Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

NLRP3 inflammasome inhibition is disrupted in a group of auto-inflammatory disease CAPS mutations

Abstract

Inflammasomes are positioned to rapidly escalate the intensity of inflammation by activating interleukin (IL)-1β, IL-18 and cell death by pyroptosis. However, negative regulation of inflammasomes remains poorly understood, as is the signaling cascade that dampens inflammasome activity. We found that rapid NLRP3 inflammasome activation was directly inhibited by protein kinase A (PKA), which was induced by prostaglandin E2 (PGE2) signaling via the PGE2 receptor E-prostanoid 4 (EP4). PKA directly phosphorylated the cytoplasmic receptor NLRP3 and attenuated its ATPase function. We found that Ser295 in human NLRP3 was critical for rapid inhibition and PKA phosphorylation. Mutations in NLRP3-encoding residues adjacent to Ser295 have been linked to the inflammatory disease CAPS (cryopyrin-associated periodic syndromes). NLRP3-S295A phenocopied the human CAPS mutants. These data suggest that negative regulation at Ser295 is critical for restraining the NLRP3 inflammasome and identify a molecular basis for CAPS-associated NLRP3 mutations.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: PGE2 suppresses activation of the NLRP3 inflammasome.
Figure 2: PGE2 selectively inhibits NLRP3 inflammasome activation.
Figure 3: PGE2 receptor EP4 and EP2 inhibit the NLRP3 inflammasome.
Figure 4: cAMP-PKA signaling mediates NLRP3 inflammasome inhibition.
Figure 5: Rapid PGE2 NLRP3 inflammasome inhibition by PKA.
Figure 6: cAMP-PKA induces NLRP3 phosphorylation.
Figure 7: NLRP3 Ser295 mediates rapid inhibition by cAMP.
Figure 8: Other CAPS mutations adjacent to Ser295 are unresponsive to rapid cAMP inhibition.

Similar content being viewed by others

References

  1. Latz, E., Xiao, T.S. & Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 13, 397–411 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Masters, S.L., Simon, A., Aksentijevich, I. & Kastner, D.L. Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease (*). Annu. Rev. Immunol. 27, 621–668 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Romberg, N. et al. Mutation of NLRC4 causes a syndrome of enterocolitis and autoinflammation. Nat. Genet. 46, 1135–1139 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Canna, S.W. et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat. Genet. 46, 1140–1146 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Strowig, T., Henao-Mejia, J., Elinav, E. & Flavell, R. Inflammasomes in health and disease. Nature 481, 278–286 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Taskén, K. & Aandahl, E.M. Localized effects of cAMP mediated by distinct routes of protein kinase A. Physiol. Rev. 84, 137–167 (2004).

    Article  PubMed  Google Scholar 

  7. Yan, Y. et al. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell 160, 62–73 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Lee, G.S. et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 492, 123–127 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sokolowska, M. et al. Prostaglandin E2 inhibits NLRP3 inflammasome activation through EP4 receptor and intracellular cyclic AMP in human macrophages. J. Immunol. 194, 5472–5487 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Serhan, C.N. & Savill, J. Resolution of inflammation: the beginning programs the end. Nat. Immunol. 6, 1191–1197 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Hirata, T. & Narumiya, S. Prostanoid receptors. Chem. Rev. 111, 6209–6230 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Kunkel, S.L., Chensue, S.W. & Phan, S.H. Prostaglandins as endogenous mediators of interleukin 1 production. J. Immunol. 136, 186–192 (1986).

    CAS  PubMed  Google Scholar 

  13. Takaishi, O. et al. Inhibition by 16,16-dimethyl prostaglandin E2 of tumor necrosis factor-alpha and interleukin-1beta production and messenger RNA expression in human monocytes stimulated by Helicobacter pylori. Dig. Dis. Sci. 44, 2405–2411 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Kunkel, S.L. et al. Prostaglandin E2 regulates macrophage-derived tumor necrosis factor gene expression. J. Biol. Chem. 263, 5380–5384 (1988).

    Article  CAS  PubMed  Google Scholar 

  15. Marotta, P., Sautebin, L. & Di Rosa, M. Modulation of the induction of nitric oxide synthase by eicosanoids in the murine macrophage cell line J774. Br. J. Pharmacol. 107, 640–641 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Takayama, K. et al. Prostaglandin E2 suppresses chemokine production in human macrophages through the EP4 receptor. J. Biol. Chem. 277, 44147–44154 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Fernandes-Alnemri, T. et al. The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation. Cell Death Differ. 14, 1590–1604 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Kayagaki, N. et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science 341, 1246–1249 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Agostini, L. et al. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity 20, 319–325 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Gloerich, M. & Bos, J.L. Epac: defining a new mechanism for cAMP action. Annu. Rev. Pharmacol. Toxicol. 50, 355–375 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Rehmann, H., Schwede, F., Døskeland, S.O., Wittinghofer, A. & Bos, J.L. Ligand-mediated activation of the cAMP-responsive guanine nucleotide exchange factor Epac. J. Biol. Chem. 278, 38548–38556 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Dalton, G.D. & Dewey, W.L. Protein kinase inhibitor peptide (PKI): a family of endogenous neuropeptides that modulate neuronal cAMP-dependent protein kinase function. Neuropeptides 40, 23–34 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Aganna, E. et al. Association of mutations in the NALP3/CIAS1/PYPAF1 gene with a broad phenotype including recurrent fever, cold sensitivity, sensorineural deafness, and AA amyloidosis. Arthritis Rheum. 46, 2445–2452 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Dodé, C. et al. New mutations of CIAS1 that are responsible for Muckle-Wells syndrome and familial cold urticaria: a novel mutation underlies both syndromes. Am. J. Hum. Genet. 70, 1498–1506 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Aróstegui, J.I. et al. Clinical and genetic heterogeneity among Spanish patients with recurrent autoinflammatory syndromes associated with the CIAS1/PYPAF1/NALP3 gene. Arthritis Rheum. 50, 4045–4050 (2004).

    Article  PubMed  CAS  Google Scholar 

  26. Feldmann, J. 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. 71, 198–203 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Neven, B. et al. Molecular basis of the spectral expression of CIAS1 mutations associated with phagocytic cell-mediated autoinflammatory disorders CINCA/NOMID, MWS, and FCU. Blood 103, 2809–2815 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Leinonen, M., Hallén, B. & Olivecrona, H. The IL-1 receptor antagonist anakinra stabilizes the NLRP3 mutation-specific risk for hearing loss in patients with severe cryopyrin-associated periodic syndromes (CAPS). Pediatr. Rheumatol. Online J. 12 (Suppl. 1), 76 (2014).

    Article  Google Scholar 

  29. Kanariou, M. et al. Twenty year follow up of a patient with a new de-novo NLRP3 mutation (S595G) and CINCA syndrome. Klin. Padiatr. 221, 379–381 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Taylor, S.S. et al. Signaling through cAMP and cAMP-dependent protein kinase: diverse strategies for drug design. Biochim. Biophys. Acta 1784, 16–26 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Duncan, J.A. et al. Cryopyrin/NALP3 binds ATP/dATP, is an ATPase, and requires ATP binding to mediate inflammatory signaling. Proc. Natl. Acad. Sci. USA 104, 8041–8046 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kayagaki, N. et al. Non-canonical inflammasome activation targets caspase-11. Nature 479, 117–121 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Ng, J. et al. Clostridium difficile toxin-induced inflammation and intestinal injury are mediated by the inflammasome. Gastroenterology 139, 542–552, 552.e1–552.e3 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Masters, S.L. et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nat. Immunol. 11, 897–904 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hentze, H., Lin, X.Y., Choi, M.S. & Porter, A.G. Critical role for cathepsin B in mediating caspase-1-dependent interleukin-18 maturation and caspase-1-independent necrosis triggered by the microbial toxin nigericin. Cell Death Differ. 10, 956–968 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Misawa, T. et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat. Immunol. 14, 454–460 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Hornung, V. et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458, 514–518 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ippagunta, S.K. et al. The inflammasome adaptor ASC regulates the function of adaptive immune cells by controlling Dock2-mediated Rac activation and actin polymerization. Nat. Immunol. 12, 1010–1016 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Susuki-Miyata, S. et al. Cross-talk between PKA-Cβ and p65 mediates synergistic induction of PDE4B by roflumilast and NTHi. Proc. Natl. Acad. Sci. USA 112, E1800–E1809 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Myeku, N. et al. Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat. Med. 22, 46–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Schmitz, T. et al. Prostaglandin E2 receptor expression in fetal baboon lung at 0.7 gestation after betamethasone exposure. Pediatr. Res. 61, 421–426 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Dey, I., Giembycz, M.A. & Chadee, K. Prostaglandin E(2) couples through EP(4) prostanoid receptors to induce IL-8 production in human colonic epithelial cell lines. Br. J. Pharmacol. 156, 475–485 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lejeune, M., Leung, P., Beck, P.L. & Chadee, K. Role of EP4 receptor and prostaglandin transporter in prostaglandin E2-induced alteration in colonic epithelial barrier integrity. Am. J. Physiol. Gastrointest. Liver Physiol. 299, G1097–G1105 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Medina, D.L. et al. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat. Cell Biol. 17, 288–299 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. He, Y. et al. 3,4-methylenedioxy-β-nitrostyrene inhibits NLRP3 inflammasome activation by blocking assembly of the inflammasome. J. Biol. Chem. 289, 1142–1150 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Shi (University of Calgary) for MSU and silica. Supported by the Canadian Institute of Health Research (MOP-340405 to K.C.; Tier 1 Canada Research Chair in Gastrointestinal Inflammation to K.C.; Team Grant in Health Challenges in Chronic Inflammation to J.A.M.), the Natural Sciences and Engineering Research Council of Canada (RGPIN-04023 to K.C.; and Canada Graduate Scholarship to L.M.) and Alberta Innovates Health Solutions (J.A.M. and L.M.).

Author information

Authors and Affiliations

Authors

Contributions

L.M., J.A.M. and K.C. conceived of and designed the experiments and wrote the paper; L.M., F.M. and J.A.M. performed the experiments and analyzed the data; and L.M., F.M., J.A.M. and K.C. contributed reagents, materials and analysis tools.

Corresponding author

Correspondence to Kris Chadee.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PGE2 does not induce degradation of NLRP3 inflammasome components but rapidly inhibits NLRP3 activation in macrophages.

(a-d) LPS-primed BMDM. (a) Lysates (LYS) from PGE2 (500 nM) treatment for 30 min. (b) The indicated concentration of PGE2 was added for 5 min followed by stimulation with (left) nigericin (5 μM for 30 min) or (right) ATP (5 mM for 45 min). (c) PGE2 (500 nM) was added 30 min after stimulation with nigericin and analyzed 50 min post initial nigericin stimulation. (d) 16,16-dimethyl PGE2 (16-DM, 500 nM) or 15-keto PGE2 (15-keto, 500 nM) were added 5 min prior to nigericin or ATP stimulation. (e-g) PMA-differentiated THP-1 cells were treated with (e,f) the indicated concentration of PGE2 for 5 min prior to stimulation with (e) nigericin (5 μM) and LPS (50 ng/ml) for 30 min or (f) ATP (5 mM) and LPS (50 ng/ml) for 45 min. (g) 16,16-dimethyl PGE2 (16-DM, 250 nM), or 15-keto PGE2 (15-keto, 250 nM) were added 5 min prior to nigericin. (b-g) Total IL-1β release in cell supernatants analyzed by ELISA. *P < 0.005 (unpaired two-tailed t test). (a-g) Data are from one experiment representative of three separate experiments with similar results, (a-d) BMDM were derived from 2 mice per experiment mixed together, (b-g) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 2 Particle-induced NLRP3 inflammasome activation is partially suppressed by PGE2, and cyclooxygenase inhibition blocks NLRP3 activation.

(a-c) BMDM were primed with LPS. (a,b) The indicated concentration of PGE2 was added 5 min before stimulation with (left) silica (250 μg/ml for 5 h) or (right) MSU (150 μg/ml for 4 h). (c) Indomethacin (50 μM) was added 30 min before stimulation with nigericin (5 μM for 30 min). (a,c) Total IL-1β release in cell supernatants analyzed by ELISA. (b,c) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). *P < 0.005 (unpaired two-tailed t test). (a-c) Data are from one experiment representative of three separate experiments with similar results, (a-c) BMDM were derived from 2 mice per experiment mixed together, (a,c) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 3 PGE2 receptors EP1 and EP3 do not inhibit, but EP4 does inhibit, the NLRP3 inflammasome.

(a-i) BMDMs primed for 3.5 h with LPS. (a,b) The indicated concentration of EP1 agonist ONO-DI-004 or (c,d) EP3 agonist ONO-AE-248 was added 5 min prior to (a,c) nigericin (5 μM for 30 min) or (b,d) ATP (5 mM for 45 min). (e-g) The indicated concentration of EP4 agonists (e left,f) CAY10598 and (e right,g) ONO-AE1-32 were added 5 min before (e) nigericin (5 μM for 30 min) or (f,g) ATP (5 mM for 45 min). (h,i) An EP4 antagonist ONO-AE3-208 (2 μM) was added for 5 min prior to addition of ONO-AE1-329 (500 nM) for 5 min and then treated with (h) nigericin (5 μM for 30 min) or (i) ATP (5 mM for 45 min). (a-i) Total IL-1β release in cell supernatants analyzed by ELISA. (a-d,f,g,i) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). *P < 0.005 (unpaired two-tailed t test). (a-i) Data are from one experiment representative of three separate experiments with similar results, (a-i) BMDM were derived from 2 mice per experiment mixed together, (a-i) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 4 PGE2 receptors EP2 and EP4 inhibit the NLRP3 inflammasome, but EP4 mediates PGE2-induced inhibition.

(a-h) BMDMs primed for 3.5 h with LPS. (a,b) The indicated concentration of EP2 agonist ONO-AE1-259-01 was added 5 min before treatment with (a) nigericin (5 μM for 30 min) or (b) ATP (5 mM 45 min). (c) The EP2 antagonist AH6809 (2 μM) was added 5 min prior to addition of ONO-AE1-259-01 followed by ATP (5 mM 45 min). (d) CAY10598 (500 nM) was added 30 min after stimulation with nigericin and analyzed 50 min post initial nigericin stimulation. (e,f) EP2 agonist ONO-AE1-259-01 or (g,h) EP4 antagonist ONO-AE3-208 was added 5 min before treatment with PGE2 (500 nM) followed by (e,g) nigericin (5 μM for 30 min) or (f,h) ATP (5 mM 45 min). (a-h). Total IL-1β release in cell supernatants analyzed by ELISA. (b,c,f,h) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). *P < 0.005 (unpaired two-tailed t test). (a-h) Data are from one experiment representative of three separate experiments with similar results, (a-h) BMDM were derived from 2 mice per experiment mixed together, (a-h) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 5 cAMP induces rapid NLRP3 inflammasome inhibition.

(a-c,e,f) BMDMs primed for 3.5 h with LPS. (a,b) The indicated concentration of forskolin was added 5 min prior to (a) nigericin (5 μM for 30 min) or (b) ATP (5 mM for 45 min). (c) db-cAMP (1 μM) was added 5 min prior to nigericin (5 μM for 30 min) or ATP (5 mM for 45 min). (d) PMA-differentiated THP-1 cells were treated for 5 min with indicated concentration of forskolin or db-cAMP prior to addition of nigericin (5 μM) and LPS (50 ng/ml) for 30 min. (e) Forskolin (50 μM) was added 30 min after stimulation with nigericin and analyzed 50 min post initial nigericin stimulation. (f) Forskolin (50 μM) or db-cAMP (1 μM) was added for 5 min prior to nigericin (5 μM for 30 min). (a-e) Total IL-1β release in cell supernatants analyzed by ELISA. (b-d,f) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). *P < 0.005 (unpaired two-tailed t test). (a-f) Data are from one experiment representative of three separate experiments with similar results, (a-c,e,f) BMDM were derived from 2 mice per experiment mixed together, (a-e) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 6 PKA inhibits the NLRP3 inflammasome in response to extracellular agonists but not particulate agonists.

(a-h) BMDMs primed for 3.5 h with LPS. (a,c) The indicated concentration of 6-Bnz-cAMP or (b) 8-CPT-2-O-Me-cAMP was added 5 min before nigericin (5 μM for 30 min). (d) 6-Bnz-cAMP (50 μM) or 8-CPT-2-O-Me-cAMP (50 μM) was added 5 min prior to treatment with MSU (125 μg/ml for 4h) or silica (250 μg/ml for 5h). (e) Rp-cAMP was added 5 min before or (f) PKI was added 30 min before (e,f) 5 min treatment with db-cAMP (1 μM) or forskolin (50 μM) and followed by nigericin (5 μM for 30 min). (g) Rp-cAMP was added 5 min before or (h) PKI was added 30 min before (g,h) 5 min treatment with PGE2 (500 nM) followed by nigericin (5 μM for 30 min). (a,b,d-h) Total IL-1β release in cell supernatants analyzed by ELISA. (c-e) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). *P < 0.005 (unpaired two-tailed t test). (a-h) Data are from one experiment representative of three separate experiments with similar results, (a-h) BMDM were derived from 2 mice per experiment mixed together, (a,b,d-h) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 7 PGE2-induced suppression of nigericin-triggered NLRP3 inflammasomes is mediated by PKA, but particulate NLRP3 agonists are not.

(a-f) BMDMs primed for 3.5 h with LPS. (a) Rp-cAMP was added 5 min before or (b) PKI was added 30 min before treatment with EP2 agonist ONO-AE1-259-01 (50 nM) for 5 min followed by nigericin (5 μM for 30 min). (c) Rp-cAMP was added 5 min before or (d) PKI was added 30 min before treatment with EP4 agonist ONO-AE1-329 (500 nM) for 5 min followed by nigericin (5 μM for 30 min). (e) PKI was added 30 min before or (f) Rp-cAMP was added 5 min before (e,f) PGE2 (500 nM) treatment for 5 min followed by addition of MSU (125 μg/ml for 4h) or silica (250 μg/ml for 5h). (a,c,e,f) Immunoblot analysis of culture supernatants (SN) and lysates (LYS). (a-f) Total IL-1β release in cell supernatants analyzed by ELISA. *P < 0.005 (unpaired two-tailed t test). (a-f) Data are from one experiment representative of three separate experiments with similar results, (a-f) BMDM were derived from 2 mice per experiment mixed together, (a-f) three replicates, each replicate is one well + s.e.m.

Supplementary Figure 8 PKA C-α and NLRP3 interact.

(a) Flow of NLRP3 ATPase assay used in Figure 6e and Figure 7e. (b,c) Immunoprecipitation of indicated protein of BMDMs primed for 3.5 h with LPS and treated for 5 min with forskolin (50 μM) in the presence or absence of PKI (1 μM). (b,c) Data are from one experiment representative of three separate experiments with similar results, (b,c) BMDM were derived from 2 mice per experiment mixed together.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 2223 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mortimer, L., Moreau, F., MacDonald, J. et al. NLRP3 inflammasome inhibition is disrupted in a group of auto-inflammatory disease CAPS mutations. Nat Immunol 17, 1176–1186 (2016). https://doi.org/10.1038/ni.3538

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3538

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing