8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      cAMP metabolism controls caspase-11 inflammasome activation and pyroptosis in sepsis

      research-article

      Read this article at

      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.

          Abstract

          Targeting immunometabolism is a strategy to prevent infection-mediated septic death.

          Abstract

          The ability of cytosolic lipopolysaccharide (LPS) to activate caspase-11–dependent nonclassical inflammasome is intricately controlled to avoid excessive inflammatory responses. However, very little is known about the regulatory role of various metabolic pathways in the control of caspase-11 activation. Here, we demonstrate that l-adrenaline can act on receptor ADRA2B to inhibit the activation of the caspase-11 inflammasome by cytosolic LPS or Escherichia coli infection in macrophages. l-adrenaline–induced cAMP production via the enzyme ADCY4 promotes protein kinase A (PKA) activation, which then blocks the caspase-11–mediated proteolytic maturation of interleukin-1β, gasdermin D (GSDMD) cleavage, and consequent DAMP release. Inhibition of PDE8A-mediated cAMP hydrolysis limits caspase-11 inflammasome activation and pyroptosis in macrophages. Consequently, pharmacological modulation of the ADRA2B-ADCY4-PDE8A-PKA axis, knockout of caspase-11 ( Casp11 −/− ), or Gsdmd inactivation ( Gsdmd I105N/I105N ) similarly protects against LPS-induced lethality in poly(I:C)-primed mice. Our results provide previously unidentified mechanistic insight into immune regulation by cAMP and represent a proof of concept that immunometabolism constitutes a potential therapeutic target in sepsis.

          Related collections

          Most cited references42

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

          Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death.

          Inflammatory caspases (caspase-1, -4, -5 and -11) are critical for innate defences. Caspase-1 is activated by ligands of various canonical inflammasomes, and caspase-4, -5 and -11 directly recognize bacterial lipopolysaccharide, both of which trigger pyroptosis. Despite the crucial role in immunity and endotoxic shock, the mechanism for pyroptosis induction by inflammatory caspases is unknown. Here we identify gasdermin D (Gsdmd) by genome-wide clustered regularly interspaced palindromic repeat (CRISPR)-Cas9 nuclease screens of caspase-11- and caspase-1-mediated pyroptosis in mouse bone marrow macrophages. GSDMD-deficient cells resisted the induction of pyroptosis by cytosolic lipopolysaccharide and known canonical inflammasome ligands. Interleukin-1β release was also diminished in Gsdmd(-/-) cells, despite intact processing by caspase-1. Caspase-1 and caspase-4/5/11 specifically cleaved the linker between the amino-terminal gasdermin-N and carboxy-terminal gasdermin-C domains in GSDMD, which was required and sufficient for pyroptosis. The cleavage released the intramolecular inhibition on the gasdermin-N domain that showed intrinsic pyroptosis-inducing activity. Other gasdermin family members were not cleaved by inflammatory caspases but shared the autoinhibition; gain-of-function mutations in Gsdma3 that cause alopecia and skin defects disrupted the autoinhibition, allowing its gasdermin-N domain to trigger pyroptosis. These findings offer insight into inflammasome-mediated immunity/diseases and also change our understanding of pyroptosis and programmed necrosis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling.

            Intracellular lipopolysaccharide from Gram-negative bacteria including Escherichia coli, Salmonella typhimurium, Shigella flexneri, and Burkholderia thailandensis activates mouse caspase-11, causing pyroptotic cell death, interleukin-1β processing, and lethal septic shock. How caspase-11 executes these downstream signalling events is largely unknown. Here we show that gasdermin D is essential for caspase-11-dependent pyroptosis and interleukin-1β maturation. A forward genetic screen with ethyl-N-nitrosourea-mutagenized mice links Gsdmd to the intracellular lipopolysaccharide response. Macrophages from Gsdmd(-/-) mice generated by gene targeting also exhibit defective pyroptosis and interleukin-1β secretion induced by cytoplasmic lipopolysaccharide or Gram-negative bacteria. In addition, Gsdmd(-/-) mice are protected from a lethal dose of lipopolysaccharide. Mechanistically, caspase-11 cleaves gasdermin D, and the resulting amino-terminal fragment promotes both pyroptosis and NLRP3-dependent activation of caspase-1 in a cell-intrinsic manner. Our data identify gasdermin D as a critical target of caspase-11 and a key mediator of the host response against Gram-negative bacteria.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores.

