Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 1.
Published in final edited form as: Int J Biochem Cell Biol. 2010 Jan 13;42(6):792–795. doi: 10.1016/j.biocel.2010.01.008

Nlrp3: an immune sensor of cellular stress and infection

Mohamed Lamkanfi 1,2, Thirumala-Devi Kanneganti 3
PMCID: PMC2862759  NIHMSID: NIHMS169866  PMID: 20079456

Abstract

Innate immune cells rely on pathogen recognition receptors such as the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family to mount an appropriate immune response against microbial threats. The NLR protein Nlrp3 senses microbial ligands, endogenous danger signals and crystalline substances in the cytosol to trigger the assembly of a large caspase-1-activating protein complex termed the Nlrp3 inflammasome. Autoproteolytic maturation of caspase-1 zymogens in the Nlrp3 inflammasome leads to maturation and extracellular release of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Gain-of-function mutations in the NOD domain of Nlrp3 are associated with auto-inflammatory disorders characterized by skin rashes and prolonged episodes of fever. In addition, decreased Nlrp3 expression was recently linked with susceptibility to Crohn’s disease in humans. In this review, we discuss recent developments on the role of the Nlrp3 inflammasome in innate immunity, its activation mechanisms and the auto-inflammatory disorders associated with deregulation of Nlrp3 inflammasome activity.

Keywords: NLR, Nlrp3, inflammasome, caspase-1, interleukin-1

Introduction

The innate immune system recognizes infections and cellular damage through pattern recognition receptors (PRRs) (Kumar et al., 2009). Recognition occurs through the detection of evolutionary conserved microbial ligands that are critical for microbial function such as flagellin and components of the bacterial cell wall or viral envelope (Kumar et al., 2009). Endogenous danger signals recognized by PRRs usually represent post-translational modifications of host proteins. The detection in an atypical location of molecules normally sequestered in a defined space represents a third way for signalling cellular dysfunction. Several classes of PRRs can be distinguished, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), RIG-I-like receptors (RLRs) and nucleotide binding and oligomerization domain (NOD)-like receptors (NLRs) (Kawai and Akira, 2006). TLRs and CLRs detect microbes on the cell surface and in endosomes, whereas RLRs and NLRs sense microbial components in intracellular compartments (Kanneganti et al., 2007b).

The discovery of NLRs as cytosolic PRRs suggested that microbes evading extracellular surveillance encounter a second line of defense in the host cytosol. Bioinformatics studies revealed the existence of 22 human NLR genes and recent gene duplications gave rise to 34 mouse NLRs (Kanneganti et al., 2007b). The NLR family member Nlrp3 (also known as Nalp3, Cryopyrin, CIAS1, PYPAF1 and CLR1.1) was originally identified as the gene mutated in patients suffering from the autosomal-dominant periodic fever syndromes familial cold autoinflammatory syndrome (FCAS), Muckle-Wells syndrome (MWS) and chronic infantile neurological cutaneous and articular syndrome (CINCA) (Hoffman et al., 2001, Feldmann et al., 2002). Our knowledge on the immune roles, signalling pathways and disease-associated mutations of Nlrp3 has dramatically increased in recent years and will be discussed below.

Structure

Nlrp3 is a protein of 1016 amino acids transcribed from the gene cias1, which is located on human chromosome 1q44 and consists of 9 coding exons (Figure 1). The architecture of Nlrp3 resembles that of a subset of plant disease-resistance (R) genes involved in the hypersensitive response against virulent plant pathogens (Lamkanfi and Dixit, 2009). Nlrp3 shares the presence of a centrally located NOD motif (also referred to as NBD or NACHT domain) with all other NLR family members (Kanneganti et al., 2007b). This NOD motif is flanked at the N-terminus by a pyrin domain to allow homotypic interactions with the bipartite adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) in the Nlrp3 inflammasome (see further). At the C-terminus, Nlrp3 contains an array of 12 leucine-rich repeat (LRR) motifs believed to be involved in modulating Nlrp3 activity and in sensing microbial ligands and endogenous alarmins (Lamkanfi and Dixit, 2009).

Figure 1.

Figure 1

Structural organization of Nlrp3. The human cias1 gene encoding Nlrp3 is localized on chromosome 1q44. The positions of the neighboring genes encoding members of the olfactory receptor family 2 (or2b11, or2w5, or2wc3) and the zinc finger protein ZNF496 is shown to scale. Nlrp3 is produced from 9 coding exons (shown as purple bars), of which the first and last also encode the 5’ and 3’ untranslated regions, respectively (shown as white bars). Nlrp3 is expressed as a protein of 1036 amino acids consisting of an N-terminal pyrin domain, a central NACHT domain and 12 C-terminal LRR motifs. The positions of the CINCA- FCAS- and MWS-associated point mutations in Nlrp3 are indicated.

