Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2020

Open Access 01-12-2020 | Review

m6A-binding proteins: the emerging crucial performers in epigenetics

Authors: Yanchun Zhao, Yuanfei Shi, Huafei Shen, Wanzhuo Xie

Published in: Journal of Hematology & Oncology | Issue 1/2020

Login to get access

Abstract

N6-methyladenosine (m6A) is a well-known post-transcriptional modification that is the most common type of methylation in eukaryotic mRNAs. The regulation of m6A is dynamic and reversible, which is erected by m6A methyltransferases (“writers”) and removed by m6A demethylases (“erasers”). Notably, the effects on targeted mRNAs resulted by m6A predominantly depend on the functions of different m6A-binding proteins (“readers”) including YT521-B homology (YTH) domain family, heterogeneous nuclear ribonucleoproteins (HNRNPs), and insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs). Indeed, m6A readers not only participate in multiple procedures of RNA metabolism, but also are involved in a variety of biological processes. In this review, we summarized the specific functions and underlying mechanisms of m6A-binding proteins in tumorigenesis, hematopoiesis, virus replication, immune response, and adipogenesis.
Literature
2.
go back to reference Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef
3.
go back to reference Perry RP, Kelley DE. Existence of methylated messenger RNA in mouse L cells. Cell. 1974;1(1):37–42.CrossRef Perry RP, Kelley DE. Existence of methylated messenger RNA in mouse L cells. Cell. 1974;1(1):37–42.CrossRef
4.
go back to reference Wei CM, Gershowitz A, Moss B. Methylated nucleotides block 5’ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379–86.PubMedCrossRef Wei CM, Gershowitz A, Moss B. Methylated nucleotides block 5’ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379–86.PubMedCrossRef
5.
go back to reference Wei CM, Moss B. Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. Biochemistry. 1977;16(8):1672–6.PubMedCrossRef Wei CM, Moss B. Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. Biochemistry. 1977;16(8):1672–6.PubMedCrossRef
6.
go back to reference Csepany T, Lin A, Baldick CJ Jr, Beemon K. Sequence specificity of mRNA N6-adenosine methyltransferase. J Biol Chem. 1990;265(33):20117–22.PubMed Csepany T, Lin A, Baldick CJ Jr, Beemon K. Sequence specificity of mRNA N6-adenosine methyltransferase. J Biol Chem. 1990;265(33):20117–22.PubMed
7.
go back to reference Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell. 2012;149(7):1635–46.PubMedPubMedCentralCrossRef Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell. 2012;149(7):1635–46.PubMedPubMedCentralCrossRef
8.
go back to reference Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature. 2016;534(7608):575–8.PubMedCrossRef Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature. 2016;534(7608):575–8.PubMedCrossRef
9.
go back to reference Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10(2):93–5.PubMedCrossRef Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10(2):93–5.PubMedCrossRef
10.
go back to reference Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.PubMedPubMedCentralCrossRef Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.PubMedPubMedCentralCrossRef
11.
go back to reference Yue Y, Liu J, Cui X, Cao J, Luo G, Zhang Z, et al. VIRMA mediates preferential m6A mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 2018;4:10.PubMedPubMedCentralCrossRef Yue Y, Liu J, Cui X, Cao J, Luo G, Zhang Z, et al. VIRMA mediates preferential m6A mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 2018;4:10.PubMedPubMedCentralCrossRef
12.
go back to reference Patil DP, Chen CK, Pickering BF, Chow A, Jackson C, Guttman M, et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 2016;537(7620):369–73.PubMedPubMedCentralCrossRef Patil DP, Chen CK, Pickering BF, Chow A, Jackson C, Guttman M, et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 2016;537(7620):369–73.PubMedPubMedCentralCrossRef
13.
go back to reference Wen J, Lv R, Ma H, Shen H, He C, Wang J, et al. Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol Cell. 2018;69(6):1028–38.e6.PubMedPubMedCentralCrossRef Wen J, Lv R, Ma H, Shen H, He C, Wang J, et al. Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol Cell. 2018;69(6):1028–38.e6.PubMedPubMedCentralCrossRef
14.
go back to reference Pendleton KE, Chen B, Liu K, Hunter OV, Xie Y, Tu BP, et al. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell. 2017;169(5):824–35.e14.PubMedPubMedCentralCrossRef Pendleton KE, Chen B, Liu K, Hunter OV, Xie Y, Tu BP, et al. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell. 2017;169(5):824–35.e14.PubMedPubMedCentralCrossRef
15.
go back to reference Warda AS, Kretschmer J, Hackert P, Lenz C, Urlaub H, Höbartner C, et al. Human METTL16 is a N6-methyladenosine (m6A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 2017;18(11):2004–14.PubMedPubMedCentralCrossRef Warda AS, Kretschmer J, Hackert P, Lenz C, Urlaub H, Höbartner C, et al. Human METTL16 is a N6-methyladenosine (m6A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 2017;18(11):2004–14.PubMedPubMedCentralCrossRef
16.
go back to reference Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885–7.PubMedPubMedCentralCrossRef Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885–7.PubMedPubMedCentralCrossRef
17.
go back to reference Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.PubMedCrossRef Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.PubMedCrossRef
18.
go back to reference Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161(6):1388–99.PubMedPubMedCentralCrossRef Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161(6):1388–99.PubMedPubMedCentralCrossRef
19.
go back to reference Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.PubMedCrossRef Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–20.PubMedCrossRef
20.
go back to reference Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 2016;7:12626.PubMedPubMedCentralCrossRef Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 2016;7:12626.PubMedPubMedCentralCrossRef
21.
go back to reference Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, et al. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 2017;27(3):315–28.PubMedPubMedCentralCrossRef Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, et al. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 2017;27(3):315–28.PubMedPubMedCentralCrossRef
22.
23.
go back to reference Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016;61(4):507–19.PubMedCrossRef Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016;61(4):507–19.PubMedCrossRef
25.
go back to reference Hsu PJ, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, et al. Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 2017;27(9):1115–27.PubMedPubMedCentralCrossRef Hsu PJ, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, et al. Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 2017;27(9):1115–27.PubMedPubMedCentralCrossRef
26.
go back to reference Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B, et al. m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun. 2019;10(1):5332.PubMedPubMedCentralCrossRef Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B, et al. m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun. 2019;10(1):5332.PubMedPubMedCentralCrossRef
27.
go back to reference Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015;162(6):1299–308.PubMedPubMedCentralCrossRef Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015;162(6):1299–308.PubMedPubMedCentralCrossRef
28.
29.
go back to reference Rajagopalan LE, Westmark CJ, Jarzembowski JA, Malter JS. hnRNP C tetramer measures RNA length to classify RNA polymerase II transcripts for export. Science. 2012;335(6076):1643–6.CrossRef Rajagopalan LE, Westmark CJ, Jarzembowski JA, Malter JS. hnRNP C tetramer measures RNA length to classify RNA polymerase II transcripts for export. Science. 2012;335(6076):1643–6.CrossRef
30.
go back to reference Zarnack K, König J, Tajnik M, Martincorena I, Eustermann S, Stévant I, et al. Direct competition between hnRNP C and U2AF65 protects the transcriptome from the exonization of Alu elements. Cell. 2013;152(3):453–66.PubMedPubMedCentralCrossRef Zarnack K, König J, Tajnik M, Martincorena I, Eustermann S, Stévant I, et al. Direct competition between hnRNP C and U2AF65 protects the transcriptome from the exonization of Alu elements. Cell. 2013;152(3):453–66.PubMedPubMedCentralCrossRef
31.
go back to reference Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L, Pan T. N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017;45(10):6051–63.PubMedPubMedCentralCrossRef Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L, Pan T. N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017;45(10):6051–63.PubMedPubMedCentralCrossRef
32.
go back to reference Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95..PubMedPubMedCentralCrossRef Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 2018;20(3):285–95..PubMedPubMedCentralCrossRef
33.
go back to reference Stoilov P, Rafalska I, Stamm S. YTH: a new domain in nuclear proteins. Trends Biochem Sci. 2002;27(10):495–7.PubMedCrossRef Stoilov P, Rafalska I, Stamm S. YTH: a new domain in nuclear proteins. Trends Biochem Sci. 2002;27(10):495–7.PubMedCrossRef
34.
go back to reference Zhang Z, Theler D, Kaminska KH, Hiller M, de la Grange P, Pudimat R, et al. The YTH domain is a novel RNA binding domain. J Biol Chem. 2010;285(19):14701–10.PubMedPubMedCentralCrossRef Zhang Z, Theler D, Kaminska KH, Hiller M, de la Grange P, Pudimat R, et al. The YTH domain is a novel RNA binding domain. J Biol Chem. 2010;285(19):14701–10.PubMedPubMedCentralCrossRef
35.
go back to reference Xu C, Wang X, Liu K, Roundtree IA, Tempel W, Li Y, et al. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol. 2014;10(11):927–9.PubMedCrossRef Xu C, Wang X, Liu K, Roundtree IA, Tempel W, Li Y, et al. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol. 2014;10(11):927–9.PubMedCrossRef
36.
go back to reference Li Y, Bedi RK, Wiedmer L, Huang D, Sledz P, Caflisch A. Flexible binding of m6A reader protein YTHDC1 to its preferred RNA motif. 2019;15(12):7004–14. Li Y, Bedi RK, Wiedmer L, Huang D, Sledz P, Caflisch A. Flexible binding of m6A reader protein YTHDC1 to its preferred RNA motif. 2019;15(12):7004–14.
37.
go back to reference Kretschmer J, Rao H, Hackert P, Sloan KE, Höbartner C, Bohnsack MT. The m6A reader protein YTHDC2 interacts with the small ribosomal subunit and the 5’-3’ exoribonuclease XRN1. RNA. 2018;24(10):1339–50.PubMedPubMedCentralCrossRef Kretschmer J, Rao H, Hackert P, Sloan KE, Höbartner C, Bohnsack MT. The m6A reader protein YTHDC2 interacts with the small ribosomal subunit and the 5’-3’ exoribonuclease XRN1. RNA. 2018;24(10):1339–50.PubMedPubMedCentralCrossRef
38.
go back to reference Wu B, Su S, Patil DP, Liu H, Gan J, Jaffrey SR, et al. Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat Commun. 2018;9(1):420.PubMedPubMedCentralCrossRef Wu B, Su S, Patil DP, Liu H, Gan J, Jaffrey SR, et al. Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat Commun. 2018;9(1):420.PubMedPubMedCentralCrossRef
39.
go back to reference Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518(7540):560–4.PubMedPubMedCentralCrossRef Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518(7540):560–4.PubMedPubMedCentralCrossRef
40.
go back to reference Bell JL, Wächter K, Mühleck B, Pazaitis N, Köhn M, Lederer M, et al. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cell Mol Life Sci. 2013;70(15):2657–75.PubMedCrossRef Bell JL, Wächter K, Mühleck B, Pazaitis N, Köhn M, Lederer M, et al. Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cell Mol Life Sci. 2013;70(15):2657–75.PubMedCrossRef
41.
go back to reference Schito L, Semenza GL. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer. 2016;2(12):758–70.PubMedCrossRef Schito L, Semenza GL. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer. 2016;2(12):758–70.PubMedCrossRef
42.
go back to reference Zhong L, Liao D, Zhang M, Zeng C, Li X, Zhang R, et al. YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 2019;442:252–61.PubMedCrossRef Zhong L, Liao D, Zhang M, Zeng C, Li X, Zhang R, et al. YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 2019;442:252–61.PubMedCrossRef
43.
go back to reference Hou J, Zhang H, Liu J, Zhao Z, Wang J, Lu Z, et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol Cancer. 2019;18(1):163.PubMedPubMedCentralCrossRef Hou J, Zhang H, Liu J, Zhao Z, Wang J, Lu Z, et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol Cancer. 2019;18(1):163.PubMedPubMedCentralCrossRef
44.
go back to reference Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018;67(6):2254–70.PubMedCrossRef Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018;67(6):2254–70.PubMedCrossRef
45.
go back to reference Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.PubMedCrossRef Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.PubMedCrossRef
47.
go back to reference Lin X, Chai G, Wu Y, Li J, Chen F, Liu J, et al. RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nat Commun. 2019;10(1):2065.PubMedPubMedCentralCrossRef Lin X, Chai G, Wu Y, Li J, Chen F, Liu J, et al. RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nat Commun. 2019;10(1):2065.PubMedPubMedCentralCrossRef
48.
go back to reference Müller S, Glaß M, Singh AK, Haase J, Bley N, Fuchs T, et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 2019;47(1):375–90.PubMedCrossRef Müller S, Glaß M, Singh AK, Haase J, Bley N, Fuchs T, et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 2019;47(1):375–90.PubMedCrossRef
49.
go back to reference Bai Y, Yang C, Wu R, Huang L, Song S, Li W, et al. YTHDF1 regulates tumorigenicity and cancer stem cell-like activity in human colorectal carcinoma. Front Oncol. 2019;9:332.PubMedPubMedCentralCrossRef Bai Y, Yang C, Wu R, Huang L, Song S, Li W, et al. YTHDF1 regulates tumorigenicity and cancer stem cell-like activity in human colorectal carcinoma. Front Oncol. 2019;9:332.PubMedPubMedCentralCrossRef
50.
go back to reference Ni W, Yao S, Zhou Y, Liu Y, Huang P, Zhou A, et al. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m6A reader YTHDF3. Mol Cancer. 2019;18:143.PubMedPubMedCentralCrossRef Ni W, Yao S, Zhou Y, Liu Y, Huang P, Zhou A, et al. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m6A reader YTHDF3. Mol Cancer. 2019;18:143.PubMedPubMedCentralCrossRef
51.
go back to reference Chen RX, Chen X, Xia LP, Zhang JX, Pan ZZ, Ma XD, et al. N6-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun. 2019;10(1):4695.PubMedPubMedCentralCrossRef Chen RX, Chen X, Xia LP, Zhang JX, Pan ZZ, Ma XD, et al. N6-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun. 2019;10(1):4695.PubMedPubMedCentralCrossRef
52.
go back to reference Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, et al. METTL3 facilitates tumor progression via an m6A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.PubMedPubMedCentralCrossRef Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, et al. METTL3 facilitates tumor progression via an m6A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18(1):112.PubMedPubMedCentralCrossRef
53.
go back to reference He H, Wu W, Sun Z, Chai L. MiR-4429 prevented gastric cancer progression through targeting METTL3 to inhibit m6A-caused stabilization of SEC62. Biochem Biophys Res Commun. 2019;517(4):581–7.PubMedCrossRef He H, Wu W, Sun Z, Chai L. MiR-4429 prevented gastric cancer progression through targeting METTL3 to inhibit m6A-caused stabilization of SEC62. Biochem Biophys Res Commun. 2019;517(4):581–7.PubMedCrossRef
55.
56.
go back to reference Shi Y, Fan S, Wu M, Zuo Z, Li X, Jiang L, et al. YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression. Nat Commun. 2019;10(1):4892.PubMedPubMedCentralCrossRef Shi Y, Fan S, Wu M, Zuo Z, Li X, Jiang L, et al. YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression. Nat Commun. 2019;10(1):4892.PubMedPubMedCentralCrossRef
57.
go back to reference Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, et al. m6A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J Hematol Oncol. 2019;12(1):135.PubMedPubMedCentralCrossRef Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, et al. m6A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J Hematol Oncol. 2019;12(1):135.PubMedPubMedCentralCrossRef
58.
go back to reference Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–45.PubMedPubMedCentralCrossRef Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–45.PubMedPubMedCentralCrossRef
59.
go back to reference Jin H, Ying X, Que B, Wang X, Chao Y, Zhang H, et al. N6-methyladenosine modification of ITGA6 mRNA promotes the development and progression of bladder cancer. EBioMedicine. 2019;47:195–207.PubMedPubMedCentralCrossRef Jin H, Ying X, Que B, Wang X, Chao Y, Zhang H, et al. N6-methyladenosine modification of ITGA6 mRNA promotes the development and progression of bladder cancer. EBioMedicine. 2019;47:195–207.PubMedPubMedCentralCrossRef
60.
go back to reference Yang F, Jin H, Que B, Chao Y, Zhang H, Ying X, et al. Dynamic m6A mRNA methylation reveals the role of METTL3-m6A-CDCP1 signaling axis in chemical carcinogenesis. Oncogene. 2019;38(24):4755–72.PubMedPubMedCentralCrossRef Yang F, Jin H, Que B, Chao Y, Zhang H, Ying X, et al. Dynamic m6A mRNA methylation reveals the role of METTL3-m6A-CDCP1 signaling axis in chemical carcinogenesis. Oncogene. 2019;38(24):4755–72.PubMedPubMedCentralCrossRef
61.
go back to reference Liu J, Eckert MA, Harada BT, Liu SM, Lu Z, Yu K, et al. m6A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat Cell Biol. 2018;20(9):1074–83.PubMedPubMedCentralCrossRef Liu J, Eckert MA, Harada BT, Liu SM, Lu Z, Yu K, et al. m6A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat Cell Biol. 2018;20(9):1074–83.PubMedPubMedCentralCrossRef
63.
go back to reference Wang X, Zhang J, Wang Y. Long noncoding RNA GAS5-AS1 suppresses growth and metastasis of cervical cancer by increasing GAS5 stability. Am J Transl Res. 2019;11(8):4909–21.PubMedPubMedCentral Wang X, Zhang J, Wang Y. Long noncoding RNA GAS5-AS1 suppresses growth and metastasis of cervical cancer by increasing GAS5 stability. Am J Transl Res. 2019;11(8):4909–21.PubMedPubMedCentral
64.
go back to reference Yang S, Wei J, Cui YH, Park G, Shah P, Deng Y, et al. m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat Commun. 2019;10(1):2782.PubMedPubMedCentralCrossRef Yang S, Wei J, Cui YH, Park G, Shah P, Deng Y, et al. m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat Commun. 2019;10(1):2782.PubMedPubMedCentralCrossRef
65.
go back to reference Jia R, Chai P, Wang S, Sun B, Xu Y, Yang Y, et al. m6A modification suppresses ocular melanoma through modulating HINT2 mRNA translation. Mol Cancer. 2019;18(1):161.PubMedPubMedCentralCrossRef Jia R, Chai P, Wang S, Sun B, Xu Y, Yang Y, et al. m6A modification suppresses ocular melanoma through modulating HINT2 mRNA translation. Mol Cancer. 2019;18(1):161.PubMedPubMedCentralCrossRef
66.
go back to reference Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, et al. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 2019;18(1):46.PubMedPubMedCentralCrossRef Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, et al. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 2019;18(1):46.PubMedPubMedCentralCrossRef
68.
go back to reference Speck NA, Gilliland DG. Core-binding factors in haematopoiesis and leukaemia. Nat Rev Cancer. 2002;2(7):502–13.PubMedCrossRef Speck NA, Gilliland DG. Core-binding factors in haematopoiesis and leukaemia. Nat Rev Cancer. 2002;2(7):502–13.PubMedCrossRef
69.
go back to reference Sonoda Y. Immunophenotype and functional characteristics of human primitive CD34-negative hematopoietic stem cells: the significance of the intra-bone marrow injection. J Autoimmun. 2008;30(3):136–44.PubMedCrossRef Sonoda Y. Immunophenotype and functional characteristics of human primitive CD34-negative hematopoietic stem cells: the significance of the intra-bone marrow injection. J Autoimmun. 2008;30(3):136–44.PubMedCrossRef
70.
go back to reference Wognum AW, Eaves AC, Thomas TE. Identification and isolation of hematopoietic stem cells. Arch Med Res. 2003;34(6):461–75.PubMedCrossRef Wognum AW, Eaves AC, Thomas TE. Identification and isolation of hematopoietic stem cells. Arch Med Res. 2003;34(6):461–75.PubMedCrossRef
71.
go back to reference Weissman IL, Shizuru JA. The origins of the identification and isolation of hematopoietic stem cells, and their capability to induce donor-specific transplantation tolerance and treat autoimmune diseases. Blood. 2008;112(9):3543–53.PubMedPubMedCentralCrossRef Weissman IL, Shizuru JA. The origins of the identification and isolation of hematopoietic stem cells, and their capability to induce donor-specific transplantation tolerance and treat autoimmune diseases. Blood. 2008;112(9):3543–53.PubMedPubMedCentralCrossRef
73.
go back to reference Zhang P, He Q, Chen D, Liu W, Wang L, Zhang C, et al. G protein-coupled receptor 183 facilitates endothelial-to-hematopoietic transition via Notch1 inhibition. Cell Res. 2015;25(10):1093–107.PubMedPubMedCentralCrossRef Zhang P, He Q, Chen D, Liu W, Wang L, Zhang C, et al. G protein-coupled receptor 183 facilitates endothelial-to-hematopoietic transition via Notch1 inhibition. Cell Res. 2015;25(10):1093–107.PubMedPubMedCentralCrossRef
74.
go back to reference Gama-Norton L, Ferrando E, Ruiz-Herguido C, Liu Z, Guiu J, Islam AB, et al. Notch signal strength controls cell fate in the haemogenic endothelium. Nat Commun. 2015;6:8510.PubMedPubMedCentralCrossRef Gama-Norton L, Ferrando E, Ruiz-Herguido C, Liu Z, Guiu J, Islam AB, et al. Notch signal strength controls cell fate in the haemogenic endothelium. Nat Commun. 2015;6:8510.PubMedPubMedCentralCrossRef
75.
go back to reference Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature. 2017;549(7671):273–6.PubMedCrossRef Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature. 2017;549(7671):273–6.PubMedCrossRef
76.
go back to reference Li Z, Qian P, Shao W, Shi H, He XC, Gogol M, et al. Suppression of m6A reader Ythdf2 promotes hematopoietic stem cell expansion. Cell Res. 2018;28(9):904–17.PubMedPubMedCentralCrossRef Li Z, Qian P, Shao W, Shi H, He XC, Gogol M, et al. Suppression of m6A reader Ythdf2 promotes hematopoietic stem cell expansion. Cell Res. 2018;28(9):904–17.PubMedPubMedCentralCrossRef
77.
go back to reference Wang H, Zuo H, Liu J, Wen F, Gao Y, Zhu X, et al. Loss of YTHDF2-mediated m6A-dependent mRNA clearance facilitates hematopoietic stem cell regeneration. Cell Res. 2018;28(10):1035–8.PubMedPubMedCentralCrossRef Wang H, Zuo H, Liu J, Wen F, Gao Y, Zhu X, et al. Loss of YTHDF2-mediated m6A-dependent mRNA clearance facilitates hematopoietic stem cell regeneration. Cell Res. 2018;28(10):1035–8.PubMedPubMedCentralCrossRef
78.
go back to reference Ye M, Zhang H, Yang H, Koche R, Staber PB, Cusan M, et al. Hematopoietic differentiation is required for initiation of acute myeloid leukemia. Cell Stem Cell. 2015;17(5):611–23.PubMedPubMedCentralCrossRef Ye M, Zhang H, Yang H, Koche R, Staber PB, Cusan M, et al. Hematopoietic differentiation is required for initiation of acute myeloid leukemia. Cell Stem Cell. 2015;17(5):611–23.PubMedPubMedCentralCrossRef
80.
go back to reference Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef
81.
go back to reference Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, et al. Targeting the RNA m6A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell. 2019;25(1):137–48.e6.PubMedPubMedCentralCrossRef Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, et al. Targeting the RNA m6A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell. 2019;25(1):137–48.e6.PubMedPubMedCentralCrossRef
82.
go back to reference Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell. 2018;172(1-2):90–105.e23.PubMedCrossRef Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell. 2018;172(1-2):90–105.e23.PubMedCrossRef
83.
go back to reference Hesser CR, Karijolich J, Dominissini D, He C, Glaunsinger BA. N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi’s sarcoma-associated herpesvirus infection. PLoS Pathog. 2018;14(4):e1006995.PubMedPubMedCentralCrossRef Hesser CR, Karijolich J, Dominissini D, He C, Glaunsinger BA. N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi’s sarcoma-associated herpesvirus infection. PLoS Pathog. 2018;14(4):e1006995.PubMedPubMedCentralCrossRef
85.
go back to reference Tan B, Liu H, Zhang S, da Silva SR, Zhang L, Meng J, et al. Viral and cellular N6-methyladenosine and N6,2’-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol. 2018;3(1):108–20.PubMedCrossRef Tan B, Liu H, Zhang S, da Silva SR, Zhang L, Meng J, et al. Viral and cellular N6-methyladenosine and N6,2’-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol. 2018;3(1):108–20.PubMedCrossRef
87.
go back to reference Lu W, Tirumuru N, St Gelais C, Koneru PC, Liu C, Kvaratskhelia M, et al. N6-methyladenosine-binding proteins suppress HIV-1 infectivity and viral production. J Biol Chem. 2018;293(34):12992–3005.PubMedPubMedCentralCrossRef Lu W, Tirumuru N, St Gelais C, Koneru PC, Liu C, Kvaratskhelia M, et al. N6-methyladenosine-binding proteins suppress HIV-1 infectivity and viral production. J Biol Chem. 2018;293(34):12992–3005.PubMedPubMedCentralCrossRef
88.
go back to reference Kennedy EM, Bogerd HP, Kornepati AV, Kang D, Ghoshal D, Marshall JB, et al. Posttranscriptional m(6)A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe. 2016;19(5):675–85.PubMedPubMedCentralCrossRef Kennedy EM, Bogerd HP, Kornepati AV, Kang D, Ghoshal D, Marshall JB, et al. Posttranscriptional m(6)A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe. 2016;19(5):675–85.PubMedPubMedCentralCrossRef
89.
go back to reference Courtney DG, Kennedy EM, Dumm RE, Bogerd HP, Tsai K, Heaton NS, et al. Epitranscriptomic enhancement of influenza A virus gene expression and replication. Cell Host Microbe. 2017;22(3):377–86.e5.PubMedPubMedCentralCrossRef Courtney DG, Kennedy EM, Dumm RE, Bogerd HP, Tsai K, Heaton NS, et al. Epitranscriptomic enhancement of influenza A virus gene expression and replication. Cell Host Microbe. 2017;22(3):377–86.e5.PubMedPubMedCentralCrossRef
90.
go back to reference Gokhale NS, McIntyre ABR, McFadden MJ, Roder AE, Kennedy EM, Gandara JA, et al. N6-methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe. 2016;20(5):654–65.PubMedPubMedCentralCrossRef Gokhale NS, McIntyre ABR, McFadden MJ, Roder AE, Kennedy EM, Gandara JA, et al. N6-methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe. 2016;20(5):654–65.PubMedPubMedCentralCrossRef
91.
go back to reference Liu J, Zhang X, Chen K, Cheng Y, Liu S, Xia M, et al. CCR7 chemokine receptor-inducible lnc-Dpf3 restrains dendritic cell migration by inhibiting HIF-1α-mediated glycolysis. Immunity. 2019;50(3):600–15.e15.PubMedCrossRef Liu J, Zhang X, Chen K, Cheng Y, Liu S, Xia M, et al. CCR7 chemokine receptor-inducible lnc-Dpf3 restrains dendritic cell migration by inhibiting HIF-1α-mediated glycolysis. Immunity. 2019;50(3):600–15.e15.PubMedCrossRef
92.
93.
go back to reference Chen YG, Chen R, Ahmad S, Verma R, Kasturi SP, Amaya L, et al. N6-methyladenosine modification controls circular RNA immunity. Mol Cell. 2019;76(1):96–109.e9.PubMedCrossRef Chen YG, Chen R, Ahmad S, Verma R, Kasturi SP, Amaya L, et al. N6-methyladenosine modification controls circular RNA immunity. Mol Cell. 2019;76(1):96–109.e9.PubMedCrossRef
94.
go back to reference Zhao BB, Guo HJ, Liu Y, Luo XY, Yang SX, Wang YT, et al. K313, a novel benzoxazole derivative, exhibits anti-inflammatory properties via inhibiting GSK3β activity in LPS-induced RAW264.7 macrophages. J Cell Biochem. 2018;119(7):5382–90.PubMedCrossRef Zhao BB, Guo HJ, Liu Y, Luo XY, Yang SX, Wang YT, et al. K313, a novel benzoxazole derivative, exhibits anti-inflammatory properties via inhibiting GSK3β activity in LPS-induced RAW264.7 macrophages. J Cell Biochem. 2018;119(7):5382–90.PubMedCrossRef
97.
go back to reference Winkler R, Gillis E, Lasman L, Safra M, Geula S, Soyris C, et al. m6A modification controls the innate immune response to infection by targeting type I interferons. Nat Immunol. 2019;20(2):173–82.PubMedCrossRef Winkler R, Gillis E, Lasman L, Safra M, Geula S, Soyris C, et al. m6A modification controls the innate immune response to infection by targeting type I interferons. Nat Immunol. 2019;20(2):173–82.PubMedCrossRef
98.
go back to reference Rubio RM, Depledge DP, Bianco C, Thompson L, Mohr I. RNA m6A modification enzymes shape innate responses to DNA by regulating interferon β. Genes Dev. 2018;32(23-24):1472–84.PubMedPubMedCentralCrossRef Rubio RM, Depledge DP, Bianco C, Thompson L, Mohr I. RNA m6A modification enzymes shape innate responses to DNA by regulating interferon β. Genes Dev. 2018;32(23-24):1472–84.PubMedPubMedCentralCrossRef
100.
101.
go back to reference Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217–21.PubMedPubMedCentralCrossRef Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217–21.PubMedPubMedCentralCrossRef
102.
go back to reference Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature. 2019;566(7743):270–4. Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature. 2019;566(7743):270–4.
104.
go back to reference Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out. Nat Rev Mol Cell Biol. 2006;7(12):885–96.PubMedCrossRef Rosen ED, MacDougald OA. Adipocyte differentiation from the inside out. Nat Rev Mol Cell Biol. 2006;7(12):885–96.PubMedCrossRef
105.
go back to reference Wu R, Liu Y, Yao Y, Zhao Y, Bi Z, Jiang Q, et al. FTO regulates adipogenesis by controlling cell cycle progression via m6A YTHDF2 dependent mechanism. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(10):1323–30.PubMedCrossRef Wu R, Liu Y, Yao Y, Zhao Y, Bi Z, Jiang Q, et al. FTO regulates adipogenesis by controlling cell cycle progression via m6A YTHDF2 dependent mechanism. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(10):1323–30.PubMedCrossRef
106.
go back to reference Wu R, Yao Y, Jiang Q, Cai M, Liu Q, Wang Y, et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m6A-YTHDF2-dependent manner. Int J Obes (Lond). 2018;42(7):1378–88.CrossRef Wu R, Yao Y, Jiang Q, Cai M, Liu Q, Wang Y, et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m6A-YTHDF2-dependent manner. Int J Obes (Lond). 2018;42(7):1378–88.CrossRef
107.
go back to reference Liu Q, Zhao Y, Wu R, Jiang Q, Cai M, Bi Z, et al. ZFP217 regulates adipogenesis by controlling mitotic clonal expansion in a METTL3-m6A dependent manner. RNA Biol. 2019;27:1–9.CrossRef Liu Q, Zhao Y, Wu R, Jiang Q, Cai M, Bi Z, et al. ZFP217 regulates adipogenesis by controlling mitotic clonal expansion in a METTL3-m6A dependent manner. RNA Biol. 2019;27:1–9.CrossRef
108.
go back to reference Wu R, Guo G, Bi Z, Liu Y, Zhao Y, Chen N, et al. m6A methylation modulates adipogenesis through JAK2-STAT3-C/EBPβ signaling. Biochim Biophys Acta Gene Regul Mech. 2019;1862(8):796–806.PubMedCrossRef Wu R, Guo G, Bi Z, Liu Y, Zhao Y, Chen N, et al. m6A methylation modulates adipogenesis through JAK2-STAT3-C/EBPβ signaling. Biochim Biophys Acta Gene Regul Mech. 2019;1862(8):796–806.PubMedCrossRef
109.
go back to reference Yao Y, Bi Z, Wu R, Zhao Y, Liu Y, Liu Q, et al. METTL3 inhibits BMSC adipogenic differentiation by targeting the JAK1/STAT5/C/EBPβ pathway via an m6A-YTHDF2-dependent manner. FASEB J. 2019;33(6):7529–44.PubMedCrossRef Yao Y, Bi Z, Wu R, Zhao Y, Liu Y, Liu Q, et al. METTL3 inhibits BMSC adipogenic differentiation by targeting the JAK1/STAT5/C/EBPβ pathway via an m6A-YTHDF2-dependent manner. FASEB J. 2019;33(6):7529–44.PubMedCrossRef
110.
go back to reference Zhong X, Yu J, Frazier K, Weng X, Li Y, Cham CM, et al. Circadian clock regulation of hepatic lipid metabolism by modulation of m6A mRNA methylation. Cell Rep. 2018;25(7):1816–28.e4.PubMedPubMedCentralCrossRef Zhong X, Yu J, Frazier K, Weng X, Li Y, Cham CM, et al. Circadian clock regulation of hepatic lipid metabolism by modulation of m6A mRNA methylation. Cell Rep. 2018;25(7):1816–28.e4.PubMedPubMedCentralCrossRef
111.
go back to reference Cai M, Liu Q, Jiang Q, Wu R, Wang X, Wang Y. Loss of m6 A on FAM134B promotes adipogenesis in porcine adipocytes through m6 A-YTHDF2-dependent way. IUBMB Life. 2019;71(5):580–6.PubMedCrossRef Cai M, Liu Q, Jiang Q, Wu R, Wang X, Wang Y. Loss of m6 A on FAM134B promotes adipogenesis in porcine adipocytes through m6 A-YTHDF2-dependent way. IUBMB Life. 2019;71(5):580–6.PubMedCrossRef
112.
go back to reference Song T, Yang Y, Wei H, Xie X, Lu J, Zeng Q, et al. Zfp217 mediates m6A mRNA methylation to orchestrate transcriptional and post-transcriptional regulation to promote adipogenic differentiation. Nucleic Acids Res. 2019;47(12):6130–44.PubMedPubMedCentralCrossRef Song T, Yang Y, Wei H, Xie X, Lu J, Zeng Q, et al. Zfp217 mediates m6A mRNA methylation to orchestrate transcriptional and post-transcriptional regulation to promote adipogenic differentiation. Nucleic Acids Res. 2019;47(12):6130–44.PubMedPubMedCentralCrossRef
113.
go back to reference Wang X, Wu R, Liu Y, Zhao Y, Bi Z, Yao Y, et al. m6A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy. 2019;26:1–15. Wang X, Wu R, Liu Y, Zhao Y, Bi Z, Yao Y, et al. m6A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy. 2019;26:1–15.
114.
go back to reference Jiang Q, Sun B, Liu Q, Cai M, Wu R, Wang F, et al. MTCH2 promotes adipogenesis in intramuscular preadipocytes via an m6A-YTHDF1-dependent mechanism. FASEB J. 2019;33(2):2971–81.PubMedCrossRef Jiang Q, Sun B, Liu Q, Cai M, Wu R, Wang F, et al. MTCH2 promotes adipogenesis in intramuscular preadipocytes via an m6A-YTHDF1-dependent mechanism. FASEB J. 2019;33(2):2971–81.PubMedCrossRef
115.
go back to reference Wang X, Sun B, Jiang Q, Wu R, Cai M, Yao Y, et al. mRNA m6A plays opposite role in regulating UCP2 and PNPLA2 protein expression in adipocytes. Int J Obes (Lond). 2018;42(11):1912–24.CrossRef Wang X, Sun B, Jiang Q, Wu R, Cai M, Yao Y, et al. mRNA m6A plays opposite role in regulating UCP2 and PNPLA2 protein expression in adipocytes. Int J Obes (Lond). 2018;42(11):1912–24.CrossRef
116.
go back to reference Yue B, Song C, Yang L, Cui R, Cheng X, Zhang Z, et al. METTL3-mediated N6-methyladenosine modification is critical for epithelialmesenchymal transition and metastasis of gastric cancer. Mol Cancer. 2019;18(1):142.PubMedPubMedCentralCrossRef Yue B, Song C, Yang L, Cui R, Cheng X, Zhang Z, et al. METTL3-mediated N6-methyladenosine modification is critical for epithelialmesenchymal transition and metastasis of gastric cancer. Mol Cancer. 2019;18(1):142.PubMedPubMedCentralCrossRef
117.
118.
go back to reference Huang Y, Yan J, Li Q, Li J, Gong S, Zhou H, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015;43(1):373–84.PubMedCrossRef Huang Y, Yan J, Li Q, Li J, Gong S, Zhou H, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015;43(1):373–84.PubMedCrossRef
Metadata
Title
m6A-binding proteins: the emerging crucial performers in epigenetics
Authors
Yanchun Zhao
Yuanfei Shi
Huafei Shen
Wanzhuo Xie
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2020
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00872-8

Other articles of this Issue 1/2020

Journal of Hematology & Oncology 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine