m6A modification: a new avenue for anti-cancer therapy

Yongtai Bai, Kai Li, Jinying Peng, Chengqi Yi

PDF(641 KB)
PDF(641 KB)
Life Medicine ›› 2023, Vol. 2 ›› Issue (1) : 7. DOI: 10.1093/lifemedi/lnad008
Review
Review

m6A modification: a new avenue for anti-cancer therapy

Author information +
History +

Abstract

To date, over 170 different kinds of chemical modifications on RNAs have been identified, some of which are involved in multiple aspects of RNA fate, ranging from RNA processing, nuclear export, translation, and RNA decay. m6A, also known as N6-methyladenosine, is a prominent internal RNA modification that is catalyzed primarily by the METTL3-METTL14-WTAP methyltransferase complex in higher eukaryotic mRNA and long noncoding RNA (lncRNA). In recent years, abnormal m6A modification has been linked to the occurrence, development, progression, and prognosis of the majority of cancers. In this review, we provide an update on the most recent m6A modification discoveries as well as the critical roles of m6A modification in cancer development and progression. We summarize the mechanisms of m6A involvement in cancer and list potential cancer therapy inhibitors that target m6A regulators such as “writer” METTL3 and “eraser” FTO.

Keywords

m6A / RNA modification / METTL3 / FTO / cancer therapy

Cite this article

Download citation ▾
Yongtai Bai, Kai Li, Jinying Peng, Chengqi Yi. m6A modification: a new avenue for anti-cancer therapy. Life Medicine, 2023, 2(1): 7 https://doi.org/10.1093/lifemedi/lnad008

References

[1]
Shi H, Wei J, He C. Where, when, and how: context-dependent functions of RNA methylation writers, readers, and erasers. Mol Cell 2019;74:640–50.
CrossRef Google scholar
[2]
He C. Grand challenge commentary: RNA epigenetics? Nat Chem Biol 2010;6:863–5.
CrossRef Google scholar
[3]
Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 2011;7:885–7.
CrossRef Google scholar
[4]
Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2013;49:18–29.
CrossRef Google scholar
[5]
Dedon PC, Begley TJ. A system of RNA modifications and biased codon use controls cellular stress response at the level of translation. Chem Res Toxicol 2014;27:330–7.
CrossRef Google scholar
[6]
Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nat Rev Cancer 2020;20:303–22.
CrossRef Google scholar
[7]
Zaccara S, Ries RJ, Jaffrey SR. Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol 2019;20:608–24.
CrossRef Google scholar
[8]
He PC, He C. m(6)A RNA methylation: from mechanisms to therapeutic potential. EMBO J 2021;40:e105977.
CrossRef Google scholar
[9]
Tanaka T, Weisblum B. Systematic difference in the methylation of ribosomal ribonucleic acid from gram-positive and gram-negative bacteria. J Bacteriol 1975;123:771–4.
CrossRef Google scholar
[10]
Lavi U, Fernandez-Munoz R, Darnell JE Jr. Content of N-6 methyl adenylic acid in heterogeneous nuclear and messenger RNA of HeLa cells. Nucleic Acids Res 1977;4:63–9.
CrossRef Google scholar
[11]
Patil DP, Chen CK, Pickering BF, et al m(6)A RNA methylation promotes Xist-mediated transcriptional repression. Nature 2016;537:369–73.
CrossRef Google scholar
[12]
Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 2014;10:93–5.
CrossRef Google scholar
[13]
Ping XL, Sun BF, Wang L, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 2014;24:177–89.
CrossRef Google scholar
[14]
Wen J, Lv R, Ma H, et al. Zc3h13 regulates nuclear RNA m(6)A methylation and mouse embryonic stem cell self-renewal. Mol Cell 2018;69:1028–1038.e6.
CrossRef Google scholar
[15]
Knuckles P, Lence T, Haussmann IU, et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m(6)A machinery component Wtap/Fl(2)D. Genes Dev 2018;32:415–29.
CrossRef Google scholar
[16]
Deng X, Su R, Weng H, et al. RNA N(6)-methyladenosine modification in cancers: current status and perspectives. Cell Res 2018;28:507–17.
CrossRef Google scholar
[17]
Yue Y, Liu J, Cui X, et al. Virma mediates preferential m(6)A mRNA methylation in 3’utr and near stop codon and associates with alternative polyadenylation. Cell Discov 2018;4:10.
CrossRef Google scholar
[18]
Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 2012;485:201–6.
CrossRef Google scholar
[19]
Ma H, Wang X, Cai J, et al. N(6-)methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA Methylation. Nat Chem Biol 2019;15:88–94.
CrossRef Google scholar
[20]
van Tran N, Ernst FGM, Hawley BR, et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res 2019;47:7719–33.
CrossRef Google scholar
[21]
Deng LJ, Deng WQ, Fan SR, et al. m6A modification: recent advances, anticancer targeted drug discovery and beyond. Mol Cancer 2022;21:52.
CrossRef Google scholar
[22]
Patil DP, Pickering BF, Jaffrey SR. Reading m(6)A in the transcriptome: m(6)A-binding proteins. Trends Cell Biol 2018;28:113–27.
CrossRef Google scholar
[23]
Xiao W, Adhikari S, Dahal U, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell 2016;61:507–19.
CrossRef Google scholar
[24]
Hsu PJ, Zhu Y, Ma H, et al. Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res 2017;27:1115–27.
CrossRef Google scholar
[25]
Han D, Liu J, Chen C, et al. Anti-tumour immunity controlled through mRNA m(6)A methylation and YTHDF1 in dendritic cells. Nature 2019;566:270–4.
CrossRef Google scholar
[26]
Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 2014;505:117–20.
CrossRef Google scholar
[27]
Li A, Chen YS, Ping XL, et al. Cytoplasmic m(6)A reader YTHDF3 promotes mRNA translation. Cell Res 2017;27:444–7.
CrossRef Google scholar
[28]
Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res 2017;27:315–28.
CrossRef Google scholar
[29]
Huang H, Weng H, Sun W, et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 2018;20:285–95.
CrossRef Google scholar
[30]
Ramesh-Kumar D, Guil S. The IGF2BP Family of RNA binding proteins links epitranscriptomics to cancer. Semin Cancer Biol 2022;86:18–31.
CrossRef Google scholar
[31]
Degrauwe N, Suva ML, Janiszewska M, et al. Imps: an RNA-binding protein family that provides a link between stem cell maintenance in normal development and cancer. Genes Dev 2016;30:2459–74.
CrossRef Google scholar
[32]
Huang H, Weng H, Chen J. m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell 2020;37:270–88.
CrossRef Google scholar
[33]
He L, Li H, Wu A, et al. Functions of N6-methyladenosine and its role in cancer. Mol Cancer 2019;18:176.
CrossRef Google scholar
[34]
Vu LP, Pickering BF, Cheng Y, et al. The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 2017;23:1369–76.
CrossRef Google scholar
[35]
Li M, Ye J, Xia Y, et al. METTL3 mediates chemoresistance by enhancing AML homing and engraftment via ITGA4. Leukemia 2022;36:2586–95.
CrossRef Google scholar
[36]
Barbieri I, Tzelepis K, Pandolfini L, et al. Promoter-bound METTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature 2017;552:126–31.
CrossRef Google scholar
[37]
Liu HT, Zou YX, Zhu WJ, et al. lncRNA THAP7-AS1, transcriptionally activated by SP1 and post-transcriptionally stabilized by METTL3-mediated m6A modification, exerts oncogenic properties by improving CUL4B entry into the nucleus. Cell Death Differ 2022;29:627–41.
CrossRef Google scholar
[38]
Lin S, Choe J, Du P, et al. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell 2016;62:335–45.
CrossRef Google scholar
[39]
Wang X, Zhao BS, Roundtree IA, et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell 2015;161:1388–99.
CrossRef Google scholar
[40]
Weng H, Huang H, Wu H, et al. Mettl14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m(6)A modification. Cell Stem Cell 2018;22:191–205.e9.
CrossRef Google scholar
[41]
Yankova E, Blackaby W, Albertella M, et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 2021;593:597–601.
CrossRef Google scholar
[42]
Li Z, Weng H, Su R, et al. FTO plays an oncogenic role in acute myeloid leukemia as a N(6)-methyladenosine RNA demethylase. Cancer Cell 2017;31:127–41.
CrossRef Google scholar
[43]
Fu X, Chin RM, Vergnes L, et al. 2-Hydroxyglutarate inhibits ATP synthase and mTOR signaling. Cell Metab 2015;22:508–15.
CrossRef Google scholar
[44]
Su R, Dong L, Li C, et al. R-2hg exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell 2018;172:90–105.e23.
CrossRef Google scholar
[45]
Qing Y, Dong L, Gao L, et al. R-2-Hydroxyglutarate attenuates aerobic glycolysis in leukemia by targeting the FTO/m(6)A/PFKP/LDHB axis. Mol Cell 2021;81:922–939.e9.
CrossRef Google scholar
[46]
Huang Y, Su R, Sheng Y, et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 2019;35:677–691.e10.
CrossRef Google scholar
[47]
Chen F, Chen Z, Guan T, et al N(6)-methyladenosine regulates mRNA stability and translation efficiency of KRT7 to promote breast cancer lung metastasis. Cancer Res 2021;81:2847–60.
CrossRef Google scholar
[48]
Cai X, Wang X, Cao C, et al. HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Lett 2018;415:11–9.
CrossRef Google scholar
[49]
Wang L, Hui H, Agrawal K, et al. m(6)A RNA methyltransferases METTL3/14 regulate immune responses to anti-PD-1 therapy. EMBO J 2020;39:e104514.
CrossRef Google scholar
[50]
Wan W, Ao X, Chen Q, et al. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N(6)-methyladenosine modification of PD-L1 mRNA in breast cancer. Mol Cancer 2022;21:60.
CrossRef Google scholar
[51]
Yin H, Zhang X, Yang P, et al. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming. Nat Commun 2021;12:1394.
CrossRef Google scholar
[52]
Wu Y, Wang Z, Han L, et al. PRMT5 regulates RNA m6A demethylation for doxorubicin sensitivity in breast cancer. Mol Ther 2022;30:2603–17.
CrossRef Google scholar
[53]
Pan X, Hong X, Li S, et al. METTL3 promotes adriamycin resistance in MCF-7 breast cancer cells by accelerating pri-microRNA-221-3p maturation in a m6A-dependent manner. Exp Mol Med 2021;53:91–102.
CrossRef Google scholar
[54]
Li Y, He X, Lu X, et al. METTL3 acetylation impedes cancer metastasis via fine-tuning its nuclear and cytosolic functions. Nat Commun 2022;13:6350.
CrossRef Google scholar
[55]
Zhang C, Chen L, Peng D, et al. METTL3 and N6-methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA-RNA hybrid accumulation. Mol Cell 2020;79:425–442.e7.
CrossRef Google scholar
[56]
Sun HL, Zhu AC, Gao Y, et al. Stabilization of ERK-phosphorylated METTL3 by USP5 increases m(6)A methylation. Mol Cell 2020;80:633–647.e7.
CrossRef Google scholar
[57]
Xiong J, He J, Zhu J, et al. Lactylation-driven METTL3-mediated RNA m(6)A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell 2022;82:1660–1677.e10.
CrossRef Google scholar
[58]
Hou G, Zhao X, Li L, et al. SUMOylation of YTHDF2 promotes mRNA degradation and cancer progression by increasing its binding affinity with m6A-modified mRNAs. Nucleic Acids Res 2021;49:2859–77.
CrossRef Google scholar
[59]
Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m(6)A-demethylation of nanog mRNA. Proc Natl Acad Sci U S A 2016;113:E2047–56.
CrossRef Google scholar
[60]
Chang G, Shi L, Ye Y, et al. YTHDF3 induces the translation of m(6) A-enriched gene transcripts to promote breast cancer brain metastasis. Cancer Cell 2020;38:857–871.e7.
CrossRef Google scholar
[61]
Raj N, Wang M, Seoane JA, et al. The Mettl3 epitranscriptomic writer amplifies p53 stress responses. Mol Cell 2022;82:2370–2384.e10.
CrossRef Google scholar
[62]
Choe J, Lin S, Zhang W, et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature 2018;561:556–60.
CrossRef Google scholar
[63]
Li Y, Gu J, Xu F, et al. Molecular characterization, biological function, tumor microenvironment association and clinical significance of m6A regulators in lung adenocarcinoma. Brief Bioinform 2021;22:bbaa225.
CrossRef Google scholar
[64]
Zhang Z, Zhang C, Yang Z, et al. m(6)A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer. J Hematol Oncol 2021;14:190.
CrossRef Google scholar
[65]
Liu J, Eckert MA, Harada BT, et al. m(6)A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat Cell Biol 2018;20:1074–83.
CrossRef Google scholar
[66]
Zhang L, Wan Y, Zhang Z, et al. IGF2BP1 overexpression stabilizes PEG10 mRNA in an m6A-dependent manner and promotes endometrial cancer progression. Theranostics 2021;11:1100–14.
CrossRef Google scholar
[67]
Han J, Wang JZ, Yang X, et al. METTL3 promote tumor proliferation of bladder cancer by accelerating pri-miR221/222 maturation in m6A-dependent manner. Mol Cancer 2019;18:110.
CrossRef Google scholar
[68]
Cheng M, Sheng L, Gao Q, et al. The m(6)A methyltransferase METTL3 promotes bladder cancer progression via AFF4/NF-κb/MYC signaling network. Oncogene 2019;38:3667–80.
CrossRef Google scholar
[69]
Wei W, Sun J, Zhang H, et al. Circ0008399 interaction with WTAP promotes assembly and activity of the m(6)A methyltransferase complex and promotes cisplatin resistance in bladder cancer. Cancer Res 2021;81:6142–56.
CrossRef Google scholar
[70]
Yang N, Wang T, Li Q, et al. HBXIP drives metabolic reprogramming in hepatocellular carcinoma cells via METTL3-mediated m6A modification of HIF-1α. J Cell Physiol 2021;236:3863–80.
CrossRef Google scholar
[71]
Chen M, Wei L, Law CT, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 2018;67:2254–70.
CrossRef Google scholar
[72]
Lin X, Chai G, Wu Y, et al. RNA m(6)A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of snail. Nat Commun 2019;10:2065.
CrossRef Google scholar
[73]
Lin Z, Niu Y, Wan A, et al. RNA m(6)A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J 2020;39:e103181.
CrossRef Google scholar
[74]
Kim GW, Imam H, Khan M, et al. HBV-induced increased N6 methyladenosine modification of PTEN RNA affects innate immunity and contributes to HCC. Hepatology 2021;73:533–47.
CrossRef Google scholar
[75]
Hou J, Zhang H, Liu J, et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol Cancer 2019;18:163.
CrossRef Google scholar
[76]
Zhang C, Huang S, Zhuang H, et al. YTHDF2 promotes the liver cancer stem cell phenotype and cancer metastasis by regulating OCT4 expression via m6A RNA methylation. Oncogene 2020;39:4507–18.
CrossRef Google scholar
[77]
Su R, Dong L, Li Y, et al. Mettl16 exerts an m(6)A-independent function to facilitate translation and tumorigenesis. Nat Cell Biol 2022;24:205–16.
CrossRef Google scholar
[78]
Dai YZ, Liu YD, Li J, et al. METTL16 promotes hepatocellular carcinoma progression through downregulating RAB11B-As1 in an m(6) A-dependent manner. Cell Mol Biol Lett 2022;27:41.
CrossRef Google scholar
[79]
Zhang C, Zhang M, Ge S, et al. Reduced m6A modification predicts malignant phenotypes and augmented Wnt/PI3K-Akt signaling in gastric cancer. Cancer Med 2019;8:4766–81.
CrossRef Google scholar
[80]
Yue B, Song C, Yang L, et al. METTL3-mediated N6-methyladenosine modification is critical for epithelial-mesenchymal transition and metastasis of gastric cancer. Mol Cancer 2019;18:142.
CrossRef Google scholar
[81]
Wang Q, Geng W, Guo H, et al. Emerging role of RNA methyltransferase METTL3 in gastrointestinal cancer. J Hematol Oncol 2020;13:57.
CrossRef Google scholar
[82]
Ge L, Zhang N, Chen Z, et al. Level of N6-methyladenosine in peripheral blood RNA: a novel predictive biomarker for gastric cancer. Clin Chem 2020;66:342–51.
CrossRef Google scholar
[83]
Wei X, Huo Y, Pi J, et al. METTL3 preferentially enhances non-m(6) A translation of epigenetic factors and promotes tumourigenesis. Nat Cell Biol 2022;24:1278–90.
CrossRef Google scholar
[84]
Lin Z, Wan AH, Sun L, et al. N6-methyladenosine demethylase FTO enhances chemo-resistance in colorectal cancer through SIVA1-mediated apoptosis. Mol Ther 2023;31:517–34.
CrossRef Google scholar
[85]
Ding C, Xu H, Yu Z, et al. RNA m(6)A demethylase ALKBH5 regulates the development of γδ T Cells. Proc Natl Acad Sci U S A 2022;119:e2203318119.
CrossRef Google scholar
[86]
Ediriweera MK, Tennekoon KH, Samarakoon SR. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: biological and therapeutic significance. Semin Cancer Biol 2019;59:147–60.
CrossRef Google scholar
[87]
Huang H, Wang Y, Kandpal M, et al. FTO-Dependent N(6)-methyladenosine modifications inhibit ovarian cancer stem cell self-renewal by blocking camp signaling. Cancer Res 2020;80:3200–14.
CrossRef Google scholar
[88]
Fukumoto T, Zhu H, Nacarelli T, et al. N(6)-methylation of adenosine of FZD10 mRNA contributes to PARP inhibitor resistance. Cancer Res 2019;79:2812–20.
CrossRef Google scholar
[89]
Liu T, Wei Q, Jin J, et al. The m6A reader YTHDF1 promotes ovarian cancer progression via augmenting EIF3C translation. Nucleic Acids Res 2020;48:3816–31.
CrossRef Google scholar
[90]
Xu F, Li J, Ni M, et al. FBW7 suppresses ovarian cancer development by targeting the N(6)-methyladenosine binding protein YTHDF2. Mol Cancer 2021;20:45.
CrossRef Google scholar
[91]
Yang S, Wei J, Cui YH, et al. m(6)A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat Commun 2019;10:2782.
CrossRef Google scholar
[92]
Liu Y, Liang G, Xu H, et al. Tumors exploit FTO- mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab 2021;33:1221–1233.e11.
CrossRef Google scholar
[93]
Cui Q, Shi H, Ye P, et al. m(6)A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep 2017;18:2622–34.
CrossRef Google scholar
[94]
Tang B, Yang Y, Kang M, et al. m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer 2020;19:3.
CrossRef Google scholar
[95]
Guo X, Li K, Jiang W, et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent Manner. Mol Cancer 2020;19:91.
CrossRef Google scholar
[96]
Du C, Lv C, Feng Y, et al. Activation of the KDM5A/miRNA-495/YTHDF2/m6A-MOB3B axis facilitates prostate cancer progression. J Exp Clin Cancer Res 2020;39:223.
CrossRef Google scholar
[97]
Liu C, Sun H, Yi Y, et al. Absolute quantification of single-Base m(6) A methylation in the mammalian transcriptome using GLORI. Nat Biotechnol 2023;41:355–66.
CrossRef Google scholar
[98]
Tassinari V, Cesarini V, Tomaselli S, et al. ADAR1 is a new target of METTL3 and plays a pro-oncogenic role in glioblastoma by an editing-independent mechanism. Genome Biol 2021;22:51.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2023 The Author(s) 2023. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(641 KB)

Accesses

Citations

Detail

Sections
Recommended

/