              Inflammatory caspases (caspases 1, 4, 5 and 11) are activated in response to microbial infection and danger signals. When activated, they cleave mouse and human gasdermin D (GSDMD) after Asp276 and Asp275, respectively, to generate an N-terminal cleavage product (GSDMD-NT) that triggers inflammatory death (pyroptosis) and release of inflammatory cytokines such as interleukin-1β. Cleavage removes the C-terminal fragment (GSDMD-CT), which is thought to fold back on GSDMD-NT to inhibit its activation. However, how GSDMD-NT causes cell death is unknown. Here we show that GSDMD-NT oligomerizes in membranes to form pores that are visible by electron microscopy. GSDMD-NT binds to phosphatidylinositol phosphates and phosphatidylserine (restricted to the cell membrane inner leaflet) and cardiolipin (present in the inner and outer leaflets of bacterial membranes). Mutation of four evolutionarily conserved basic residues blocks GSDMD-NT oligomerization, membrane binding, pore formation and pyroptosis. Because of its lipid-binding preferences, GSDMD-NT kills from within the cell, but does not harm neighbouring mammalian cells when it is released during pyroptosis. GSDMD-NT also kills cell-free bacteria in vitro and may have a direct bactericidal effect within the cytosol of host cells, but the importance of direct bacterial killing in controlling in vivo infection remains to be determined.
                Bookmark

                Author and article information

                Journal
                Sci Adv
                Sci Adv
                SciAdv
                advances
                Science Advances
                American Association for the Advancement of Science
                2375-2548
                May 2019
                22 May 2019
                : 5
                : 5
                : eaav5562
                Affiliations
                [1 ]The Third Affiliated Hospital, Protein Modification and Degradation Laboratory, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510510, China.
                [2 ]Department of Infectious Diseases and State Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
                [3 ]State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Research Institute for Traffic Medicine of People’s Liberation Army, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
                [4 ]Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
                [5 ]Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
                [6 ]Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France.
                [7 ]Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.
                [8 ]Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.
                [9 ]Université Pierre et Marie Curie, 75006 Paris, France.
                [10 ]Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France.
                [11 ]Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
                [12 ]Department of Women’s and Children’s Health, Karolinska University Hospital, 17176 Stockholm, Sweden.
                [13 ]Laboratory of Emergency Medicine, North Shore University Hospital and The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
                [14 ]Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
                Author notes
                [* ]Corresponding author. Email: rui.kang@ 123456utsw.edu (R.K.); daolin.tang@ 123456utsw.edu (D.T.); jiangjx@ 123456cta.cq.cn (J.J.)
                Author information
                http://orcid.org/0000-0002-0353-592X
                http://orcid.org/0000-0002-9640-9083
                http://orcid.org/0000-0003-2017-1934
                http://orcid.org/0000-0002-9334-4405
                http://orcid.org/0000-0002-0211-9000
                http://orcid.org/0000-0003-0907-5976
                http://orcid.org/0000-0002-1903-6180
                http://orcid.org/0000-0003-2725-1574
                Article
                aav5562
                10.1126/sciadv.aav5562
                6531004
                31131320
                c7f92f75-8587-4ecc-92ed-385d7cc66a5f
                Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).

                This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

                History
                : 27 September 2018
                : 11 April 2019
                Funding
                Funded by: doi http://dx.doi.org/10.13039/100000052, NIH Office of the Director;
                Award ID: R01GM115366, R01CA160417, and R01GM127791
                Funded by: doi http://dx.doi.org/10.13039/100000052, NIH Office of the Director;
                Award ID: R01CA211070
                Funded by: doi http://dx.doi.org/10.13039/100000052, NIH Office of the Director;
                Award ID: R01AT005076 and R01GM063075
                Funded by: doi http://dx.doi.org/10.13039/100000052, NIH Office of the Director;
                Award ID: R01GM044100 and R35GM127027
                Categories
                Research Article
                Research Articles
                SciAdv r-articles
                Immunology
                Immunology
                Custom metadata
                Fritzie Benzon

                Comments

                Comment on this article