).
AIM2 inflammasome
AIM2 is a ALR family of a non-NLR cytosolic protein that can activate caspase-1 and subsequently cytokines (80-82). AIM2 is different from other inflammasomes due to its ability of detecting DNA from its characteristic HIN200 domain (25, 80). Normally, DNA is not present in the cytosol of eukaryotic cells, hence detection of pathogenic DNA from multiple origins or even self-DNA can trigger AIM2 inflammasome responses (25). Recent studies have also showed detection of RNA by AIM2 (83). AIM2 consists of a N-terminal PYD domain and C-terminal HIN200 domain. The N-terminal PYD domain interacts with ASC, while the HIN200 domain recognizes DNA sequences. The optimal length of DNA sequences to be recognized by AIM2 is approximately 80 base pairs (84). In normal inactive state, AIM2HIN confines AIM2PYD domain, restricting its ability to form PYD-PYD interactions (84). The binding of dsDNA to AIM2HIN by oligonucleotide/oligosaccharide binding folds of HIN or independent interactions, release PYD domain further enabling it to form PYD-PYD interactions (85). Binding of AIM2PYD to ASC completes the formation of an active inflammasome assembly to perform downstream actions. However, few studies contradicted the autoinhibitory model of AIM2PYD activation, the studies suggested the oligomerization-driven activation of AIM2PYD assembly (86, 87). The regulation of AIM2 activation and assembly has been through PYD-only proteins (POP) (88). POP1 and POP2 binds to ASCPYD, whereas POP3 binds directly to AIM2PYD. Binding of POP3 to AIM2PYD blocks PYD-PYD interactions with ASC and subsequent activation of caspase-1 and cytokine release. Similarly, CARD-only proteins (COPs) and decoy proteins belonging to the ALR family can regulate activation of AIM2 inflammasome assembly (89).
Pyrin inflammasome
Pyrin is primarily expressed in neutrophils, monocytes, eosinophils and dendritic cells. Human pyrin contains four functional domains (90): 1) PYD domain (1-92) at N-terminal end which is responsible for interaction with ASC and subsequently activation of caspase-1 and cytokine release. 2) A zinc finger B-box domain (370-412) and 3) a coiled-coil (CC) domain (420-440). Both these domains are involved in the oligomerization of pyrin and organization of cytoskeleton. 4) The C-terminal B30.2/SPRY domain is responsible for direct interactions with caspase-1 and with the other inflammasome components (91).
Activation of pyrin has been associated with the disruption in actin filaments and cytoskeleton assembly (92). Pyrin activation is triggered on pathogen-induced modifications in Rho guanosine triphosphatases (Rho GTPases; RhoA) of host cells resulting in formation of pyrin-ASC inflammasome complex (93). However, pyrin does not directly sense the modifications in RhoA but the resulting activation of downstream RhoA signals (94). RhoA modifications can disrupt actin cytoskeleton assembly triggering activation of pyrin inflammasome assembly. Studies using the regulators of actin cytoskeleton assembly like WDR1, vinblastine or colchicine showed changes in the formation of pyrin inflammasome assembly (95, 96). Individuals deficient in wdr1 gene showed increase in the actin polymerization and suffer with IL-18 dependent autoinflammatory phenotype and thrombocytopenia (96–98). These studies highlight the engagement of inflammasome components with structural organization of cells. Moreover, bacterial toxins can activate pyrin inflammasome (99), however very few have studied the association between pyrin inflammasome and microbiome-mediated intestinal inflammation (100–102).
Other NLR inflammasomes
Besides the above discussed inflammasomes, over the year’s scientists have also identified additional inflammasomes. NLRP7 has been shown to activate in response to microbial acylated lipopeptides and induce ASC-dependent formation of inflammasome assembly (103). NLRP9 is a other inflammasome originally linked to the reproductive system, however recent studies have highlighted NLRP9’s role in the intestine on rotavirus infection (104). NLRP2 and NLRP5 are also other inflammasomes associated with the early embryonic arrest and infertility (105).
ROLE OF INFLAMMASOME IN IBD
Essential microbiota residing in the human intestinal tract is not part of the immune system. Hence, even a minor invasion of this microbiota through a tightly regulated intestinal barrier can trigger cellular inflammatory responses (Figure 3). The gut inflammasome primary regulates the host defense responses to microbial pathogens limiting their ingress through the intestinal tract. This does mean the possibilities of their involvement in the progression of IBD.
Experimental animal studies
Several studies demonstrate the protective effects of gut inflammasome in the development of IBD. Mice lacking components of inflammasome assembly NLRP3, NLRC4, IL-1β, Caspase-1/11, and ASC showed more susceptibility to colitis with exacerbation in clinical disease symptoms, and increased mortality when challenged with chemically induced colitis compared to their wild-type (WT) mates (106–112). Caspase-1−/− and pycard−/− (ASC containing CARD) mice demonstrated severe acute and chronic dextran sulfate sodium (DSS)-induced colitis with significantly increased morbidity, mortality and clinical disease score compared to the WT colitis mice (106). Further studies from other groups reiterated these observations and showed that in the absence of NLRP3 or even the downstream components of NLRP3 pathway like ASC and caspase-1 are sufficient to exacerbate colitis condition (107, 113). These studies also concluded that both the activation of inflammasome components and subsequent release of cytokines are equally important in attenuating colitis. Subsequently, another study showed a significantly lower intestinal levels of IL-10 and TGF-β in nlrp3−/− colitis mice (108). Both IL-10 and TGF-β play prominent roles in downregulating intestinal inflammation (114). The neutrophils isolated from nlrp3−/− mice displayed increased apoptosis and impaired chemotaxis to neutrophil chemotaxis factors (keratinocyte-derived chemokine (KC), leukotriene B4, WKTMVm and C5). Concurrent observation of impaired neutrophil migration in Nlrp3−/− mice was observed in another study that showed decreased KC-induced release of intracellular calcium elevation, Rac activation, and actin assembly formation in nlrp3−/− neutrophils (115). Defective neutrophil chemotaxis has been observed in individuals with CD and can be responsible for impaired innate immune response towards microbial invasion (116). Nlrp3−/− and caspase-1−/− colitis mice showed loss of intestinal barrier integrity (106, 107). Caspase-1−/− mice were extremely susceptible to develop colitis and continued to lose their body weight with severe hematochezia (110). Caspase-1−/− mice showed reduced claudin-3 expression and intestinal epithelial cell (IEC) proliferation.
Caspases dysfunction has been associated with intestinal epithelial inflammation and disruption of epithelial barrier function and carry a direct ability to regulate IL-18 and IL-1β cytokines. Caspase-11−/− mice showed increased susceptibility to acute DSS-colitis, increased mortality and morbidity and impaired epithelial barrier permeability compared to their WT mates (117–119). Moreover, the caspase-1 expression was increased in caspase-11−/− mice and can confer to the levels of IL-1β in these animals. The role of TRIF was not observed in caspase-11 mediated effects on intestinal inflammation, however IFN-γ induced STAT1 expression and phosphorylation was corelated with the severity of DSS-colitis mice and partially dependent on caspase-11 (119, 120). STAT1 signaling in IECs was observed to play a role in protective effects of caspase-11 in colitis (119). Further study established that the caspase-1 induced STAT1 activation in IEC was independent of IFN-γ but dependent on LPS/IL-1β-mediated activation (120). This presents a new paradigm in caspase-11 mediated STAT1 activation in IECs via IL-1β released from non-canonical inflammasome.
IL-18 contributes towards gut microbiota development, intestinal barrier function, and mucosal renewal (121). Recent studies implicate that IL-18 expression is expressed only in intestinal epithelium and this expression is independent of NLRP3 activation but dependent on Caspase-1 (122, 123). In concordance with this observation, previous studies have attributed colonic epithelial assault to significantly ablated levels of mature IL-18 in caspase-1/11−/− colitis mice (107, 117). Susceptibility to develop colitis and colorectal cancer was reported in IL-18−/− mice (2, 124–126). On the contrary, on treating caspase-11−/− colitis mice with recombinant IL-18, animals showed significant attenuation in colitis disease pathology. These studies highlight the importance of IL-18 in imparting the protection from intestinal inflammation.
However, recent studies consider NLRP6 as a critical player in the maintenance of gut homeostasis, mucosal renewal, and proliferation, but not IL-18 (127–129). Gene knockdown studies presents the protective role of NLRP6 against intestinal inflammation. NLRP6 is essentially expressed in IECs and impaired expression of nlrp6 develops severe colitis in mice challenged with DSS compared to their WT mates (129, 130). Nlrp6−/− mice exhibited changes in terminal ileum, colonic crypt hyperplasia, and increased size of Peyer’s patches with formation of germinal centers. Colonic explants from nlrp6−/− mice also produced significantly less IL-18. However, recent study showed no change in the expression of IL-18 despite the differential NLRP6 expression in human colonic biopsies from CD and UC patients (131). In agreement to this study, Normand et al. (132) showed no NLRP6-dependent changes in the IL-18 expression of colonic biopsies from nlrp6−/− and WT mice. This source of IL-18 can be from other inflammasomes or cells adjacent to intestinal microenvironment. Seregin et al. (133) showed that NLRP6 from lamina propria cells and not IECs was upregulated in response to DSS-treatment in IL-10−/− mice. Moreover, adoptive transfer of Ly6Chi monocytes from WT mice to nlrp6−/− mice reduced DSS-induced mortality, intestinal injury, intestinal permeability and inflammation. The adoptively transferred Ly6Chi monocytes migrated to the colon. The study concluded NLRP6-mediated IL-18 secretion by inflammatory monocytes recruited to colon in response to DSS-induced intestinal assault. These monocytes, in turn, upregulate TNFα and promote epithelial repair and recovery. The study presents a whole new paradigm of NLRP6-mediated protective effects in intestinal inflammation and homeostasis. The correlation between TNF-α and NLRP6 remains to be fully elucidated. Furthermore, the underlying mechanisms of association between IL-18 and NLRP6 is more complex. Elucidating their specific roles is important although challenging. NLRP6 deficient mice also displayed reduced intestinal mucus layer (IML) (134), a common feature in the IBD patients (135). While this notion was contradicted by Volk et al. (136), they showed that the IML formation and function are independent of NLRP6 activation (136).
The importance of GSDMD is continuously emerging as a regulator of downstream processes in inflammasome pathway. However, little is known on the roles of GSDMD in the context of intestinal inflammation. The distribution of GSDMD protein was observed in the lamina propria myeloid cells and E-cadherin+ IECs (137). DSS-induced assault to intestinal epithelium in mice significantly increased the expression and activation of GSDMD in both myeloid cells and IECs promoting pyroptosis in colitis (137). Role of GSDMD was protective in DSS-colitis mice, as the condition got exacerbated in gsdmd−/− mice independent of the microbiota influence. The study outlined GSDMD as a negative regulator of cyclic GMP–AMP synthase (cGAS)-dependent inflammation and cGAS-inhibitor RU.521 treatment attenuated colitis severity in gsdmd−/− mice.
Immunosuppressive activity of IL-10 is critical in restoring anti-inflammatory pathways in IBD. Individuals with IL-10R mutations spontaneously develop CD (138). IL-10 deficient mice spontaneously develop colitis and are even an experimental animal model to study colitis. IL-10 has been correlated with NLRP3 upregulation in the intestinal mucosa. Liu et al. (139) showed elevated NLRP3 activity in intestinal epithelial cells and colonic macrophages of IL-10−/− mice. NLRP3 expression was increased in IL-10−/− mice even before colitis onset suggesting a colitogenic role of NLRP3. Likewise, IL-10k/o chronic colitis mice showed significantly increased colonic IL-1β protein levels that got reversed following inhibition of inflammasome activation with IL-1R antagonist or caspase-1 inhibitors (140).
