Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 1.
Published in final edited form as: Immunol Rev. 2023 Dec 25;322(1):259–282. doi: 10.1111/imr.13292

The discovery of NLRP3 and its function in cryopyrin-associated periodic syndromes and innate immunity

Christopher D Putnam 1, Lori Broderick 2,3, Hal M Hoffman 1,2,3
PMCID: PMC10950545  NIHMSID: NIHMS1953893  PMID: 38146057

Summary

From studies of individual families to global collaborative efforts, the NLRP3 inflammasome is now recognized to be a key regulator of innate immunity. Activated by a panoply of pathogen-associated and endogenous triggers, NLRP3 serves as an intracellular sensor that drives carefully coordinated assembly of the inflammasome, and downstream inflammation mediated by IL-1 and IL-18. Initially discovered as the cause of the autoinflammatory spectrum of cryopyrin-associated periodic syndrome (CAPS), NLRP3 is now also known to play a role in more common diseases including cardiovascular disease, gout, and liver disease. We have seen cohesion in results from clinical studies in CAPS patients, ex vivo studies of human cells and murine cells, and in vivo murine models leading to our understanding of the downstream pathways, cytokine secretion, and cell death pathways that has solidified the role of autoinflammation in the pathogenesis of human disease. Recent advances in our understanding of the structure of the inflammasome have provided ways for us to visualize normal and mutant protein function and pharmacologic inhibition. The subsequent development of targeted therapies successfully used in the treatment of patients with CAPS completes the bench to bedside translational loop which has defined the study of this unique protein.

Keywords: cryopyrin-associated periodic syndrome, familial cold autoinflammatory syndrome, inflammasome, NLRP3

1 |. INTRODUCTION

By the mid-20th century, three categories of diseases were known to be caused by the immune system: immunodeficiency, allergy, and autoimmunity. Prior to the 1900s, studies of the immune system largely focused on host–microbe interactions and immune deficiencies. These studies built on the work of Edward Jenner, Louis Pasteur, Emil von Behring, and Shibasabura Kitasato,1,2 but were fundamentally attempts at understanding immune responses that were described as early as Hippocrates. In the 1900s, experimental descriptions of autoimmunity, allergy, and anaphylaxis not only expanded knowledge of diseases involving the immune system, but also led to a paradigm shift acknowledging that the immune system was capable of self-damage.3,4 Despite this hard-won knowledge, rare patients and families with systemic inflammatory disorders were recognized that violated this established dogma, as they failed to fit into the established categories of immune system diseases. These patients were characterized by non-infectious episodes of inflammation, without high-titer autoantibodies or antigen-specific T cells. In the late 20th century, these disorders were termed autoinflammatory syndromes to distinguish them from autoimmune disorders and were molecularly characterized by the identification of the genes whose defects caused four landmark hereditary periodic fever syndromes: TNFRSF1A in TNFR1-associated periodic syndromes (TRAPS), MEFV in familial Mediterranean fever (FMF), MVK in Hyper-IgD syndrome, and NLRP3 in cryopyrin-associated periodic syndrome (CAPS).58 This review will cover the progress made in our understanding of the role of NLRP3 in human disease beginning with families experiencing unusual inflammatory symptoms and extending to over a quarter of a century of translational approaches using genetics, molecular biology, cellular and mouse immunology, structural analysis and modeling, and targeted pharmacology.

2 |. THE CLINICAL FEATURES OF CAPS AS AN AUTOINFLAMMATORY DISEASE SPECTRUM

CAPS, as it is now recognized, encompasses three previously identified syndromes: familial cold autoinflammatory syndrome (FCAS), Muckle–Wells syndrome (MWS), and neonatal-onset multisystem inflammatory disease (NOMID), which is also called chronic infantile neurologic cutaneous articular (CINCA) syndrome (Figure 1, Table 1). Although each of these syndromes has distinct characteristics, patients with FCAS/MWS and MWS/NOMID overlap phenotypes have been reported, which suggested these disorders existed on a spectrum of inflammatory disease even before the NLRP3 gene was identified.911

FIGURE 1.

FIGURE 1

Clinical manifestations of NLRP3 spectrum disease. CAPS, cryopyrin-associated periodic syndrome; FCAS, familial cold autoinflammatory syndrome; ICP, intracranial pressure; MWS, Muckle–Wells syndrome; NOMID/CINCA, neonatal-onset multisystem inflammatory disease/chronic infantile neurologic cutaneous articular syndrome. Created with Biorender.com.

TABLE 1.

Clinical characteristics of CAPS spectrum disease (modified from Welzel & Kuemmerle-Deschner176).

Mild phenotype (FCAS) Moderate phenotype (MWS) Severe phenotype (CINCA/NOMID) Atypical CAPS170,171
Disease onset <6 months–adulthood Early childhood–adulthood Perinatal Early childhood–adulthood
Family history Often positive Often positive Often negative (sporadic de novo mutations) Often positive
Inflammatory flares Cold induced Yes, may have continuous disease symptoms Continuous disease symptoms Yes
Duration of inflammatory flares 30 min–72 h 1–3 Days ± subclinical Persistent inflammation Variable
Cold trigger Yes Possible Rare Rare
Dermatological manifestations Cold-induced neutrophilic urticaria Neutrophilic urticaria Neutrophilic urticaria Less frequent
Fever 6–24 h after cold exposure possible Particularly in childhood Yes No
Ocular manifestation Conjunctivitis Conjunctivitis, episcleritis, optic disc edema/papilledema Conjunctivitis, episcleritis, optic disc edema/papilledema Conjunctivitis, episcleritis, papilledema
Musculoskeletal manifestations Myalgia, arthralgia Myalgia, arthralgia, oligoarthritis Myalgia, arthralgia, polyarthritis. Epiphyseal bony overgrowth, limb-length discrepancies, contractures Myalgia, arthralgia
Hearing loss No Yes Yes Yes
Central nervous system manifestations Headache Headache, intermittent aseptic meningitis Headache, chronic aseptic meningitis, increased intracranial pressure, brain atrophy Headache, interm
Amyloidosis Rare Yes Yes No

2.1 |. Familial cold autoinflammatory syndrome (FCAS)

FCAS was described in 1940 by Kile and Rusk who studied a unique family with affected members, equally male and female across five generations, exhibiting recurrent episodes of urticarial-like rash, limb pain, and fever following generalized cold exposure.12 The proband described a lifelong history of burning, rather than pruritic, erythema beginning 30 min after going outside in cold damp weather, followed by fever to 101–103 °F, and joint stiffness lasting 6–8 h following mild cold exposure or 24 h following more extended cold exposures. Symptoms could be induced by immersion of her arm in cold water and were resistant to therapies for allergic urticaria commonly used at the time. Additional families with similar phenotypes and elevations in acute phase reactants were reported over the next 6 decades, variably described as cold hypersensitivity, familial cold urticaria (FCU), and finally FCAS13,14 in an attempt to differentiate this chronic, inherited, systemic inflammatory disorder from the more common acquired cold urticaria.15

2.2 |. Muckle–Wells syndrome

While the cold-induced urticaria uniquely defined the family described by Kile and Rusk, additional descriptions of families with apparent autosomal dominantly inherited urticarial disorders began to appear in the literature. In 1962, Muckle and Wells reported a family with similar features of non-infectious episodes of urticaria-like rash, limb pain, and fever with rigors, which they described as “aguey bouts” which lasted approximately 36 h.16 In contrast to FCAS patients, this family developed progressive bilateral sensorineural hearing loss beginning in childhood, and end stage renal disease secondary to AA amyloidosis beginning in adulthood, with laboratory evaluations showing anemia, increased serum immunoglobulins and elevated inflammatory markers.16 Similar to FCAS patients, these patients failed to respond to therapy with antihistamines or daily steroids. Muckle and Wells noted the similarities of recurrent skin rash and amyloidosis between MWS patients and FMF patients,17 in what may be one of the first attempts to phenotypically link autoinflammatory syndromes.

2.3 |. CINCA/NOMID

Chronic infantile neurologic cutaneous articular (CINCA) syndrome was first described in 1980 by Prieur who reported three unrelated children presenting in the neonatal period with a chronic urticarial-like rash, neutrophilia and significant involvement of the large joints. Described as “bread crumb appearance” on radiograph, the inflammatory joint disease involved the epiphyses and patella and resulted in physical deformation and disability.18 Extensive neurologic disease including seizures secondary to chronic sterile meningitis increased intracranial pressure and developmental delay were also described.18 Progressive deafness and visual impairment further added to the morbidity for this disorder. Initially termed chronic meningo-cutaneo-articular syndrome in children, other patients with similar phenotypes were subsequently described, and the disorder was referred to as either CINCA or NOMID.1921

3 |. IDENTIFICATION, CLONING, AND CHARACTERIZATION OF THE NLRP3 GENE AS THE CAUSE OF CAPS

A fortuitous meeting with families that described a lifelong history of cold-induced rash, fever and arthralgias, became the turning point for the field of inflammasome biology. We used DNA from a total of 69 individuals from five families, including the initial proband described in 1940, and used positional cloning to link a defined region on chromosome 1q43-q44 to FCAS,11 the same area previously linked to MWS.22 The team then screened predicted exons and flanking intronic sequences in this region, identifying missense mutations in FCAS and MWS patients in a gene initially labeled CIAS1 (for cold-induced autoinflammatory syndrome 1).6 Using the nascent Human Genome Project, we identified seven exons by homology and two additional exons by sequencing of RT–PCR products. This method also revealed extensive alternative splicing in the 3′ end of the gene, the relevance of which would not come to light for several years. Northern blot analysis demonstrated that transcripts of this newly discovered gene were present at a low level in peripheral blood leukocytes and had little or no expression in other tissues. This was consistent with the expression patterns seen in MEFV, establishing a link between these recurrent inflammatory disorders on a molecular level. The name of the disorder was changed to FCAS to highlight the genetic and phenotypic similarities.6 A year later, CIAS1 (later termed NLRP3) mutations were also identified as the cause of NOMID.23,24 Since then, research focused on the molecular basis for these rare diseases has been the work of many scientists around the world in addition to our laboratory.

3.1 |. Genotype and phenotype of a monogenic NLRP3 associated disease

Since the initial description of the NLRP3 gene in 2001, over 250 variants have been described worldwide.2528 There appears to be significant genotype–phenotype correlation in that certain mutations are often associated with specific disease spectrum severity, although different mutations affecting the same amino acid can cause different diseases along the CAPS spectrum (Figure 2).9 One complication in the literature is that two different methionines have been used as the initiating amino acid. The NLRP3 sequence from the originally used methionine starts with the amino acid sequence MASTR (e.g., NP_004886.2 and NP_001230062.1). The NLRP3 sequence has also been redescribed as beginning with an upstream in-frame methionine so that the protein starts with the sequence MKMASTR (e.g., NP_004886.3); however, this upstream methionine is not universally conserved in mammals. This ambiguity means that the same mutation in different publications can be given a position that differs by 2, for example p.L359W and p.L361W refer to the same mutation. In this review, we exclusively use the original nomenclature; hence, we will name the above mutation p.L359W.

FIGURE 2.

FIGURE 2

NLRP3 mutations in CAPS and undefined autoinflammatory disease. Amino acid positions mutated in NLRP3-mediated autoinflammatory disease with the associated amino acid substitution placed according to the resulting phenotype. Black dots indicate the number of distinct missense mutations associated with each position. Regions A–G are local NLRP3 regions containing multiple missense mutations often affecting the same amino acid. The Δ−309 at G307 corresponds to the p.G307_F309del mutation. Data from the Infevers database.25

The majority of patients in the CAPS spectrum have autosomal dominant NLRP3 missense mutations; however, there are mutation-negative patients with classic CAPS symptoms and pathology. At least some of these patients can be explained by the presence of somatic mutations that were not detected by standard sequencing, with some patients ultimately having mutations in NLRP3, and a few other patients with CAPS phenotype were determined to have NLRC4 mutations.2933 We have also identified a promoter variant in one NLRP3 mutation-negative patient associated with increased NLRP3 expression,34 providing a potential novel disease mechanism. An extensive pathway-targeted candidate gene approach12 and further linkage studies have not yet identified additional disease genes.

3.2 |. Low-penetrance mutations

Our initial description of NLRP3 mutations in FCAS included a variant (p.V198M) that was not initially observed in a panel of healthy controls.6 However, additional sequencing revealed that this variant and two other variants (p.R488K and p.Q703K) that had been initially observed in CAPS phenotypes were also identified in healthy controls as well as patients with atypical clinical presentations. The higher frequency of these variants in different populations and the genetic data is not supportive of classifying them as pathogenic variants; however, there are clinical and functional data to suggest that they have an intermediate phenotype between pathogenic CAPS and healthy controls.35 The phenotypic differences between the classic CAPS mutations and low-penetrance mutations provide insight into the unique role of NLRP3 in modulating the immune response. Subsequently, detailed evaluations uniting molecular experiments and structural biology have been instrumental in our understanding of how NLRP3 responds to an array of diverse stimuli and directs formation of the cytoplasmic multiprotein platform known as the inflammasome.

4 |. INFLAMMASOMES: NOMENCLATURE, STRUCTURE, AND FUNCTION

The NLRP3 protein, originally named cryopyrin for the N-terminal pyrin domain and link to cold-induced symptoms,6 belongs to the nucleotide-binding domain and leucine-rich repeat (LRR) containing (NLR) family of proteins. This family is characterized by diverse N-terminal domain(s), a central nucleotide-binding NACHT (for Neuronal Apoptosis inhibitor protein CIITA, HET-E and TP1) domain,36 and a C-terminal Leucine-Rich Repeat (LRR) domain (Figure 3). NLR proteins are classified into five subfamilies on the basis of the N-terminal domain:37 (1) NLRA proteins like CIITA have an Acidic transactivating domain, (2) NLRB proteins like NAIP have a Baculovirus Inhibitor of apoptosis protein Repeat (BIR) domain, (3) NLRC proteins like NOD1 have a CARD (CAspase Recruitment Domain), (4) NLRP proteins like NLRP3 have a PYD (Pyrin domain; Figure 3), and (5) NLRX proteins like NLRX1 lack significant N-terminal homologies. The human genome encodes 22 NLR proteins, many of which are associated with diseases driven by inflammation.38

FIGURE 3.

FIGURE 3

NLRP3 gene structure and CAPS mutations. Protein domains shown at top are the pyrin domain (PYD), the NACHT domain, and the leucine-rich repeat (LRR) domain. The NACHT domain is further subdivided into the FISNA (fish-specific NACHT associated) domain, nucleotide-binding domain (NBD), HD1, the winged-helix domain (WH), and HD2. Positions of mutations in different diseases are shown as hashes in the middle. Exon structure is shown at the bottom, with the non-coding exon 1 omitted from the diagram. Regions commonly mutagenized in autoinflammatory disease (regions A–G) are highlighted in red.

4.1 |. Similarities and differences among innate immune modulators of the inflammasome

Infection by pathogens and other environmental cues give rise to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) that induce the innate immune system. These PAMPs and DAMPs are thought to trigger the assembly of NLR proteins into large molecular weight complexes, termed inflammasomes after a pivotal study by Jorg Tshopp’s group describing a lipopolysaccharide (LPS)-inducible assembly of NLRP1 with apoptosis related speck-like protein containing CARD (ASC), caspase-1, and caspase-5.39 Inflammasome formation has been verified for multiple NLR proteins, including NLRC4, NLRP1, NLRP3, and NLRP7.3944 Inflammasome formation by NLRP3 appears to be driven by two signals: The first signal is “priming,” and the second signal is “activation”. Signal 1 leads to NF-κB-driven expression of genes encoding NLR proteins and proinflammatory cytokines, such as IL-1β and IL-18, as well as deubiquitination of the LRR domain by BRCC3,4547 the recognition of LPS by Toll-like receptor (TLR) proteins, and binding of proinflammatory cytokines to their cognate receptors. Once expressed, inactive NLRP3 appears to be stabilized in the cytosol through interactions with the SCF ubiquitin ligase subunit SGT1 and heat shock protein 90 (HSP90),48 similar to the interaction for pathogen-sensing NLR proteins in plants.49 Signal 2 includes a large variety of triggers for NLR protein assembly into inflammasomes and the maturation and release of the proinflammatory cytokines.

Assembled inflammasomes form 10- and 11-subunit disks comprised of the NACHT and LRR domains (for NLRP3, see Figure 4).4143 The N-terminal pyrin domains of NLRP inflammasomes can form 5-symmetric helical filaments alone,42,50 and electron microscopy suggests that inflammasome assembly generates a 10-or 11-subunit NLRP pyrin domain proto-filament capable of nucleating a filament made up of the pyrin domains of the ASC adapter protein.51 ASC is comprised of two domains, an N-terminal pyrin domain and a C-terminal CARD domain, which recruits pro-caspase 1 through a homotypic interaction between the CARD domains of ASC and pro-caspase 1 (Figure 4).5153 Activation of pro-caspase 1 molecules is likely achieved through increased local concentration mediated by ASC binding followed by trans-cleavage and dimerization of the mature caspase 1 subunits. Mature caspase 1 then cleaves the pro-inflammatory cytokines, leading to their release into the extracellular matrix for signaling, and cleaves and activates gasdermin D (GSDMD). The inflammasome-triggered formation of ASC filaments serves as a signal amplification mechanism to drive the innate immune response.54

FIGURE 4.

FIGURE 4

A unified model for the formation of the active NLRP3 inflammasome. Predicted NLRP3 assembly states and inflammasome intermediates (top) are correlated with likely cellular localization of these assemblies (green highlighted regions, bottom).

The PAMPs and DAMPs that activate some NLR proteins have been identified; however, the mechanism of activation remains unclear for most of these proteins. NLRC4 responds to bacterial flagellins and the inner rod components of bacterial type III secretion systems; NLRP1 responds to dsRNA and dsDNA, associated with viral infections, and NLRP7 responds to microbial lipopeptides.40,55,56 The precise mechanism by which these molecular patterns activate inflammasome formation remains poorly understood for most NLR proteins. NLRP1 assembly appears to be triggered by proteolytic cleavage of the N-terminus that is induced by a variety of factors, including viral proteases, viral replication, and ribotoxic stress, rather than by direct interaction with PAMPs and DAMPs.57 In contrast, NLRP6 may form inflammasomes through a liquid–liquid phase separation after directly interacting with viral RNA molecules or lipoteichoic acid during infection by Gram-positive bacteria.58 Similarly, NLRC4 utilizes an accessory subunit, one of the NLR family apoptosis inhibitory proteins (NAIPs, a single gene in humans but present in 6 copies in mice) that provides ligand specificity56,5961 and promotes inflammasome formation by nucleating a disk containing 1 NAIP protein and 10 NLRC4 proteins.43 It has recently been suggested that NLRP3 also forms a non-canonical inflammasome that also contains NLRC4, the RNA helicase DDX17, ASC, and caspase-1 in response to the presence of short interspersed nuclear element (SINE) RNA molecules.62

4.2 |. NLRP3 as a unique innate immune sensor of cellular stress

Remarkably, the NLRP3 inflammasome is unique in that it is responsive to a wide range of activating agents, including uric acid crystals, alum, silica, cholesterol crystals, extracellular adenosine triphosphate (ATP), hyaluronan, amyloid-β fibrils, mitochondrial reactive oxygen species (ROS) generation, and lowered intracellular potassium concentrations such as by nigericin or gramicidin treatment.6366 It seems unlikely that NLRP3 can directly detect each of these DAMPs, and a unifying feature for these agents has been suggested to be increased potassium efflux from the cell.67,68 Consistent with this, inhibition of potassium efflux by the mammalian ketone metabolite β-hydroxybutyrate suppresses NLRP3 activity.69

Reduced potassium levels appear to be an indirect activator of NLRP3 assembly.70 Several of the treatments shown to activate NLRP3, including extracellular ATP and potassium efflux triggered by nigericin or gramicidin, lead to the disassembly of the trans-Golgi network. NLRP3 is recruited to vesicles in this dispersed trans-Golgi network (dTGN) through the charged interaction between the phosphatidylinositol-4-phosphate lipid and the linker between the NLRP3 pyrin and NACHT domains and a region of the fish-specific NACHT associated (FISNA) subdomain (Figure 3).71,72 It has been proposed that the inactive “cage” form of NLRP3 could be the state at which NLRP3 binds to the dTGN vesicles71; however, constitutively activated CAPS mutant proteins are also capable of spontaneously forming membrane-bound NLRP3 puncta,70 suggesting that active forms can also bind to membranes. Remarkably, two potassium-efflux independent NLRP3 activators, imiquimod and CL097, also promote the formation of dTGN vesicles and NLRP3 conformational changes similar to potassium efflux70,72; however, it is currently unclear why the requirement for potassium efflux for NLRP3 activation varies among different treatments that disrupt the trans-Golgi network.

Even after full or partial assembly of NLRP3 on dTGN vesicles, several lines of evidence indicate that NLRP3 activity is further controlled through subcellular localization. First, NLRP3 activation in mice requires binding of the serine/threonine protein kinase NEK7 (centrosomal NIMA-related kinase 7) by the NLRP3 LRR domain (Figures 4 and 5).7375 Importantly, in humans, this activation appears to be independent of the NEK7 kinase activity.75 While NEK7 binding would be predicted to disrupt the inactive NLRP3 “cage” structures (Figure 4),50,71,76 NEK7 is primarily localized at the microtubule organizing center (MTOC), and activated NLRP3 is moved by microtubule retrograde transport from the trans-Golgi network to the MTOC.77,78 This transport is responsible for the formation of ASC specks, which are large helical fibrils that typically form at one site per cell and are convenient experimental markers for NLRP3 activation, as they can be visualized by fluorescence microscopy.79 The requirements for the NLRP3 retrograde transport are reminiscent of the retrograde transport of aggresomes, which are large molecular aggregates that are resistant to proteosome degradation and are degraded by autophagy at the MTOC.80,81 A relocalization requirement likely explains why partial, but not complete, defects in inflammasome activity are caused by defects in the dynein adapter histone deacetylase 6 (HDAC6), HDAC6 inhibitors, defects in microtubule-affinity reducing kinase 4 (MARK4), dynein disruption, and loss of the aggresome component vimentin.77,8291 Cargo recruited for transport by HDAC6 is typically ubiquitinated,92 and extensive studies have indicated that NLRP3 inflammasome components are regulated both positively and negatively by ubiquitination.93 A relocalization requirement also explains why the microtubule depolymerizing agent colchicine is useful in treating symptoms of gout, which is an NLRP3-mediated disease; colchicine blocks both ASC speck formation and IL-1β release driven by monosodium urea crystals.9496

FIGURE 5.

FIGURE 5

NLRP3 LRR domain interactions. (A and B) Leucine-rich repeats encoded by different exons are depicted in alternating red and yellow. An insertion within the LRR domain that binds the concave face of the LRR domain in the inactive NLRP3 decameric “cage” (A) but is unstructured in the active NLRP3-NEK7 disk (B) is colored in blue. (C) LRR domain-mediated interactions (red and blue) make up the interactions in the inactive decameric “cage” assembly. (D) LRR domain-mediated interactions (blue) in the active disk assembly are with NEK7; the NLRP3 disk is only held together by interactions between the NACHT domains (red), primarily involving the FISNA (FIS) subdomain.

It is currently unclear what assembly state NLRP3 is in during transport to the MTOC; however, a small molecule inhibitor of NLRP3, MCC950/CRID3, which likely prevents the inactive-to-active conformational change in NLRP3 subunits and subsequent inflammasome disc assembly, blocks ASC speck formation,97 suggesting that, at minimum, the active conformation of NLRP3 is required. Similarly, the NLRP3-ASC interaction appears to be required. In the presence of ASC, activated NLRP3 is localized to a single speck; however, in the absence of ASC, activated NLRP3 is distributed in dTGN-associated puncta.70,86 This suggests that ASC is required for retrograde transport or for retention of NLRP3 at the MTOC, possibly through recruitment of ubiquitinated ASC by HDAC6. ASC is known to be modified by linear ubiquitin chains via the linear ubiquitin chain assembly complex (LUBEC), and two LUBEC subunits, HOIL-1 and SHARPIN, promote inflammasome activity.98,99 ASC is also subject to modification by K63-linked ubiquitin chains via the TRAF3 and Peli1 E3 ubiquitin ligases.56,99101 Remarkably, TRAF3, Peli1, and HOIL-1 were shown to promote ASC speck formation;56,99,100 however, it is not yet clear if these proteins act at the step of retrograde transport.

The complex choreography required for the formation of active NLRP3 inflammasomes at the MTOC may have arisen due to the ability of NLRP3 to respond to a wide variety of inflammatory activators. Because the proximal signal for activation, likely disruption of the trans-Golgi network, is only indirectly tied to infection, preventing inappropriate activation and being able to rapidly silence the inflammasome to return to a basal state are potentially more crucial for NLRP3-mediated inflammatory responses than for inflammasomes triggered by direct interaction with PAMPs/DAMPs. Remarkably, both NLRP3 and pyrin inflammasomes localize to the MTOC, whereas absent in melanoma 2 (AIM2) inflammasomes do not.77 Activation by MTOC recruitment and binding of the MTOC-resident protein NEK7 localizes activated NLRP3 inflammasomes where they could be rapidly degraded by autophagy. Consistent with this view, autophagosome formation is induced by many NLRP3 stimuli, autophagosomes partially colocalize with activated NLRP3 inflammasomes, loss of autophagosome function increases inflammasome activity, and K63-linked ubiquitin chain modification leads to autophagosome recruitment of ASC.102

5 |. ALTERNATIVE SPLICING OF THE NLRP3 MRNA

We originally observed extensive alternative splicing of the 3′ end of the NLRP3 gene when sequencing RT-PCR products during the initial cloning suggesting yet another mechanism that could regulate NLRP3 inflammasome function.6 More recently, we collaborated with Eike Latz to leverage high throughput Illumina sequencing to characterize NLRP3 mRNAs from human monocyte-derived macrophages to more closely analyze splice variants.103 Exons 5–9, which encode most of the LRR domain, have a remarkable structure. Each of these exons are 171 bp in length, precisely encode two leucine-rich repeats of 28 and 29 amino acids, and are in the same reading frame (Figure 5A). Each leucine-rich repeat is comprised of a sheet-turn-helix fold where the conserved leucine positions form a hydrophobic core that packs laterally with adjacent repeats to generate a structure that can be described as an arc or a solenoid. The effect of these features means that loss of any one or any combination of these exons will result in an NLRP3 protein with a shortened LRR domain that is predicted to still fold. This patten is shared by other NLR proteins but is distinct from TLR proteins in which the entire LRR domain-coding region exists in a single exon. Alternative splicing of NLRP3, and potentially other NLR proteins, is reminiscent of the adaptive immune system of jawless fish that rely upon lymphocyte receptors whose structure is varied through somatic recombination between short LRR-encoding exons.104

The NLRP3 LRR domain is known to interact with a variety of proteins, including SGT1, NEK7, and other NLRP3 molecules (Figure 5C,D). The most common NLRP3 splice form, NLRP3Δexon5 (previously termed NLRP3Δexon4), would be predicted to disrupt key interactions in the NLRP3 inactive cage structures as well as observed interactions with NEK7. NLRP3Δexon5,7 was also observed less commonly in this study,103 and we as well as Martinon and colleagues105 have observed yet other splice variants. Loss of exon 5 disrupted NEK7 binding and prevented both ASC speck formation and release of IL-1β in response to multiple NLRP3 activators, including nigericin.103 Similarly, expression of constructs lacking exons 5, 6, 8, or 10 (but not lacking exons 7 or 9) prevented nigericin activation of the inflammasome, suppressed auto-activation by the CAPS mutation p.R260W, reduced affinity for SGT1, and failed to trigger ASC polymerization.105 The inactive variants were unable to act as dominant negative mutants,105 consistent with the fact that they are present at modest levels in normal cells.103

Although these splice variants could arise by chance, the relative inefficiency of the splice site acceptors at exon 5 (and exon 7) that promote the more common splice variants103 suggest that these splice variants might have a biological role. One possibility is that these splice variants might promote inflammasome activation in response to other PAMP/DAMP signals or in combination with activating proteins other than NEK7. Remarkably, one nonsense mutation, p.R554X, results in a complete loss of the LRR domain and is associated with an atypical cold-induced autoinflammatory syndrome;106 however, this mutation is unusual as truncations of the LRR domain did not induce an inflammatory response in mice but could support one in response to nigericin.107 Another possibility is that these splice variants are normally inactive but generate stable proteins that fine-tune the inflammatory response.

This complex, multi-level regulation may be required to modulate the formation of NLRP3 inflammasomes and NLRP3-mediated immune responses, which may ultimately prevent chronic inflammation and tissue damage. The identification of patients with CAPS, however, highlights the risks of uncontrolled NLRP3-mediated inflammation and provides additional insights into NLPR3 folding, activation, and key models to link these molecular models with human disease.

6 |. CAPS MUTATIONS CAUSE A GAIN-OF-FUNCTION PHENOTYPE

CAPS mutations in NLRP3 are autosomal dominant and result in increased inflammasome activity, including caspase-1 activation, IL-1β cleavage and release, and inflammatory cell death, which has been observed in in vitro studies using cell lines transfected with plasmids carrying mutant NLRP3 or patient cells.108111 These studies also demonstrate that CAPS mutations lead to spontaneous formation of NLRP3-ASC specks, and their release into the supernatant causes phagocytosis and perpetuation of inflammasome-mediated inflammation.112,113 Similarly, peripheral blood mononuclear cells isolated from patients with CAPS show increased inflammasome activity with IL-1β and IL-18 secretion compared to healthy human controls at baseline or with minimal stimulation such as low dose LPS.114,115

6.1 |. Cold and NLRP3 in FCAS

The cold specificity of NLRP3 activation in FCAS led to the development of ex vivo, in vitro and murine models to further study the role of NLRP3 and the CAPS phenotype (Table 2). Ex vivo, adherent monocytes derived from the peripheral blood of patients with FCAS demonstrate significant release of IL-1β with as short as 1 h of incubation at 32°C, suggesting the presence of pre-transcribed and pre-translated pro-IL-1β in the presence of mutations in NLRP3. Prolonged incubation at this reduced temperature leads to release of IL-1β, IL-6, and TNF-alpha, consistent with perpetuation of an inflammatory cascade. Cold-induced IL-1β release could be inhibited by caspase-1 inhibition, confirming a role for inflammasome activation in cold-related symptoms. In addition, anakinra failed to prevent early IL-1β release but significantly reduced the late-phase transcription and release of all cytokines, consistent with a pro-IL-1 autoinflammatory feedback loop.116 This ex vivo system of “intrinsic” NLRP3 activation has become a key model for pre-clinical studies of therapeutics targeting the NLRP3 inflammasome.

TABLE 2.

Knockin and knockout murine models (modified from Hoffman177).

Human phenotype Human mutation Mouse mutation Design Mouse phenotype References
FCAS L353P L351P Conditional knockin Perinatal lethality, ex vivo cold sensitivity 114
JAX:017970
MWS A352V A350V Conditional knockin Growth defect, rash, early death as juvenile 114
JAX:017969
MWS R260W R258W Constitutive knockin Rash 123
NOMID D303N D301N Conditional knockin Growth defect, rash, early death, osteoporosis 132
JAX:017971
NOMID D303N n/a Syntenic knockin Adult-onset arthritis, osteoporosis 178
NOMID N477K N475K Conditional knockin Rash, growth defect, early death, amyloidosis 133
n/a n/a Deletion exons 1–2 Constitutive knockout Fertile, healthy 126,127
n/a n/a Insertion into intron 2 Constitutive knockout Fertile, healthy 95
n/a n/a LacZ Neo replacement of exons 4–9 Constitutive knockout Fertile, healthy 129
n/a n/a LacZ Neo replacement of exons 1–9 Constitutive knockout Fertile, healthy 129
n/a n/a Neo replacement of exons 1–9 Constitutive knockout Fertile, healthy 179
JAX:021302
n/a n/a Neo targeted deletion of exon 4 Conditional knockout Fertile, healthy 130

Abbreviations: FCAS, Familial cold autoinflammatory syndrome; MWS, Muckle Wells syndrome; n/a, not applicable.

More recently, Karasawa and colleagues investigated the long-standing question of how cold induces NLRP3 activation in FCAS.117 Using transfected cell lines, the authors showed that cells expressing FCAS-associated mutations p.L353P and p.Y563N formed cryo-sensitive aggregates that act as a scaffold for inflammasome activation.117 Aggregate formation was dependent on caspase-1 mediated calcium influx, rather than potassium efflux, suggesting a possible divergence in inflammasome activation between wild-type and mutant NLRP3, which has implications for therapy. Additional studies have implicated the involvement of heat shock protein 70 (HSP70) in the mechanism of cold induction observed in FCAS-associated mutant NLRP3.118

6.2 |. How do CAPS mutations cause NLRP3 activation?

Most CAPS mutations affect exon 4 of NLRP3 (previously called exon 3 prior to the identification of a short non-coding 5′ exon103), which encodes the NACHT domain (Figure 3). Analysis of disease-causing NLRP3 missense mutations in the Infevers database25 reveals seven protein regions (regions A–G) that are affected by many missense mutations in CAPS and other autoinflammatory diseases (Figure 2). In many cases, multiple missense mutations have been identified for the same residue in these regions.

The recent availability of active and inactive NLRP3 structures provides an opportunity to decipher the effects of these disease-causing mutations.42,50,76,119,120 Remarkably, the NACHT domains in the human and mouse NLRP3 “cage” assemblies do not contact each other.50,71,76 None of the mutations in the NACHT domain nor those in other NLRP3 domains affect the protein–protein contacts in the inactive assembly. Similarly, the protein–protein interfaces in the active NLRP3 disk conformation are not affected by most of the mutations, excepting p.D211N and p.H213R (NACHT-NACHT interface) and p.E567G/K/Q, p.R777C, and p.R918Q (LRR-NEK7 interface).42 Thus, most of the mutations likely do not stabilize or destabilize the protein–protein interactions in either the active or inactive conformations.

The NLRP3 protein undergoes a large closed-to-open conformational change when transitioning between the inactive and active states (Figure 6A,B). This motion can be described as a rigid body hinge motion between the FISNA-N BD-HD1 N-terminal subdomains of the NACHT domain and the WH-HD2-LRR portions of the protein. Strikingly, the commonly mutated regions A, B, and C form a single surface on the NBD, consistent with the predictions we made 15 years ago,9 and interact in the inactive conformation with the commonly mutated regions F (WH subdomain) and G (HD2 subdomain) (Figure 6C). The interface delineated by the regions A–C, F and G in the inactive conformation is where the NLRP3 inhibitor MCC950/CRID3 binds.50,71,121 This inhibitor is thought to function by stabilizing the inactive conformation by binding a pocket that is only present in the inactive conformation. This interface is lost in the active conformation, and many of the residues in regions A–C, F and G become more solvent exposed (Figure 6D). Similarly, the commonly mutated regions D and E are part of an interface of the HD1 domain with the HD2 and LRR domains that is present in the inactive conformation but undergoes substantial alteration in the active conformation (Figure 6C). Thus, a shared feature of each of the regions commonly mutated in autoinflammatory diseases (Figure 2) is that they lie at domain-domain interfaces that are lost or substantially changed in the conversion of inactive NLRP3 to active NLRP3 (Figure 6C,D).

FIGURE 6.

FIGURE 6

Many CAPS mutations lie at intradomain interfaces in the inactive conformation. (A) Distance difference matrix calculated by determining the differences between all Cα-Cα distances for the active NLRP3 disk conformation (PDB id 7pzc;174) and the inactive NLRP3 “cage” conformation (PDB id 8ej4;42). Similar results were obtained using other structures with the inactive NLRP3 conformation. Regions that do not move relative to each other have low (light colored) differences whereas regions that do have high (red colored) differences. The FISNA-NBD-HD1 and WH-HD2-LRR regions of NLRP3 behave as two rigid bodies whose relative motion can be described as a hinge. (B) Superposition of the active and inactive conformations using the FISNA-NBD-HD1 portion of the molecule, which shows the relative motion of the WH-HD2-LRR rigid body; structures of intermediate conformations were generated using ChimeraX.175 (C and D) Residues commonly mutated in autoinflammatory disease (regions A–G, see Figures 2 and 3) are shown as spheres in the inactive (C) and active (D) NLRP3 conformations. (E) Local environmental change (LEC) scores calculated for the conformational change between the inactive NLRP3 “cage” conformation and the active NLRP3 disk conformation with a radius cutoff of 5 Å. High scores indicate large changes to the local environment of the residue, and small scores indicate little or no change. Solid black circles are residues that are mutated in CAPS and undefined autoinflammatory disease. Open gray circles are other residues. The commonly mutated regions A–G are indicated by horizontal bars. (F) CAPS mutations that lie at interdomain boundaries in the inactive conformation (top) likely function to destabilize the inactive conformation and drive the NLRP3 conformation equilibrium to the active conformation (bottom).

To systematically explore all of the identified mutations and not just those at regions A–G, we identified domain–domain interface residues whose environment changed between the active and inactive conformation, using a local environmental change (LEC) score that we devised. This score is calculated by finding all atoms within a 5 Å cutoff in both conformations and calculating the average absolute difference between the distances in both conformations. As expected, the commonly mutated regions A–G were robustly identified by this analysis as well as some of the other mutated residues (Figure 6E). This analysis, however, indicated that there were some mutations associated with gain-of-function disease whose local environment was not substantially altered in the inactive-to-active conformational change (Figure 6E).

These data suggest the hypothesis that many of the mutations cause disease by disrupting the domain-domain interactions that stabilize the inactive state (Figure 6F). A shift in the equilibrium of the NLRP3 conformation to the active state predicts that the mutations would reduce or eliminate the need for a second activating signal such as potassium efflux. This hypothesis is consistent with multiple experimental results. First, CAPS mutations have reduced crosslinking to the MCC950/CRID3 inhibitor, which binds the inactive conformation.122 Second, in the absence of stimulation, the p.R260W, pA352V, and p.L353P mutant proteins form dispersed puncta that are similar to those formed by wild-type NLRP3 in the presence of stimulation.70 Third, the p.R260W mutation did not require potassium efflux for activation,123 but retained the requirement for NEK7 binding,73 while other mutations such as p.L353P appear to be more dependent on calcium influx.117 Fourth, more missense mutations are known to cause severe disease than mild disease, which could be consistent with a mechanism involving disruption of the inactive conformation, instead of stabilization of the active conformation if this bias is not due to the increased likelihood of patients with severe phenotypes to undergo more extensive genetic analysis. In general, mutations giving rise to different disease severity (FCAS vs. MWS vs. NOMID) are not distinguished by their position on the NLRP3 protein; disease severity may be a consequence of how strongly individual mutations affect the NLRP3 conformational equilibria and affect the sensitivity to intracellular ion flux.

Some disease-causing mutations do not affect residues that directly lie at the interfaces that change during the conformational change (Figure 6E). These mutations may drive a gain-of-function phenotype through one of several mechanisms. Some of these mutations might destabilize the inactive state indirectly by affecting the local protein structure, which may include adjacent interface residues. Other mutations might locally disrupt the structure and recruit protein chaperones; the NLRP3-chaperone interactions have the potential to promote the active, open conformation due to steric collisions with NLRP3 domains in the inactive, closed conformation. Finally, the small number of mutations that lie at the protein–protein interfaces in the active conformation could function by directly stabilizing the active, signaling state.

7 |. CONSERVATION OF NLRP3 ACROSS SPECIES

The NLRP3 gene sequence is strongly conserved among primate and non-primate mammals, particularly in the pyrin and NACHT domains, which was used to PCR amplify and clone portions of NLRP3 from multiple mammalian species.124 Analysis of the ratio of the rates of non-synonymous mutations (dN) to synonymous mutations (dS) within each branch in the NLRP3 phylogenic tree is less than 1.124 This result indicates that NLRP3 is subject to purifying selection during mammalian evolution, which is consistent with its central role in mediating innate immune responses to a wide variety of signals. An important observation of this early work, which has borne out by the subsequent sequencing of mammalian genomes from essentially every order of Mammalia, is that disease-causing mutations tend to affect highly conserved residues (Figure 7). Importantly, human and mouse NLRP3 share both extensive sequence conservation and patterns of expression.124 These similarities make the mouse a useful species to study NLRP3.

FIGURE 7.

FIGURE 7

Conservation of NLRP3 across species. Alignment of NLRP3 residues mutated in inflammatory diseases are highlighted in yellow if identical to the human sequence or red if identical to a disease-causing mutation. Sequences were sampled from species in different mammalian orders (Homo sapiens, human, Primates; Mus musculus, mouse, Rodentia; Dasypus novemcinctus, armadillo, Xenarthra; Tupaia chinensis, tree shrew, Scandentia; Galeopterus variegatus, colugos, Dermoptera; Bos taurus, cow, Cetartiodactyla; Erinaceaus europaens, hedgehog, Eulipotyphia; Loxodonta africana, elephant, Proboscidea; Equus caballus, horse, Perissodactyla; Rhinolophus ferrumequinum, horseshoe bat, Chiroptera; Canis lupis familiarus, dog, Carnivora; Orycteropus afer afer, aardvark, Tubulidentata; Oryctolagus cuniculus, rabbit, Lagomorpha). Mutation data from the Infevers database.25

7.1 |. The mouse is an excellent model to study NLRP3 and CAPS

Mice have been used for the study of immunity for decades, initially for in vivo experiments that cannot be performed in humans, and later to determine specific gene/protein function using recombinant technology allowing for the study of specific gene knockouts. However, since the beginning, disagreements have arisen between human and mouse immunologists as significant differences exist, particularly in lymphocyte biology, but also in innate immune responses.125 Despite these differences in immune function and phenotype, mice remain extremely useful as models for human immune diseases, and this holds true for the study of NLRP3 and CAPS, with some caveats.

7.2 |. Deletion of Nlrp3 in mice

In 2006, several landmark papers were published by various groups95,126,127 using Nlrp3 knockout mice. These studies revealed that NLRP3 was a unique and multifunctional protein with diverse activators including urate crystals, RNA, and toxins, with significant roles in several inflammatory and infectious diseases including gout, contact dermatitis, and Salmonella. These mice have continued to be studied over the last 17 years to expand our knowledge of the increasing number of roles for NLRP3 in the pathophysiology of diseases involving almost every organ. In fact, descriptions of conditions where NLRP3 does not play a role are the exception, rather than the rule.128

To better understand the function of the LRR region, we developed a unique mouse model with deletion of only the C-terminal LRR domain of NLRP3 in collaboration with Regeneron Pharmaceuticals. We showed that the LRR domain is necessary for NLRP3 function in both in vivo models (i.e., gout) and in ex vivo responses to several known activators by comparing this LRR deletion mouse to a full Nlrp3 knockout.129 In order to understand the role of NLRP3 in specific cells and tissues in different disease models, we also developed and published the first conditional Nlrp3 knockout mouse in collaboration with Ariel Feldstein, showing that loss of Nlrp3 expression in myeloid cells is crucial for the development of acute and chronic liver disease.130 The availability of the conditional Nlrp3 knockout combined with increased accessibility of specific Cre mice has allowed for increased understanding of the role of NLRP3 in non-hematopoietic cells and in other immune cells outside of the myeloid compartment.130

7.3 |. CAPS associated gain-of-function mutations in Nlrp3 in mice produce a CAPS phenotype

While deletion of Nlrp3 in mice established the role of NLRP3 in numerous mouse disease models, knockout mice did not provide direct information related to CAPS, where patients have gain-of-function mutations in NLRP3. Although we had access to CAPS patient cells and already had three effective therapies for patients, questions remained concerning disease mechanisms, particularly the response to cold in FCAS patients.131 Pre-clinical models to test novel therapies were also desirable. Therefore, in collaboration with Susannah Brydges and Dan Kastner, James Mueller and I generated three different conditional mutant Nlrp3 mouse models to cover the mild, moderate, and severe aspects of the CAPS spectrum and attempt to understand differences between the phenotypes observed with different mutations.114

The conditional nature of the mutations in these mice was fortuitous and allowed for a variety of useful experimental manipulations. Our initial approach was to recreate heterozygous mutations in all cells similar to most CAPS patients using a universal Cre, resulting in significant systemic inflammation, poor growth, and reduced survival in all of the mutant models. Laboratory evaluation showed neutrophilia in blood and elevation of many proinflammatory cytokines in serum including IL-1β, IL-18, IL-6, IL-8, and granulocyte-colony stimulating factor (G-CSF), but not TNF. Histologic analysis showed neutrophilia in skin, joints, conjunctiva, and the meninges, but not lungs, heart, kidneys, and gut consistent with the tissues known to be affected in CAPS. Neutrophilia was also observed in bone marrow, liver, and spleen, and muscle. Similar to CAPS patients, IL-1β and IL-6 protein expression was increased in affected skin.114 The primary difference between human and mouse CAPS was the overall increased severity of disease in these mice, with all of the mutations showing mortality ranging between birth and 6 weeks of life. However, the most unexpected observation was the reversal of the spectrum of severity in that the mice with the FCAS mutation (correlating to human p.L353P) were the most severely affected, and the mice with the NOMID mutation were the least severely affected, highlighting unexplained differences between humans and mice.114,132 This curious pattern was extended with the later development of a mouse using the same construct and strategy but inserting a mutation with a more severe human NOMID phenotype. This mouse developed by Chiesa and colleagues was less severely affected than the NOMID mouse we generated, living long enough to observe the chronic sequela of systemic amyloidosis observed in some CAPS patients.133 A similar reversed pattern of severity was also observed in our three mice bred to a tamoxifen-inducible Cre in which injecting these adult mice with tamoxifen resulted in systemic inflammation, rash, weight loss, and death.134,135 While the mechanism of reversal in severity between humans and mice with mutations in NLRP3 is still unknown, the spectrum of severity in murine CAPS allow us to choose different models depending on the desired study or expected effect of a genetic or pharmacologic intervention.

7.4 |. Nlrp3 mutant mice reveal myeloid cells and neutrophils as mediating CAPS

To determine the role of specific immune cells in CAPS, we adopted two strategies. First, we bred our universal mutant mice to mice lacking specific cell types (i.e., T cells, Rag1 or mast cells, KitWsh). The progeny of these crosses did not have altered phenotypes, suggesting that T cells and mast cells do not play significant roles in mouse CAPS. Second, we crossed mice whose NLRP3 mutant expression was Cre dependent with mice whose Cre expression was cell-specific, including lysozyme Cre (myeloid cells), Mcpt5 Cre (mast cells), and MRP8 Cre (neutrophils). Restriction of Nlrp3 expression to myeloid cells resulted in a similar phenotype to universal expression.114 This result suggested that myeloid cells are the primary drivers of CAPS phenotype and was consistent with observations in patients whose complete and typical CAPS phenotypes are due somatic mosaicism and expression of NLRP3 mutations primarily in myeloid cells. Similarly, breeding our Nlrp3 mutant mice to MRP8 Cre mice resulted in a phenotype similar to the lysozyme Cre mice suggesting an important role for neutrophils in mouse CAPS.136,137 In contrast, breeding the mutant CAPS mouse to Mcpt5 Cre had no apparent phenotype indicating a minimal role for mast cells.136 despite the urticarial nature of the CAPS rash and previous data suggesting a role for mast cells in another mouse model.138

7.5 |. Nlrp3 mutant mice support both IL-1 dependent and independent pathophysiologies in CAPS

The discovery of NLRP3 as a platform for caspase 1 activation and the success of IL-1 targeted therapies in patients with CAPS pointed toward a major pathologic role for IL-1β, so it was natural that we would investigate the effect of blocking the IL-1 pathway downstream using genetic and pharmacologic approaches in murine models. Therefore, we bred our Nlrp3 mutant mice to IL-1β and IL-1R knockouts.114,139 While there was some rescue, we still observed persistent rash and decreased growth and survival in the first few weeks of life in these mice. Interestingly, we observed some differences between IL-1β and IL-1R knockouts suggesting a possible role for IL-1α in mouse CAPS, which remains to be investigated. The MWS mutant IL-1β or IL-1R deficient mice that survived past 6 weeks had long-lasting scarring due to rash, but they had near normal growth and survival, more similar to human CAPS patients treated with IL-1 targeted therapy. We also treated mutant pups with a mouse rilonacept and showed partial rescue similar to the early rescue of the IL-1 targeted genetic approaches.114 We and others have also treated mutant adult inducible mice with recombinant IL-1RA (anakinra) with partial rescue of phenotype, but this was not as effective as an IL-1RA gene therapy in this model.140

The identification of IL-1 independent mechanisms in the pathophysiology of murine CAPS directed our attention to other pro-inflammatory cytokines, beginning with IL-18, another inflammasome-associated cytokine requiring caspase 1 for cleavage and activation. Mutant mice treated with an IL-18 binding protein demonstrated significant improvement in rash, weight gain, survival, and liver pathology.141 Similarly, mutant Nlrp3 mice on an IL-18 or IL-18R deficient background revealed a strong phenotypic rescue in pups with complete resolution of rash, and normal growth and survival through the first 2 months of life in the MWS model. However, we did observe a later onset phenotype after 8 weeks of age characterized by weight loss, systemic and tissue inflammation, and ultimately reduced survival suggesting that at least in MWS mice, IL-18 drives the early CAPS phenotype and IL-1 drives the later CAPS phenotype. The logical next step was to generate an Nlrp3 mutant mouse on an IL-1β/IL-18 double knockout background to investigate a synergistic role for these cytokines, hypothesizing that the phenotypic rescue would be better than each cytokine alone. While our hypothesis was correct, to our surprise, we did identify IL-1/IL-18 independent mechanisms involved and demonstrated that these were at least partially driven by NLRP3-dependent pyroptotic cell death.139

Using similar strategies, we also evaluated other pro-inflammatory cytokines besides those specifically regulated by NLRP3 including the downstream cytokines TNF, IL-17, and IL-6. Historically, treating CAPS patients with TNF targeted therapy did not have beneficial effects, but treating our Nlrp3 mutant mice with etanercept, a soluble TNF receptor, resulted in an unexpected rescue.142 Consistently, breeding the Nlrp3 mutant mice to TNF knockout mice also improved our previously reported primary outcomes in the pups including early tissue inflammation and fibrosis,142,143 but similar to mutant IL-18 knockout mice, this rescue was short-lived as we observed the development of a late onset inflammatory phenotype. While IL-17 plays a major role in several inflammatory diseases and can be driven by IL-1β, we observed little change in the mouse CAPS phenotype when we bred our mutant mice on an IL-17a deficient background.143 Similarly, IL-6, another well-known IL-1 driven pro-inflammatory cytokine had no obvious effect when deleted from the Nlrp3 mutant mice,144 consistent with the lack of clinical effect when a patient with CAPS was treated with an IL-6 targeted therapy.145

We have used similar genetic and pharmacologic approaches to tease apart upstream inflammasome pathways. Breeding our Nlrp3 mutant mice on an ASC-deficient background resulted in the only complete and long-lasting phenotypic rescue that we have identified to date.114 We have observed time-limited clinical improvement in our mice bred on caspase 1 knockout backgrounds, suggesting that several inflammasome independent pathways are driven by NLRP3. Finally, we and others have treated our mice with NLRP3 inhibitors demonstrating significant efficacy and providing the pre-clinical data for ongoing and planned clinical trials in CAPS patients.97,146

8 |. CAPS AND NLRP3 ARE A CONSUMMATE EXAMPLE OF BEDSIDE TO BENCH AND BACK

The hallmark of autoinflammatory diseases has been a nearly side-by-side understanding of patient clinical phenotypes, human genetics and molecular modeling, and translation to therapy in patients. Over the last several decades, the collective contributions from physicians, scientists, and patients around the world have resulted in the approval of therapies blocking IL-1-induced inflammation in CAPS, with ongoing industry investment pursuing upstream targets, including NLRP3.

8.1 |. Early IL-1 targeted therapy in CAPS

The observation that cold exposure led to reproducible and easily observed inflammatory symptoms and laboratory changes in patients with FCAS led to an investigator-initiated trial in a small number of patients to determine whether pre-treatment with an anti-IL-1 therapeutic could prevent the NLRP3-specific cold-induced symptoms.116 At the time, the only approved drug in the IL-1 pathway was anakinra, a recombinant version of the naturally occurring IL-1R antagonist. This short acting injectable medicine was developed for the treatment of sepsis, but approved only for rheumatoid arthritis at the time. Self-appointed the “anakinra chicks,” three women with FCAS underwent purposeful cold exposure—45 min in a laboratory cold room—with and without pre-treatment with two doses of anakinra. Cold exposure prior to treatment led to the typical pattern of fever, rash, arthralgias, with rapid increases in WBC and serum IL-6 within hours. Symptoms failed to develop when patients were pre-treated for 24 h with anakinra. This somewhat unconventional approach demonstrating that IL-1 blockade could successfully reduce acute inflammatory symptoms in patients with gain-of-function NLRP3 mutations was similar to results observed in a more traditional study measuring chronic symptom and laboratory inflammation control in two patients with the more severe MWS.147 These two proof of concept trials were the basis of a more complex and extremely successful clinical trial in patients with NOMID leading to ultimate approval by the FDA.148

8.2 |. Currently approved IL-1 targeted therapies in CAPS

To date, three drugs targeting the IL-1 pathway are approved by the FDA and EMA for the treatment of CAPS spectrum disorders, namely anakinra, rilonacept, a recombinant IL-1R that binds to and inhibits IL-1α, IL-1β, and IL-1RA, and canakinumab, a human monoclonal antibody that binds specifically to IL-1β.149,150 While each drug has different structure and mechanisms including the ability to block IL-1α and very different pharmacokinetics, they all have similar safety profiles and are well tolerated by patients. Decisions of which medicine to choose are often based on patient preference, availability, and costs/insurance limitations. Combined with their success in treating these rare disorders, the expansion of diseases implicating IL-1 in inflammatory disease pathogenesis has made this pathway a lucrative target for the pharmaceutical industry. Beyond inhibition of IL-1 and the IL-1R, development of other biologics and small molecule inhibitors targeting the different steps from NLRP3 assembly to IL-1 release is an active area, with CAPS patients often playing an integral part in the process (Table 3).

TABLE 3.

Integral role of CAPS patients in clinical trials.

NCT Number Study Status Conditions Interventions
NCT05186051 Completed CAPS ZYIL1 capsule
NCT00991146 Completed CAPS Canakinumab
NCT04868968 Completed FCAS DFV890
NCT00288704 Completed FCAS, MWS Rilonacept vs. Placebo
NCT01302860 Completed CAPS ACZ885
NCT00465985 Completed MWS ACZ885 vs. Placebo
NCT01105507 Completed CAPS Canakinumab (ACZ885D)
NCT00214851 Completed FCAS Kineret (anakinra)
NCT01576367 Completed CAPS BIOLOGICAL: ACZ885
NCT00685373 Completed CAPS Canakinumab (ACZ885)
NCT04086602 Completed CAPS IZD334 vs. Placebo
NCT01045772 Completed MWS Rilonacept
NCT03923140 Recruiting CAPS Tranilast
NCT05812781 Recruiting CAPS VTX2735
NCT05670301 Recruiting Autoinflammatory Disease, incl CAPS Cytokine and lipidomic profiling

Abbreviations: CAPS, cryopyrin-associated periodic syndrome; FCAS, Familial cold autoinflammatory syndrome; MWS, Muckle Wells syndrome.

8.3 |. Other potential drug targets in the NLRP3 pathway

Despite the efficacy of IL-1 drugs in CAPS and other inflammatory disease, there has been a keen interest in other targets for a variety of reasons including finding oral available treatments and theoretical advantages of targeting specific mechanisms upstream of IL-1 and the IL-1R in order to preserve some IL-1 function through other inflammasomes. For example, specific inhibition of the NLRP3 pathway may reduce the risk of infections associated with more general IL-1 blockade.151 There was early interest in the blocking the enzymatic function of caspase 1 as a strategy to block IL-1 release resulting in inhibitors with reasonable specificity reaching later stages of development. We demonstrated that one of these compounds VX-765 was very effective at blocking ex vivo IL-1β release from FCAS patient cells.115 In vivo studies were initiated, but never completed nor reported. There has also been steady interest in the signaling pathways downstream of IL-1R such as MEK, IRAK4, and MK2 with initiation of pre-clinical studies in CAPS; however, the latter is the only published report.152 Other strategies to block the NLRP3 inflammasome pathway that are being studied and developed include ASC, NEK7, and gasdermin D but the largest focus has been on inhibiting NLRP3 directly beginning with the most prevalent NLRP3 targeting molecule initially called CRID3 (now known as MCC950).97 While clinical development in this drug was halted years ago, it is a commonly used effective tool compound153 and has been used to guide development of dozens of related and unrelated compounds currently in different stages of development. Many of these drugs have been studied in CAPS mice and patient cells,146 and a few have been administered in vivo to CAPS patients.154 In addition to small molecule inhibitors of NLRP3, we have also studied anti sense therapy as a novel approach to the treatment of CAPS and other NLRP3 driven diseases.155 (Table 4).

TABLE 4.

NLRP3 targeted drugs in development.

Drug Mechanism Clinical trial
BMS-986299 Agonist NCT03444753
Dapansutrile Small molecular inhibitor NCT03595371, NCT04540120
IZD334 Small molecule inhibitor NCT04086602
ZYIL1 Small molecule inhibitor NCT04731324
IZD174 Small molecule inhibitor NCT04338997
AC-201 Small molecule inhibitor NCT02287818
Tranilast Small molecule inhibitor NCT05130892, NCT03923140
Oridonin Small molecule inhibitor NCT05130892
VTX2735 Small molecule inhibitor NCT05812781
DFV890 Small molecule inhibitor NCT05552469, NCT04886258

9 |. EXPANDING ROLE FOR NLRP3 IN HUMAN DISEASE

The discovery of NLRP3 began with CAPS, but the extent of the roles that NLRP3 plays in the etiology of other diseases has extended well beyond this monogenic autoinflammatory disease. The first complex disease shown to be mediated by NLRP3 was gout, as monosodium urate (MSU) was the first of many crystals shown to activate the NLRP3 inflammasome leading to IL-1β mediated inflammation.95 Early in vitro studies were later confirmed in vivo using NLRP3 deficient mice in models of gout.129 This was later translated to humans in studies using IL-1 inhibitors in acute and chronic gout,156 that preceded clinical trials of oral small molecule NLRP3 inhibitors currently in development.

International interest in the role of IL-1 mediated inflammation quickly linked the NLRP3 inflammasome to other common disorders. Early data suggested a role for the IL-1 pathway in nonalcoholic fatty liver disease (NAFLD) based on mouse models with IL-1RA deficient mice that developed worse liver pathology.157 We hypothesized that NLRP3 drives the IL-1 mediated inflammation which was supported by liver pathology in our NLRP3 gain-of-function (CAPS) mice including neutrophilic inflammation, hepatic stellate cell activation, collagen formation, and fibrosis that was similar to that observed in diet-induced NAFLD mouse models. We also showed that diet-induced NAFLD models were protected in mice that were deficient for NLRP3.158,159 This began a now decade long investigation into the upstream and downstream NLRP3-d riven mechanisms leading to NAFLD and the use of known and novel oral NLRP3 inhibitors for murine NAFLD, with a goal of translation to human patients.160,161

In cardiovascular disease, the role of IL-1, macrophages and inflammation in coronary artery disease, atherosclerosis and myocardial infarction was similarly recognized162 with multiple investigators aiming to reduce morbidity and mortality by targeting the IL-1β pathway. In one of the largest studies to date, the CANTOS trial showed a modest but significant reduction in nonfatal myocardial infarction, nonfatal stroke, or cardiovascular death in patients with a history of previous myocardial infarction and elevated C-reactive protein (CRP) who received canakinumab compared to placebo.163 A specific role of NLRP3 activation in the mechanisms of ischemic heart disease and ventricular dysfunction was shown using our inducible mutant Nlrp3 mice.164 In pediatrics, the role of NLRP3-mediated IL-1 signaling was also shown to play a key role in the development of aneurysms in Kawasaki disease, the most common cause of acquired cardiac disease in children in the United States.165,166 These studies suggest that NLRP3 may be also be an appropriate therapeutic target across the spectrum of human heart disease.

Beyond these disorders, the role of NLRP3 and its downstream effects through IL-1 have been implicated in nearly every organ in humans. While some are perhaps more obvious given clear associations with known inflammasome triggers such as in silicosis167 or asbestosis,168 others such as neurodegenerative disorders169 and ovarian aging153 have been linked to excess NLRP3 activation and expression (Figure 8). For these challenging disorders, inhibitors of NLRP3 have been shown to reduce inflammation in vitro and in vivo, lending new opportunities for intervention.

FIGURE 8.

FIGURE 8

Expanding role of NLRP3 in human disease. The NLRP3 inflammasome has been linked via expression or mechanistic studies to numerous, more common human diseases including neurologic, ophthalmologic, cardiopulmonary, hepatic, renal, osteal and joint, as well as disorders of metabolism and reproduction. CNS, central nervous system; COPD, chronic obstructive pulmonary disease; HEENT, head, eyes, ears, nose, and throat; NAFLD, nonalcoholic fatty liver disease; NASH, Nonalcoholic steatohepatitis. Figure created with Biorender.com.

10 |. ONGOING QUESTIONS

Translationally, CAPS has demonstrated a true bench to bedside circle, with the identification of NLRP3, our understanding of the inflammasome and IL-1 release, and subsequent use of targeted therapies leading to heightened understanding of innate immune mechanisms, as well as providing a much-needed therapy to CAPS patients and others with IL-1 mediated disease. However, the expansion of NLRP3 association with other disorders, and increased sequencing of patients with signs and symptoms of autoinflammation, but features of non-monogenic disease has led to new challenges. For example, patients with NLRP3 variants and atypical CAPS phenotypes, including patients with variants in the LRR domains, may have hearing loss, with less prominent systemic inflammatory symptoms or urticarial-like rash.170,171 Furthermore, increased recognition of low-penetrance variants,35 oligogenic or digenic presentations,172 somatic mosaicism2932 and “mutation-negative” patients continue to be clinically challenging in both the diagnosis and long-term therapeutic management. These cases further highlight the need for molecular clinical testing to determine whether a variant or collection of variants are pathogenic, and to determine severity of the phenotype. Understanding how these complex genetic presentations lead to disease will be important to fully appreciate the role of NLRP3 in regulating innate immunity.

Additionally, with NLRP3-mutation positive patients as proof-of-principle that inflammasome-induced inflammation could be successfully targeted with anti-IL-1 therapies, there was excitement that many more common diseases could similarly be treated with one of the three approved compounds: anakinra, canakinumab, or rilonacept. In theory, the different mechanisms of action would allow better understanding of inflammation in these disorders, as it has for CAPS. In clinical experience, however, the results and successes have not necessarily been as clear, likely due to polygenic and environmental contributions to these disorders, as well as incompletely understood intra-IL-1 family member regulatory mechanisms. Harnessing this pathway, rather than simply inhibiting it, could be equally useful in more complex, evolving diseases such as cancer173 where precise timing of turning on and turning off inflammation may better direct immune responses, inhibit disease progression, and identify adjunct therapies.

11 |. FUTURE DIRECTIONS AND CONCLUSIONS

Although we now have effective biologic therapies for CAPS, there remains an unmet medical need for less expensive small molecule inhibitors that target the inflammasome directly or the downstream inflammatory pathways of IL-1. Such targeted therapy would likely have significant impact on the treatment of more common inflammasome-mediated disorders as well. While the last several decades have led to significant advances in our understanding of the role of the inflammasome in innate immunity and human disease, unanswered questions remain concerning CAPS pathophysiology including the unique tissue involvement of these systemic inflammatory disorders, the specific hematopoietic and somatic cells involved in autoinflammatory disease pathogenesis, and the best way to target NLRP3 involvement in related but polygenic disorders. The integration of patients, physicians, and scientists will continue to be integral to understanding how one unique protein can carefully orchestrate the innate immune response.

ACKNOWLEDGMENTS

This work was supported by funding from the National Institutes of Health R01DK113592, R01HL140898, R01AI155869, P01HL152958 (HMH). The authors wish to dedicate this review to A. Fowler and B. Souther in recognition of their lifelong enthusiasm for understanding NLRP3 and CAPS.

Funding information

National Institutes of Health, Grant/Award Number: P01HL152958, R01AI155869, R01DK113592 and R01HL140898

Footnotes

CONFLICT OF INTEREST STATEMENT

LB is a site PI for Novartis, Inc. HMH is a speaker and consultant for Novartis, Inc., and has research collaborations with Regeneron, Inc., Jecure, Inc., and Zomagen, Inc.

DATA AVAILABILITY STATEMENT

The data that support the findings of this study are available at publicly available repositories.

The NLRP3 mutations in CAPS and undefined autoinflammatory diseases were derived from the Infevers database at https://infevers.umai-montpellier.fr/web/index.php. The structures of NLRP3, ASC pyrin domain, Caspase 1 CARD domain were downloaded from the RSCB Protein database at https://www.rcsb.org/ with accessions 5fna, 6npy, 7keu, 7pzc, 7pzd, 7zgu, and 8ej4. The sequences of the mammalian NLRP3 homologs were downloaded from the NCBI protein sequence database at https://www.ncbi.nlm.nih.gov/protein/ with accessions MK829787.1, NM_145827.4, XM_006162028.3, XM_010807177.3, XM_023540331.1, XM_033095764.1, XM_03544093.1, NM_004895.5, XM_004481057.3, XM_008579966.1, XM_016193428.1, XM_023618258.1, XM_036904180.2, and XM_042782806.1. The clinical trial information were downloaded from https://clinicaltrials.gov/ with accessions NCT05186051, NCT00991146, NCT04868968, NCT00288704, NCT01302860, NCT00465985, NCT01105507, NCT00214851, NCT01576367, NCT00685373, NCT04086602, NCT01045772, NCT03923140, NCT05812781, NCT05670301. NCT03444753, NCT03595371, NCT04540120, NCT04086602, NCT04731324, NCT04338997, NCT02287818, NCT05130892, NCT03923140, NCT05130892, NCT05812781, NCT05552469, and NCT04886258.

REFERENCES

  • 1.Kaufmann SH. Immunology’s foundation: the 100-year anniversary of the Nobel prize to Paul Ehrlich and Elie Metchnikoff. Nat Immunol. 2008;9(7):705–712. [DOI] [PubMed] [Google Scholar]
  • 2.Riedel S Edward Jenner and the history of smallpox and vaccination. Proceedings (Baylor University. Medical Center). 2005;18(1):21–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mackay IR. Travels and travails of autoimmunity: a historical journey from discovery to rediscovery. Autoimmun Rev. 2010;9(5):A251–A258. [DOI] [PubMed] [Google Scholar]
  • 4.Ring J, Grosber M, Brockow K, Bergmann K-C. Anaphylaxis. In: Bergmann KC, Ring J, eds. History of Allergy. Vol 100. S. Karger AG; 2014. [DOI] [PubMed] [Google Scholar]
  • 5.French FMFC. A candidate gene for familial Mediterranean fever. Nat Genet. 1997;17(1):25–31. [DOI] [PubMed] [Google Scholar]
  • 6.Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. 2001;29(3):301–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.International_FMF_Consortium. Ancient missense mutations in a new member of the RoRet gene family are likely to cause familial Mediterranean fever. Cell. 1997;90(4):797–807. [DOI] [PubMed] [Google Scholar]
  • 8.McDermott MF, Aksentijevich I, Galon J, et al. Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell. 1999;97(1):133–144. [DOI] [PubMed] [Google Scholar]
  • 9.Aksentijevich I, Putnam CD, Remmers EF, et al. The clinical continuum of cryopyrinopathies: novel CIAS1 mutations in north American patients and a new cryopyrin model. Arthritis Rheum. 2007;56(4):1273–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hassink SG, Goldsmith DP. Neonatal onset multisystem inflammatory disease. Arthritis Rheum. 1983;26(5):668–673. [DOI] [PubMed] [Google Scholar]
  • 11.Hoffman HM, Wright FA, Broide DH, Wanderer AA, Kolodner RD. Identification of a locus on chromosome 1q44 for familial cold urticaria. Am J Hum Genet. 2000;66(5):1693–1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kile RL, Rusk HA. A case of cold urticaria with an unusual family history. JAMA. 1940;114(12):1067–1068. [Google Scholar]
  • 13.Shepard MK. Cold hypersensitivity. Birth Defects Orig Artic Ser. 1971;7(8):352. [PubMed] [Google Scholar]
  • 14.Urano Y, Shikiji T, Sasaki S, Fukuhara K, Arase S. An unusual reaction to cold: a sporadic case of familial polymorphous cold eruption? Br J Dermatol. 1998;139(3):504–507. [DOI] [PubMed] [Google Scholar]
  • 15.Hoffman HM, Wanderer AA, Broide DH. Familial cold autoinflammatory syndrome: phenotype and genotype of an autosomal dominant periodic fever. J Allergy Clin Immunol. 2001;108(4):615–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Muckle TJ, Wells M. Urticaria, deafness and amyloidosis: a new heredo-familial syndrome. Q J Med. 1962;31(2):235–248. [PubMed] [Google Scholar]
  • 17.Heller H, Sohar E, Sherf L. Familial Mediterranean fever. AMA Arch Intern Med. 1958;102(1):50–71. [DOI] [PubMed] [Google Scholar]
  • 18.Prieur AM, Griscelli C. Chronic meningo-c utaneo-articular syndrome in children. Rev Rhum mal Osteoartic. 1980;47(11):645–649. [PubMed] [Google Scholar]
  • 19.de Boeck H, Scheerlinck T, Otten J. The CINCA syndrome: a rare cause of chronic arthritis and multisystem inflammatory disorders. Acta Orthop Belg. 2000;66(5):433–437. [PubMed] [Google Scholar]
  • 20.Dollfus H, Hafner R, Hofmann HM, et al. Chronic infantile neurological cutaneous and articular/neonatal onset multisystem inflammatory disease syndrome: ocular manifestations in a recently recognized chronic inflammatory disease of childhood. Arch Ophthalmol. 2000;118(10):1386–1392. [DOI] [PubMed] [Google Scholar]
  • 21.Prieur AM. A recently recognised chronic inflammatory disease of early onset characterised by the triad of rash, central nervous system involvement and arthropathy. Clin Exp Rheumatol. 2001;19(1):103–106. [PubMed] [Google Scholar]
  • 22.Cuisset L, Drenth JP, Berthelot JM, et al. Genetic linkage of the Muckle-Wells syndrome to chromosome 1q44. Am J Hum Genet. 1999;65(4):1054–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Aksentijevich I, Nowak M, Mallah M, et al. De novo CIAS1 mutations, cytokine activation, and evidence for genetic heterogeneity in patients with neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases. Arthritis Rheum. 2002;46(12):3340–3348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Feldmann J, Prieur AM, Quartier P, et al. Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am J Hum Genet. 2002;71(1):198–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Milhavet F, Cuisset L, Hoffman HM, et al. The infevers autoinflammatory mutation online registry: update with new genes and functions. Hum Mutat. 2008;29(6):803–808. [DOI] [PubMed] [Google Scholar]
  • 26.Sarrauste de Menthiere C, Terriere S, Pugnere D, Ruiz M, Demaille J, Touitou I. INFEVERS: the registry for FMF and hereditary inflammatory disorders mutations. Nucleic Acids Res. 2003;31(1):282–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Touitou I, Lesage S, McDermott M, et al. Infevers: an evolving mutation database for auto-inflammatory syndromes. Hum Mutat. 2004;24(3):194–198. [DOI] [PubMed] [Google Scholar]
  • 28.Van Gijn ME, Ceccherini I, Shinar Y, et al. New workflow for classification of genetic variants’ pathogenicity applied to hereditary recurrent fevers by the international study Group for Systemic Autoinflammatory Diseases (INSAID). J Med Genet. 2018;55(8):530–537. [DOI] [PubMed] [Google Scholar]
  • 29.Kawasaki Y, Oda H, Ito J, et al. Identification of a high-frequency somatic NLRC4 mutation as a cause of autoinflammation by pluripotent cell-based phenotype dissection. Arthritis Rheumatol. 2017;69(2):447–459. [DOI] [PubMed] [Google Scholar]
  • 30.Kontzias A, Zarabi SK, Calabrese C, et al. Somatic mosaicism in adult-onset TNF receptor-associated periodic syndrome (TRAPS). Mol Genet Genomic Med. 2019;7(8):e791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mensa-Vilaro A, Teresa Bosque M, Magri G, et al. Brief report: late-onset Cryopyrin-associated periodic syndrome due to myeloid-restricted somatic NLRP3 mosaicism. Arthritis Rheumatol. 2016;68(12):3035–3041. [DOI] [PubMed] [Google Scholar]
  • 32.Rowczenio DM, Gomes SM, Arostegui JI, et al. Late-onset Cryopyrin-associated periodic syndromes caused by somatic NLRP3 mosaicism-UK single center experience. Front Immunol. 2017;8:1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kitamura A, Sasaki Y, Abe T, Kano H, Yasutomo K. An inherited mutation in NLRC4 causes autoinflammation in human and mice. J Exp Med. 2014;211(12):2385–2396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Anderson JP, Mueller JL, Misaghi A, et al. Initial description of the human NLRP3 promoter. Genes Immun. 2008;9(8):721–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kuemmerle-Deschner JB, Verma D, Endres T, et al. Clinical and molecular phenotypes of low-penetrance variants of NLRP3: diagnostic and therapeutic challenges. Arthritis Rheumatol. 2017;69(11):2233–2240. [DOI] [PubMed] [Google Scholar]
  • 36.Koonin EV, Aravind L. The NACHT family -a new group of predicted NTPases implicated in apoptosis and MHC transcription activation. Trends Biochem Sci. 2000;25(5):223–224. [DOI] [PubMed] [Google Scholar]
  • 37.Ting JP, Lovering RC, Alnemri ES, et al. The NLR gene family: a standard nomenclature. Immunity. 2008;28(3):285–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhong Y, Kinio A, Saleh M. Functions of NOD-like receptors in human diseases. Front Immunol. 2013;4:333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–426. [DOI] [PubMed] [Google Scholar]
  • 40.Khare S, Dorfleutner A, Bryan NB, et al. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity. 2012;36(3):464–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hu Z, Zhou Q, Zhang C, et al. Structural and biochemical basis for induced self-propagation of NLRC4. Science. 2015;350(6259):399–404. [DOI] [PubMed] [Google Scholar]
  • 42.Xiao L, Magupalli VG, Wu H. Cryo-EM structures of the active NLRP3 inflammasome disc. Nature. 2023;613(7944):595–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zhang L, Chen S, Ruan J, et al. Cryo-EM structure of the activated NAIP2-NLRC4 inflammasome reveals nucleated polymerization. Science. 2015;350(6259):404–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Poyet JL, Srinivasula SM, Tnani M, Razmara M, Fernandes-Alnemri T, Alnemri ES. Identification of Ipaf, a human caspase-1-activating protein related to Apaf-1. J Biol Chem. 2001;276(30): 28309–28313. [DOI] [PubMed] [Google Scholar]
  • 45.Bauernfeind FG, Horvath G, Stutz A, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F, Alnemri ES. Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. J Biol Chem. 2012;287(43):36617–36622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Py BF, Kim MS, Vakifahmetoglu-Norberg H, Yuan J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol Cell. 2013;49(2):331–338. [DOI] [PubMed] [Google Scholar]
  • 48.Mayor A, Martinon F, De Smedt T, Petrilli V, Tschopp J. A crucial function of SGT1 and HSP90 in inflammasome activity links mammalian and plant innate immune responses. Nat Immunol. 2007;8(5):497–503. [DOI] [PubMed] [Google Scholar]
  • 49.Kadota Y, Shirasu K, Guerois R. NLR sensors meet at the SGT1-HSP90 crossroad. Trends Biochem Sci. 2010;35(4):199–207. [DOI] [PubMed] [Google Scholar]
  • 50.Hochheiser IV, Behrmann H, Hagelueken G, et al. Directionality of PYD filament growth determined by the transition of NLRP3 nucleation seeds to ASC elongation. Sci Adv. 2022;8(19):eabn7583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lu A, Magupalli VG, Ruan J, et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014;156(6):1193–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Srinivasula SM, Poyet JL, Razmara M, Datta P, Zhang Z, Alnemri ES. The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. J Biol Chem. 2002;277(24):21119–21122. [DOI] [PubMed] [Google Scholar]
  • 53.Vajjhala PR, Mirams RE, Hill JM. Multiple binding sites on the pyrin domain of ASC protein allow self-association and interaction with NLRP3 protein. J Biol Chem. 2012;287(50):41732–41743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dick MS, Sborgi L, Ruhl S, Hiller S, Broz P. ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 2016;7:11929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhou JY, Sarkar MK, Okamura K, Harris JE, Gudjonsson JE, Fitzgerald KA. Activation of the NLRP1 inflammasome in human keratinocytes by the dsDNA mimetic poly(dA:dT). Proc Natl Acad Sci U S A. 2023;120(5):e2213777120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhao Y, Yang J, Shi J, et al. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature. 2011;477(7366):596–600. [DOI] [PubMed] [Google Scholar]
  • 57.Barnett KC, Li S, Liang K, Ting JP. A 360 degrees view of the inflammasome: mechanisms of activation, cell death, and diseases. Cell. 2023;186(11):2288–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Shen C, Li R, Negro R, et al. Phase separation drives RNA virus-induced activation of the NLRP6 inflammasome. Cell. 2021;184(23):5759–5774 e5720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Diez E, Lee SH, Gauthier S, et al. Birc1e is the gene within the Lgn1 locus associated with resistance to legionella pneumophila. Nat Genet. 2003;33(1):55–60. [DOI] [PubMed] [Google Scholar]
  • 60.Growney JD, Dietrich WF. High-resolution genetic and physical map of the Lgn1 interval in C57BL/6J implicates Naip2 or Naip5 in legionella pneumophila pathogenesis. Genome Res. 2000;10(8):1158–1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kofoed EM, Vance RE. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity. Nature. 2011;477(7366):592–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang SB, Narendran S, Hirahara S, et al. DDX17 is an essential mediator of sterile NLRC4 inflammasome activation by retrotransposon RNAs. Sci Immunol. 2021;6(66):eabi4493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gasse P, Riteau N, Charron S, et al. Uric acid is a danger signal activating NALP3 inflammasome in lung injury inflammation and fibrosis. Am J Respir Crit Care Med. 2009;179(10):903–913. [DOI] [PubMed] [Google Scholar]
  • 64.Halle A, Hornung V, Petzold GC, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9(8):857–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mariathasan S, Weiss DS, Newton K, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440(7081):228–232. [DOI] [PubMed] [Google Scholar]
  • 66.Yamasaki K, Muto J, Taylor KR, et al. NLRP3/cryopyrin is necessary for interleukin-1beta (IL-1beta) release in response to hyaluronan, an endogenous trigger of inflammation in response to injury. J Biol Chem. 2009;284(19):12762–12771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM, Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 2013;38(6):1142–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Petrilli V, Papin S, Dostert C, Mayor A, Martinon F, Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 2007;14(9):1583–1589. [DOI] [PubMed] [Google Scholar]
  • 69.Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21(3):263–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chen J, Chen ZJ. PtdIns4P on dispersed trans-Golgi network mediates NLRP3 inflammasome activation. Nature. 2018;564(7734):71–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ohto U, Kamitsukasa Y, Ishida H, et al. Structural basis for the oligomerization-mediated regulation of NLRP3 inflammasome activation. Proc Natl Acad Sci USA. 2022;119(11):e2121353119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tapia-Abellan A, Angosto-Bazarra D, Alarcon-Vila C, et al. Sensing low intracellular potassium by NLRP3 results in a stable open structure that promotes inflammasome activation. Sci Adv. 2021;7(38):eabf4468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.He Y, Zeng MY, Yang D, Motro B, Nunez G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature. 2016;530(7590):354–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schmid-Burgk JL, Chauhan D, Schmidt T, et al. A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) screen identifies NEK7 as an essential component of NLRP3 Inflammasome activation. J Biol Chem. 2016;291(1):103–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shi H, Wang Y, Li X, et al. NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component. Nat Immunol. 2016;17(3):250–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Andreeva L, David L, Rawson S, et al. NLRP3 cages revealed by full-length mouse NLRP3 structure control pathway activation. Cell. 2021;184(26):6299–6312 e6222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Magupalli VG, Negro R, Tian Y, et al. HDAC6 mediates an aggresome-like mechanism for NLRP3 and pyrin inflammasome activation. Science. 2020;369(6510):eaas8995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wu J, Fernandes-Alnemri T, Alnemri ES. Involvement of the AIM2, NLRC4, and NLRP3 inflammasomes in caspase-1 activation by listeria monocytogenes. J Clin Immunol. 2010;30(5):693–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hoss F, Rodriguez-Alcazar JF, Latz E. Assembly and regulation of ASC specks. Cell Mol Life Sci. 2017;74(7):1211–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kopito RR. Aggresomes, inclusion bodies and protein aggregation. Trends Cell Biol. 2000;10(12):524–530. [DOI] [PubMed] [Google Scholar]
  • 81.Ouyang H, Ali YO, Ravichandran M, et al. Protein aggregates are recruited to aggresome by histone deacetylase 6 via unanchored ubiquitin C termini. J Biol Chem. 2012;287(4):2317–2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xu S, Chen H, Ni H, Dai Q. Targeting HDAC6 attenuates nicotine-induced macrophage pyroptosis via NF-kappaB/NLRP3 pathway. Atherosclerosis. 2021;317:1–9. [DOI] [PubMed] [Google Scholar]
  • 83.Yan S, Wei X, Jian W, et al. Pharmacological inhibition of HDAC6 attenuates NLRP3 inflammatory response and protects dopaminergic neurons in experimental models of Parkinson’s disease. Front Aging Neurosci. 2020;12:78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yang Q, Li S, Zhou Z, et al. HDAC6 inhibitor Cay10603 inhibits high glucose-induced oxidative stress, inflammation and apoptosis in retinal pigment epithelial cells via regulating NF-kappaB and NLRP3 inflammasome pathway. Gen Physiol Biophys. 2020;39(2): 169–177. [DOI] [PubMed] [Google Scholar]
  • 85.Cao Z, Gu Z, Lin S, et al. Attenuation of NLRP3 Inflammasome activation by Indirubin-derived PROTAC targeting HDAC6. ACS Chem Biol. 2021;16(12):2746–2751. [DOI] [PubMed] [Google Scholar]
  • 86.Chen Q, Wang Y, Jiao F, et al. HDAC6 Inhibitor ACY1215 inhibits the activation of NLRP3 inflammasome in acute liver failure by regulating the ATM/F-Actin signalling pathway. J Cell Mol Med. 2021;25(15):7218–7228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hou Q, Kan S, Wang Z, et al. Inhibition of HDAC6 with CAY10603 ameliorates diabetic kidney disease by suppressing NLRP3 Inflammasome. Front Pharmacol. 2022;13:938391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bockstiegel J, Wurnig SL, Engelhardt J, Enns J, Hansen FK, Weindl G. Pharmacological inhibition of HDAC6 suppresses NLRP3 inflammasome-mediated IL-1beta release. Biochem Pharmacol. 2023;215:115693. [DOI] [PubMed] [Google Scholar]
  • 89.dos Santos G, Rogel MR, Baker MA, et al. Vimentin regulates activation of the NLRP3 inflammasome. Nat Commun. 2015;6:6574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Li X, Thome S, Ma X, et al. MARK4 regulates NLRP3 positioning and inflammasome activation through a microtubule-dependent mechanism. Nat Commun. 2017;8:15986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yue K, Sun S, Jia G, et al. First-in-c lass hydrazide-based HDAC6 selective inhibitor with potent Oral anti-i nflammatory activity by attenuating NLRP3 Inflammasome activation. J Med Chem. 2022;65(18):12140–12162. [DOI] [PubMed] [Google Scholar]
  • 92.Zhu Y, Feng M, Wang B, et al. New insights into the non-enzymatic function of HDAC6. Biomed Pharmacother. 2023;161:114438. [DOI] [PubMed] [Google Scholar]
  • 93.Lopez-Castejon G Control of the inflammasome by the ubiquitin system. FEBS J. 2020;287(1):11–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dalbeth N, Lauterio TJ, Wolfe HR. Mechanism of action of colchicine in the treatment of gout. Clin Ther. 2014;36(10):1465–1479. [DOI] [PubMed] [Google Scholar]
  • 95.Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–241. [DOI] [PubMed] [Google Scholar]
  • 96.Misawa T, Takahama M, Kozaki T, et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol. 2013;14(5):454–460. [DOI] [PubMed] [Google Scholar]
  • 97.Coll RC, Robertson AA, Chae JJ, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3):248–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gurung P, Lamkanfi M, Kanneganti TD. Cutting edge: SHARPIN is required for optimal NLRP3 inflammasome activation. J Immunol. 2015;194(5):2064–2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Zhang L, Ko CJ, Li Y, et al. Peli1 facilitates NLRP3 inflammasome activation by mediating ASC ubiquitination. Cell Rep. 2021;37(4):109904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Rodgers MA, Bowman JW, Fujita H, et al. The linear ubiquitin assembly complex (LUBAC) is essential for NLRP3 inflammasome activation. J Exp Med. 2014;211(7):1333–1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Guan K, Wei C, Zheng Z, et al. MAVS promotes Inflammasome activation by targeting ASC for K63-linked ubiquitination via the E3 ligase TRAF3. J Immunol. 2015;194(10):4880–4890. [DOI] [PubMed] [Google Scholar]
  • 102.Shi CS, Shenderov K, Huang NN, et al. Activation of autophagy by inflammatory signals limits IL-1beta production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hoss F, Mueller JL, Rojas Ringeling F, et al. Alternative splicing regulates stochastic NLRP3 activity. Nat Commun. 2019;10(1):3238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Boehm T, Hirano M, Holland SJ, Das S, Schorpp M, Cooper MD. Evolution of alternative adaptive immune Systems in Vertebrates. Annu Rev Immunol. 2018;36:19–42. [DOI] [PubMed] [Google Scholar]
  • 105.Theodoropoulou K, Spel L, Zaffalon L, Delacretaz M, Hofer M, Martinon F. NLRP3 leucine-rich repeats control induced and spontaneous inflammasome activation in cryopyrin-associated periodic syndrome. J Allergy Clin Immunol. 2023;151(1):222–232.e229. [DOI] [PubMed] [Google Scholar]
  • 106.Jeru I, Hayrapetyan H, Duquesnoy P, Sarkisian T, Amselem S. PYPAF1 nonsense mutation in a patient with an unusual autoinflammatory syndrome: role of PYPAF1 in inflammation. Arthritis Rheum. 2006;54(2):508–514. [DOI] [PubMed] [Google Scholar]
  • 107.Hafner-Bratkovic I, Susjan P, Lainscek D, et al. NLRP3 lacking the leucine-rich repeat domain can be fully activated via the canonical inflammasome pathway. Nat Commun. 2018;9(1):5182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Duncan JA, Bergstralh DT, Wang Y, et al. Cryopyrin/NALP3 binds ATP/dATP, is an ATPase, and requires ATP binding to mediate inflammatory signaling. Proc Natl Acad Sci USA. 2007;104(19): 8041–8046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.O’Connor W Jr, Harton JA, Zhu X, Linhoff MW, Ting JP. Cutting edge: CIAS1/cryopyrin/PYPAF1/NALP3/CATERPILLER 1.1 is an inducible inflammatory mediator with NF-kappa B suppressive properties. J Immunol. 2003;171(12):6329–6333. [DOI] [PubMed] [Google Scholar]
  • 110.Willingham SB, Allen IC, Bergstralh DT, et al. NLRP3 (NALP3, Cryopyrin) facilitates in vivo caspase-1 activation, necrosis, and HMGB1 release via inflammasome-dependent and -independent pathways. J Immunol. 2009;183(3):2008–2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Yu JW, Wu J, Zhang Z, et al. Cryopyrin and pyrin activate caspase-1, but not NF-kappaB, via ASC oligomerization. Cell Death Differ. 2006;13(2):236–249. [DOI] [PubMed] [Google Scholar]
  • 112.Baroja-Mazo A, Martin-Sanchez F, Gomez AI, et al. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014;15(8):738–748. [DOI] [PubMed] [Google Scholar]
  • 113.Franklin BS, Bossaller L, De Nardo D, et al. The adaptor ASC has extracellular and ‘prionoid’ activities that propagate inflammation. Nat Immunol. 2014;15(8):727–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Brydges SD, Mueller JL, McGeough MD, et al. Inflammasome-mediated disease animal models reveal roles for innate but not adaptive immunity. Immunity. 2009;30(6):875–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Stack JH, Beaumont K, Larsen PD, et al. IL-converting en-zyme/caspase-1 inhibitor VX-765 blocks the hypersensitive response to an inflammatory stimulus in monocytes from familial cold autoinflammatory syndrome patients. J Immunol. 2005;175(4):2630–2634. [DOI] [PubMed] [Google Scholar]
  • 116.Hoffman HM, Rosengren S, Boyle DL, et al. Prevention of cold-associated acute inflammation in familial cold autoinflammatory syndrome by interleukin-1 receptor antagonist. Lancet. 2004;364(9447):1779–1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Karasawa T, Komada T, Yamada N, et al. Cryo-sensitive aggregation triggers NLRP3 inflammasome assembly in cryopyrin-associated periodic syndrome. Elife. 2022;11:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Raghawan AK, Ramaswamy R, Swarup G. Cold-induced loss of interaction with HSC70 triggers inflammasome activity of familial cold autoinflammatory syndrome-causing mutants of NLRP3. Biochem Biophys Res Commun. 2023;641:42–49. [DOI] [PubMed] [Google Scholar]
  • 119.Sharif H, Wang L, Wang WL, et al. Structural mechanism for NEK7-licensed activation of NLRP3 inflammasome. Nature. 2019;570(7761):338–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Behavioral Dantzer R., physiological and functional aspects of stereotyped behavior: a review and a re-interpretation. J Anim Sci. 1986;62(6):1776–1786. [DOI] [PubMed] [Google Scholar]
  • 121.Dekker C, Mattes H, Wright M, et al. Crystal structure of NLRP3 NACHT domain with an inhibitor defines mechanism of Inflammasome inhibition. J Mol Biol. 2021;433(24):167309. [DOI] [PubMed] [Google Scholar]
  • 122.Vande Walle L, Stowe IB, Sacha P, et al. MCC950/CRID3 potently targets the NACHT domain of wild-type NLRP3 but not disease-associated mutants for inflammasome inhibition. PLoS Biol. 2019;17(9):e3000354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Meng G, Zhang F, Fuss I, Kitani A, Strober W. A mutation in the Nlrp3 gene causing inflammasome hyperactivation potentiates Th17 cell-dominant immune responses. Immunity. 2009;30(6):860–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Anderson JP, Mueller JL, Rosengren S, et al. Structural, expression, and evolutionary analysis of mouse CIAS1. Gene. 2004;338(1):25–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172(5):2731–2738. [DOI] [PubMed] [Google Scholar]
  • 126.Kanneganti TD, Ozoren N, Body-Malapel M, et al. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature. 2006;440(7081):233–236. [DOI] [PubMed] [Google Scholar]
  • 127.Sutterwala FS, Ogura Y, Szczepanik M, et al. Critical role for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1. Immunity. 2006;24(3):317–327. [DOI] [PubMed] [Google Scholar]
  • 128.Fusco R, Siracusa R, Genovese T, Cuzzocrea S, Di Paola R. Focus on the role of NLRP3 Inflammasome in diseases. Int J Mol Sci. 2020;21(12):4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Hoffman HM, Scott P, Mueller JL, et al. Role of the leucine-rich repeat domain of cryopyrin/NALP3 in monosodium urate crystal-induced inflammation in mice. Arthritis Rheum. 2010;62(7):2170–2179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kaufmann B, Kui L, Reca A, et al. Cell-specific deletion of NLRP3 Inflammasome identifies myeloid cells as key drivers of liver inflammation and fibrosis in murine Steatohepatitis. Cell Mol Gastroenterol Hepatol. 2022;14(4):751–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rosengren S, Mueller JL, Anderson JP, et al. Monocytes from familial cold autoinflammatory syndrome patients are activated by mild hypothermia. J Allergy Clin Immunol. 2007;119(4):991–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Bonar SL, Brydges SD, Mueller JL, et al. Constitutively activated NLRP3 inflammasome causes inflammation and abnormal skeletal development in mice. PLoS ONE. 2012;7(4):e35979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Bertoni A, Carta S, Baldovini C, et al. A novel knock-in mouse model of cryopyrin-associated periodic syndromes with development of amyloidosis: therapeutic efficacy of proton pump inhibitors. J Allergy Clin Immunol. 2020;145(1):368–378 e313. [DOI] [PubMed] [Google Scholar]
  • 134.Inzaugarat ME, Johnson CD, Holtmann TM, et al. NLR family pyrin domain-containing 3 Inflammasome activation in hepatic stellate cells induces liver fibrosis in mice. Hepatology. 2019;69(2):845–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Calcagno DM, Chu A, Gaul S, et al. NOD-like receptor protein 3 activation causes spontaneous inflammation and fibrosis that mimics human NASH. Hepatology. 2022;76(3):727–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Stackowicz J, Gaudenzio N, Serhan N, et al. Neutrophil-specific gain-of-function mutations in Nlrp3 promote development of cryopyrin-associated periodic syndrome. J Exp Med. 2021;218(10):e20201466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Kaufmann B, Leszczynska A, Reca A, et al. NLRP3 activation in neutrophils induces lethal autoinflammation, liver inflammation, and fibrosis. EMBO Rep. 2022;23(11):e54446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Nakamura Y, Franchi L, Kambe N, Meng G, Strober W, Nunez G. Critical role for mast cells in interleukin-1beta-driven skin inflammation associated with an activating mutation in the nlrp3 protein. Immunity. 2012;37(1):85–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Brydges SD, Broderick L, McGeough MD, Pena CA, Mueller JL, Hoffman HM. Divergence of IL-1, IL-18, and cell death in NLRP3 inflammasomopathies. J Clin Invest. 2013;123(11):4695–4705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Colantuoni M, Jofra Hernandez R, Pettinato E, et al. Constitutive IL-1RA production by modified immune cells protects against IL-1-mediated inflammatory disorders. Sci Transl Med. 2023;15(698): eade3856. [DOI] [PubMed] [Google Scholar]
  • 141.Knorr J, Kaufmann B, Inzaugarat ME, et al. Interleukin-18 signaling promotes activation of hepatic stellate cells in mouse liver fibrosis. Hepatology. 2023;77(6):1968–1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.McGeough MD, Wree A, Inzaugarat ME, et al. TNF regulates transcription of NLRP3 inflammasome components and inflammatory molecules in cryopyrinopathies. J Clin Invest. 2017;127(12): 4488–4497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wree A, McGeough MD, Inzaugarat ME, et al. NLRP3 inflammasome driven liver injury and fibrosis: roles of IL-17 and TNF in mice. Hepatology. 2018;67(2):736–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.McGeough MD, Pena CA, Mueller JL, et al. Cutting edge: IL-6 is a marker of inflammation with no direct role in inflammasome-mediated mouse models. J Immunol. 2012;189(6):2707–2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Matsubara T, Hasegawa M, Shiraishi M, et al. A severe case of chronic infantile neurologic, cutaneous, articular syndrome treated with biologic agents. Arthritis Rheum. 2006;54(7):2314–2320. [DOI] [PubMed] [Google Scholar]
  • 146.Ambrus-Aikelin G, Takeda K, Joetham A, et al. JT002, a small molecule inhibitor of the NLRP3 inflammasome for the treatment of autoinflammatory disorders. Sci Rep. 2023;13(1):13524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hawkins PN, Lachmann HJ, Aganna E, McDermott MF. Spectrum of clinical features in Muckle-Wells syndrome and response to anakinra. Arthritis Rheum. 2004;50(2):607–612. [DOI] [PubMed] [Google Scholar]
  • 148.Goldbach-Mansky R, Dailey NJ, Canna SW, et al. Neonatal-onset multisystem inflammatory disease responsive to interleukin-1beta inhibition. N Engl J Med. 2006;355(6):581–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov. 2012;11(8):633–652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lachmann HJ, Lowe P, Felix SD, et al. In vivo regulation of interleukin 1beta in patients with cryopyrin-associated periodic syndromes. J Exp Med. 2009;206(5):1029–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.LaRock CN, Todd J, LaRock DL, et al. IL-1beta is an innate immune sensor of microbial proteolysis. Sci Immunol. 2016;1(2):eaah3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wang C, Hockerman S, Jacobsen EJ, et al. Selective inhibition of the p38alpha MAPK-M K2 axis inhibits inflammatory cues including inflammasome priming signals. J Exp Med. 2018;215(5):1315–1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Navarro-Pando JM, Alcocer-Gomez E, Castejon-Vega B, et al. Inhibition of the NLRP3 inflammasome prevents ovarian aging. Sci Adv. 2021;7(1):eabc7409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hissaria P, Kansagra K, Patel H, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of ZY-IL1 in three patients with Cryopyrin-associated periodic syndromes. Clin Pharmacol Drug Dev. 2023:1–8. [DOI] [PubMed] [Google Scholar]
  • 155.Kaufmann B, de Los Reyes Jimenez M, Booshehri LM, et al. Antisense oligonucleotide therapy decreases IL-1beta expression and prolongs survival in mutant Nlrp3 mice. J Immunol 2023;211(2):287–294. [DOI] [PubMed] [Google Scholar]
  • 156.So A, Dumusc A, Nasi S. The role of IL-1 in gout: from bench to bedside. Rheumatology (Oxford). 2018;57(suppl_1):i12–i19. [DOI] [PubMed] [Google Scholar]
  • 157.Isoda K, Sawada S, Ayaori M, et al. Deficiency of interleukin-1 receptor antagonist deteriorates fatty liver and cholesterol metabolism in hypercholesterolemic mice. J Biol Chem. 2005;280(8):7002–7009. [DOI] [PubMed] [Google Scholar]
  • 158.Wree A, Eguchi A, McGeough MD, et al. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology. 2014;59(3):898–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Wree A, McGeough MD, Pena CA, et al. NLRP3 inflammasome activation is required for fibrosis development in NAFLD. J Mol Med (Berl). 2014;92(10):1069–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Mridha AR, Wree A, Robertson AAB, et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J Hepatol. 2017;66(5):1037–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Povero D, Lazic M, McBride C, et al. Pharmacology of a potent and novel inhibitor of the NOD-like receptor pyrin domain-c ontaining protein 3 (NLRP3) Inflammasome that attenuates development of nonalcoholic Steatohepatitis and liver fibrosis. J Pharmacol Exp Ther. 2023;386(2):242–258. [DOI] [PubMed] [Google Scholar]
  • 162.Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352(16):1685–1695. [DOI] [PubMed] [Google Scholar]
  • 163.Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with Canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–1131. [DOI] [PubMed] [Google Scholar]
  • 164.Toldo S, Mezzaroma E, McGeough MD, et al. Independent roles of the priming and the triggering of the NLRP3 inflammasome in the heart. Cardiovasc Res. 2015;105(2):203–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lee Y, Wakita D, Dagvadorj J, et al. IL-1 signaling is critically required in stromal cells in Kawasaki disease Vasculitis mouse model: role of both IL-1alpha and IL-1beta. Arterioscler Thromb Vasc Biol. 2015;35(12):2605–2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Wakita D, Kurashima Y, Crother TR, et al. Role of Interleukin-1 signaling in a mouse model of Kawasaki disease-associated abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol. 2016;36(5):886–897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Lam M, Mansell A, Tate MD. Another one fights the dust: targeting the NLRP3 Inflammasome for the treatment of silicosis. Am J Respir Cell Mol Biol. 2022;66(6):601–611. [DOI] [PubMed] [Google Scholar]
  • 168.Franko A, Goricar K, Kovac V, Dodic-Fikfak M, Dolzan V. NLRP3 and CARD8 polymorphisms influence risk for asbestos-related diseases. J Med Biochem. 2020;39(1):91–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Barczuk J, Siwecka N, Lusa W, Rozpedek-Kaminska W, Kucharska E, Majsterek I. Targeting NLRP3-mediated Neuroinflammation in Alzheimer’s disease treatment. Int J Mol Sci. 2022;23(16): 1879–1895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Fayand A, Cescato M, Le Corre L, et al. Pathogenic variants in the NLRP3 LRR domain at position 861 are responsible for a boost-dependent atypical CAPS phenotype. J Allergy Clin Immunol. 2023;152:1303–1311.e1. [DOI] [PubMed] [Google Scholar]
  • 171.Nakanishi H, Kawashima Y, Kurima K, et al. NLRP3 mutation and cochlear autoinflammation cause syndromic and nonsyndromic hearing loss DFNA34 responsive to anakinra therapy. Proc Natl Acad Sci U S A. 2017;114(37):E7766–E7775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Schnappauf O, Aksentijevich I. Current and future advances in genetic testing in systemic autoinflammatory diseases. Rheumatology (Oxford). 2019;58(Suppl 6):vi44–vi55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Zhao H, Wu L, Yan G, et al. Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther. 2021;6(1):263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Hochheiser IV, Pilsl M, Hagelueken G, et al. Structure of the NLRP3 decamer bound to the cytokine release inhibitor CRID3. Nature. 2022;604(7904):184–189. [DOI] [PubMed] [Google Scholar]
  • 175.Pettersen EF, Goddard TD, Huang CC, et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 2021;30(1):70–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Welzel T, Kuemmerle-Deschner JB. Diagnosis and Management of the Cryopyrin-Associated Periodic Syndromes (CAPS): what do we know today? J Clin Med. 2021;10(1):128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Hoffman HM. Autoinflammatory disease: new mouse models and therapies. J Allergy Clin Immunol. 2020;145(1):116–118. [DOI] [PubMed] [Google Scholar]
  • 178.Snouwaert JN, Nguyen M, Repenning PW, et al. An NLRP3 mutation causes Arthropathy and osteoporosis in humanized mice. Cell Rep. 2016;17(11):3077–3088. [DOI] [PubMed] [Google Scholar]
  • 179.Kovarova M, Hesker PR, Jania L, et al. NLRP1-dependent pyroptosis leads to acute lung injury and morbidity in mice. J Immunol. 2012;189(4):2006–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available at publicly available repositories.

The NLRP3 mutations in CAPS and undefined autoinflammatory diseases were derived from the Infevers database at https://infevers.umai-montpellier.fr/web/index.php. The structures of NLRP3, ASC pyrin domain, Caspase 1 CARD domain were downloaded from the RSCB Protein database at https://www.rcsb.org/ with accessions 5fna, 6npy, 7keu, 7pzc, 7pzd, 7zgu, and 8ej4. The sequences of the mammalian NLRP3 homologs were downloaded from the NCBI protein sequence database at https://www.ncbi.nlm.nih.gov/protein/ with accessions MK829787.1, NM_145827.4, XM_006162028.3, XM_010807177.3, XM_023540331.1, XM_033095764.1, XM_03544093.1, NM_004895.5, XM_004481057.3, XM_008579966.1, XM_016193428.1, XM_023618258.1, XM_036904180.2, and XM_042782806.1. The clinical trial information were downloaded from https://clinicaltrials.gov/ with accessions NCT05186051, NCT00991146, NCT04868968, NCT00288704, NCT01302860, NCT00465985, NCT01105507, NCT00214851, NCT01576367, NCT00685373, NCT04086602, NCT01045772, NCT03923140, NCT05812781, NCT05670301. NCT03444753, NCT03595371, NCT04540120, NCT04086602, NCT04731324, NCT04338997, NCT02287818, NCT05130892, NCT03923140, NCT05130892, NCT05812781, NCT05552469, and NCT04886258.

RESOURCES