2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: implication 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

          Accumulating evidence shows that N6-methyladenine (m 6A) modulators contribute to the etiology and progression of colorectal cancer (CRC). However, the exact mechanisms of m 6A reader involved in glycolytic metabolism remain vague. This article aimed to crosstalk the m 6A reader with glycolytic metabolism and reveal a new mechanism for the progression of CRC.

          Methods

          The relationship between candidate lncRNA and m 6A reader was analyzed by bioinformatics, ISH and IHC assays. In vivo and in vitro studies (including MTT, CFA, trans-well, apoptosis, western blot, qRT-PCR and xenograft mouse models) were utilized to explore the biological functions of these indicators. Lactate detection, ATP activity detection and ECAR assays were used to verify the biological function of the downstream target. The bioinformatics, RNA stability, RIP experiments and RNA pull-down assays were used to explore the potential molecular mechanisms.

          Results

          We identified that the crosstalk of the m 6A reader IMP2 with long-noncoding RNA (lncRNA) ZFAS1 in an m 6A modulation-dependent manner, subsequently augmented the recruitment of Obg-like ATPase 1 (OLA1) and adenosine triphosphate (ATP) hydrolysis and glycolysis during CRC proliferation and progression. Specifically, IMP2 and ZFAS1 are significantly overexpressed with elevated m 6A levels in CRC cells and paired CRC cohorts ( n = 144). These indicators could be independent biomarkers for CRC prognostic prediction. Notably, IMP2 regulated ZFAS1 expression and enhanced CRC cell proliferation, colony formation, and apoptosis inhibition; thus, it was oncogenic. Mechanistically, ZFAS1 is modified at adenosine +843 within the RGGAC/RRACH element in an m 6A-dependent manner. Thus, direct interaction between the KH3–4 domain of IMP2 and ZFAS1 where IMP2 serves as a reader for m 6A-modified ZFAS1 and promotes the RNA stability of ZFAS1 is critical for CRC development. More importantly, stabilized ZFAS1 recognizes the OBG-type functional domain of OLA1, which facilitated the exposure of ATP-binding sites (NVGKST, 32–37), enhanced its protein activity, and ultimately accelerated ATP hydrolysis and the Warburg effect.

          Conclusions

          Our findings reveal a new cancer-promoting mechanism, that is, the critical modulation network underlying m 6A readers stabilizes lncRNAs, and they jointly promote mitochondrial energy metabolism in the pathogenesis of CRC.

          Supplementary Information

          The online version contains supplementary material available at 10.1186/s13045-021-01204-0.

          Related collections

          Most cited references40

          • 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

            Recognition of RNA N 6 -methyladenosine by IGF2BP Proteins Enhances mRNA Stability and Translation

            N 6-methyladenosine (m6A) is the most prevalent modification in eukaryotic messenger RNAs (mRNAs) and is interpreted by its readers, such as YTH domain-containing proteins, to regulate mRNA fate. Here we report the insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs; including IGF2BP1/2/3) as a distinct family of m6A readers that target thousands of mRNA transcripts through recognizing the consensus GG(m6A)C sequence. In contrast to the mRNA-decay-promoting function of YTHDF2, IGF2BPs promote the stability and storage of their target mRNAs (e.g., MYC) in an m6A-depedent manner under normal and stress conditions and thus affect gene expression output. Moreover, the K homology (KH) domains of IGF2BPs are required for their recognition of m6A and are critical for their oncogenic functions. Our work therefore reveals a different facet of the m6A-reading process that promotes mRNA stability and translation, and highlights the functional importance of IGF2BPs as m6A readers in post-transcriptional gene regulation and cancer biology.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              m(6)A RNA methylation promotes XIST-mediated transcriptional repression.

              The long non-coding RNA X-inactive specific transcript (XIST) mediates the transcriptional silencing of genes on the X chromosome. Here we show that, in human cells, XIST is highly methylated with at least 78 N(6)-methyladenosine (m(6)A) residues-a reversible base modification of unknown function in long non-coding RNAs. We show that m(6)A formation in XIST, as well as in cellular mRNAs, is mediated by RNA-binding motif protein 15 (RBM15) and its paralogue RBM15B, which bind the m(6)A-methylation complex and recruit it to specific sites in RNA. This results in the methylation of adenosine nucleotides in adjacent m(6)A consensus motifs. Furthermore, we show that knockdown of RBM15 and RBM15B, or knockdown of methyltransferase like 3 (METTL3), an m(6)A methyltransferase, impairs XIST-mediated gene silencing. A systematic comparison of m(6)A-binding proteins shows that YTH domain containing 1 (YTHDC1) preferentially recognizes m(6)A residues on XIST and is required for XIST function. Additionally, artificial tethering of YTHDC1 to XIST rescues XIST-mediated silencing upon loss of m(6)A. These data reveal a pathway of m(6)A formation and recognition required for XIST-mediated transcriptional repression.
                Bookmark

                Author and article information

                Contributors
                18002438598@163.com
                han_cmu@163.com
                18253254674@163.com
                18304025798@163.com
                18102489307@163.com
                liyalun55116@sina.com
                qcchen@cmu.edu.cn
                ywf3209@163.com
                hemiao_cmu@126.com
                znwang@cmu.edu.cn
                wuhz@cmu.edu.cn
                minjieweicmu@163.com
                Journal
                J Hematol Oncol
                J Hematol Oncol
                Journal of Hematology & Oncology
                BioMed Central (London )
                1756-8722
                7 November 2021
                7 November 2021
                2021
                : 14
                : 188
                Affiliations
                [1 ]GRID grid.412449.e, ISNI 0000 0000 9678 1884, Department of Pharmacology, School of Pharmacy, , China Medical University, ; Shenyang, 110122 People’s Republic of China
                [2 ]GRID grid.412449.e, ISNI 0000 0000 9678 1884, Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation; Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, , Ministry of Education; China Medical University, ; Shenyang, 110122 People’s Republic of China
                [3 ]Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, Liaoning Province People’s Republic of China
                [4 ]GRID grid.412636.4, Department of Anorectal Surgery, , First Affiliated Hospital of China Medical University, ; Shenyang, 110001 People’s Republic of China
                [5 ]GRID grid.412636.4, Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, , Ministry of Education, First Affiliated Hospital of China Medical University, ; Shenyang, 110001 People’s Republic of China
                Author information
                http://orcid.org/0000-0002-5744-1603
                Article
                1204
                10.1186/s13045-021-01204-0
                8574039
                34743750
                04ecd1a3-7e22-4ad9-a1f3-b55bab86f16c
                © The Author(s) 2021

                Open AccessThis 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
                : 27 August 2021
                : 22 October 2021
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100001809, National Natural Science Foundation of China;
                Award ID: 81872905
                Award ID: 31828005
                Award ID: 82073884
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100007765, Shenyang Science and Technology Bureau;
                Award ID: 20-204-4-22
                Award ID: 19-109-4-09
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2021

                Oncology & Radiotherapy
                m6a methylation,imp2,atp-hydrolyzing and glycolysis,zfas1,ola1,colorectal cancer

                Comments

                Comment on this article