18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Epigenetic modification of m 6A regulator proteins in cancer

      review-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

          Divergent N 6-methyladenosine (m 6A) modifications are dynamic and reversible posttranscriptional RNA modifications that are mediated by m 6A regulators or m 6A RNA methylation regulators, i.e., methyltransferases (“writers”), demethylases (“erasers”), and m 6A-binding proteins (“readers”). Aberrant m 6A modifications are associated with cancer occurrence, development, progression, and prognosis. Numerous studies have established that aberrant m 6A regulators function as either tumor suppressors or oncogenes in multiple tumor types. However, the functions and mechanisms of m 6A regulators in cancer remain largely elusive and should be explored. Emerging studies suggest that m 6A regulators can be modulated by epigenetic modifications, namely, ubiquitination, SUMOylation, acetylation, methylation, phosphorylation, O-GlcNAcylation, ISGylation, and lactylation or via noncoding RNA action, in cancer. This review summarizes the current roles of m 6A regulators in cancer. The roles and mechanisms for epigenetic modification of m 6A regulators in cancer genesis are segregated. The review will improve the understanding of the epigenetic regulatory mechanisms of m 6A regulators.

          Related collections

          Most cited references134

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

          Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq.

          An extensive repertoire of modifications is known to underlie the versatile coding, structural and catalytic functions of RNA, but it remains largely uncharted territory. Although biochemical studies indicate that N(6)-methyladenosine (m(6)A) is the most prevalent internal modification in messenger RNA, an in-depth study of its distribution and functions has been impeded by a lack of robust analytical methods. Here we present the human and mouse m(6)A modification landscape in a transcriptome-wide manner, using a novel approach, m(6)A-seq, based on antibody-mediated capture and massively parallel sequencing. We identify over 12,000 m(6)A sites characterized by a typical consensus in the transcripts of more than 7,000 human genes. Sites preferentially appear in two distinct landmarks--around stop codons and within long internal exons--and are highly conserved between human and mouse. Although most sites are well preserved across normal and cancerous tissues and in response to various stimuli, a subset of stimulus-dependent, dynamically modulated sites is identified. Silencing the m(6)A methyltransferase significantly affects gene expression and alternative splicing patterns, resulting in modulation of the p53 (also known as TP53) signalling pathway and apoptosis. Our findings therefore suggest that RNA decoration by m(6)A has a fundamental role in regulation of gene expression.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            m6A-dependent regulation of messenger RNA stability

            N6 -methyladenosine (m6A) is the most prevalent internal (non-cap) modification present in the messenger RNA (mRNA) of all higher eukaryotes 1,2 . Although essential to cell viability and development 3–5 , the exact role of m6A modification remains to be determined. The recent discovery of two m6A demethylases in mammalian cells highlighted the importance of m6A in basic biological functions and disease 6–8 . Here we show that m6A is selectively recognized by the human YTH domain family 2 (YTHDF2) protein to regulate mRNA degradation. We identified over 3,000 cellular RNA targets of YTHDF2, most of which are mRNAs, but which also include non-coding RNAs, with a conserved core motif of G(m6A)C. We further establish the role of YTHDF2 in RNA metabolism, showing that binding of YTHDF2 results in the localization of bound mRNA from the translatable pool to mRNA decay sites, such as processing bodies 9 . The C-terminal domain of YTHDF2 selectively binds to m6A-containing mRNA whereas the N-terminal domain is responsible for the localization of the YTHDF2-mRNA complex to cellular RNA decay sites. Our results indicate that the dynamic m6A modification is recognized by selective-binding proteins to affect the translation status and lifetime of mRNA.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              N6-Methyladenosine in Nuclear RNA is a Major Substrate of the Obesity-Associated FTO

              We report here that FTO (fat mass and obesity-associated protein) exhibits efficient oxidative demethylation activity of abundant N 6-methyladenosine (m6A) residues in RNA in vitro. FTO knockdown with siRNA led to an increased level of m6A in mRNA, whereas overexpression of FTO resulted in a decreased level of m6A in human cells. We further show that FTO partially colocalizes with nuclear speckles, supporting m6A in nuclear RNA as a physiological substrate of FTO.
                Bookmark

                Author and article information

                Contributors
                wjh@imm.ac.cn
                chenz@stjohns.edu
                whongquan@alu.fudan.edu.cn
                Journal
                Mol Cancer
                Mol Cancer
                Molecular Cancer
                BioMed Central (London )
                1476-4598
                30 June 2023
                30 June 2023
                2023
                : 22
                : 102
                Affiliations
                [1 ]GRID grid.11135.37, ISNI 0000 0001 2256 9319, Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, , Peking University Aerospace School of Clinical Medicine, ; Beijing, 100049 China
                [2 ]GRID grid.452708.c, ISNI 0000 0004 1803 0208, Hunan Provincial Key Laboratory of Hepatobiliary Disease Research, Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, , The Second Xiangya Hospital of Central South University, ; Changsha, 410008, China
                [3 ]GRID grid.264091.8, ISNI 0000 0001 1954 7928, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, , St. John’s University, ; Queens, NY 11439 USA
                [4 ]GRID grid.506261.6, ISNI 0000 0001 0706 7839, Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, , Chinese Academy of Medical Sciences and Peking Union Medical College, ; Beijing, 100050 China
                [5 ]GRID grid.411918.4, ISNI 0000 0004 1798 6427, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, , Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, ; Tianjin, 300060 China
                Article
                1810
                10.1186/s12943-023-01810-1
                10311752
                37391814
                d3ef963e-aeb6-4187-aecd-48b7ad405833
                © The Author(s) 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 4 April 2023
                : 19 June 2023
                Funding
                Funded by: Science Foundation of CASIC
                Award ID: 2020-LCYL-009
                Funded by: Hygiene and Health Development Scientific Research Fostering Plan of Haidian District Beijing
                Award ID: HP2021-19-50701
                Funded by: Science Foundation of ASCH
                Award ID: YN202104
                Funded by: Science Foundation of AMHT
                Award ID: 2022YK01
                Categories
                Review
                Custom metadata
                © BioMed Central Ltd., part of Springer Nature 2023

                Oncology & Radiotherapy
                cancer,n6-methyladenosine methylation,rna modification,m6a regulators,m6a methylation enzymes

                Comments

                Comment on this article