Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

RNA-modifying enzymes and their function in a chromatin context

Abstract

Exciting research has connected specific RNA modifications to chromatin, providing evidence for co-transcriptional deposition and function in gene regulation. Here we review insights gained from studying the co-transcriptional roles of RNA modifications, and their influence in normal and disease contexts. We also discuss how the availability of novel technical approaches could raise the translational potential of targeting RNA-modifying enzymes for the treatment of disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Similar content being viewed by others

References

  1. Xhemalce, B., Dawson, M.A. & Bannister, A.J. Histone modifications. in Reviews in Cell Biology and Molecular Medicine (ed. R.A. Meyers) (2011).

  2. Grembecka, J. et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol. 8, 277–284 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. McCabe, M. T. et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492, 108–112 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Cheung, N. et al. Targeting aberrant epigenetic networks mediated by PRMT1 and KDM4C in acute myeloid leukemia. Cancer Cell 29, 32–48 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hu, H., Qian, K., Ho, M. C. & Zheng, Y. G. Small molecule inhibitors of protein arginine methyltransferases. Expert Opin. Investig. Drugs 25, 335–358 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Dawson, M. A. et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Schenk, T. et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat. Med. 18, 605–611 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Eckschlager, T., Plch, J., Stiborova, M. & Hrabeta, J. Histone deacetylase inhibitors as anticancer drugs. Int. J. Mol. Sci. 18, E1414 (2017).

  10. Nickerson, J. A., Krochmalnic, G., Wan, K. M. & Penman, S. Chromatin architecture and nuclear RNA. Proc. Natl Acad. Sci. USA 86, 177–181 (1989).

  11. Bernstein, E. & Allis, C. D. RNA meets chromatin. Genes Dev. 19, 1635–1655 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Mercer, T. R. & Mattick, J. S. Structure and function of long noncoding RNAs in epigenetic regulation. Nat. Struct. Mol. Biol. 20, 300–307 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Rinn, J. L. & Chang, H. Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 81, 145–166 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Tsai, M. C. et al. Long noncoding RNA as modular scaffold of histone modification complexes. Science 329, 689–693 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chu, C. et al. Systematic discovery of Xist RNA binding proteins. Cell 161, 404–416 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McHugh, C. A. et al. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 521, 232–236 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jégu, T. et al. Xist RNA antagonizes the SWI/SNF chromatin remodeler BRG1 on the inactive X chromosome. Nat. Struct. Mol. Biol. 26, 96–109 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Frye, M., Jaffrey, S. R., Pan, T., Rechavi, G. & Suzuki, T. RNA modifications: what have we learned and where are we headed? Nat. Rev. Genet. 17, 365–372 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Saletore, Y. et al. The birth of the Epitranscriptome: deciphering the function of RNA modifications. Genome Biol. 13, 175 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schwartz, S. Cracking the epitranscriptome. RNA 22, 169–174 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Meyer, K. D. et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 149, 1635–1646 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Khoddami, V. & Cairns, B. R. Identification of direct targets and modified bases of RNA cytosine methyltransferases. Nat. Biotechnol. 31, 458–464 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Khoddami, V. & Cairns, B. R. Transcriptome-wide target profiling of RNA cytosine methyltransferases using the mechanism-based enrichment procedure Aza-IP. Nat. Protoc. 9, 337–361 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Khoddami, V. et al. Transcriptome-wide profiling of multiple RNA modifications simultaneously at single-base resolution. Proc. Natl Acad. Sci. USA 116, 6784–6789 (2019).

  26. Linder, B. et al. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat. Methods 12, 767–772 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, K. et al. High-resolution N6-methyladenosine (m6A) map using photo-crosslinking-assisted m6A sequencing. Angew. Chem. Int. Edn Engl. 54, 1587–1590 (2015).

    Article  CAS  Google Scholar 

  28. Garcia-Campos, M. A. et al. Deciphering the “m6A Code” via antibody-independent quantitative profiling. Cell 178, 731–747.e716 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Zhang, Z. et al. Single-base mapping of m6A by an antibody-independent method. Sci. Adv. 5, eaax0250, https://doi.org/10.1126/sciadv.aax0250 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Clark, W. C., Evans, M. E., Dominissini, D., Zheng, G. & Pan, T. tRNA base methylation identification and quantification via high-throughput sequencing. RNA 22, 1771–1784 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Edelheit, S., Schwartz, S., Mumbach, M. R., Wurtzel, O. & Sorek, R. Transcriptome-wide mapping of 5-methylcytidine RNA modifications in bacteria, archaea, and yeast reveals m5C within archaeal mRNAs. PLoS Genet. 9, e1003602 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Carlile, T. M. et al. Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells. Nature 515, 143–146 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Schwartz, S. et al. Transcriptome-wide mapping reveals widespread dynamic-regulated pseudouridylation of ncRNA and mRNA. Cell 159, 148–162 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cao, X. & Limbach, P. A. Enhanced detection of post-transcriptional modifications using a mass-exclusion list strategy for RNA modification mapping by LC-MS/MS. Anal. Chem. 87, 8433–8440 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wetzel, C. & Limbach, P. A. Mass spectrometry of modified RNAs: recent developments. Analyst 141, 16–23 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Ross, R., Cao, X., Yu, N. & Limbach, P. A. Sequence mapping of transfer RNA chemical modifications by liquid chromatography tandem mass spectrometry. Methods 107, 73–78 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Montanaro, L. et al. Dyskerin expression influences the level of ribosomal RNA pseudo-uridylation and telomerase RNA component in human breast cancer. J. Pathol. 210, 10–18 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Xhemalce, B., Robson, S. C. & Kouzarides, T. Human RNA methyltransferase BCDIN3D regulates microRNA processing. Cell 151, 278–288 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jiang, Q. et al. ADAR1 promotes malignant progenitor reprogramming in chronic myeloid leukemia. Proc. Natl Acad. Sci. USA 110, 1041–1046 (2013).

  40. Blanco, S. et al. Stem cell function and stress response are controlled by protein synthesis. Nature 534, 335–340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Vu, L. P. et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat. Med. 23, 1369–1376 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hart, T. et al. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell 163, 1515–1526 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Steinhart, Z. et al. Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors. Nat. Med. 23, 60–68 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903.e815 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Behan, F. M. et al. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens. Nature 568, 511–516 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Tessarz, P. & Kouzarides, T. Histone core modifications regulating nucleosome structure and dynamics. Nat. Rev. Mol. Cell Biol. 15, 703–708 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Dawson, M. A., Kouzarides, T. & Huntly, B. J. Targeting epigenetic readers in cancer. N. Engl. J. Med. 367, 647–657 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11, 384–400 (2012).

    Article  CAS  PubMed  Google Scholar 

  50. Dawson, M. A. et al. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature 461, 819–822 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mews, P. et al. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature 546, 381–386 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Katoh, Y. et al. Methionine adenosyltransferase II serves as a transcriptional corepressor of Maf oncoprotein. Mol. Cell 41, 554–566 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Cheng, J. X. et al. RNA cytosine methylation and methyltransferases mediate chromatin organization and 5-azacytidine response and resistance in leukaemia. Nat. Commun. 9, 1163 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Huang, H. et al. Histone H3 trimethylation at lysine 36 guides m6A RNA modification co-transcriptionally. Nature 567, 414–419 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Barbieri, I. et al. Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature 552, 126–131 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rottman, F., Shatkin, A. J. & Perry, R. P. Sequences containing methylated nucleotides at the 5′ termini of messenger RNAs: possible implications for processing. Cell 3, 197–199 (1974).

    Article  CAS  PubMed  Google Scholar 

  58. Narayan, P. & Rottman, F. M. An in vitro system for accurate methylation of internal adenosine residues in messenger RNA. Science 242, 1159–1162 (1988).

    Article  CAS  PubMed  Google Scholar 

  59. Csepany, T., Lin, A., Baldick, C. J. Jr. & Beemon, K. Sequence specificity of mRNA N6-adenosine methyltransferase. J. Biol. Chem. 265, 20117–20122 (1990).

    CAS  PubMed  Google Scholar 

  60. Wang, Y. et al. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat. Cell Biol. 16, 191–198 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Liu, J. et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat. Chem. Biol. 10, 93–95 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Patil, D. P. et al. m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature 537, 369–373 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Knuckles, P. & Bühler, M. Adenosine methylation as a molecular imprint defining the fate of RNA. FEBS Lett. 592, 2845–2859 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yue, Y. et al. VIRMA mediates preferential m6A mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 4, 10 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Wen, J. et al. Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol. Cell 69, 1028–1038.e1026 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Aguilo, F. et al. Coordination of m6A mRNA methylation and gene transcription by ZFP217 regulates pluripotency and reprogramming. Cell Stem Cell 17, 689–704 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Pendleton, K. E. et al. The U6 snRNA m6A Methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 169, 824–835.e814 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Warda, A. S. et al. Human METTL16 is a N 6-methyladenosine (m6A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 18, 2004–2014 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Geula, S. et al. Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 347, 1002–1006 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Knuckles, P. et al. RNA fate determination through cotranscriptional adenosine methylation and microprocessor binding. Nat. Struct. Mol. Biol. 24, 561–569 (2017).

    Article  CAS  PubMed  Google Scholar 

  71. Wang, X. et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Alarcón, C. R. et al. HNRNPA2B1 is a mediator of m6A-dependent nuclear RNA processing events. Cell 162, 1299–1308 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Huang, H. et al. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 20, 285–295 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Śledź, P. & Jinek, M. Structural insights into the molecular mechanism of the m6A writer complex. eLife 5, e18434 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Wang, X. et al. Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature 534, 575–578 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Wang, P., Doxtader, K. A. & Nam, Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol. Cell 63, 306–317 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ping, X. L. et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24, 177–189 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Slobodin, B. et al. Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 169, 326–337.e312 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Xiang, Y. et al. RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 543, 573–576 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bertero, A. et al. The SMAD2/3 interactome reveals that TGFβ controls m6A mRNA methylation in pluripotency. Nature 555, 256–259 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Desrosiers, R., Friderici, K. & Rottman, F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc. Natl Acad. Sci. USA 71, 3971–3975 (1974).

  82. Dubin, D. T. & Taylor, R. H. The methylation state of poly A-containing messenger RNA from cultured hamster cells. Nucleic Acids Res. 2, 1653–1668 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Motorin, Y., Lyko, F. & Helm, M. 5-methylcytosine in RNA: detection, enzymatic formation and biological functions. Nucleic Acids Res. 38, 1415–1430 (2010).

    Article  CAS  PubMed  Google Scholar 

  84. Schaefer, M. et al. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev. 24, 1590–1595 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hussain, S. et al. NSun2-mediated cytosine-5 methylation of vault noncoding RNA determines its processing into regulatory small RNAs. Cell Rep. 4, 255–261 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Yang, X. et al. 5-Methylcytosine promotes mRNA export — NSUN2 as the methyltransferase and ALYREF as an m5C reader. Cell Res. 27, 606–625 (2017).

  87. Frye, M. et al. Genomic gain of 5p15 leads to over-expression of Misu (NSUN2) in breast cancer. Cancer Lett. 289, 71–80 (2010).

    Article  CAS  PubMed  Google Scholar 

  88. Fumagalli, D. et al. Principles governing A-to-I RNA editing in the breast cancer transcriptome. Cell Rep. 13, 277–289 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Rosenthal, J. J. The emerging role of RNA editing in plasticity. J. Exp. Biol. 218, 1812–1821 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Zipeto, M. A. et al. ADAR1 activation drives leukemia stem cell self-renewal by impairing Let-7 biogenesis. Cell Stem Cell 19, 177–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Wang, Q., Zhang, Z., Blackwell, K. & Carmichael, G. G. Vigilins bind to promiscuously A-to-I-edited RNAs and are involved in the formation of heterochromatin. Curr. Biol. 15, 384–391 (2005).

    Article  CAS  PubMed  Google Scholar 

  92. Jeronimo, C. et al. Systematic analysis of the protein interaction network for the human transcription machinery reveals the identity of the 7SK capping enzyme. Mol. Cell 27, 262–274 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Shuman, S. Transcriptional networking cap-tures the 7SK RNA 5′-γ-methyltransferase. Mol. Cell 27, 517–519 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Xue, Y., Yang, Z., Chen, R. & Zhou, Q. A capping-independent function of MePCE in stabilizing 7SK snRNA and facilitating the assembly of 7SK snRNP. Nucleic Acids Res. 38, 360–369 (2010).

    Article  CAS  PubMed  Google Scholar 

  95. Shelton, S. B. et al. Crosstalk between the RNA methylation and histone-binding activities of MePCE regulates P-TEFb activation on chromatin. Cell Rep. 22, 1374–1383 (2018).

    Article  CAS  PubMed  Google Scholar 

  96. Aregger, M. & Cowling, V. H. Regulation of mRNA capping in the cell cycle. RNA Biol. 14, 11–14 (2017).

    Article  PubMed  Google Scholar 

  97. Alexandrov, A., Martzen, M. R. & Phizicky, E. M. Two proteins that form a complex are required for 7-methylguanosine modification of yeast tRNA. RNA 8, 1253–1266 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Shaheen, R. et al. Mutation in WDR4 impairs tRNA m7G46 methylation and causes a distinct form of microcephalic primordial dwarfism. Genome Biol. 16, 210 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Lin, S. et al. Mettl1/Wdr4-mediated m7G tRNA methylome is required for normal mRNA translation and embryonic stem cell self-renewal and differentiation. Mol. Cell 71, 244–255.e245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Pandolfini, L. et al. METTL1 promotes let-7 microRNA processing via m7G methylation. Mol. Cell 74, 1278–1290.e1279 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Zhang, L. S. et al. Transcriptome-wide mapping of internal N7-methylguanosine methylome in mammalian mRNA. Mol. Cell 74, 1304–1316.e1308 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Choe, J. et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature 561, 556–560 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691.e610 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Lin, S., Choe, J., Du, P., Triboulet, R. & Gregory, R. I. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 62, 335–345 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ma, J. Z. et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methyladenosine-dependent primary microRNA processing. Hepatology 65, 529–543 (2017).

  106. Su, R. et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell 172, 90–105.e123 (2018).

    Article  CAS  PubMed  Google Scholar 

  107. Han, D. et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature 566, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Rapino, F. et al. Codon-specific translation reprogramming promotes resistance to targeted therapy. Nature 558, 605–609 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tony Kouzarides.

Ethics declarations

Competing interests

T.K. is a co-founder of Abcam and Storm Therapeutics. O.R. is an employee of Storm Therapeutics.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tzelepis, K., Rausch, O. & Kouzarides, T. RNA-modifying enzymes and their function in a chromatin context. Nat Struct Mol Biol 26, 858–862 (2019). https://doi.org/10.1038/s41594-019-0312-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0312-0

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer