15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Structural mechanism for NEK7-licensed NLRP3 inflammasome activation

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          The NLRP3 inflammasome can be activated by diverse stimuli, including nigericin, uric acid crystals, amyloid-β fibrils, and extracellular ATP. The mitotic kinase NEK7 licenses NLRP3 inflammasome assembly and activation in the interphase. Here we report a 3.8-Å cryo-electron microscopy structure of inactive human NLRP3 in complex with NEK7. The earring-shaped NLRP3 consists of curved leucine-rich repeat (LRR) and globular NACHT domains, whereas the C-terminal lobe of NEK7 nestles against both NLRP3 domains. Structural recognition between NLRP3 and NEK7 is confirmed by mutagenesis both in vitro and in cells. Modelling of an active NLRP3-NEK7 conformation based on the NLRC4 inflammasome predicts an additional contact between an NLRP3-bound NEK7 and a neighbouring NLRP3. Mutations on this interface abolish the ability of NEK7 or NLRP3 to rescue NLRP3 activation in NEK7 KO or NLRP3 KO cells. Taken together, these data suggest that NEK7 bridges adjacent NLRP3 subunits with bipartite interactions to mediate NLRP3 inflammasome activation.

          Related collections

          Most cited references27

          • Record: found
          • Abstract: not found
          • Article: not found

          Targeting the NLRP3 inflammasome in inflammatory diseases

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component

            The NLRP3 inflammasome responds to microbes and danger signals by processing and activating proinflammatory cytokines including IL-1β and IL-18. We show that NLRP3 inflammasome activation is restricted to interphase of the cell cycle by NEK7, a serine/threonine kinase previously implicated in mitosis. NLRP3 inflammasome activation requires NEK7, which binds to the NLRP3 leucine-rich repeat domain in a kinase-independent manner downstream from the induction of mitochondrial ROS. This interaction is necessary for NLRP3-ASC complex formation, ASC oligomerization, and caspase-1 activation. NEK7 promotes the NLRP3-dependent cellular inflammatory response to intraperitoneal monosodium urate challenge, and the development of experimental autoimmune encephalitis in mice. Our findings suggest NEK7 serves as a cellular switch that enforces mutual exclusivity between the inflammasome response and cell division.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity.

              NLRP3 is an important pattern recognition receptor involved in mediating inflammasome activation in response to viral and bacterial infections as well as various proinflammatory stimuli associated with tissue damage or malfunction. Upon activation, NLRP3 assembles a multimeric inflammasome complex comprising the adaptor ASC and the effector pro-caspase-1 to mediate the activation of caspase-1. Although NLRP3 expression is induced by the NF-κB pathway, the posttranscriptional molecular mechanism controlling the activation of NLRP3 remains elusive. Using both pharmacological and molecular approaches, we show that the activation of NLRP3 inflammasome is regulated by a deubiquitination mechanism. We further identify the deubiquitinating enzyme, BRCC3, as a critical regulator of NLRP3 activity by promoting its deubiquitination and characterizing NLRP3 as a substrate for the cytosolic BRCC3-containing BRISC complex. Our results elucidate a regulatory mechanism involving BRCC3-dependent NLRP3 regulation and highlight NLRP3 ubiquitination as a potential therapeutic target for inflammatory diseases. Copyright © 2013 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Journal
                0410462
                6011
                Nature
                Nature
                Nature
                0028-0836
                1476-4687
                5 June 2019
                12 June 2019
                June 2019
                12 December 2019
                : 570
                : 7761
                : 338-343
                Affiliations
                [1 ]Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
                [2 ]Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115
                [3 ]State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Center for Quantitative Biology, Peking University, Beijing, 100871, China
                [4 ]Intel Parallel Computing Center for Structural Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
                [5 ]Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
                [6 ]Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109
                [7 ]These authors contributed equally.
                Author notes

                Author Contributions H.W., L.W. and H.S. conceived the study. L.W. designed constructs. L.W., Q.Q. and H.S. purified the complexes, and H.S., L.W. and Q.Q. made cryo-grids for data collection. H.S., L.W., W.L.W., Q.Q., Z.W. and Y.M. collected data. H.S., W.L.W. and Q.Q. analysed cryo-EM data and H.W. and Y.M. supervised data processing. H.S. performed initial model building and refinement, and W.L.W. and Y.M. performed additional model fitting and refinement. H.S. designed mutants for in vitro and cell based assays. V.G.M. performed all cell-based assays. L.A. and A.V.H. performed NEK7 mutant assays. G.N. provided reagents for cell based assays and valuable discussions. H.W., H.S. and Y.M. wrote the manuscript, and all authors provided comments on the manuscript.

                Author Information Reprints and permissions information is available at www.nature.com/reprints. The authors declare competing financial interests: L.W. and A.V.H are employees and H.W. is co-founder of SMOC Therapeutics; other authors declare no competing interests. Readers are welcome to comment on the online version of the paper. Correspondence and requests for materials should be addressed to H.W. ( wu@ 123456crystal.harvard.edu ) or Y.M. ( ymao@ 123456pku.edu.cn ).

                Article
                NIHMS1529572
                10.1038/s41586-019-1295-z
                6774351
                31189953
                a98f40c2-7166-4fec-a2d0-5ba601871dcd

                Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms

                History
                Categories
                Article

                Uncategorized
                Uncategorized

                Comments

                Comment on this article