Recently, the CD risk factor, immunity related GTPase M (IRGM) has been shown as a negative regulator of NLRP3 activation. Irgm1−/− mice showed aggravated DSS-colitis development compared to their Irgm1+/+ littermate controls. Expression of NLRP3, IL-1β, TNF-α, and IL-18 genes was also significantly increased in Irgm−/− animals compared to their littermate controls (141). Recent studies showed novel role of IRGM2 as an inhibitor of caspase-11 when challenged with bacterial LPS (142, 143). The Irgm2−/− mice when challenged with endotoxemia exhibited increased serum levels of TNF-α, IL-1β and IL-18 cytokines, increased weight loss and mortality (142). However, LPS challenged Irgm2−/− capsase-11−/− mice did not show increased serum levels of TNF-α, IL-1β and IL-18 cytokines and less mortality. The LPS primed Irgm−/− bone marrow derived macrophages (BMDM) showed pyroptosis, reduced cell viability, IL-1β and IL-18 secretion and ASC speck formation. All these effects were abolished on ectopic expression of Irgm2 in Irgm2−/− BDBM or on deletion of caspase-11. Moreover, the observed effects of IRGM2 were independent of NLRP3 activation (142, 143). Further studies are warranted to explore the role of IRGM in IBD pathogenesis and utilize IRGM as a therapeutic target for treating intestinal inflammation. NEK7 a component of NLRP3 is also shown to modulate NLRP3 activation and subsequently induce pyroptosis in DSS-induced chronic colitis in mice (144).
Effect on intestinal microbiota
Deficiencies in the inflammasome pathway may have stunted innate immune response to microbiota and promote intestinal assault. Inflammasome pathway may drive specific alterations in the intestinal microbiota, which in turn promote disease in individuals by either exposing them to these pro-colitogenic microbiota or disruption in their normal gut epithelial barrier function due to a variety of inflammatory insults. NLRC4 and interleukin-1 receptor (IL-1R) deficient mice exhibited a reduced immune response to salmonella and Citrobacter rodentium infections respectively (145, 146). Similarly, mice lacking NLRP3, NLRC4, caspase-1, and ASC showed higher sensitivity to C. rodentium, Clostridium difficile infections and establishment of Akkermansia muciniphila in nlrp6−/− mice (130, 147, 148). Moreover, in-spite of housing in the same environment, nlrp3−/− mice displayed differences in intestinal microbiota than WT mice with detection of potentially pathogenic bacterium from species Enterobacteriaceae and Mycobacterium (108). Elinav et al. (129) showed that colonic microbiota of NLRP6, IL-18, ASC, and caspase-1 deficient mice dominated colitiogenic TM7 and Prevotellaceae (Bacteroidetes phyla) bacterial strains. However, another study contradicted these observations using littermate-controlled nlrp6−/− mice and ex-germ-free littermate-controlled asc−/− mice (149). Importantly, recent studies have also contradicted the assumption that inflammasomes can influence shaping of intestinal microbiota. Lemire et al. (150) showed that NLRP6 had no dominant influence in shaping the structure of intestinal bacterial community and both WT and nlrp6−/− littermates exhibited similar microbial community. Different from this point of view of whether inflammasomes disrupts normal gut microbiota structure, Meng et al. (123) observed improvement in colitis and colorectal cancer in NLRP3R258W mutation experimental mice. NLRP3R258W mutation has been associated with the hyperactivation of inflammasome, autoinflammatory disorders and excessive production of IL-1β. If NLRP3 increases susceptibility to intestinal inflammation as discussed in another section of this review, NLRP3R258W mutation should exacerbate the colitis condition. However, intrinsically pro-inflammatory condition and intestinal assault due to the hyperactivation of NLRP3R258W mutation was compensated by the reshaping of local microbiota and induction of T-regulatory milieu. The findings open an interesting new avenue of inflammasome research in intestinal disorders.
Studies contradicting protective role of inflammasome in intestinal inflammation
Although most studies support the notion that inflammasomes activation reduced the pathology in IBD, several studies contradict this view. Proponents of this theory demonstrated the detrimental effects of inflammasome activation in IBD. Animal studies showed no influence of NLRP6-ASC in curating the gut microbiota and susceptibility to DSS-induced colitis in NLRP6 and ASC deficient mice compared to the WT mice (149, 150). Thus, these studies undermine the protective effects of the inflammasome activation in IBD as otherwise suggested elsewhere (129). In another study, NLRP6 aggravated the allogeneic immune-mediated gastrointestinal graft-versus-host disease independent of changes in the composition of gut microbiome (151). Continued observations showed that mice lacking caspases-1 or NLRP3 exhibited significantly less severe clinical and pathological symptoms of colitis than their WT counterparts (152–154). These mice also showed reduced levels of IL-1β and IL-18 cytokines in the gut suggesting a prominent role of these cytokines in the progression of inflammasome-mediated intestinal inflammation. Recently, Fan et al. (155) showed no effect of increased caspase-11 activity on the severity of spontaneous chronic DSS-colitis in IL-10−/− mice. The effect of caspase-11 on the severity of chronic colitis was also not pronounced in double knockout caspase-11−/− IL-10−/− mice. However, the different effects of caspase-11 were observed between acute and chronic DSS models of colitis over a repeated and longer period of experimental follow-up. The investigators could not specify exact reasons for this discrepancy in their research outcomes. Another study observed improvement in the clinical DSS-colitis symptoms, reduced mortality and reduced goblet cell loss in caspase-1−/− mice compared to their WT mates normalized for intestinal microbiota (156). Contradicting observations on the protective role of GSDMD in colitis is published recently (157). The severity of acute DSS-induced colitis got attenuated in the gsdmd−/− mice. Other than the widely recognized pyroptosis inducing role of GSDMD, this study reported a novel non-pyroptotic role of GSDMD. GSDMD was shown to form IL-1β–containing small extracellular vesicles in IECs. Following a stimulated activation of caspase-8 and subsequent cleavage of GSDMD, a GSDMD-NEDD4 complex is assembled on the NLRP3/caspase-8 complex to produce mature IL-1β. This complete GSDMD-complex is loaded on LC3-II+ vesicles to release out of the cell and release mature IL-1β.
Human studies
In concordance with the above findings, human research also highlights the role of the inflammasome in IBD. Peripheral blood mononuclear cells from 60% of CD patients showed higher NLRP3 activation compared to the cells from 28.6% of control individuals (158). Similarly, colonic biopsies from quiescent or active UC and CD patients showed upregulated expression of NLRP3, IL-1β, caspase-1, and ASC (159). The colon biopsies from UC and CD patients reported 131-fold and 3.9 fold increase of NLRP6 expression in ileal CD and colonic CD patients respectively (131). The NLRP6 was localized to the IECs, myofibroblasts, neutrophils, and monocytic lineage cells of the lamina propria in CD patients evaluated. The colon biopsies from similar patients also showed increase in the mRNA levels of IL-1β, nlrp1, nlrp3, nlrc4, nlrp12 and aim2. NLRP3 was expressed by the intestinal neutrophils and lamina propria cells but not in the epithelial cell layer of active UC patients (139, 159). A genome-wide study suggested an association between NLRP7 signaling and inflammasome formation during IBD pathogenesis (160). Human studies showed upregulated inflammasome activity in CD patients with higher levels of IL-1β and IL-18 in intestinal cells (112). A study published in 1999, showed increased levels of IL-18 mRNA in the colon biopsies from CD patients (161). The localization of mature IL-18 was more in the IECs and LPMCs of CD patients. Individuals with human immunodeficiency virus (HIV) infection showed a concomitant increase in the circulating levels of IL-18 and LPS and reduced levels of IL-18 binding protein (IL-18BP) compared to the control subjects (162, 163). These individuals often demonstrate intestinal inflammation and compromised intestinal epithelial integrity. The disbalance between the circulating levels of IL-18 and IL-18BP in CD patients can be one reason behind intestinal inflammation and compromised barrier integrity (164). This discrepancy among the serum levels of IL-18 and IL-18BP was also reported in children with IBD (165). Similarly, excessive level of IL-1β is considered to increase susceptibility for intestinal inflammation. In patients with CARD8 mutation and IL-10 receptor abnormalities, IL-1β was shown to trigger intestinal inflammation independent of NLRP3 (112, 166, 167). Blocking IL-1β successfully suppressed inflammation (166). In a recent study, the nucleotide polymorphism in IL-18 and IL-1β genes among CD and UC patients has been corelated with a poor response to anti-TNF-α therapy (168).
In summary, research evidence suggests differential roles of inflammasome regulation in IBD (Table 1). Recurrent intestinal tissue injury and prolonged inflammation can switch the protective effects of inflammasomes to detrimental effects. Moreover, the shaping of intestinal microbiota structure can have a fundamental role in differential research outcomes from these studies.
Table 1:
Experimental animal studies | Human studies | |
---|---|---|
NLR family proteins | • Attenuation or exacerbation of colitis • Maintenance of epithelial barrier integrity, repair and recovery • Gut homeostasis and mucosal renewal • Maintenance of intestinal mucus layer • Alteration or no effect on microbiota • Induction of T-cell activation |
• Increased severity to intestinal inflammation • Increased activation in CD and UC patients • Increased expression in intestinal epithelial cells, neutrophils and lamina propria cells |
Caspases | • Attenuation of colitis • Maintenance of intestinal epithelial barrier integrity • Release of IL-18 and IL-1β cytokines • Regulation of STAT1 signaling in IECs |
• Increased activation in CD and UC patients |
IL-18 | • Attenuation or exacerbation of colitis • Controlling the outgrowth of colitogenic bacteria and maintenance of intestinal microbiota • Maintenance of intestinal barrier function and mucosal renewal • Induces release of other cytokines |
• Increased severity to intestinal inflammation • Increased activation in CD patients • Maintenance of intestinal barrier integrity • Increased expression in intestinal epithelial cells and lamina propria cells |
IL-1β | • No dominant role in intestinal inflammation • Activation of T-cell immune response |
• Increased activation in CD and UC patients • Increased severity to intestinal inflammation |
UC: Ulcerative colitis, CD: Crohn’s disease, IECs: Intestinal epithelial cells
INFLAMMASOME INHIBITORY MOLECULES AS A THERAPEUTIC APPROACH IN IBD
To date, several physiological, pharmacological, and inhibitors of inflammasome have been discovered. These inhibitors target either upstream or downstream effects of the inflammasome pathways and block NLRP3, TLR receptors, DAMPs, PAMPS, P2X7 receptor, ASC oligomerization, NACTH as well as inflammasome associated cytokines (Table 2).
Table 2:
Category of inhibitors | Drug/molecule | Experimental outcome/mechanism of action/clinical trial status | Reference |
---|---|---|---|
NLRP3 inhibitors | MCC950 | • Significantly ameliorated colitis in mice • Reduced ASC oligomerization and colon levels of IL-1β and IL-18 cytokines • Binding to NACTH domain Walker B motif • Inhibits ATP hydrolysis |
(169, 170, 174, 176) |
Oridonin | • Attenuated TNBS-induced CD and colitis in mice • Inhibits NLRP3 activation via covalently binding to NACTH domain and blocks interaction with NEK7 |
(178–180) |
|
Tranilast | • Directly inhibit NLRP3 by binding NACHT domain and block oligomerization • Attenuated gouty arthritis, cryopyrin-associated autoinflammatory syndromes, and type 2 diabetes |
(208) | |
OLT1177 | • Reduced ATPase activity of recombinant NLRP3 protein • Prevented NLRP3-ASC and Nlrp3-caspase-1 interactions • Reduced caspase-1 activity, IL-1β and IL-18 |
(209) | |
β-hydroxybutyrate | • Attenuates Muckle-Wells Syndrome, Familial Cold Autoinflammatory syndrome and Gout flares • Inhibits NLRP3 by blocking K+ efflux, reducing ASC oligomerization and speck formation • Reduces IL-1β and IL-18 cytokine maturation |
(210, 211) | |
NSAIDs | • Fenamate inhibited IL-1β release and ASC speck formation | (212) | |
Dopamine | • Regulate NLRP3 inflammasome activation • Ameliorated neurotoxin-induced neuroinflammation, LPS-induced systemic inflammation, and monosodium urate crystal-induced peritoneal inflammation in mice |
(213) | |
Micro RNA | miR-223 | • Post-transcriptional regulator of NLRP3 • Nanoparticle-mediated overexpression of miR-223 ameliorated colitis |
(186) |
Antioxidant | Curcumin | • Reduces K+ efflux, ROS, and cathepsin B • Alleviated DSS-induced colitis in mice • Combined treatment of curcumin with mesalamine increased clinical remission in patients with colitis |
(190, 191) |
Antagonist | Anakinra | • IL-1β receptor antagonist • Initiated Phase-2 clinical trial for colitis |
(203) |
Pyroptosis inhibitor |
Disulfiram | • Blocks GSDMD-mediated pore formation and IL-1β • Reduced mortality in LPS-induced sepsis in mice |
(205) |
Dimethyl fumarate | • Inhibited GSDMD interaction with caspases • Ameliorated multiple sclerosis and familial Mediterranean fever in mice |
(214) | |
VX-765 | • Inhibited atheroma and progression of atherosclerosis in mice and ameliorated multiple sclerosis • Reduced pyroptosis and IL-1β processing by caspase-1 |
(206, 207) |
DSS: dextran sulfate sodium, TNBS: trinitrobenzene sulfonic acid, ROS: reactive oxygen species, NSAIDs: Non-steroidal anti-inflammatory drugs, VRAC: volume-regulated anion channel, GSDMD: gasdermin-D, LPS: lipopolysaccharide, CD: Crohn’s disease
Inhibitors that directly block NLRP3 or reduce the levels of NLRP3 are the ones that are primarily evaluated for inhibiting inflammasome. A pharmacological inhibitor of both canonical and non-canonical activation of NLRP3 can benefit by binding to the NACTH domain of the Walker B motif and by inhibiting ATP hydrolysis (169–172). Such an inhibitor, MCC950 has completed Phase-1 clinical trials for rheumatoid arthritis and can be developed as a potential treatment for IBD (173). MCC950 treatment alleviated the experimental chronic or acute colitis in mice by inhibiting ASC oligomerization, caspase-1 dependent activation of IL-1β, IL-18, and reducing the levels of pro-inflammatory cytokines (174, 175). In NOD2k/o mice, MCC950 treatment significantly decreased the clinical and pathological symptoms of DSS-induced colitis (176). A recent study also showed that combinational therapy of metformin and MCC950 significantly ameliorated the DSS-induced colitis in mice (177). Treated animals showed reduced clinical and histopathological symptoms of colitis along with reduced TLR4/NF-kB signaling (effect of metformin) and inhibition of NLRP3 activation and caspase-1 activity (effect of MCC950) (177). The positive outcomes from these studies project MCC950 as a potential treatment for IBD. However, more studies are required in this direction. Oridonin, another plant-derived product and covalent inhibitor of NLRP3 was shown to suppress experimental colitis in mice (178–180).
MicroRNAs (miR’s) can be used as post-transcriptional regulators of NLRP3 inflammasomes by negatively regulating NLRP3 protein production and activation (181). However, the role of miR-223 has been ambiguous in IBD (182–185). Intestinal biopsies from active IBD patients showed increased expression of miR-233 (186, 187). On the contrary, in mir-223-/y mice, colitis was exacerbated with increased clinical disease pathology (184, 186). The condition got reversed following treatment with nanoparticle loaded hyperexpression of miR-223 resulting in reduced expression of NLRP3 and IL-1β (186). This suggested that, further studies are warranted before exploring miR-223 as a treatment for IBD.
Antioxidants are shown to reverse NLRP3 activation (188). Curcumin a hydrophobic polyphenol derived from turmeric has been investigated as a treatment for IBD (189). Curcumin treatment reduced clinical symptoms of murine DSS-colitis (190). DSS-induced ROS release, K+ efflux, and activation of cathepsin B were inhibited in curcumin-treated colitis mice (190). Clinical studies have also reported the therapeutic effects of curcumin in colitis patients (191, 192). However, one study showed no added advantage of curcumin treatment over placebo in randomized control trials (193). Similarly, flavonoid VI-16, plumercin, procyanidin, dimethyl fumarate, and caffeic acid phenethyl ester are found to be therapeutically effective in ameliorating experimental mouse models of colitis. The effects appear to be by inhibition of ROS release and NLRP3 (194–198).
Several other molecules inhibiting caspase-1, IL-1β, GSDMD, PAMP, and DAMP have also been proposed for regulating inflammasome activation in immune-mediated disorders. L-sulforaphane, a phytochemical derived from broccoli and a potential caspase-1 inhibitor is currently in clinical trials for several immune-mediated disorders like asthma, allergy, and rhinitis (199). Sulforaphane can also be explored for the treatment of IBD (200). Canakinumab (IL-1β antibody) and Anakinra (IL-1β receptor antagonist) are FDA approved therapies for cryopyrin-associated periodic syndromes and can be explored for the potential treatment for IBD (201, 202). Recently a Phase-2 clinical trial (Trial registration # ISRCTN43717130) investigates the use of anakinra in combination with corticosteroid treatment in patients with acute UC (203). Another study showed that IL-1Ra loaded alginate/chitosan microcapsule administration alleviated the clinical and pathological symptoms of DSS-colitis in mice (204). An FDA-approved drug disulfiram has been shown to inhibit pyroptosis in LPS-primed THP-1 macrophages by blocking both caspases and GSDMD-mediated pore formation (205). Disulfiram treatment improved survival rates, reduced TNF-α and IL-6 secretion and reduced GSDMD activation against LPS-induced experimental sepsis in mice (205). VX-765 is a specific inhibitor of caspase-1 used against myocardial infraction. VX-765 treatment reduced the caspase-1 activity, levels of p20 and IL-1β and inhibited pyroptosis in vascular smooth muscle cells treated with OxLDL (206). VX-765 also inhibited proinflammatory caspases and subsequent pyroptosis to reduce inflammation in experimental autoimmune encephalomyelitis (207). These studies provide evidence that inhibitors of pyroptosis (GSDMD and caspases inhibitors) can be used to reduce inflammation, however further studies are required to evaluate these drugs against intestinal inflammation.
FUTURE PERSPECTIVES
The function of the inflammasome in IBD is complex with no conclusive outcomes from clinical and experimental studies. This complexity of the inflammasome pathway and underexplored IBD pathology makes it difficult to completely understand the association between inflammasome activation and intestinal inflammation. Experimental approaches should be revisited using suitable and advance animal models with appropriate littermate controls, genetic background, considering the role of dietary components, housing conditions, and microbiota composition to establish the critical relationship between inflammasome and IBD. The development of advanced 3D organoid models may provide a niche in studying the role of the inflammasome in IBD. For now, targeting inflammasome appears to be a promising and lucrative treatment approach for IBD; clinical and long-term safety trials with a larger sample size should be conducted to determine the therapeutic efficacy of inflammasome inhibitory compounds in IBD. Advancing our understandings on the inflammasomes will aid in discovering novel pharmacological inhibitors. However, the variability in patient responses to therapeutics in IBD and combinational therapeutic efficacy of inflammasome targeting drugs along with the on-going remission following treatments is an important question to be answered. Furthermore, we do not know if abrupt and complete shutting of the inflammasome pathway in the gut may lead to severe side effects or even serious exacerbation of the inflammatory condition. The development of pharmacological drugs targeting upstream and downstream inflammasome pathways in the gut and clinical studies rationalizing the optimal required dosage is required. Overall, it is important to develop inflammasome-targeted IBD therapeutics and bring them to the clinic, which will benefit millions of patients who are suffering from IBD and colitis and will be a rewarding effort.
ACKNOWLEDGEMENTS:
The financial supports by National Institutes of Health (NIH), USA grant # RO1AI116441 (RK), and Blazer Foundation of Rockford, USA grant # 344524, are gratefully acknowledged. Biorender Inc. software was used to create the figures.
NONSTANDARD ABBREVIATIONS:
- IBD
Inflammatory bowel disease
- CD
Crohn’s disease
- UC
Ulcerative colitis
- 5-ASA
5-aminosalicylic acid
- PAMPs
pathogen-associated molecular patterns
- DAMPs
danger-associated molecular patterns
- PRR
pattern recognition receptors
- NLR
nucleotide-binding domain and leucine-rich repeat receptors
- NLRP
NLR pyrin domain-containing family
- NLRC4
NLR family caspase recruitment domain-containing protein-4
- AIM2
double-stranded DNA sensors absent in melanoma-2
- NAIP
NLR apoptosis inhibitory protein
- CARD
caspase activation and recruitment domain
- ASC
apoptosis-associated speck-like protein containing CARD
- PYD
amino-terminal pyrin domain
- NACTH
N-terminal central nucleotide-binding and oligomerization domain
- LRR
carboxy-terminal leucine-rich repeat domain
- MAPK
mitogen-activated protein kinase
- ATP
adenosine triphosphate
- GBP
guanylate binding proteins
- TRIF
TIR domain-containing adapter-inducing interferon-β
- RIPK
receptor interacting protein kinases
- FADD
Fas-associated protein with death domain
- DSS
dextran sulfate sodium
- IRGM
immunity-related GTPase M
Footnotes
CONFLICT OF INTEREST STATEMENT: The authors report no conflicts of interest.
REFERENCES
- 1.Collaborators, G. B. D. I. B. D. (2020) The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 5, 17–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Man SM (2018) Inflammasomes in the gastrointestinal tract: infection, cancer and gut microbiota homeostasis. Nat Rev Gastroenterol Hepatol 15, 721–737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kirsner JB (2001) Historical origins of current IBD concepts. World J Gastroenterol 7, 175–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Samuel W (1859) Morbid appearances in the intestine of Miss Bankes. Med Times Gazette 2, 2 [Google Scholar]
- 5.Crohn BB, Ginzburg L, and Oppenheimer GD (1984) Landmark article Oct 15, 1932. Regional ileitis. A pathological and clinical entity. By Burril B. Crohn, Leon Ginzburg, and Gordon D. Oppenheimer. JAMA 251, 73–79 [DOI] [PubMed] [Google Scholar]
- 6.de Campos Silva EF, Baima JP, de Barros JR, Tanni SE, Schreck T, Saad-Hossne R, and Sassaki LY (2020) Risk factors for ulcerative colitis-associated colorectal cancer: A retrospective cohort study. Medicine (Baltimore) 99, e21686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Khatri V, Amdare N, Tarnekar A, Goswami K, and Reddy MV (2015) Brugia malayi cystatin therapeutically ameliorates dextran sulfate sodium-induced colitis in mice. J Dig Dis 16, 585–594 [DOI] [PubMed] [Google Scholar]
- 8.Feuerstein JD, Moss AC, and Farraye FA (2019) Ulcerative Colitis. Mayo Clin Proc 94, 1357–1373 [DOI] [PubMed] [Google Scholar]
- 9.Torres J, Mehandru S, Colombel JF, and Peyrin-Biroulet L (2017) Crohn’s disease. Lancet 389, 1741–1755 [DOI] [PubMed] [Google Scholar]
- 10.Hugot JP, Alberti C, Berrebi D, Bingen E, and Cezard JP (2003) Crohn’s disease: the cold chain hypothesis. Lancet 362, 2012–2015 [DOI] [PubMed] [Google Scholar]
- 11.Mak WY, Zhao M, Ng SC, and Burisch J (2020) The epidemiology of inflammatory bowel disease: East meets west. J Gastroenterol Hepatol 35, 380–389 [DOI] [PubMed] [Google Scholar]
- 12.Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, Panaccione R, Ghosh S, Wu JCY, Chan FKL, Sung JJY, and Kaplan GG (2018) Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 390, 2769–2778 [DOI] [PubMed] [Google Scholar]
- 13.De Vroey B, and Colombel JF (2011) IBD in 2010: optimizing treatment and minimizing adverse events. Nat Rev Gastroenterol Hepatol 8, 74–76 [DOI] [PubMed] [Google Scholar]
- 14.Morrison G, Headon B, and Gibson P (2009) Update in inflammatory bowel disease. Aust Fam Physician 38, 956–961 [PubMed] [Google Scholar]
- 15.Verstockt B, Ferrante M, Vermeire S, and Van Assche G (2018) New treatment options for inflammatory bowel diseases. J Gastroenterol 53, 585–590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Frolkis AD, Dykeman J, Negron ME, Debruyn J, Jette N, Fiest KM, Frolkis T, Barkema HW, Rioux KP, Panaccione R, Ghosh S, Wiebe S, and Kaplan GG (2013) Risk of surgery for inflammatory bowel diseases has decreased over time: a systematic review and meta-analysis of population-based studies. Gastroenterology 145, 996–1006 [DOI] [PubMed] [Google Scholar]
- 17.Peyrin-Biroulet L, Loftus EV Jr., Colombel JF, and Sandborn WJ (2010) The natural history of adult Crohn’s disease in population-based cohorts. Am J Gastroenterol 105, 289–297 [DOI] [PubMed] [Google Scholar]
- 18.Mishra R, Dhawan P, Srivastava AS, and Singh AB (2020) Inflammatory bowel disease: Therapeutic limitations and prospective of the stem cell therapy. World J Stem Cells 12, 1050–1066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Allegretti JR, Barnes EL, and Cameron A (2015) Are patients with inflammatory bowel disease on chronic immunosuppressive therapy at increased risk of cervical high-grade dysplasia/cancer? A meta-analysis. Inflamm Bowel Dis 21, 1089–1097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Long MD, Martin CF, Pipkin CA, Herfarth HH, Sandler RS, and Kappelman MD (2012) Risk of melanoma and nonmelanoma skin cancer among patients with inflammatory bowel disease. Gastroenterology 143, 390–399 e391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wasan SK, Coukos JA, and Farraye FA (2011) Vaccinating the inflammatory bowel disease patient: deficiencies in gastroenterologists knowledge. Inflamm Bowel Dis 17, 2536–2540 [DOI] [PubMed] [Google Scholar]
- 22.Melmed GY, Ippoliti AF, Papadakis KA, Tran TT, Birt JL, Lee SK, Frenck RW, Targan SR, and Vasiliauskas EA (2006) Patients with inflammatory bowel disease are at risk for vaccine-preventable illnesses. Am J Gastroenterol 101, 1834–1840 [DOI] [PubMed] [Google Scholar]
- 23.de Zoete MR, Palm NW, Zhu S, and Flavell RA (2014) Inflammasomes. Cold Spring Harb Perspect Biol 6, a016287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Amarante-Mendes GP, Adjemian S, Branco LM, Zanetti LC, Weinlich R, and Bortoluci KR (2018) Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front Immunol 9, 2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zheng D, Liwinski T, and Elinav E (2020) Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discov 6, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sharif H, Wang L, Wang WL, Magupalli VG, Andreeva L, Qiao Q, Hauenstein AV, Wu Z, Nunez G, Mao Y, and Wu H (2019) Structural mechanism for NEK7-licensed activation of NLRP3 inflammasome. Nature 570, 338–343 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang L, Chen S, Ruan J, Wu J, Tong AB, Yin Q, Li Y, David L, Lu A, Wang WL, Marks C, Ouyang Q, Zhang X, Mao Y, and Wu H (2015) Cryo-EM structure of the activated NAIP2-NLRC4 inflammasome reveals nucleated polymerization. Science 350, 404–409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Maekawa S, Ohto U, Shibata T, Miyake K, and Shimizu T (2016) Crystal structure of NOD2 and its implications in human disease. Nat Commun 7, 11813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yang X, Yang F, Wang W, Lin G, Hu Z, Han Z, Qi Y, Zhang L, Wang J, Sui SF, and Chai J (2018) Structural basis for specific flagellin recognition by the NLR protein NAIP5. Cell Res 28, 35–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Boucher D, Monteleone M, Coll RC, Chen KW, Ross CM, Teo JL, Gomez GA, Holley CL, Bierschenk D, Stacey KJ, Yap AS, Bezbradica JS, and Schroder K (2018) Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. J Exp Med 215, 827–840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, Schroder GF, Fitzgerald KA, Wu H, and Egelman EH (2014) Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell 156, 1193–1206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dick MS, Sborgi L, Ruhl S, Hiller S, and Broz P (2016) ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun 7, 11929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shi J, Gao W, and Shao F (2017) Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem Sci 42, 245–254 [DOI] [PubMed] [Google Scholar]
- 34.Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, and Shao F (2015) Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 [DOI] [PubMed] [Google Scholar]
- 35.Liu Z, Wang C, Yang J, Chen Y, Zhou B, Abbott DW, and Xiao TS (2020) Caspase-1 Engages Full-Length Gasdermin D through Two Distinct Interfaces That Mediate Caspase Recruitment and Substrate Cleavage. Immunity 53, 106–114 e105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC, and Shao F (2016) Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535, 111–116 [DOI] [PubMed] [Google Scholar]
- 37.Martin-Sanchez F, Diamond C, Zeitler M, Gomez AI, Baroja-Mazo A, Bagnall J, Spiller D, White M, Daniels MJ, Mortellaro A, Penalver M, Paszek P, Steringer JP, Nickel W, Brough D, and Pelegrin P (2016) Inflammasome-dependent IL-1beta release depends upon membrane permeabilisation. Cell Death Differ 23, 1219–1231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Prochnicki T, Mangan MS, and Latz E (2016) Recent insights into the molecular mechanisms of the NLRP3 inflammasome activation. F1000Res 5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kelley N, Jeltema D, Duan Y, and He Y (2019) The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci 20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Vanaja SK, Russo AJ, Behl B, Banerjee I, Yankova M, Deshmukh SD, and Rathinam VAK (2016) Bacterial Outer Membrane Vesicles Mediate Cytosolic Localization of LPS and Caspase-11 Activation. Cell 165, 1106–1119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mazgaeen L, and Gurung P (2020) Recent Advances in Lipopolysaccharide Recognition Systems. Int J Mol Sci 21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Baker PJ, Boucher D, Bierschenk D, Tebartz C, Whitney PG, D’Silva DB, Tanzer MC, Monteleone M, Robertson AA, Cooper MA, Alvarez-Diaz S, Herold MJ, Bedoui S, Schroder K, and Masters SL (2015) NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur J Immunol 45, 2918–2926 [DOI] [PubMed] [Google Scholar]
- 43.Ruhl S, and Broz P (2015) Caspase-11 activates a canonical NLRP3 inflammasome by promoting K(+) efflux. Eur J Immunol 45, 2927–2936 [DOI] [PubMed] [Google Scholar]
- 44.Santos JC, Boucher D, Schneider LK, Demarco B, Dilucca M, Shkarina K, Heilig R, Chen KW, Lim RYH, and Broz P (2020) Human GBP1 binds LPS to initiate assembly of a caspase-4 activating platform on cytosolic bacteria. Nat Commun 11, 3276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wandel MP, Kim BH, Park ES, Boyle KB, Nayak K, Lagrange B, Herod A, Henry T, Zilbauer M, Rohde J, MacMicking JD, and Randow F (2020) Guanylate-binding proteins convert cytosolic bacteria into caspase-4 signaling platforms. Nat Immunol 21, 880–891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gomes MTR, Cerqueira DM, Guimaraes ES, Campos PC, and Oliveira SC (2019) Guanylate-binding proteins at the crossroad of noncanonical inflammasome activation during bacterial infections. J Leukoc Biol 106, 553–562 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Balakrishnan A, Karki R, Berwin B, Yamamoto M, and Kanneganti TD (2018) Guanylate binding proteins facilitate caspase-11-dependent pyroptosis in response to type 3 secretion system-negative Pseudomonas aeruginosa. Cell Death Discov 4, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gaidt MM, Ebert TS, Chauhan D, Schmidt T, Schmid-Burgk JL, Rapino F, Robertson AA, Cooper MA, Graf T, and Hornung V (2016) Human Monocytes Engage an Alternative Inflammasome Pathway. Immunity 44, 833–846 [DOI] [PubMed] [Google Scholar]
- 49.Mezzasoma L, Talesa VN, Romani R, and Bellezza I (2020) ANP and BNP Exert Anti-Inflammatory Action via NPR-1/cGMP Axis by Interfering with Canonical, Non-Canonical, and Alternative Routes of Inflammasome Activation in Human THP1 Cells. Int J Mol Sci 22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Martinez-Garcia JJ, Martinez-Banaclocha H, Angosto-Bazarra D, de Torre-Minguela C, Baroja-Mazo A, Alarcon-Vila C, Martinez-Alarcon L, Amores-Iniesta J, Martin-Sanchez F, Ercole GA, Martinez CM, Gonzalez-Lisorge A, Fernandez-Pacheco J, Martinez-Gil P, Adriouch S, Koch-Nolte F, Lujan J, Acosta-Villegas F, Parrilla P, Garcia-Palenciano C, and Pelegrin P (2019) P2X7 receptor induces mitochondrial failure in monocytes and compromises NLRP3 inflammasome activation during sepsis. Nat Commun 10, 2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cao F, Hu LQ, Yao SR, Hu Y, Wang DG, Fan YG, Pan GX, Tao SS, Zhang Q, Pan HF, and Wu GC (2019) P2X7 receptor: A potential therapeutic target for autoimmune diseases. Autoimmun Rev 18, 767–777 [DOI] [PubMed] [Google Scholar]
- 52.Verma D, Fekri SZ, Sigurdardottir G, Bivik Eding C, Sandin C, and Enerback C (2020) Enhanced Inflammasome Activity in Patients with Psoriasis Promotes Systemic Inflammation. J Invest Dermatol [DOI] [PubMed] [Google Scholar]
- 53.Jin T, Curry J, Smith P, Jiang J, and Xiao TS (2013) Structure of the NLRP1 caspase recruitment domain suggests potential mechanisms for its association with procaspase-1. Proteins 81, 1266–1270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Faustin B, Lartigue L, Bruey JM, Luciano F, Sergienko E, Bailly-Maitre B, Volkmann N, Hanein D, Rouiller I, and Reed JC (2007) Reconstituted NALP1 inflammasome reveals two-step mechanism of caspase-1 activation. Mol Cell 25, 713–724 [DOI] [PubMed] [Google Scholar]
- 55.Taabazuing CY, Griswold AR, and Bachovchin DA (2020) The NLRP1 and CARD8 inflammasomes. Immunol Rev 297, 13–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.D’Osualdo A, Weichenberger CX, Wagner RN, Godzik A, Wooley J, and Reed JC (2011) CARD8 and NLRP1 undergo autoproteolytic processing through a ZU5-like domain. PLoS One 6, e27396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Finger JN, Lich JD, Dare LC, Cook MN, Brown KK, Duraiswami C, Bertin J, and Gough PJ (2012) Autolytic proteolysis within the function to find domain (FIIND) is required for NLRP1 inflammasome activity. J Biol Chem 287, 25030–25037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Levinsohn JL, Newman ZL, Hellmich KA, Fattah R, Getz MA, Liu S, Sastalla I, Leppla SH, and Moayeri M (2012) Anthrax lethal factor cleavage of Nlrp1 is required for activation of the inflammasome. PLoS Pathog 8, e1002638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sandstrom A, Mitchell PS, Goers L, Mu EW, Lesser CF, and Vance RE (2019) Functional degradation: A mechanism of NLRP1 inflammasome activation by diverse pathogen enzymes. Science 364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chui AJ, Okondo MC, Rao SD, Gai K, Griswold AR, Johnson DC, Ball DP, Taabazuing CY, Orth EL, Vittimberga BA, and Bachovchin DA (2019) N-terminal degradation activates the NLRP1B inflammasome. Science 364, 82–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gai K, Okondo MC, Rao SD, Chui AJ, Ball DP, Johnson DC, and Bachovchin DA (2019) DPP8/9 inhibitors are universal activators of functional NLRP1 alleles. Cell Death Dis 10, 587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Okondo MC, Johnson DC, Sridharan R, Go EB, Chui AJ, Wang MS, Poplawski SE, Wu W, Liu Y, Lai JH, Sanford DG, Arciprete MO, Golub TR, Bachovchin WW, and Bachovchin DA (2017) DPP8 and DPP9 inhibition induces pro-caspase-1-dependent monocyte and macrophage pyroptosis. Nat Chem Biol 13, 46–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhong FL, Robinson K, Teo DET, Tan KY, Lim C, Harapas CR, Yu CH, Xie WH, Sobota RM, Au VB, Hopkins R, D’Osualdo A, Reed JC, Connolly JE, Masters SL, and Reversade B (2018) Human DPP9 represses NLRP1 inflammasome and protects against autoinflammatory diseases via both peptidase activity and FIIND domain binding. J Biol Chem 293, 18864–18878 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Levy M, Shapiro H, Thaiss CA, and Elinav E (2017) NLRP6: A Multifaceted Innate Immune Sensor. Trends Immunol 38, 248–260 [DOI] [PubMed] [Google Scholar]
- 65.Janani C, and Ranjitha Kumari BD (2015) PPAR gamma gene--a review. Diabetes Metab Syndr 9, 46–50 [DOI] [PubMed] [Google Scholar]
- 66.Nie H, Hu Y, Guo W, Wang W, Yang Q, Dong Q, Tang Y, Li Q, and Tang Z (2020) miR-331-3p Inhibits Inflammatory Response after Intracerebral Hemorrhage by Directly Targeting NLRP6. Biomed Res Int 2020, 6182464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zheng D, Kern L, and Elinav E (2020) The NLRP6 inflammasome. Immunology [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mukherjee S, Kumar R, Tsakem Lenou E, Basrur V, Kontoyiannis DL, Ioakeimidis F, Mosialos G, Theiss AL, Flavell RA, and Venuprasad K (2020) Deubiquitination of NLRP6 inflammasome by Cyld critically regulates intestinal inflammation. Nat Immunol 21, 626–635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tian X, Li R, Liu C, Liu F, Yang LJ, Wang SP, and Wang CL (2020) NLRP6-caspase 4 inflammasome activation in response to cariogenic bacterial lipoteichoic acid in human dental pulp inflammation. Int Endod J [DOI] [PubMed] [Google Scholar]
- 70.Leng F, Yin H, Qin S, Zhang K, Guan Y, Fang R, Wang H, Li G, Jiang Z, Sun F, Wang DC, and Xie C (2020) NLRP6 self-assembles into a linear molecular platform following LPS binding and ATP stimulation. Sci Rep 10, 198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hara H, Seregin SS, Yang D, Fukase K, Chamaillard M, Alnemri ES, Inohara N, Chen GY, and Nunez G (2018) The NLRP6 Inflammasome Recognizes Lipoteichoic Acid and Regulates Gram-Positive Pathogen Infection. Cell 175, 1651–1664 e1614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Shen C, Lu A, Xie WJ, Ruan J, Negro R, Egelman EH, Fu TM, and Wu H (2019) Molecular mechanism for NLRP6 inflammasome assembly and activation. Proc Natl Acad Sci U S A 116, 2052–2057 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lu A, Li Y, Yin Q, Ruan J, Yu X, Egelman E, and Wu H (2015) Plasticity in PYD assembly revealed by cryo-EM structure of the PYD filament of AIM2. Cell Discov 1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gram AM, Wright JA, Pickering RJ, Lam NL, Booty LM, Webster SJ, and Bryant CE (2020) Salmonella Flagellin Activates NAIP/NLRC4 and Canonical NLRP3 Inflammasomes in Human Macrophages. J Immunol [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Reyes Ruiz VM, Ramirez J, Naseer N, Palacio NM, Siddarthan IJ, Yan BM, Boyer MA, Pensinger DA, Sauer JD, and Shin S (2017) Broad detection of bacterial type III secretion system and flagellin proteins by the human NAIP/NLRC4 inflammasome. Proc Natl Acad Sci U S A 114, 13242–13247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Miao EA, Mao DP, Yudkovsky N, Bonneau R, Lorang CG, Warren SE, Leaf IA, and Aderem A (2010) Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasome. Proc Natl Acad Sci U S A 107, 3076–3080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bauer R, and Rauch I (2020) The NAIP/NLRC4 inflammasome in infection and pathology. Mol Aspects Med 76, 100863. [DOI] [PubMed] [Google Scholar]
- 78.Broz P, von Moltke J, Jones JW, Vance RE, and Monack DM (2010) Differential requirement for Caspase-1 autoproteolysis in pathogen-induced cell death and cytokine processing. Cell Host Microbe 8, 471–483 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Duncan JA, and Canna SW (2018) The NLRC4 Inflammasome. Immunol Rev 281, 115–123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Burckstummer T, Baumann C, Bluml S, Dixit E, Durnberger G, Jahn H, Planyavsky M, Bilban M, Colinge J, Bennett KL, and Superti-Furga G (2009) An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol 10, 266–272 [DOI] [PubMed] [Google Scholar]
- 81.Fernandes-Alnemri T, Yu JW, Datta P, Wu J, and Alnemri ES (2009) AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458, 509–513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E, and Fitzgerald KA (2009) AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458, 514–518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhang H, Luo J, Alcorn JF, Chen K, Fan S, Pilewski J, Liu A, Chen W, Kolls JK, and Wang J (2017) AIM2 Inflammasome Is Critical for Influenza-Induced Lung Injury and Mortality. J Immunol 198, 4383–4393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jin T, Perry A, Jiang J, Smith P, Curry JA, Unterholzner L, Jiang Z, Horvath G, Rathinam VA, Johnstone RW, Hornung V, Latz E, Bowie AG, Fitzgerald KA, and Xiao TS (2012) Structures of the HIN domain:DNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity 36, 561–571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wang L, Sun L, Byrd KM, Ko CC, Zhao Z, and Fang J (2020) AIM2 Inflammasome’s First Decade of Discovery: Focus on Oral Diseases. Front Immunol 11, 1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Morrone SR, Matyszewski M, Yu X, Delannoy M, Egelman EH, and Sohn J (2015) Assembly-driven activation of the AIM2 foreign-dsDNA sensor provides a polymerization template for downstream ASC. Nat Commun 6, 7827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Maruzuru Y, Koyanagi N, Kato A, and Kawaguchi Y (2020) Role of the DNA Binding Activity of Herpes Simplex Virus 1 VP22 in Evading AIM2-Dependent Inflammasome Activation Induced by the Virus. J Virol [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Khare S, Ratsimandresy RA, de Almeida L, Cuda CM, Rellick SL, Misharin AV, Wallin MC, Gangopadhyay A, Forte E, Gottwein E, Perlman H, Reed JC, Greaves DR, Dorfleutner A, and Stehlik C (2014) The PYRIN domain-only protein POP3 inhibits ALR inflammasomes and regulates responses to infection with DNA viruses. Nat Immunol 15, 343–353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wang B, Bhattacharya M, Roy S, Tian Y, and Yin Q (2020) Immunobiology and structural biology of AIM2 inflammasome. Mol Aspects Med 76, 100869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Schnappauf O, Chae JJ, Kastner DL, and Aksentijevich I (2019) The Pyrin Inflammasome in Health and Disease. Front Immunol 10, 1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Papin S, Cuenin S, Agostini L, Martinon F, Werner S, Beer HD, Grutter C, Grutter M, and Tschopp J (2007) The SPRY domain of Pyrin, mutated in familial Mediterranean fever patients, interacts with inflammasome components and inhibits proIL-1beta processing. Cell Death Differ 14, 1457–1466 [DOI] [PubMed] [Google Scholar]
- 92.Jamilloux Y, Magnotti F, Belot A, and Henry T (2018) The pyrin inflammasome: from sensing RhoA GTPases-inhibiting toxins to triggering autoinflammatory syndromes. Pathog Dis 76 [DOI] [PubMed] [Google Scholar]
- 93.Xu H, Yang J, Gao W, Li L, Li P, Zhang L, Gong YN, Peng X, Xi JJ, Chen S, Wang F, and Shao F (2014) Innate immune sensing of bacterial modifications of Rho GTPases by the Pyrin inflammasome. Nature 513, 237–241 [DOI] [PubMed] [Google Scholar]
- 94.Heilig R, and Broz P (2018) Function and mechanism of the pyrin inflammasome. Eur J Immunol 48, 230–238 [DOI] [PubMed] [Google Scholar]
- 95.Gao W, Yang J, Liu W, Wang Y, and Shao F (2016) Site-specific phosphorylation and microtubule dynamics control Pyrin inflammasome activation. Proc Natl Acad Sci U S A 113, E4857–4866 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kile BT, Panopoulos AD, Stirzaker RA, Hacking DF, Tahtamouni LH, Willson TA, Mielke LA, Henley KJ, Zhang JG, Wicks IP, Stevenson WS, Nurden P, Watowich SS, and Justice MJ (2007) Mutations in the cofilin partner Aip1/Wdr1 cause autoinflammatory disease and macrothrombocytopenia. Blood 110, 2371–2380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kim ML, Chae JJ, Park YH, De Nardo D, Stirzaker RA, Ko HJ, Tye H, Cengia L, DiRago L, Metcalf D, Roberts AW, Kastner DL, Lew AM, Lyras D, Kile BT, Croker BA, and Masters SL (2015) Aberrant actin depolymerization triggers the pyrin inflammasome and autoinflammatory disease that is dependent on IL-18, not IL-1beta. J Exp Med 212, 927–938 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Standing AS, Malinova D, Hong Y, Record J, Moulding D, Blundell MP, Nowak K, Jones H, Omoyinmi E, Gilmour KC, Medlar A, Stanescu H, Kleta R, Anderson G, Nanthapisal S, Gomes SM, Klein N, Eleftheriou D, Thrasher AJ, and Brogan PA (2017) Autoinflammatory periodic fever, immunodeficiency, and thrombocytopenia (PFIT) caused by mutation in actin-regulatory gene WDR1. J Exp Med 214, 59–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jing W, Lo Pilato J, Kay C, and Man SM (2021) Activation mechanisms of inflammasomes by bacterial toxins. Cell Microbiol, e13309 [DOI] [PubMed] [Google Scholar]
- 100.Sharma D, Malik A, Guy CS, Karki R, Vogel P, and Kanneganti TD (2018) Pyrin Inflammasome Regulates Tight Junction Integrity to Restrict Colitis and Tumorigenesis. Gastroenterology 154, 948–964 e948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Alimov I, Menon S, Cochran N, Maher R, Wang Q, Alford J, Concannon JB, Yang Z, Harrington E, Llamas L, Lindeman A, Hoffman G, Schuhmann T, Russ C, Reece-Hoyes J, Canham SM, and Cai X (2019) Bile acid analogues are activators of pyrin inflammasome. J Biol Chem 294, 3359–3366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Loeven NA, Medici NP, and Bliska JB (2020) The pyrin inflammasome in host-microbe interactions. Curr Opin Microbiol 54, 77–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Khare S, Dorfleutner A, Bryan NB, Yun C, Radian AD, de Almeida L, Rojanasakul Y, and Stehlik C (2012) An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity 36, 464–476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mullins B, and Chen J (2020) NLRP9 in innate immunity and inflammation. Immunology [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mu J, Wang W, Chen B, Wu L, Li B, Mao X, Zhang Z, Fu J, Kuang Y, Sun X, Li Q, Jin L, He L, Sang Q, and Wang L (2019) Mutations in NLRP2 and NLRP5 cause female infertility characterised by early embryonic arrest. J Med Genet 56, 471–480 [DOI] [PubMed] [Google Scholar]
- 106.Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, Herfarth HH, Jobin C, and Ting JP (2010) The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med 207, 1045–1056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, and Kanneganti TD (2010) The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity 32, 379–391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hirota SA, Ng J, Lueng A, Khajah M, Parhar K, Li Y, Lam V, Potentier MS, Ng K, Bawa M, McCafferty DM, Rioux KP, Ghosh S, Xavier RJ, Colgan SP, Tschopp J, Muruve D, MacDonald JA, and Beck PL (2011) NLRP3 inflammasome plays a key role in the regulation of intestinal homeostasis. Inflamm Bowel Dis 17, 1359–1372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, McDowell J, Paskind M, Rodman L, Salfeld J, and et al. (1995) Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell 80, 401–411 [DOI] [PubMed] [Google Scholar]
- 110.Dupaul-Chicoine J, Yeretssian G, Doiron K, Bergstrom KS, McIntire CR, LeBlanc PM, Meunier C, Turbide C, Gros P, Beauchemin N, Vallance BA, and Saleh M (2010) Control of intestinal homeostasis, colitis, and colitis-associated colorectal cancer by the inflammatory caspases. Immunity 32, 367–378 [DOI] [PubMed] [Google Scholar]
- 111.Ranson N, Kunde D, and Eri R (2017) Regulation and Sensing of Inflammasomes and Their Impact on Intestinal Health. Int J Mol Sci 18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Mao L, Kitani A, Strober W, and Fuss IJ (2018) The Role of NLRP3 and IL-1beta in the Pathogenesis of Inflammatory Bowel Disease. Front Immunol 9, 2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Itani S, Watanabe T, Nadatani Y, Sugimura N, Shimada S, Takeda S, Otani K, Hosomi S, Nagami Y, Tanaka F, Kamata N, Yamagami H, Tanigawa T, Shiba M, Tominaga K, Fujiwara Y, and Arakawa T (2016) NLRP3 inflammasome has a protective effect against oxazolone-induced colitis: a possible role in ulcerative colitis. Sci Rep 6, 39075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Tatiya-Aphiradee N, Chatuphonprasert W, and Jarukamjorn K (2018) Immune response and inflammatory pathway of ulcerative colitis. J Basic Clin Physiol Pharmacol 30, 1–10 [DOI] [PubMed] [Google Scholar]
- 115.Inoue Y, Shirasuna K, Kimura H, Usui F, Kawashima A, Karasawa T, Tago K, Dezaki K, Nishimura S, Sagara J, Noda T, Iwakura Y, Tsutsui H, Taniguchi S, Yanagisawa K, Yada T, Yasuda Y, and Takahashi M (2014) NLRP3 regulates neutrophil functions and contributes to hepatic ischemia-reperfusion injury independently of inflammasomes. J Immunol 192, 4342–4351 [DOI] [PubMed] [Google Scholar]
- 116.Wera O, Lancellotti P, and Oury C (2016) The Dual Role of Neutrophils in Inflammatory Bowel Diseases. J Clin Med 5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Williams TM, Leeth RA, Rothschild DE, McDaniel DK, Coutermarsh-Ott SL, Simmons AE, Kable KH, Heid B, and Allen IC (2015) Caspase-11 attenuates gastrointestinal inflammation and experimental colitis pathogenesis. Am J Physiol Gastrointest Liver Physiol 308, G139–150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Demon D, Kuchmiy A, Fossoul A, Zhu Q, Kanneganti TD, and Lamkanfi M (2014) Caspase-11 is expressed in the colonic mucosa and protects against dextran sodium sulfate-induced colitis. Mucosal Immunol 7, 1480–1491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Oficjalska K, Raverdeau M, Aviello G, Wade SC, Hickey A, Sheehan KM, Corr SC, Kay EW, O’Neill LA, Mills KH, and Creagh EM (2015) Protective role for caspase-11 during acute experimental murine colitis. J Immunol 194, 1252–1260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Flood B, Manils J, Nulty C, Flis E, Kenealy S, Barber G, Fay J, Mills KHG, Kay EW, and Creagh EM (2019) Caspase-11 regulates the tumour suppressor function of STAT1 in a murine model of colitis-associated carcinogenesis. Oncogene 38, 2658–2674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Williams MA, O’Callaghan A, and Corr SC (2019) IL-33 and IL-18 in Inflammatory Bowel Disease Etiology and Microbial Interactions. Front Immunol 10, 1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Song-Zhao GX, Srinivasan N, Pott J, Baban D, Frankel G, and Maloy KJ (2014) Nlrp3 activation in the intestinal epithelium protects against a mucosal pathogen. Mucosal Immunol 7, 763–774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Yao X, Zhang C, Xing Y, Xue G, Zhang Q, Pan F, Wu G, Hu Y, Guo Q, Lu A, Zhang X, Zhou R, Tian Z, Zeng B, Wei H, Strober W, Zhao L, and Meng G (2017) Remodelling of the gut microbiota by hyperactive NLRP3 induces regulatory T cells to maintain homeostasis. Nat Commun 8, 1896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zaki MH, Vogel P, Body-Malapel M, Lamkanfi M, and Kanneganti TD (2010) IL-18 production downstream of the Nlrp3 inflammasome confers protection against colorectal tumor formation. J Immunol 185, 4912–4920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Takagi H, Kanai T, Okazawa A, Kishi Y, Sato T, Takaishi H, Inoue N, Ogata H, Iwao Y, Hoshino K, Takeda K, Akira S, Watanabe M, Ishii H, and Hibi T (2003) Contrasting action of IL-12 and IL-18 in the development of dextran sodium sulphate colitis in mice. Scand J Gastroenterol 38, 837–844 [DOI] [PubMed] [Google Scholar]
- 126.Dinarello CA (2018) Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol Rev 281, 8–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wlodarska M, Thaiss CA, Nowarski R, Henao-Mejia J, Zhang JP, Brown EM, Frankel G, Levy M, Katz MN, Philbrick WM, Elinav E, Finlay BB, and Flavell RA (2014) NLRP6 inflammasome orchestrates the colonic host-microbial interface by regulating goblet cell mucus secretion. Cell 156, 1045–1059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chen GY, Liu M, Wang F, Bertin J, and Nunez G (2011) A functional role for Nlrp6 in intestinal inflammation and tumorigenesis. J Immunol 186, 7187–7194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, and Flavell RA (2011) NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145, 745–757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Seregin SS, Golovchenko N, Schaf B, Chen J, Pudlo NA, Mitchell J, Baxter NT, Zhao L, Schloss PD, Martens EC, Eaton KA, and Chen GY (2017) NLRP6 Protects Il10(−/−) Mice from Colitis by Limiting Colonization of Akkermansia muciniphila. Cell Rep 19, 733–745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ranson N, Veldhuis M, Mitchell B, Fanning S, Cook AL, Kunde D, and Eri R (2018) Nod-Like Receptor Pyrin-Containing Protein 6 (NLRP6) Is Up-regulated in Ileal Crohn’s Disease and Differentially Expressed in Goblet Cells. Cell Mol Gastroenterol Hepatol 6, 110–112 e118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Normand S, Delanoye-Crespin A, Bressenot A, Huot L, Grandjean T, Peyrin-Biroulet L, Lemoine Y, Hot D, and Chamaillard M (2011) Nod-like receptor pyrin domain-containing protein 6 (NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon injury. Proc Natl Acad Sci U S A 108, 9601–9606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Seregin SS, Golovchenko N, Schaf B, Chen J, Eaton KA, and Chen GY (2017) NLRP6 function in inflammatory monocytes reduces susceptibility to chemically induced intestinal injury. Mucosal Immunol 10, 434–445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Yin J, Sheng B, Yang K, Sun L, Xiao W, and Yang H (2019) The protective roles of NLRP6 in intestinal epithelial cells. Cell Prolif 52, e12555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Swidsinski A, Loening-Baucke V, Theissig F, Engelhardt H, Bengmark S, Koch S, Lochs H, and Dorffel Y (2007) Comparative study of the intestinal mucus barrier in normal and inflamed colon. Gut 56, 343–350 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Volk JK, Nystrom EEL, van der Post S, Abad BM, Schroeder BO, Johansson A, Svensson F, Javerfelt S, Johansson MEV, Hansson GC, and Birchenough GMH (2019) The Nlrp6 inflammasome is not required for baseline colonic inner mucus layer formation or function. J Exp Med 216, 2602–2618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ma C, Yang D, Wang B, Wu C, Wu Y, Li S, Liu X, Lassen K, Dai L, and Yang S (2020) Gasdermin D in macrophages restrains colitis by controlling cGAS-mediated inflammation. Sci Adv 6, eaaz6717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, Perro M, Diestelhorst J, Allroth A, Murugan D, Hatscher N, Pfeifer D, Sykora KW, Sauer M, Kreipe H, Lacher M, Nustede R, Woellner C, Baumann U, Salzer U, Koletzko S, Shah N, Segal AW, Sauerbrey A, Buderus S, Snapper SB, Grimbacher B, and Klein C (2009) Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med 361, 2033–2045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Liu L, Dong Y, Ye M, Jin S, Yang J, Joosse ME, Sun Y, Zhang J, Lazarev M, Brant SR, Safar B, Marohn M, Mezey E, and Li X (2017) The Pathogenic Role of NLRP3 Inflammasome Activation in Inflammatory Bowel Diseases of Both Mice and Humans. J Crohns Colitis 11, 737–750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhang J, Fu S, Sun S, Li Z, and Guo B (2014) Inflammasome activation has an important role in the development of spontaneous colitis. Mucosal Immunol 7, 1139–1150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Mehto S, Jena KK, Nath P, Chauhan S, Kolapalli SP, Das SK, Sahoo PK, Jain A, Taylor GA, and Chauhan S (2019) The Crohn’s Disease Risk Factor IRGM Limits NLRP3 Inflammasome Activation by Impeding Its Assembly and by Mediating Its Selective Autophagy. Mol Cell 73, 429–445 e427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Finethy R, Dockterman J, Kutsch M, Orench-Rivera N, Wallace GD, Piro AS, Luoma S, Haldar AK, Hwang S, Martinez J, Kuehn MJ, Taylor GA, and Coers J (2020) Dynamin-related Irgm proteins modulate LPS-induced caspase-11 activation and septic shock. EMBO Rep 21, e50830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Eren E, Planes R, Bagayoko S, Bordignon PJ, Chaoui K, Hessel A, Santoni K, Pinilla M, Lagrange B, Burlet-Schiltz O, Howard JC, Henry T, Yamamoto M, and Meunier E (2020) Irgm2 and Gate-16 cooperatively dampen Gram-negative bacteria-induced caspase-11 response. EMBO Rep 21, e50829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chen X, Liu G, Yuan Y, Wu G, Wang S, and Yuan L (2019) NEK7 interacts with NLRP3 to modulate the pyroptosis in inflammatory bowel disease via NF-kappaB signaling. Cell Death Dis 10, 906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Broz P, Newton K, Lamkanfi M, Mariathasan S, Dixit VM, and Monack DM (2010) Redundant roles for inflammasome receptors NLRP3 and NLRC4 in host defense against Salmonella. J Exp Med 207, 1745–1755 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lebeis SL, Powell KR, Merlin D, Sherman MA, and Kalman D (2009) Interleukin-1 receptor signaling protects mice from lethal intestinal damage caused by the attaching and effacing pathogen Citrobacter rodentium. Infect Immun 77, 604–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Liu Z, Zaki MH, Vogel P, Gurung P, Finlay BB, Deng W, Lamkanfi M, and Kanneganti TD (2012) Role of inflammasomes in host defense against Citrobacter rodentium infection. J Biol Chem 287, 16955–16964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hasegawa M, Kamada N, Jiao Y, Liu MZ, Nunez G, and Inohara N (2012) Protective role of commensals against Clostridium difficile infection via an IL-1beta-mediated positive-feedback loop. J Immunol 189, 3085–3091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Mamantopoulos M, Ronchi F, Van Hauwermeiren F, Vieira-Silva S, Yilmaz B, Martens L, Saeys Y, Drexler SK, Yazdi AS, Raes J, Lamkanfi M, McCoy KD, and Wullaert A (2017) Nlrp6- and ASC-Dependent Inflammasomes Do Not Shape the Commensal Gut Microbiota Composition. Immunity 47, 339–348 e334 [DOI] [PubMed] [Google Scholar]
- 150.Lemire P, Robertson SJ, Maughan H, Tattoli I, Streutker CJ, Platnich JM, Muruve DA, Philpott DJ, and Girardin SE (2017) The NLR Protein NLRP6 Does Not Impact Gut Microbiota Composition. Cell Rep 21, 3653–3661 [DOI] [PubMed] [Google Scholar]
- 151.Toubai T, Fujiwara H, Rossi C, Riwes M, Tamaki H, Zajac C, Liu C, Mathew AV, Byun J, Oravecz-Wilson K, Matsuda I, Sun Y, Peltier D, Wu J, Chen J, Seregin S, Henig I, Kim S, Brabbs S, Pennathur S, Chen G, and Reddy P (2019) Host NLRP6 exacerbates graft-versus-host disease independent of gut microbial composition. Nat Microbiol 4, 800–812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Siegmund B, Lehr HA, Fantuzzi G, and Dinarello CA (2001) IL-1 beta -converting enzyme (caspase-1) in intestinal inflammation. Proc Natl Acad Sci U S A 98, 13249–13254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Bauer C, Duewell P, Lehr HA, Endres S, and Schnurr M (2012) Protective and aggravating effects of Nlrp3 inflammasome activation in IBD models: influence of genetic and environmental factors. Dig Dis 30 Suppl 1, 82–90 [DOI] [PubMed] [Google Scholar]
- 154.Bauer C, Duewell P, Mayer C, Lehr HA, Fitzgerald KA, Dauer M, Tschopp J, Endres S, Latz E, and Schnurr M (2010) Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by the NLRP3 inflammasome. Gut 59, 1192–1199 [DOI] [PubMed] [Google Scholar]
- 155.Fan TJ, Tchaptchet SY, Arsene D, Mishima Y, Liu B, Sartor RB, Carroll IM, Miao EA, Fodor AA, and Hansen JJ (2018) Environmental Factors Modify the Severity of Acute DSS Colitis in Caspase-11-Deficient Mice. Inflamm Bowel Dis 24, 2394–2403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Blazejewski AJ, Thiemann S, Schenk A, Pils MC, Galvez EJC, Roy U, Heise U, de Zoete MR, Flavell RA, and Strowig T (2017) Microbiota Normalization Reveals that Canonical Caspase-1 Activation Exacerbates Chemically Induced Intestinal Inflammation. Cell Rep 19, 2319–2330 [DOI] [PubMed] [Google Scholar]
- 157.Bulek K, Zhao J, Liao Y, Rana N, Corridoni D, Antanaviciute A, Chen X, Wang H, Qian W, Miller-Little WA, Swaidani S, Tang F, Willard BB, McCrae K, Kang Z, Dubyak GR, Cominelli F, Simmons A, Pizarro TT, and Li X (2020) Epithelial-derived gasdermin D mediates nonlytic IL-1beta release during experimental colitis. J Clin Invest 130, 4218–4234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Lazaridis LD, Pistiki A, Giamarellos-Bourboulis EJ, Georgitsi M, Damoraki G, Polymeros D, Dimitriadis GD, and Triantafyllou K (2017) Activation of NLRP3 Inflammasome in Inflammatory Bowel Disease: Differences Between Crohn’s Disease and Ulcerative Colitis. Dig Dis Sci 62, 2348–2356 [DOI] [PubMed] [Google Scholar]
- 159.Ranson N, Veldhuis M, Mitchell B, Fanning S, Cook AL, Kunde D, and Eri R (2018) NLRP3-Dependent and -Independent Processing of Interleukin (IL)-1beta in Active Ulcerative Colitis. Int J Mol Sci 20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Onoufriadis A, Stone K, Katsiamides A, Amar A, Omar Y, de Lange KM, Taylor K, Barrett JC, Pollok R, Hayee B, Mansfield JC, Sanderson JD, Simpson MA, Mathew CG, and Prescott NJ (2018) Exome Sequencing and Genotyping Identify a Rare Variant in NLRP7 Gene Associated With Ulcerative Colitis. J Crohns Colitis 12, 321–326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Pizarro TT, Michie MH, Bentz M, Woraratanadharm J, Smith MF Jr., Foley E, Moskaluk CA, Bickston SJ, and Cominelli F (1999) IL-18, a novel immunoregulatory cytokine, is up-regulated in Crohn’s disease: expression and localization in intestinal mucosal cells. J Immunol 162, 6829–6835 [PubMed] [Google Scholar]
- 162.Allam O, Samarani S, Mehraj V, Jenabian MA, Tremblay C, Routy JP, Amre D, and Ahmad A (2018) HIV induces production of IL-18 from intestinal epithelial cells that increases intestinal permeability and microbial translocation. PLoS One 13, e0194185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Ludwiczek O, Kaser A, Novick D, Dinarello CA, Rubinstein M, and Tilg H (2005) Elevated systemic levels of free interleukin-18 (IL-18) in patients with Crohn’s disease. Eur Cytokine Netw 16, 27–33 [PubMed] [Google Scholar]
- 164.Dinarello CA, Novick D, Kim S, and Kaplanski G (2013) Interleukin-18 and IL-18 binding protein. Front Immunol 4, 289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Leach ST, Messina I, Lemberg DA, Novick D, Rubenstein M, and Day AS (2008) Local and systemic interleukin-18 and interleukin-18-binding protein in children with inflammatory bowel disease. Inflamm Bowel Dis 14, 68–74 [DOI] [PubMed] [Google Scholar]
- 166.Shouval DS, Biswas A, Kang YH, Griffith AE, Konnikova L, Mascanfroni ID, Redhu NS, Frei SM, Field M, Doty AL, Goldsmith JD, Bhan AK, Loizides A, Weiss B, Yerushalmi B, Yanagi T, Lui X, Quintana FJ, Muise AM, Klein C, Horwitz BH, Glover SC, Bousvaros A, and Snapper SB (2016) Interleukin 1beta Mediates Intestinal Inflammation in Mice and Patients With Interleukin 10 Receptor Deficiency. Gastroenterology 151, 1100–1104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Mao L, Kitani A, Similuk M, Oler AJ, Albenberg L, Kelsen J, Aktay A, Quezado M, Yao M, Montgomery-Recht K, Fuss IJ, and Strober W (2018) Loss-of-function CARD8 mutation causes NLRP3 inflammasome activation and Crohn’s disease. J Clin Invest 128, 1793–1806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Bank S, Julsgaard M, Abed OK, Burisch J, Broder Brodersen J, Pedersen NK, Gouliaev A, Ajan R, Nytoft Rasmussen D, Honore Grauslund C, Roug S, Galsgaard J, Sprogoe Hoyer Finsen D, Lindby K, Danish IBDGWG, Sorensen J, Larsen L, Rohr Andersen M, Brandslund I, Thomassen M, Green A, Bo Bojesen A, Bek Sorensen S, Vogel U, and Andersen V (2019) Polymorphisms in the NFkB, TNF-alpha, IL-1beta, and IL-18 pathways are associated with response to anti-TNF therapy in Danish patients with inflammatory bowel disease. Aliment Pharmacol Ther 49, 890–903 [DOI] [PubMed] [Google Scholar]
- 169.Tapia-Abellan A, Angosto-Bazarra D, Martinez-Banaclocha H, de Torre-Minguela C, Ceron-Carrasco JP, Perez-Sanchez H, Arostegui JI, and Pelegrin P (2019) MCC950 closes the active conformation of NLRP3 to an inactive state. Nat Chem Biol 15, 560–564 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Vande Walle L, Stowe IB, Sacha P, Lee BL, Demon D, Fossoul A, Van Hauwermeiren F, Saavedra PHV, Simon P, Subrt V, Kostka L, Stivala CE, Pham VC, Staben ST, Yamazoe S, Konvalinka J, Kayagaki N, and Lamkanfi M (2019) MCC950/CRID3 potently targets the NACHT domain of wild-type NLRP3 but not disease-associated mutants for inflammasome inhibition. PLoS Biol 17, e3000354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Coll RC, Hill JR, Day CJ, Zamoshnikova A, Boucher D, Massey NL, Chitty JL, Fraser JA, Jennings MP, Robertson AAB, and Schroder K (2019) MCC950 directly targets the NLRP3 ATP-hydrolysis motif for inflammasome inhibition. Nat Chem Biol 15, 556–559 [DOI] [PubMed] [Google Scholar]
- 172.Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Nunez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, and O’Neill LA (2015) A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21, 248–255 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Caseley EA, Poulter JA, Rodrigues F, Immunome Project Consortium for Autoinflammatory, D., and McDermott MF (2020) Inflammasome inhibition under physiological and pharmacological conditions. Genes Immun 21, 211–223 [DOI] [PubMed] [Google Scholar]
- 174.Perera AP, Fernando R, Shinde T, Gundamaraju R, Southam B, Sohal SS, Robertson AAB, Schroder K, Kunde D, and Eri R (2018) MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice. Sci Rep 8, 8618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Wu D, Wu K, Zhu Q, Xiao W, Shan Q, Yan Z, Wu J, Deng B, Xue Y, Gong W, Lu G, and Ding Y (2018) Formononetin Administration Ameliorates Dextran Sulfate Sodium-Induced Acute Colitis by Inhibiting NLRP3 Inflammasome Signaling Pathway. Mediators Inflamm 2018, 3048532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Umiker B, Lee HH, Cope J, Ajami NJ, Laine JP, Fregeau C, Ferguson H, Alves SE, Sciammetta N, Kleinschek M, and Salmon M (2019) The NLRP3 inflammasome mediates DSS-induced intestinal inflammation in Nod2 knockout mice. Innate Immun 25, 132–143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Saber S, and El-Kader EMA (2020) Novel complementary coloprotective effects of metformin and MCC950 by modulating HSP90/NLRP3 interaction and inducing autophagy in rats. Inflammopharmacology [DOI] [PubMed] [Google Scholar]
- 178.He H, Jiang H, Chen Y, Ye J, Wang A, Wang C, Liu Q, Liang G, Deng X, Jiang W, and Zhou R (2018) Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat Commun 9, 2550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wang S, Zhang Y, Saas P, Wang H, Xu Y, Chen K, Zhong J, Yuan Y, Wang Y, and Sun Y (2015) Oridonin’s therapeutic effect: suppressing Th1/Th17 simultaneously in a mouse model of Crohn’s disease. J Gastroenterol Hepatol 30, 504–512 [DOI] [PubMed] [Google Scholar]
- 180.Liu QQ, Wang HL, Chen K, Wang SB, Xu Y, Ye Q, and Sun YW (2016) Oridonin derivative ameliorates experimental colitis by inhibiting activated T-cells and translocation of nuclear factor-kappa B. J Dig Dis 17, 104–112 [DOI] [PubMed] [Google Scholar]
- 181.Yuan X, Berg N, Lee JW, Le TT, Neudecker V, Jing N, and Eltzschig H (2018) MicroRNA miR-223 as regulator of innate immunity. J Leukoc Biol 104, 515–524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Wani S, Ka Man Law I, and Pothoulakis C (2020) Role and Mechanisms of Exosomal miRNAs in IBD Pathophysiology. Am J Physiol Gastrointest Liver Physiol [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wang H, Chao K, Ng SC, Bai AH, Yu Q, Yu J, Li M, Cui Y, Chen M, Hu JF, and Zhang S (2016) Pro-inflammatory miR-223 mediates the cross-talk between the IL23 pathway and the intestinal barrier in inflammatory bowel disease. Genome Biol 17, 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Zhou H, Xiao J, Wu N, Liu C, Xu J, Liu F, and Wu L (2015) MicroRNA-223 Regulates the Differentiation and Function of Intestinal Dendritic Cells and Macrophages by Targeting C/EBPbeta. Cell Rep 13, 1149–1160 [DOI] [PubMed] [Google Scholar]
- 185.Li M, Zhao J, Cao M, Liu R, Chen G, Li S, Xie Y, Xie J, Cheng Y, Huang L, Su M, Xu Y, Zheng M, Zou K, Geng L, Xu W, and Gong S (2020) Mast cells-derived MiR-223 destroys intestinal barrier function by inhibition of CLDN8 expression in intestinal epithelial cells. Biol Res 53, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Neudecker V, Haneklaus M, Jensen O, Khailova L, Masterson JC, Tye H, Biette K, Jedlicka P, Brodsky KS, Gerich ME, Mack M, Robertson AAB, Cooper MA, Furuta GT, Dinarello CA, O’Neill LA, Eltzschig HK, Masters SL, and McNamee EN (2017) Myeloid-derived miR-223 regulates intestinal inflammation via repression of the NLRP3 inflammasome. J Exp Med 214, 1737–1752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Valmiki S, Ahuja V, and Paul J (2017) MicroRNA exhibit altered expression in the inflamed colonic mucosa of ulcerative colitis patients. World J Gastroenterol 23, 5324–5332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Hu Y, Mao K, Zeng Y, Chen S, Tao Z, Yang C, Sun S, Wu X, Meng G, and Sun B (2010) Tripartite-motif protein 30 negatively regulates NLRP3 inflammasome activation by modulating reactive oxygen species production. J Immunol 185, 7699–7705 [DOI] [PubMed] [Google Scholar]
- 189.Shao BZ, Wang SL, Pan P, Yao J, Wu K, Li ZS, Bai Y, and Linghu EQ (2019) Targeting NLRP3 Inflammasome in Inflammatory Bowel Disease: Putting out the Fire of Inflammation. Inflammation 42, 1147–1159 [DOI] [PubMed] [Google Scholar]
- 190.Gong Z, Zhao S, Zhou J, Yan J, Wang L, Du X, Li H, Chen Y, Cai W, and Wu J (2018) Curcumin alleviates DSS-induced colitis via inhibiting NLRP3 inflammsome activation and IL-1beta production. Mol Immunol 104, 11–19 [DOI] [PubMed] [Google Scholar]
- 191.Iqbal U, Anwar H, and Quadri AA (2018) Use of Curcumin in Achieving Clinical and Endoscopic Remission in Ulcerative Colitis: A Systematic Review and Meta-analysis. Am J Med Sci 356, 350–356 [DOI] [PubMed] [Google Scholar]
- 192.Wang Y, Tang Q, Duan P, and Yang L (2018) Curcumin as a therapeutic agent for blocking NF-kappaB activation in ulcerative colitis. Immunopharmacol Immunotoxicol 40, 476–482 [DOI] [PubMed] [Google Scholar]
- 193.Grammatikopoulou MG, Gkiouras K, Theodoridis X, Asteriou E, Forbes A, and Bogdanos DP (2018) Oral Adjuvant Curcumin Therapy for Attaining Clinical Remission in Ulcerative Colitis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Zhao Y, Guo Q, Zhu Q, Tan R, Bai D, Bu X, Lin B, Zhao K, Pan C, Chen H, and Lu N (2019) Flavonoid VI-16 protects against DSS-induced colitis by inhibiting Txnip-dependent NLRP3 inflammasome activation in macrophages via reducing oxidative stress. Mucosal Immunol 12, 1150–1163 [DOI] [PubMed] [Google Scholar]
- 195.Rapa SF, Waltenberger B, Di Paola R, Adesso S, Siracusa R, Peritore AF, D’Amico R, Autore G, Cuzzocrea S, Stuppner H, and Marzocco S (2020) Plumericin prevents intestinal inflammation and oxidative stress in vitro and in vivo. FASEB J 34, 1576–1590 [DOI] [PubMed] [Google Scholar]
- 196.Chen L, You Q, Hu L, Gao J, Meng Q, Liu W, Wu X, and Xu Q (2017) The Antioxidant Procyanidin Reduces Reactive Oxygen Species Signaling in Macrophages and Ameliorates Experimental Colitis in Mice. Front Immunol 8, 1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Li S, Takasu C, Lau H, Robles L, Vo K, Farzaneh T, Vaziri ND, Stamos MJ, and Ichii H (2020) Dimethyl Fumarate Alleviates Dextran Sulfate Sodium-Induced Colitis, through the Activation of Nrf2-Mediated Antioxidant and Anti-inflammatory Pathways. Antioxidants (Basel) 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Dai G, Jiang Z, Sun B, Liu C, Meng Q, Ding K, Jing W, and Ju W (2020) Caffeic Acid Phenethyl Ester Prevents Colitis-Associated Cancer by Inhibiting NLRP3 Inflammasome. Front Oncol 10, 721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Mazarakis N, Snibson K, Licciardi PV, and Karagiannis TC (2020) The potential use of l-sulforaphane for the treatment of chronic inflammatory diseases: A review of the clinical evidence. Clin Nutr 39, 664–675 [DOI] [PubMed] [Google Scholar]
- 200.Deng Z, Rong Y, Teng Y, Mu J, Zhuang X, Tseng M, Samykutty A, Zhang L, Yan J, Miller D, Suttles J, and Zhang HG (2017) Broccoli-Derived Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase. Mol Ther 25, 1641–1654 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Dhimolea E (2010) Canakinumab. MAbs 2, 3–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Giacomelli R, Sota J, Ruscitti P, Campochiaro C, Colafrancesco S, Dagna L, Iacono D, Iannone F, Lopalco G, Sfriso P, and Cantarini L (2020) The treatment of adult-onset Still’s disease with anakinra, a recombinant human IL-1 receptor antagonist: a systematic review of literature. Clin Exp Rheumatol [DOI] [PubMed] [Google Scholar]
- 203.Thomas MG, Bayliss C, Bond S, Dowling F, Galea J, Jairath V, Lamb C, Probert C, Timperley-Preece E, Watson A, Whitehead L, Williams JG, Parkes M, Kaser A, and Raine T (2019) Trial summary and protocol for a phase II randomised placebo-controlled double-blinded trial of Interleukin 1 blockade in Acute Severe Colitis: the IASO trial. BMJ Open 9, e023765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Cao J, Cheng J, Xi S, Qi X, Shen S, and Ge Y (2019) Alginate/chitosan microcapsules for in-situ delivery of the protein, interleukin-1 receptor antagonist (IL-1Ra), for the treatment of dextran sulfate sodium (DSS)-induced colitis in a mouse model. Eur J Pharm Biopharm 137, 112–121 [DOI] [PubMed] [Google Scholar]
- 205.Hu JJ, Liu X, Xia S, Zhang Z, Zhang Y, Zhao J, Ruan J, Luo X, Lou X, Bai Y, Wang J, Hollingsworth LR, Magupalli VG, Zhao L, Luo HR, Kim J, Lieberman J, and Wu H (2020) FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol 21, 736–745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Li Y, Niu X, Xu H, Li Q, Meng L, He M, Zhang J, Zhang Z, and Zhang Z (2020) VX-765 attenuates atherosclerosis in ApoE deficient mice by modulating VSMCs pyroptosis. Exp Cell Res 389, 111847. [DOI] [PubMed] [Google Scholar]
- 207.McKenzie BA, Mamik MK, Saito LB, Boghozian R, Monaco MC, Major EO, Lu JQ, Branton WG, and Power C (2018) Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci U S A 115, E6065–E6074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Huang Y, Jiang H, Chen Y, Wang X, Yang Y, Tao J, Deng X, Liang G, Zhang H, Jiang W, and Zhou R (2018) Tranilast directly targets NLRP3 to treat inflammasome-driven diseases. EMBO Mol Med 10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Marchetti C, Swartzwelter B, Gamboni F, Neff CP, Richter K, Azam T, Carta S, Tengesdal I, Nemkov T, D’Alessandro A, Henry C, Jones GS, Goodrich SA, St Laurent JP, Jones TM, Scribner CL, Barrow RB, Altman RD, Skouras DB, Gattorno M, Grau V, Janciauskiene S, Rubartelli A, Joosten LAB, and Dinarello CA (2018) OLT1177, a beta-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses the metabolic cost of inflammation. Proc Natl Acad Sci U S A 115, E1530–E1539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn A, Alnemri E, and Dixit VD (2015) The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21, 263–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Goldberg EL, Asher JL, Molony RD, Shaw AC, Zeiss CJ, Wang C, Morozova-Roche LA, Herzog RI, Iwasaki A, and Dixit VD (2017) beta-Hydroxybutyrate Deactivates Neutrophil NLRP3 Inflammasome to Relieve Gout Flares. Cell Rep 18, 2077–2087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Daniels MJ, Rivers-Auty J, Schilling T, Spencer NG, Watremez W, Fasolino V, Booth SJ, White CS, Baldwin AG, Freeman S, Wong R, Latta C, Yu S, Jackson J, Fischer N, Koziel V, Pillot T, Bagnall J, Allan SM, Paszek P, Galea J, Harte MK, Eder C, Lawrence CB, and Brough D (2016) Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nat Commun 7, 12504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Yan Y, Jiang W, Liu L, Wang X, Ding C, Tian Z, and Zhou R (2015) Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell 160, 62–73 [DOI] [PubMed] [Google Scholar]
- 214.Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, Li S, Wang B, Nemmara VV, Wilson R, Jiang Z, Khalighinejad F, Muneeruddin K, Shaffer SA, Dutta R, Ionete C, Pesiridis S, Yang S, Thompson PR, and Fitzgerald KA (2020) Succination inactivates gasdermin D and blocks pyroptosis. Science 369, 1633–1637 [DOI] [PMC free article] [PubMed] [Google Scholar]