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

      Inhibition of HDAC2 sensitises antitumour therapy by promoting NLRP3/GSDMD‐mediated pyroptosis in colorectal cancer

      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

          Background

          Although numerous studies have indicated that activated pyroptosis can enhance the efficacy of antitumour therapy in several tumours, the precise mechanism of pyroptosis in colorectal cancer (CRC) remains unclear.

          Methods

          Pyroptosis in CRC cells treated with antitumour agents was assessed using various techniques, including Western blotting, lactate dehydrogenase release assay and microscopy analysis. To uncover the epigenetic mechanisms that regulate NLRP3, chromatin changes and NLRP3 promoter histone modifications were assessed using Assay for Transposase‐Accessible Chromatin using sequencing and RNA sequencing. Chromatin immunoprecipitation‒quantitative polymerase chain reaction was used to investigate the NLRP3 transcriptional regulatory mechanism. Additionally, xenograft and patient‐derived xenograft models were constructed to validate the effects of the drug combinations.

          Results

          As the core molecule of the inflammasome, NLRP3 expression was silenced in CRC, thereby limiting gasdermin D (GSDMD)‐mediated pyroptosis. Supplementation with NLRP3 can rescue pyroptosis induced by antitumour therapy. Overexpression of HDAC2 in CRC silences NLRP3 via epigenetic regulation. Mechanistically, HDAC2 suppressed chromatin accessibility by eliminating H3K27 acetylation. HDAC2 knockout promotes H3K27ac‐mediated recruitment of the BRD4‐p‐P65 complex to enhance NLRP3 transcription. Inhibiting HDAC2 by Santacruzamate A in combination with classic antitumour agents (5‐fluorouracil or regorafenib) in CRC xenograft‐bearing animals markedly activated pyroptosis and achieved a significant therapeutic effect. Clinically, HDAC2 is inversely correlated with H3K27ac/p‐P65/NLRP3 and is a prognostic factor for CRC patients.

          Conclusion

          Collectively, our data revealed a crucial role for HDAC2 in inhibiting NLRP3/GSDMD‐mediated pyroptosis in CRC cells and highlighted HDAC2 as a potential therapeutic target for antitumour therapy.

          Highlights

          • Silencing of NLRP3 limits the GSDMD‐dependent pyroptosis in colorectal cancer.

          • HDAC2‐mediated histone deacetylation leads to epigenetic silencing of NLRP3.

          • HDAC2 suppresses the NLRP3 transcription by inhibiting the formation of H3K27ac/BRD4/p‐P65 complex.

          • Targeting HDAC2 activates pyroptosis and enhances therapeutic effect.

          Abstract

          Highlights

          • Silencing of NLRP3 limits the GSDMD‐dependent pyroptosis in colorectal cancer.

          • HDAC2‐mediated histone deacetylation leads to epigenetic silencing of NLRP3.

          • HDAC2 suppresses the NLRP3 transcription by inhibiting the formation of H3K27ac/BRD4/p‐P65 complex.

          • Targeting HDAC2 activates pyroptosis and enhances therapeutic effect.

          Related collections

          Most cited references49

          • 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

            Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position.

            We describe an assay for transposase-accessible chromatin using sequencing (ATAC-seq), based on direct in vitro transposition of sequencing adaptors into native chromatin, as a rapid and sensitive method for integrative epigenomic analysis. ATAC-seq captures open chromatin sites using a simple two-step protocol with 500-50,000 cells and reveals the interplay between genomic locations of open chromatin, DNA-binding proteins, individual nucleosomes and chromatin compaction at nucleotide resolution. We discovered classes of DNA-binding factors that strictly avoided, could tolerate or tended to overlap with nucleosomes. Using ATAC-seq maps of human CD4(+) T cells from a proband obtained on consecutive days, we demonstrated the feasibility of analyzing an individual's epigenome on a timescale compatible with clinical decision-making.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a Gasdermin

              Pyroptosis is a form of cell death that is critical for immunity. It can be induced by the canonical caspase-1 inflammasomes or by activation of caspase-4, -5 and -11 by cytosolic lipopolysaccharide. The caspases cleave gasdermin D (GSDMD) in its middle linker to release autoinhibition on its gasdermin-N domain, which executes pyroptosis via its pore-forming activity. GSDMD belongs to a gasdermin family that shares the pore-forming domain. The functions and mechanisms of activation of other gasdermins are unknown. Here we show that GSDME, which was originally identified as DFNA5 (deafness, autosomal dominant 5), can switch caspase-3-mediated apoptosis induced by TNF or chemotherapy drugs to pyroptosis. GSDME was specifically cleaved by caspase-3 in its linker, generating a GSDME-N fragment that perforates membranes and thereby induces pyroptosis. After chemotherapy, cleavage of GSDME by caspase-3 induced pyroptosis in certain GSDME-expressing cancer cells. GSDME was silenced in most cancer cells but expressed in many normal tissues. Human primary cells exhibited GSDME-dependent pyroptosis upon activation of caspase-3 by chemotherapy drugs. Gsdme-/- (also known as Dfna5-/-) mice were protected from chemotherapy-induced tissue damage and weight loss. These findings suggest that caspase-3 activation can trigger necrosis by cleaving GSDME and offer new insights into cancer chemotherapy.
                Bookmark

                Author and article information

                Contributors
                yaoyuanfei@hrbmu.edu.cn
                liuchao@hrbmu.edu.cn
                yanqiaozhang@ems.hrbmu.edu.cn
                Journal
                Clin Transl Med
                Clin Transl Med
                10.1002/(ISSN)2001-1326
                CTM2
                Clinical and Translational Medicine
                John Wiley and Sons Inc. (Hoboken )
                2001-1326
                28 May 2024
                June 2024
                : 14
                : 6 ( doiID: 10.1002/ctm2.v14.6 )
                : e1692
                Affiliations
                [ 1 ] Department of Gastrointestinal Medical Oncology Harbin Medical University Cancer Hospital Harbin China
                [ 2 ] Key Laboratory of Tumor Immunology in Heilongjiang Harbin China
                [ 3 ] Clinical Research Center for Colorectal Cancer in Heilongjiang Harbin China
                [ 4 ] Department of Radiation Oncology Sun Yat‐Sen University Cancer Center Guangzhou China
                [ 5 ] Phase I Clinical Research Center The Affiliated Hospital of Qingdao University Qingdao China
                [ 6 ] Department of Orthopedic Surgery Harbin Medical University Cancer Hospital Harbin China
                Author notes
                [*] [* ] Correspondence

                Yanqiao Zhang, Chao Liu and Yuanfei Yao, Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150001, China.

                Email: yanqiaozhang@ 123456ems.hrbmu.edu.cn ; liuchao@ 123456hrbmu.edu.cn ; yaoyuanfei@ 123456hrbmu.edu.cn

                Author information
                https://orcid.org/0000-0002-6206-8364
                Article
                CTM21692
                10.1002/ctm2.1692
                11131357
                38804602
                9057deb8-7efc-4cff-8116-ccf4c45f50d1
                © 2024 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

                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
                : 04 April 2024
                : 26 December 2023
                : 27 April 2024
                Page count
                Figures: 9, Tables: 0, Pages: 22, Words: 10566
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: U22A20330
                Award ID: 82173233
                Award ID: 82373372
                Award ID: 82102858
                Award ID: 82102988
                Funded by: Key Project of Research and Development Plan in Heilongjiang Province
                Award ID: 2022ZX06C01
                Funded by: Natural Science Funding of Heilongjiang
                Award ID: YQ2022H017
                Funded by: Harbin Medical University Cancer Hospital , doi 10.13039/100014418;
                Award ID: BJQN2021‐01
                Funded by: Top Young Talents Project of Harbin Medical University Cancer Hospital
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                June 2024
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.4.3 mode:remove_FC converted:28.05.2024

                Medicine
                colorectal cancer,h3k27ac,hdac2,nlrp3,pyroptosis
                Medicine
                colorectal cancer, h3k27ac, hdac2, nlrp3, pyroptosis

                Comments

                Comment on this article