Nlrp3 expression and activation

Nlrp3 expression is detected mainly in the cytosol of granulocytes, monocytes, dendritic cells, T and B cells, epithelial cells and osteoblasts (McCall et al., 2008, Kummer et al., 2007), suggesting an important role in the primary defense mechanisms of the body against microbial threats. Thus, most studies characterizing the role of Nlrp3 signaling in immunity have been conducted in cells of the immune system.

Given that many molecules without obvious homology can induce Nlrp3 activation, this process is widely believed to involve the generation/activation of a secondary messenger. However, the precise nature of this factor and the way it leads to Nlrp3 activation are still debated. Nevertheless, several mutually non-exclusive mechanisms have been suggested (Figure 2) including K+ efflux, the generation of reactive oxygen species and lysosomal destabilization (Lamkanfi and Dixit, 2009).

Figure 2.

Figure 2

Overview of Nlrp3 signalling pathways. Assembly and activation of caspase-1 within the cytosolic Nlrp3 inflammasome complex is induced by several stimuli including binding of ATP to the purinergic receptor P2X7, the ionophores nigericin and maitotoxin, β-amyloid and crystalline substances. Nlrp3 has been proposed to sense potassium efflux, the generation of ROS by the NADPH oxidase complex and lysosomal membrane disruption in response to these stimuli. Once activated in the Nlrp3 inflammasome, caspase-1 cleaves its substrates IL-1β, IL-18 and caspase-7 into the biologically active forms.

Biological functions

Nlrp3 was initially shown to assemble a large (700 kDa) multiprotein complex coined the “inflammasome” that was sufficient to trigger activation of the cystein protease caspase-1 under certain in vitro conditions (Martinon et al., 2004). Once activated, caspase-1 processes the precursor forms of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 to generate biologically active IL-1β and IL-18 (Kanneganti et al., 2007b, Lamkanfi and Dixit, 2009). The bipartite adaptor protein ASC plays a central role in the interaction between Nlrp3 and caspase-1 in the inflammasome (Figure 2). As a consequence, caspase-1 activation and the secretion of IL-1β and IL-18 are abolished in ASC-deficient macrophages (Mariathasan et al., 2004). ASC has a specific role in caspase-1 activation because secretion of the cytokines TNF-α and IL-6 is not affected by ASC deficiency.

Studies in mice with a gene-targeted deletion in cias1 demonstrated that Nlrp3-dependent caspase-1 activation is stimulus-dependent under physiological conditions (Kanneganti et al., 2006, Mariathasan et al., 2006, Sutterwala et al., 2006). The Nlrp3 inflammasome is responsible for caspase-1 activation in macrophages and dendritic cells infected with Staphylococcus aureus (Mariathasan et al., 2006), but bacterial pathogens such as Salmonella typhimurium, Legionella pneumophila, Pseudomonas aeruginosa, Shigella flexneri and Francisella tularensis activate caspase-1 independently of the Nlrp3 inflammasome (Lamkanfi and Dixit, 2009, Kanneganti et al., 2007b). Notably, the Nlrp3 inflammasome plays a critical role in the host response against influenza virus (Thomas et al., 2009, Ichinohe et al., 2009, Allen et al., 2009) and the fungal pathogen Candida albicans (Gross et al., 2009, Hise et al., 2009, Joly et al., 2009). The Nlrp3 inflammasome also drives the inflammatory response in skin keratinocytes exposed to various skin irritants such as ultraviolet B irradiation and chemicals inducing contact hypersensitivity (Sutterwala et al., 2006, Feldmeyer et al., 2007).

Activation of the Nlrp3 inflammasome in cultured macrophages is also achieved with millimolar concentrations of ATP provided the cells are pre-exposed to TLR ligands such as lipopolysaccharide (LPS), to bacterial or viral nucleic acids, or to fungal cell wall components (Kanneganti et al., 2006, Mariathasan et al., 2006, Sutterwala et al., 2006, Kanneganti et al., 2007a, Muruve et al., 2008, Lamkanfi et al., 2009). ATP triggers opening of the non-selective cation channel of the purinergic P2X7 receptor. The shellfish toxin maitotoxin and the bacterial ionophore nigericin can substitute for ATP in the activation of caspase-1 via Nlrp3 (Mariathasan et al., 2006).

Alzheimer’s disease-assoicated amyloid deposits and medically-relevant crystals such as monosodium urate (MSU), calcium pyrophosphate dihydrate (CPPD), crystalline asbestos and silica all induce Nlrp3-dependent activation of caspase-1 in LPS-primed macrophages (Kanneganti et al., 2007b, Lamkanfi and Dixit, 2009). Also aluminium adjuvants activate the Nlrp3 inflammasome in LPS-pretreated macrophages, suggesting that the Nlrp3 inflammasome may mediate antibody production with alum-containing vaccines (Lamkanfi and Dixit, 2009).

Clinical implications

Polymorphisms in regulatory elements that cause decreased Nlrp3 expression and IL-1β production were recently linked with increased susceptibility to Crohn’s disease in humans (Villani et al., 2009). In addition, gain-of-function mutations in and around the Nlrp3 NACHT domain (Figure 1) cause three auto-inflammatory conditions of which the primary symptoms are urticarial skin rashes and prolonged episodes of fever in the absence of any apparent infection. These hereditary periodic-fever syndromes are MWS, FACS and CINCA (Hoffman et al., 2001, Feldmann et al., 2002). Patients suffering from one of these disorders – which are collectively referred to as the Cryopyrin/Nlrp3-associated periodic syndromes (CAPS) - can further present with arthralgia, headaches, elevated spinal fluid pressure, cognitive deficits, sensorineural hearing loss and renal amyloidosis (Hoffman et al., 2001, Feldmann et al., 2002). The CAPS-associated mutations result in constitutively active forms of Nlrp3, which drive continuous caspase-1 activation (Dowds et al., 2004). As a consequence, mononuclear cells from CAPS patients spontaneously secrete IL-1β and IL-18, resulting in the clinical manifestations of these syndromes (Agostini et al., 2004). Analysis of mice expressing CAPS-associated Nlrp3 mutations confirmed the role of the inflammasome and showed that CAPS symptoms are partially dependent on IL-1β (Brydges et al., 2009, Meng et al., 2009).

Until recently, the therapeutic options for patients with CAPS disorders have been limited. Thanks to substantial advances in our understanding of the molecular mechanisms underlying these disorders, the efficacy of new therapeutic strategies targeting IL-1 production and signalling are underway. The safety and efficacy of the IL-1 receptor antagonist anakinra (Kineret, Amgen), the human IL-1β neutralizing monoclonal antibody canakinumab (Ilaris, Novartis) and rilonacept (Arcalyst, Regeneron), a fusion protein of the ligand-binding domains of human IL-1 receptor (IL-1R1) and IL-1 receptor accessory protein (IL-1RAcP) with the Fc portion of a human immunoglobulin G1 (IgG1), have already been demonstrated (Hawkins et al., 2003, Hoffman et al., 2004, Hoffman et al., 2008, Lachmann et al., 2009). Caspase-1 inhibitors represent a fourth mechanism to prevent overproduction of IL-1β in CAPS patients. VX-765 (Vertex Pharmaceuticals) is an orally active caspase-1 inhibitor that inhibited IL-1β secretion from LPS-stimulated peripheral blood mononuclear cells of FCAS patients (Stack et al., 2005). Thus, the Nlrp3 research field represents a prime example of how basic research on the mechanisms governing immunity pave the way for new therapies.

Acknowledgements

We apologize to colleagues whose work was not cited here owing to space limitations. This work was supported by National Institute of Health Grant AR056296, a Cancer Center Support Grant (CCSG 2 P30 CA 21765), Centers of Excellence for Influenza Research and Surveillance (CEIRS) project and the American Lebanese Syrian Associated Charities (ALSAC) to T-D.K. ML is supported by the Fonds voor Wetenschappelijk Onderzoek-Vlaanderen.

Abbreviations

ASC

apoptosis-associated speck-like protein containing a CARD

BIR

baculovirus IAP repeat

CAPS

Cryopyrin/Nlrp3-associated periodic syndromes

CARD

caspase recruitment domain

CLR

C-type lectin receptor

Ig

immunoglobulin

IL

interleukin

IL-1R1

IL-1 receptor 1

IL-1RAcP

IL-1 receptor accessory protein

LPS

lipopolysaccharide

LRR

leucine-rich repeat

NLR

NOD-like receptor

NOD

nucleotide-binding and oligomerization domain

PRR

pattern recognition receptor

TLR

Toll-like receptor

RLR

RIG-I-like receptor

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 2004;20:319–325. doi: 10.1016/s1074-7613(04)00046-9. [DOI] [PubMed] [Google Scholar]
  2. Allen IC, Scull MA, Moore CB, Holl EK, McElvania-TeKippe E, Taxman DJ, Guthrie EH, Pickles RJ, Ting JP. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 2009;30:556–565. doi: 10.1016/j.immuni.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Brydges SD, Mueller JL, McGeough MD, Pena CA, Misaghi A, Gandhi C, Putnam CD, Boyle DL, Firestein GS, Horner AA, Soroosh P, Watford WT, O'Shea JJ, Kastner DL, Hoffman HM. Inflammasome-mediated disease animal models reveal roles for innate but not adaptive immunity. Immunity. 2009;30:875–887. doi: 10.1016/j.immuni.2009.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Dowds TA, Masumoto J, Zhu L, Inohara N, Nunez G. Cryopyrin-induced interleukin 1beta secretion in monocytic cells: enhanced activity of disease-associated mutants and requirement for ASC. J Biol Chem. 2004;279:21924–21928. doi: 10.1074/jbc.M401178200. [DOI] [PubMed] [Google Scholar]
  5. Feldmann J, Prieur AM, Quartier P, Berquin P, Certain S, Cortis E, Teillac-Hamel D, Fischer A, de Saint Basile G. 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;71:198–203. doi: 10.1086/341357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Feldmeyer L, Keller M, Niklaus G, Hohl D, Werner S, Beer HD. The inflammasome mediates UVB-induced activation and secretion of interleukin-1beta by keratinocytes. Curr Biol. 2007;17:1140–1145. doi: 10.1016/j.cub.2007.05.074. [DOI] [PubMed] [Google Scholar]
  7. Gross O, Poeck H, Bscheider M, Dostert C, Hannesschlager N, Endres S, Hartmann G, Tardivel A, Schweighoffer E, Tybulewicz V, Mocsai A, Tschopp J, Ruland J. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature. 2009;459:433–436. doi: 10.1038/nature07965. [DOI] [PubMed] [Google Scholar]
  8. Hawkins PN, Lachmann HJ, McDermott MF. Interleukin-1-receptor antagonist in the Muckle-Wells syndrome. N Engl J Med. 2003;348:2583–2584. doi: 10.1056/NEJM200306193482523. [DOI] [PubMed] [Google Scholar]
  9. Hise AG, Tomalka J, Ganesan S, Patel K, Hall BA, Brown GD, Fitzgerald KA. An essential role for the NLRP3 inflammasome in host defense against the human fungal pathogen Candida albicans. Cell Host Microbe. 2009;5:487–497. doi: 10.1016/j.chom.2009.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. 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;29:301–305. doi: 10.1038/ng756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Hoffman HM, Rosengren S, Boyle DL, Cho JY, Nayar J, Mueller JL, Anderson JP, Wanderer AA, Firestein GS. Prevention of cold-associated acute inflammation in familial cold autoinflammatory syndrome by interleukin-1 receptor antagonist. Lancet. 2004;364:1779–1785. doi: 10.1016/S0140-6736(04)17401-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Hoffman HM, Throne ML, Amar NJ, Sebai M, Kivitz AJ, Kavanaugh A, Weinstein SP, Belomestnov P, Yancopoulos GD, Stahl N, Mellis SJ. Efficacy and safety of rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated periodic syndromes: results from two sequential placebo-controlled studies. Arthritis Rheum. 2008;58:2443–2452. doi: 10.1002/art.23687. [DOI] [PubMed] [Google Scholar]
  13. Ichinohe T, Lee HK, Ogura Y, Flavell R, Iwasaki A. Inflammasome recognition of influenza virus is essential for adaptive immune responses. J Exp Med. 2009;206:79–87. doi: 10.1084/jem.20081667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Joly S, Ma N, Sadler JJ, Soll DR, Cassel SL, Sutterwala FS. Cutting edge: Candida albicans hyphae formation triggers activation of the Nlrp3 inflammasome. J Immunol. 2009;183:3578–3581. doi: 10.4049/jimmunol.0901323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, Taraporewala ZF, Miller D, Patton JT, Inohara N, Nunez G. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem. 2006;281:36560–36568. doi: 10.1074/jbc.M607594200. [DOI] [PubMed] [Google Scholar]
  16. Kanneganti TD, Lamkanfi M, Kim YG, Chen G, Park JH, Franchi L, Vandenabeele P, Nunez G. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity. 2007a;26:433–443. doi: 10.1016/j.immuni.2007.03.008. [DOI] [PubMed] [Google Scholar]
  17. Kanneganti TD, Lamkanfi M, Nunez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007b;27:549–559. doi: 10.1016/j.immuni.2007.10.002. [DOI] [PubMed] [Google Scholar]
  18. Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006;13:816–825. doi: 10.1038/sj.cdd.4401850. [DOI] [PubMed] [Google Scholar]
  19. Kumar H, Kawai T, Akira S. Pathogen recognition in the innate immune response. Biochem J. 2009;420:1–16. doi: 10.1042/BJ20090272. [DOI] [PubMed] [Google Scholar]
  20. Kummer JA, Broekhuizen R, Everett H, Agostini L, Kuijk L, Martinon F, van Bruggen R, Tschopp J. Inflammasome components NALP 1 and 3 show distinct but separate expression profiles in human tissues suggesting a site-specific role in the inflammatory response. J Histochem Cytochem. 2007;55:443–452. doi: 10.1369/jhc.6A7101.2006. [DOI] [PubMed] [Google Scholar]
  21. Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB, Leslie KS, Hachulla E, Quartier P, Gitton X, Widmer A, Patel N, Hawkins PN. Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med. 2009;360:2416–2425. doi: 10.1056/NEJMoa0810787. [DOI] [PubMed] [Google Scholar]
  22. Lamkanfi M, Dixit VM. Inflammasomes: guardians of cytosolic sanctity. Immunol Rev. 2009;227:95–105. doi: 10.1111/j.1600-065X.2008.00730.x. [DOI] [PubMed] [Google Scholar]
  23. Lamkanfi M, Malireddi RK, Kanneganti TD. Fungal zymosan and mannan activate the cryopyrin inflammasome. J Biol Chem. 2009;284:20574–20581. doi: 10.1074/jbc.M109.023689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Mariathasan S, Newton K, Monack DM, Vucic D, French DM, Lee WP, Roose-Girma M, Erickson S, Dixit VM. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004;430:213–218. doi: 10.1038/nature02664. [DOI] [PubMed] [Google Scholar]
  25. Mariathasan S, Weiss DS, Newton K, McBride J, O'Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–232. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
  26. Martinon F, Agostini L, Meylan E, Tschopp J. Identification of bacterial muramyl dipeptide as activator of the NALP3/cryopyrin inflammasome. Curr Biol. 2004;14:1929–1934. doi: 10.1016/j.cub.2004.10.027. [DOI] [PubMed] [Google Scholar]
  27. McCall SH, Sahraei M, Young AB, Worley CS, Duncan JA, Ting JP, Marriott I. Osteoblasts express NLRP3, a nucleotide-binding domain and leucine-rich repeat region containing receptor implicated in bacterially induced cell death. J Bone Miner Res. 2008;23:30–40. doi: 10.1359/JBMR.071002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. 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;30:860–874. doi: 10.1016/j.immuni.2009.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, Parks RJ, Tschopp J. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452:103–107. doi: 10.1038/nature06664. [DOI] [PubMed] [Google Scholar]
  30. Stack JH, Beaumont K, Larsen PD, Straley KS, Henkel GW, Randle JC, Hoffman HM. IL-converting enzyme/caspase-1 inhibitor VX-765 blocks the hypersensitive response to an inflammatory stimulus in monocytes from familial cold autoinflammatory syndrome patients. J Immunol. 2005;175:2630–2634. doi: 10.4049/jimmunol.175.4.2630. [DOI] [PubMed] [Google Scholar]
  31. Sutterwala FS, Ogura Y, Szczepanik M, Lara-Tejero M, Lichtenberger GS, Grant EP, Bertin J, Coyle AJ, Galan JE, Askenase PW, Flavell RA. Critical role for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1. Immunity. 2006;24:317–327. doi: 10.1016/j.immuni.2006.02.004. [DOI] [PubMed] [Google Scholar]
  32. Thomas PG, Dash P, Aldridge JR, Jr, Ellebedy AH, Reynolds C, Funk AJ, Martin WJ, Lamkanfi M, Webby RJ, Boyd KL, Doherty PC, Kanneganti TD. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 2009;30:566–575. doi: 10.1016/j.immuni.2009.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Villani AC, Lemire M, Fortin G, Louis E, Silverberg MS, Collette C, Baba N, Libioulle C, Belaiche J, Bitton A, Gaudet D, Cohen A, Langelier D, Fortin PR, Wither JE, Sarfati M, Rutgeerts P, Rioux JD, Vermeire S, Hudson TJ, Franchimont D. Common variants in the NLRP3 region contribute to Crohn's disease susceptibility. Nat Genet. 2009;41:71–76. doi: 10.1038/ng285. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES