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

      NOD1 induces pyroptotic cell death to aggravate liver ischemia‐reperfusion injury in mice

      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

          Nucleotide‐binding oligomerization domain 1 (NOD1) can direct the release of inflammatory factors and influence autophagy and apoptosis in hepatic ischemia‐reperfusion injury (IRI) in mice. As pyroptosis is involved in a number of inflammatory reactions, in this report, we investigated the potential for NOD1 to affect pyroptosis. We found that an increased expression of NOD1 during IRI was related to activation of the pyroptotic signaling pathway. With NOD1 activation, cleavage fragments of Caspase‐1, gasdermin D (GSDMD), and interleukin (IL)‐1β were all increased. Moreover, downregulation of NOD1 expression in AML12 cells exerted an opposite effect. Expression levels of cleaved‐Caspase‐1 and cleaved‐GSDMD decreased after exposure to IRI and the number of cell membrane pores and apoptotic or pyroptotic cells decreased, along with the contents of inflammatory factors and lactate dehydrogenase in the supernatants of AML12 cells. Based on these findings, we conclude that NOD1 aggravates the pyroptotic cell death associated with hepatic ischemia‐reperfusion injury in a mouse model via the Caspase‐1/GSDMD axis. These findings help to alleviate pyroptotic cell death during liver transplantation or resection, providing new insights into novel protective therapies for liver IRI.

          Abstract

          NOD1 is involved in apoptosis and autophagy during hepatic ischemia‐reperfusion. In this paper, we investigated its relationship with pyroptosis. The results showed that activation of NOD1 significantly aggravated pyroptotic cell death, resulting in liver injury, which has positive significance for further research on how to reduce liver ischemia‐reperfusion injury during liver transplantation or resection by affecting NOD1.

          Related collections

          Most cited references29

          • 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

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

              Pyroptosis: host cell death and inflammation.

              Eukaryotic cells can initiate several distinct programmes of self-destruction, and the nature of the cell death process (non-inflammatory or proinflammatory) instructs responses of neighbouring cells, which in turn dictates important systemic physiological outcomes. Pyroptosis, or caspase 1-dependent cell death, is inherently inflammatory, is triggered by various pathological stimuli, such as stroke, heart attack or cancer, and is crucial for controlling microbial infections. Pathogens have evolved mechanisms to inhibit pyroptosis, enhancing their ability to persist and cause disease. Ultimately, there is a competition between host and pathogen to regulate pyroptosis, and the outcome dictates life or death of the host.
                Bookmark

                Author and article information

                Contributors
                zgl_022@126.com
                caijinzhen@qdu.edu.cn
                Journal
                MedComm (2020)
                MedComm (2020)
                10.1002/(ISSN)2688-2663
                MCO2
                MedComm
                John Wiley and Sons Inc. (Hoboken )
                2688-2663
                31 August 2022
                September 2022
                : 3
                : 3 ( doiID: 10.1002/mco2.v3.3 )
                : e170
                Affiliations
                [ 1 ] Department of Gastroenterology Tianjin First Central Hospital The First Central Clinical College Tianjin Medical University Tianjin China
                [ 2 ] Department of internal medicine Wangdingdi Hospital Nankai District Tianjin China
                [ 3 ] Department of General Surgery Jiaozuo People's Hospital Xinxiang Medical University Jiaozuo China
                [ 4 ] Department of organ transplantation Organ Transplant Center Affiliated Hospital of Qingdao University Qingdao China
                Author notes
                [*] [* ] Correspondence

                Guoliang Zhang, Department of Gastroenterology, Tianjin First Central Hospital, Tianjin 300192, China.

                Email: zgl_022@ 123456126.com

                Jinzhen Cai, Organ Transplant Center, Affiliated Hospital of Qingdao University, Qingdao 266000, China.

                Email: caijinzhen@ 123456qdu.edu.cn

                Author information
                https://orcid.org/0000-0001-8615-2440
                Article
                MCO2170
                10.1002/mco2.170
                9433815
                36092860
                932e5f3b-6bb4-4d5f-b516-d8dd5832ae0d
                © 2022 The Authors. MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 26 April 2022
                : 04 February 2022
                : 28 April 2022
                Page count
                Figures: 5, Tables: 0, Pages: 11, Words: 5132
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 81670600
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                September 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.7 mode:remove_FC converted:01.09.2022

                hepatocytes,ischemia‐reperfusion,nod1,pyroptosis
                hepatocytes, ischemia‐reperfusion, nod1, pyroptosis

                Comments

                Comment on this article