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.

  • Review Article
  • Published:

Role of RNA modifications in cancer

Abstract

Specific chemical modifications of biological molecules are an efficient way of regulating molecular function, and a plethora of downstream signalling pathways are influenced by the modification of DNA and proteins. Many of the enzymes responsible for regulating protein and DNA modifications are targets of current cancer therapies. RNA epitranscriptomics, the study of RNA modifications, is the new frontier of this arena. Despite being known since the 1970s, eukaryotic RNA modifications were mostly identified on transfer RNA and ribosomal RNA until the last decade, when they have been identified and characterized on mRNA and various non-coding RNAs. Increasing evidence suggests that RNA modification pathways are also misregulated in human cancers and may be ideal targets of cancer therapy. In this Review we highlight the RNA epitranscriptomic pathways implicated in cancer, describing their biological functions and their connections to the 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

Fig. 1: Internal RNA modifications.
Fig. 2: Distribution of the seven post-transcriptional modifications on different RNA subtypes.
Fig. 3: The N6-methyladenosine machinery and its roles in cancer.
Fig. 4: The main functions of RNA-modifying enzymes in cancer beyond those of the N6-methyladenosine modification.
Fig. 5: 5′ cap structures present on different RNA subtypes.
Fig. 6: RNA modification writers, readers and erasers can have either promoting or inhibitory effects on the hallmarks of cancer.

Similar content being viewed by others

References

  1. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Li, S. & Mason, C. E. The pivotal regulatory landscape of RNA modifications. Annu. Rev. Genomics Hum. Genet. 15, 127–150 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Zhang, J., Yang, P. L. & Gray, N. S. Targeting cancer with small molecule kinase inhibitors. Nat. Rev. Cancer 9, 28–39 (2009).

    Article  PubMed  CAS  Google Scholar 

  4. Dor, Y. & Cedar, H. Principles of DNA methylation and their implications for biology and medicine. Lancet 392, 777–786 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Mohammad, H. P., Barbash, O. & Creasy, C. L. Targeting epigenetic modifications in cancer therapy: erasing the roadmap to cancer. Nat. Med. 25, 403–418 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Gebert, L. F. R. & MacRae, I. J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 20, 21–37 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yao, R.-W., Wang, Y. & Chen, L.-L. Cellular functions of long noncoding RNAs. Nat. Cell Biol. 21, 542–551 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Zhao, B. S., Roundtree, I. A. & He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 18, 31–42 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Shelton, S. B., Reinsborough, C. & Xhemalce, B. Who watches the watchmen: roles of RNA modifications in the RNA interference pathway. PLoS Genet. 12, e1006139 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Esteller, M. & Pandolfi, P. P. The epitranscriptome of noncoding RNAs in cancer. Cancer Discov. 7, 359–368 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Helm, M. & Motorin, Y. Detecting RNA modifications in the epitranscriptome: predict and validate. Nat. Rev. Genet. 18, 275–291 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Shubert, C. Epitranscriptomics: RNA revisited. Science 364, 693–695 (2019).

    Google Scholar 

  14. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fazi, F. & Fatica, A. Interplay between N6-methyladenosine (m6A) and non-coding RNAs in cell development and cancer. Front. Cell Dev. Biol. 7, 116 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Alarcón, C. R., Lee, H., Goodarzi, H., Halberg, N. & Tavazoie, S. F. N6-methyladenosine marks primary microRNAs for processing. Nature 519, 482–485 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Maden, B. E. H. Identification of the locations of the methyl groups in 18S ribosomal RNA from Xenopus laevis and man. J. Mol. Biol. 189, 681–699 (1986).

    Article  CAS  PubMed  Google Scholar 

  18. Maden, B. E. H. Locations of methyl groups in 28S rRNA of xenopus laevis and man: clustering in the conserved core of molecule. J. Mol. Biol. 201, 289–314 (1988).

    Article  CAS  PubMed  Google Scholar 

  19. Liu, J. et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat. Chem. Biol. 10, 93–95 (2014). In this study, the METTL3–METTL14 complex is identified as the main m 6A writer in mammalian cells.

    Article  CAS  PubMed  Google Scholar 

  20. Pendleton, K. E. et al. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 169, 824–835.e14 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. van Tran, N. et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 47, 7719–7733 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Ma, H. et al. N6-Methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat. Chem. Biol. 15, 88–94 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Perry, R. P. & Kelley, D. E. Existence of methylated messenger RNA in mouse L cells. Cell 1, 37–42 (1974).

    Article  CAS  Google Scholar 

  24. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012). This important study maps for the first time m 6A modifications on the transcriptome, highlighting the importance of this modification for mRNA regulation and starting the new field of epitranscriptomics.

    Article  CAS  PubMed  Google Scholar 

  25. 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 

  26. Jia, G. et al. N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011). In this study, the first m 6A eraser is identified. This highlights the dynamic nature of this modification on mRNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zheng, G. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Mauer, J. et al. Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 541, 371–375 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Liao, S., Sun, H. & Xu, C. YTH domain: a family of N6-methyladenosine (m6A) readers. Genomics Proteomics Bioinformatics 16, 99–107 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. 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 

  32. Wu, R. et al. A novel m6A reader Prrc2a controls oligodendroglial specification and myelination. Cell Res. 29, 23–41 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Wu, B. et al. Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat. Commun. 9, 420 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Sun, L. et al. RNA structure maps across mammalian cellular compartments. Nat. Struct. Mol. Biol. 26, 322–330 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Du, H. et al. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4–NOT deadenylase complex. Nat. Commun. 7, 12626 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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). In this study, METTL3 is found to increase MYC translation and sustain the growth of AML cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Barbieri, I. et al. Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature 552, 126–131 (2017). This study identifies METTL3 as a potential target in AML and identifies a function of this enzyme that is dependent on its recruitment to chromatin.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Weng, H. et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A modification. Cell Stem Cell 22, 191–205.e9 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. 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 

  41. Chen, M. et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 67, 2254–2270 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Lin, X. et al. RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nat. Commun. 10, 2065 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Ma, J. et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N 6-methyladenosine-dependent primary MicroRNA processing. Hepatology 65, 529–543 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Liu, J. et al. m6A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat. Cell Biol. 20, 1074–1083 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cui, Q. et al. m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 18, 2622–2634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Visvanathan, A. et al. Essential role of METTL3-mediated m6A modification in glioma stem-like cells maintenance and radioresistance. Oncogene 37, 522–533 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Li, F. et al. N6-methyladenosine modulates nonsense-mediated mRNA decay in human glioblastoma. Cancer Res. 79, 5785–5798 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Brown, J. A., Kinzig, C. G., DeGregorio, S. J. & Steitz, J. A. Methyltransferase-like protein 16 binds the 3′-terminal triple helix of MALAT1 long noncoding RNA. Proc. Natl Acad. Sci. USA 113, 14013–14018 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sun, Y. & Ma, L. New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers 11, E216 (2019).

    Article  PubMed  CAS  Google Scholar 

  50. Iles, M. M. et al. A variant in FTO shows association with melanoma risk not due to BMI. Nat. Genet. 45, 428–432 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Yang, S. et al. m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat. Commun. 10, 2782 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Li, Z. et al. FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell 31, 127–141 (2017). This article describes the discovery of the oncogenic function of the m 6A demethylase FTO in AML.

    Article  PubMed  CAS  Google Scholar 

  53. Elkashef, S. M. et al. IDH mutation, competitive inhibition of FTO, and RNA methylation. Cancer Cell 31, 619–620 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ward, P. S. et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting α-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Su, R. et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell 172, 90–105.e23 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691.e10 (2019). This article describes the first small-molecule inhibitor of the m 6A pathway, which demonstrates efficacy in mouse models of AML.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mauer, J. et al. FTO controls reversible m6Am RNA methylation during snRNA biogenesis. Nat. Chem. Biol. 15, 340–347 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, S. et al. m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell 31, 591–606.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang, C. et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc. Natl Acad. Sci. USA 113, E2047–E2056 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Tanabe, A. et al. RNA helicase YTHDC2 promotes cancer metastasis via the enhancement of the efficiency by which HIF-1α mRNA is translated. Cancer Lett. 376, 34–42 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Paris, J. et al. Targeting the RNA m6A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell 25, 137–148.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Han, D. et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature 566, 270–274 (2019). In this study, YTHDF1 is identified as a critical suppressor of antigen presentation in dendritic cells and its depletion could enhance the effect of immune checkpoint inhibitors in melanoma.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Degrauwe, N., Suvà, M.-L., Janiszewska, M., Riggi, N. & Stamenkovic, I. IMPs: an RNA-binding protein family that provides a link between stem cell maintenance in normal development and cancer. Genes. Dev. 30, 2459–2474 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Weidensdorfer, D. et al. Control of c-myc mRNA stability by IGF2BP1-associated cytoplasmic RNPs. RNA 15, 104 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Müller, S. et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 47, 375–390 (2019).

    Article  PubMed  CAS  Google Scholar 

  66. Bohnsack, K., Höbartner, C. & Bohnsack, M. Eukaryotic 5-methylcytosine (m5C) RNA methyltransferases: mechanisms, cellular functions, and links to disease. Genes 10, 102 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  67. Trixl, L. & Lusser, A. The dynamic RNA modification 5-methylcytosine and its emerging role as an epitranscriptomic mark. Wiley Interdiscip. Rev. RNA 10, e1510 (2019).

    Article  PubMed  CAS  Google Scholar 

  68. Schosserer, M. et al. Methylation of ribosomal RNA by NSUN5 is a conserved mechanism modulating organismal lifespan. Nat. Commun. 6, 6158 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Squires, J. E. et al. Widespread occurrence of 5-methylcytosine in human coding and non-coding RNA. Nucleic Acids Res. 40, 5023–5033 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Legrand, C. et al. Statistically robust methylation calling for whole-transcriptome bisulfite sequencing reveals distinct methylation patterns for mouse RNAs. Genome Res. 27, 1589–1596 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Song, C. X., Yi, C. & He, C. Mapping recently identified nucleotide variants in the genome and transcriptome. Nat. Biotechnol. 30, 1107–1116 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Huang, T., Chen, W., Liu, J., Gu, N. & Zhang, R. Genome-wide identification of mRNA 5-methylcytosine in mammals. Nat. Struct. Mol. Biol. 26, 230–388 (2019). This study determines the abundance of high-confidence m 5C modification sites within eukaryotic mRNAs.

    Article  CAS  Google Scholar 

  73. 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). In this study, the first function of mRNA m 5C and the first RNA m 5C reader are identified.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. 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 

  75. Freeman, J. W. et al. Identification and characterization of a human proliferation-associated nucleolar antigen with a molecular weight of 120,000 expressed in early G1 phase. Cancer Res. 48, 1244–1251 (1988).

    CAS  PubMed  Google Scholar 

  76. Bantis, A. et al. Expression of p120, Ki-67 and PCNA as proliferation biomarkers in imprint smears of prostate carcinoma and their prognostic value. Cytopathology 15, 25–31 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Saijo, Y. et al. Expression of nucleolar protein p120 predicts poor prognosis in patients with stage I lung adenocarcinoma. Ann. Oncol. 12, 1121–1125 (2001).

    Article  CAS  PubMed  Google Scholar 

  78. Frye, M. & Watt, F. M. The RNA methyltransferase Misu (NSun2) mediates Myc-induced proliferation and is upregulated in tumors. Curr. Biol. 16, 971–981 (2006). This article describes the discovery of NSUN2 as a MYC target and oncogenic factor.

    Article  CAS  PubMed  Google Scholar 

  79. Li, Y. et al. Novel long noncoding RNA NMR promotes tumor progression via NSUN2 and BPTF in esophageal squamous cell carcinoma. Cancer Lett. 430, 57–66 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. Chen, X. et al. 5-methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs. Nat. Cell Biol. 21, 978–990 (2019).

    Article  CAS  PubMed  Google Scholar 

  81. Campbell, T. M., Castro, M. A. A., de Oliveira, K. G., Ponder, B. A. J. & Meyer, K. B. ERα binding by transcription factors NFIB and YBX1 enables FGFR2 signaling to modulate estrogen responsiveness in breast cancer. Cancer Res. 78, 410–421 (2018).

    Article  CAS  PubMed  Google Scholar 

  82. Yamashita, T., Higashi, M., Momose, S., Morozumi, M. & Tamaru, J.-I. Nuclear expression of Y box binding-1 is important for resistance to chemotherapy including gemcitabine in TP53-mutated bladder cancer. Int. J. Oncol. 51, 579–586 (2017).

    Article  CAS  PubMed  Google Scholar 

  83. 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 

  84. Saikia, M., Fu, Y., Pavon-Eternod, M., He, C. & Pan, T. Genome-wide analysis of N1-methyl-adenosine modification in human tRNAs. RNA 16, 1317–1327 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Sloan, K. E. et al. Tuning the ribosome: the influence of rRNA modification on eukaryotic ribosome biogenesis and function. RNA Biol. 14, 1138–1152 (2017).

    Article  PubMed  Google Scholar 

  86. Safra, M. et al. The m1A landscape on cytosolic and mitochondrial mRNA at single-base resolution. Nature 551, 251–255 (2017). This study presents the genome-wide mapping of m 1A, showing limited abundance of this modification on mRNA.

    Article  CAS  PubMed  Google Scholar 

  87. Dominissini, D. et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature 530, 441–446 (2016). This study presents the genome-wide mapping of m 1A in mammalian cells, showing specific topology and association with high translation levels.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhou, H. et al. Evolution of a reverse transcriptase to map N1-methyladenosine in human messenger RNA. Nat. Methods 16, 1281–1288 (2019). This study through the development of a new technique to detect m 1A determines the topology of this modification on eukaryotic mRNAs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Dai, X., Wang, T., Gonzalez, G. & Wang, Y. Identification of YTH domain-containing proteins as the readers for N 1-methyladenosine in RNA. Anal. Chem. 90, 6380–6384 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Chen, Z. et al. Transfer RNA demethylase ALKBH3 promotes cancer progression via induction of tRNA-derived small RNAs. Nucleic Acids Res. 47, 2533–2545 (2019).

    Article  CAS  PubMed  Google Scholar 

  91. Liu, F. et al. ALKBH1-mediated trna demethylation regulates translation. Cell 167, 816–828.e16 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Woo, H.-H. & Chambers, S. K. Human ALKBH3-induced m1A demethylation increases the CSF-1 mRNA stability in breast and ovarian cancer cells. Biochim. Biophys. Acta Gene Regul. Mech. 1862, 35–46 (2019).

    Article  CAS  PubMed  Google Scholar 

  93. Konishi, N. et al. High expression of a new marker PCA-1 in human prostate carcinoma. Clin. Cancer Res. 11, 5090–5097 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Nakao, S. et al. Design and synthesis of prostate cancer antigen-1 (PCA-1/ALKBH3) inhibitors as anti-prostate cancer drugs. Bioorg. Med. Chem. Lett. 24, 1071–1074 (2014).

    Article  CAS  PubMed  Google Scholar 

  95. Duncan, T. et al. Reversal of DNA alkylation damage by two human dioxygenases. Proc. Natl Acad. Sci. USA 99, 16660–16665 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ramanathan, A., Robb, G. B. & Chan, S.-H. mRNA capping: biological functions and applications. Nucleic Acids Res. 44, 7511–7526 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Tomikawa, C. 7-Methylguanosine modifications in transfer RNA (tRNA). Int. J. Mol. Sci. 19, E4080 (2018).

    Article  PubMed  Google Scholar 

  98. Pandolfini, L. et al. METTL1 promotes let-7 microRNA processing via m7G methylation. Mol. Cell 74, 1278–1290.e9 (2019). In this study, internal m 7G is identified on mature and pre-miRNAs, including let-7, where it is required for pre-let-7 processing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhang, L. S. et al. Transcriptome-wide mapping of internal N7-methylguanosine methylome in mammalian mRNA. Mol. Cell 74, 1304–1316.e8 (2019). This article identifies internal m 7G on mRNA and describes the transcriptome-wide mapping of the modification.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. 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 

  101. Haag, S., Kretschmer, J. & Bohnsack, M. T. WBSCR22/Merm1 is required for late nuclear pre-ribosomal RNA processing and mediates N7-methylation of G1639 in human 18S rRNA. RNA 21, 180–187 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Malbec, L. et al. Dynamic methylome of internal mRNA N7-methylguanosine and its regulatory role in translation. Cell Res. 29, 927–941 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Cartlidge, R. A. et al. The tRNA methylase METTL1 is phosphorylated and inactivated by PKB and RSK in vitro and in cells. EMBO J. 24, 1696–1705 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Ping, Y. et al. Identifying core gene modules in glioblastoma based on multilayer factor-mediated dysfunctional regulatory networks through integrating multi-dimensional genomic data. Nucleic Acids Res. 43, 1997–2007 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Tian, Q.-H. et al. METTL1 overexpression is correlated with poor prognosis and promotes hepatocellular carcinoma via PTEN. J. Mol. Med. 97, 1535–1545 (2019).

    Article  CAS  PubMed  Google Scholar 

  106. Okamoto, M. et al. tRNA modifying enzymes, NSUN2 and METTL1, determine sensitivity to 5-fluorouracil in HeLa cells. PLoS Genet. 10, e1004639 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Gustavsson, M. & Ronne, H. Evidence that tRNA modifying enzymes are important in vivo targets for 5-fluorouracil in yeast. RNA 14, 666–674 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Tsukamoto, T., Shibagaki, Y., Niikura, Y. & Mizumoto, K. Cloning and characterization of three human cDNAs encoding mRNA (guanine-7-)-methyltransferase, an mRNA cap methylase. Biochem. Biophys. Res. Commun. 251, 27–34 (1998).

    Article  CAS  PubMed  Google Scholar 

  109. Posternak, V., Ung, M. H., Cheng, C. & Cole, M. D. MYC mediates mRNA cap methylation of canonical Wnt/β-catenin signaling transcripts by recruiting CDK7 and RNA methyltransferase. Mol. Cancer Res. 15, 213–224 (2017).

    Article  CAS  PubMed  Google Scholar 

  110. Franke, J., Gehlen, J. & Ehrenhofer-Murray, A. E. Hypermethylation of yeast telomerase RNA by the snRNA and snoRNA methyltransferase Tgs1. J. Cell Sci. 121, 3553–3560 (2008).

    Article  CAS  PubMed  Google Scholar 

  111. 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 

  112. Singh, R. & Reddy, R. Gamma-monomethyl phosphate: a cap structure in spliceosomal U6 small nuclear RNA. Proc. Natl Acad. Sci. USA 86, 8280–8283 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. 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 

  114. Martinez, A. et al. Human BCDIN3D monomethylates cytoplasmic histidine transfer RNA. Nucleic Acids Res. 45, 5423–5436 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Davis, F. F. & Allen, F. W. Ribonucleic acids from yeast which contain a fifth nucleotide. J. Biol. Chem. 227, 907–915 (1957).

    CAS  PubMed  Google Scholar 

  116. Penzo, M., Guerrieri, A. N., Zacchini, F., Treré, D. & Montanaro, L. RNA pseudouridylation in physiology and medicine: for better and for worse. Genes 8, E301 (2017).

    Article  PubMed  CAS  Google Scholar 

  117. 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 

  118. 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 

  119. Li, X. et al. Chemical pulldown reveals dynamic pseudouridylation of the mammalian transcriptome. Nat. Chem. Biol. 11, 592–597 (2015). This article describes the transcriptome-wide mapping of pseudouridines in human cells.

    Article  CAS  PubMed  Google Scholar 

  120. Penzo, M. & Montanaro, L. Turning uridines around: role of rRNA pseudouridylation in ribosome biogenesis and ribosomal function. Biomolecules 8, E38 (2018).

    Article  PubMed  CAS  Google Scholar 

  121. Bohnsack, M. T. & Sloan, K. E. Modifications in small nuclear RNAs and their roles in spliceosome assembly and function. Biol. Chem. 399, 1265–1276 (2018).

    Article  CAS  PubMed  Google Scholar 

  122. Chen, J.-L., Opperman, K. K. & Greider, C. W. A critical stem-loop structure in the CR4-CR5 domain of mammalian telomerase RNA. Nucleic Acids Res. 30, 592–597 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Carlile, T. M. et al. mRNA structure determines modification by pseudouridine synthase 1. Nat. Chem. Biol. 15, 966–974 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Zhao, X. et al. Regulation of nuclear receptor activity by a pseudouridine synthase through posttranscriptional modification of steroid receptor RNA activator. Mol. Cell 15, 549–558 (2004).

    Article  CAS  PubMed  Google Scholar 

  125. Jana, S., Hsieh, A. C. & Gupta, R. Reciprocal amplification of caspase-3 activity by nuclear export of a putative human RNA-modifying protein, PUS10 during TRAIL-induced apoptosis. Cell Death Dis. 8, e3093–e3093 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Heiss, N. S. et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat. Genet. 19, 32–38 (1998).

    Article  CAS  PubMed  Google Scholar 

  127. Ruggero, D. et al. Dyskeratosis congenita and cancer in mice deficient in ribosomal RNA modification. Science 299, 259–262 (2003). This study characterizes DKC1 as a tumour suppressor in a mouse model of dyskeratosis congenita, showing that the oncogenic effect of DKC1 loss-of-function mutations is caused by a lack of pseudouridylation of rRNA.

    Article  CAS  PubMed  Google Scholar 

  128. Montanaro, L. et al. Novel dyskerin-mediated mechanism of p53 inactivation through defective mRNA translation. Cancer Res. 70, 4767–4777 (2010).

    Article  CAS  PubMed  Google Scholar 

  129. Sieron, P. et al. DKC1 overexpression associated with prostate cancer progression. Br. J. Cancer 101, 1410 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Penzo, M. et al. Dyskerin and TERC expression may condition survival in lung cancer patients. Oncotarget 6, 21755–21760 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Menezes, M. R., Balzeau, J. & Hagan, J. P. 3′ RNA uridylation in epitranscriptomics, gene regulation, and disease. Front. Mol. Biosci. 5, 61 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Martin, G. & Keller, W. RNA-specific ribonucleotidyl transferases. RNA 13, 1834–1849 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yu, C. et al. Star-PAP, a poly(A) polymerase, functions as a tumor suppressor in an orthotopic human breast cancer model. Cell Death Dis. 8, e2582–e2582 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Zhu, D., Lou, Y., He, Z. & Ji, M. Nucleotidyl transferase TUT1 inhibits lipogenesis in osteosarcoma cells through regulation of microRNA-24 and microRNA-29a. Tumor Biol. 35, 11829–11835 (2014).

    Article  CAS  Google Scholar 

  135. Lim, J. et al. Uridylation by TUT4 and TUT7 marks mRNA for degradation. Cell 159, 1365–1376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Heo, I. et al. Mono-uridylation of pre-microRNA as a key step in the biogenesis of group II let-7 microRNAs. Cell 151, 521–532 (2012).

    Article  CAS  PubMed  Google Scholar 

  137. Thornton, J. E., Chang, H.-M., Piskounova, E. & Gregory, R. I. Lin28-mediated control of let-7 microRNA expression by alternative TUTases Zcchc11 (TUT4) and Zcchc6 (TUT7). RNA 18, 1875–1885 (2012). In this study, the LIN28-dependent uridylation of let-7 mediated by TUT4 and TUT7 and its effect on let-7 expression levels is discovered.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Wei, T. & Zhao, W.-X. The Wnt-catenin pathway represses let-7 microRNA expression through transactivation of Lin28 to augment breast cancer stem cell expansion. Artic. J. Cell Sci. 126, 2877–2889 (2013).

    Article  CAS  Google Scholar 

  139. Alajez, N. M. et al. Lin28b promotes head and neck cancer progression via modulation of the insulin-like growth factor survival pathway. Oncotarget 3, 1641–1652 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  140. Balzeau, J., Menezes, M. R., Cao, S. & Hagan, J. P. The LIN28/let-7 pathway in cancer. Front. Genet. 8, 31 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Lin, S. & Gregory, R. I. Identification of small molecule inhibitors of Zcchc11 TUTase activity. RNA Biol. 12, 792–800 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  142. Chang, H.-M., Triboulet, R., Thornton, J. E. & Gregory, R. I. A role for the Perlman syndrome exonuclease Dis3l2 in the Lin28-let-7 pathway. Nature 497, 244–248 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Astuti, D. et al. Germline mutations in DIS3L2 cause the Perlman syndrome of overgrowth and Wilms tumor susceptibility. Nat. Genet. 44, 277–284 (2012).

    Article  CAS  PubMed  Google Scholar 

  144. Ustianenko, D. et al. Mammalian DIS3L2 exoribonuclease targets the uridylated precursors of let-7 miRNAs. RNA 19, 1632–1638 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Hunter, R. W. et al. Loss of Dis3l2 partially phenocopies Perlman syndrome in mice and results in up-regulation of Igf2 in nephron progenitor cells. Genes. Dev. 32, 903–908 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Chen, S. et al. Apolipoprotein B-48 is the product of a messenger RNA with an organ-specific in-frame stop codon. Science 238, 363–366 (1987).

    Article  CAS  PubMed  Google Scholar 

  147. Powell, L. M. et al. A novel form of tissue-specific RNA processing produces apolipoprotein-B48 in intestine. Cell 50, 831–840 (1987).

    Article  CAS  PubMed  Google Scholar 

  148. Rebagliati, M. R. & Melton, D. A. Antisense RNA injections in fertilized frog eggs reveal an RNA duplex unwinding activity. Cell 48, 599–605 (1987).

    Article  CAS  PubMed  Google Scholar 

  149. Bass, B. L. & Weintraub, H. An unwinding activity that covalently modifies its double-stranded RNA substrate. Cell 55, 1089–1098 (1988).

    Article  CAS  PubMed  Google Scholar 

  150. Bass, B. L. & Weintraub, H. A developmentally regulated activity that unwinds RNA duplexes. Cell 48, 607–613 (1987).

    Article  CAS  PubMed  Google Scholar 

  151. Nishikura, K. A-to-I editing of coding and non-coding RNAs by ADARs. Nat. Rev. Mol. Cell Biol. 17, 83–96 (2016).

    Article  CAS  PubMed  Google Scholar 

  152. Herbert, A. ADAR and immune silencing in cancer. Trends Cancer 5, 272–282 (2019).

    Article  CAS  PubMed  Google Scholar 

  153. Chung, H. et al. Human ADAR1 prevents endogenous RNA from triggering translational shutdown. Cell 172, 811–824.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Ishizuka, J. J. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 565, 43–48 (2019). In this study, ADAR1 depletion is shown to sensitize cells to immunotherapy and to prevent resistance, indicating that ADAR1 inhibition is a viable strategy to increase the efficacy of immunotherapies.

    Article  CAS  PubMed  Google Scholar 

  155. Weiss, S. A., Wolchok, J. D. & Sznol, M. Immunotherapy of melanoma: facts and hopes. Clin. Cancer Res. 25, 5191–5201 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Doroshow, D. B. et al. Immunotherapy in non-small cell lung cancer: facts and hopes. Clin. Cancer Res. 25, 4592–4602 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  157. Adams, S. et al. Current landscape of immunotherapy in breast cancer: a review. JAMA Oncol. 5, 1205–1214 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Chen, L. et al. Recoding RNA editing of AZIN1 predisposes to hepatocellular carcinoma. Nat. Med. 19, 209–216 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Okugawa, Y. et al. Enhanced AZIN1 RNA editing and overexpression of its regulatory enzyme ADAR1 are important prognostic biomarkers in gastric cancer. J. Transl Med. 16, 366 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Shigeyasu, K. et al. AZIN1 RNA editing confers cancer stemness and enhances oncogenic potential in colorectal cancer. JCI Insight 3, 99976 (2018).

    Article  PubMed  Google Scholar 

  161. Chen, W. et al. A-to-I RNA editing of BLCAP lost the inhibition to STAT3 activation in cervical cancer. Oncotarget 8, 39417–39429 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  162. Lazzari, E. et al. Alu-dependent RNA editing of GLI1 promotes malignant regeneration in multiple myeloma. Nat. Commun. 8, 1922 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  163. Amin, E. M. et al. The RNA-editing enzyme ADAR promotes lung adenocarcinoma migration and invasion by stabilizing FAK. Sci. Signal. 10, eaah3941 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. 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 

  165. Shoshan, E. et al. Reduced adenosine-to-inosine miR-455-5p editing promotes melanoma growth and metastasis. Nat. Cell Biol. 17, 311–321 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Zhang, M. et al. RNA editing derived epitopes function as cancer antigens to elicit immune responses. Nat. Commun. 9, 3919 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Galeano, F. et al. ADAR2-editing activity inhibits glioblastoma growth through the modulation of the CDC14B/Skp2/p21/p27 axis. Oncogene 32, 998–1009 (2013).

    Article  CAS  PubMed  Google Scholar 

  168. Tomaselli, S. et al. Modulation of microRNA editing, expression and processing by ADAR2 deaminase in glioblastoma. Genome Biol. 16, 5 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Wang, C. et al. Characterizing the role of PCDH9 in the regulation of glioma cell apoptosis and invasion. J. Mol. Neurosci. 52, 250–260 (2014).

    Article  CAS  PubMed  Google Scholar 

  170. Cesarini, V. et al. ADAR2/miR-589-3p axis controls glioblastoma cell migration/invasion. Nucleic Acids Res. 46, 2045–2059 (2018).

    Article  CAS  PubMed  Google Scholar 

  171. Kodama, T. et al. ADAM12 is selectively overexpressed in human glioblastomas and is associated with glioblastoma cell proliferation and shedding of heparin-binding epidermal growth factor. Am. J. Pathol. 165, 1743–1753 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Zhang, Y. et al. ADAR3 expression is an independent prognostic factor in lower-grade diffuse gliomas and positively correlated with the editing level of GRIA2Q607R. Cancer Cell Int. 18, 196 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Fu, L. et al. RNA editing of SLC22A3 drives early tumor invasion and metastasis in familial esophageal cancer. Proc. Natl Acad. Sci. USA 114, E4631–E4640 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Chen, Y.-B. et al. ADAR2 functions as a tumor suppressor via editing IGFBP7 in esophageal squamous cell carcinoma. Int. J. Oncol. 50, 622–630 (2017).

    Article  CAS  PubMed  Google Scholar 

  175. Deng, W. et al. Trm9-catalyzed tRNA modifications regulate global protein expression by codon-biased translation. PLoS Genet. 11, e1005706 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Nedialkova, D. D. & Leidel, S. A. Optimization of codon translation rates via tRNA modifications maintains proteome integrity. Cell 161, 1606–1618 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Rapino, F., Delaunay, S., Zhou, Z., Chariot, A. & Close, P. tRNA modification: is cancer having a wobble? Trends Cancer 3, 249–252 (2017).

    Article  CAS  PubMed  Google Scholar 

  178. Rapino, F. et al. Codon-specific translation reprogramming promotes resistance to targeted therapy. Nature 558, 605–609 (2018). In this study, it is shown that U34 wobble-modifying enzymes are highly expressed in BRAF V600E-driven melanomas and their inhibition can restore sensitivity to BRAF inhibition in BRAF inhibitor-resistant melanomas.

    Article  CAS  PubMed  Google Scholar 

  179. Delaunay, S. et al. Elp3 links tRNA modification to IRES-dependent translation of LEF1 to sustain metastasis in breast cancer. J. Exp. Med. 213, 2503–2523 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Nguyen, A. et al. Wnt pathway component LEF1 mediates tumor cell invasion and is expressed in human and murine breast cancers lacking ErbB2 (her-2/neu) overexpression. Int. J. Oncol. 27, 949–956 (2005).

    CAS  PubMed  Google Scholar 

  181. Ladang, A. et al. Elp3 drives Wnt-dependent tumor initiation and regeneration in the intestine. J. Exp. Med. 212, 2057–2075 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Schapira, M. Structural chemistry of human RNA methyltransferases. ACS Chem. Biol. 11, 575–582 (2016).

    Article  CAS  PubMed  Google Scholar 

  183. 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 

  184. Pugh, C. S., Borchardt, R. T. & Stone, H. O. Sinefungin, a potent inhibitor of virion mRNA(guanine-7-)-methyltransferase, mRNA(nucleoside-2’-)-methyltransferase, and viral multiplication. J. Biol. Chem. 253, 4075–4077 (1978).

    CAS  PubMed  Google Scholar 

  185. Stein, E. M. et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 131, 2662–2669 (2018).

    Google Scholar 

  186. Wang, L. et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat. Commun. 4, 2035 (2013).

    Article  PubMed  CAS  Google Scholar 

  187. Hashizume, R. et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med. 20, 1394–1396 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Véliz, E. A., Easterwood, L. H. M. & Beal, P. A. Substrate analogues for an RNA-editing adenosine deaminase: mechanistic investigation and inhibitor design. J. Am. Chem. Soc. 125, 10867–10876 (2003).

    Article  PubMed  CAS  Google Scholar 

  189. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  190. Wolpaw, A. J. & Dang, C. V. Exploiting metabolic vulnerabilities of cancer with precision and accuracy. Trends Cell Biol. 28, 201–212 (2018).

    Article  CAS  PubMed  Google Scholar 

  191. Rizos, H. et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin. Cancer Res. 20, 1965–1977 (2014).

    Article  CAS  PubMed  Google Scholar 

  192. Keith, G. Mobilities of modified ribonucleotides on two-dimensional cellulose thin-layer chromatography. Biochimie 77, 142–144 (1995).

    Article  CAS  PubMed  Google Scholar 

  193. Liu, N. et al. Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA 19, 1848–1856 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  195. Suzuki, T., Ueda, H., Okada, S. & Sakurai, M. Transcriptome-wide identification of adenosine-to-inosine editing using the ICE-seq method. Nat. Protoc. 10, 715–732 (2015).

    Article  CAS  PubMed  Google Scholar 

  196. Motorin, Y., Muller, S., Behm-Ansmant, I. & Branlant, C. Identification of modified residues in RNAs by reverse transcription-based methods. Methods Enzymol. 425, 21–53 (2007).

    Article  CAS  PubMed  Google Scholar 

  197. 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 

  198. 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 

  199. Meyer, K. D. DART-seq: an antibody-free method for global m6A detection. Nat. Methods 16, 1275–1280 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Zhang, Z. et al. Single-base mapping of m6A by an antibody-independent method. Sci. Adv. 5, eaax0250 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors apologize to colleagues whose work was not acknowledged owing to space limitations. The authors thank A. Bannister for critically reading the manuscript and for the insightful discussions. Work in the Kouzarides laboratory is supported by grants from Cancer Research UK (grant reference RG17001) and the European Research Council (project number 268569), in addition to benefiting from core support from the Wellcome Trust (Core Grant reference 092096) and Cancer Research UK (grant reference C6946/A14492). I.B. is funded by a Kay Kendall Leukaemia Fund project grant (grant reference RG88664) and Cancer Research UK (grant reference RG86786).

Author information

Authors and Affiliations

Authors

Contributions

I.B. researched data for the article. T.K. contributed substantially to discussion of the content. I.B. and T.K contributed equally to writing the article and to reviewing and editing the manuscript.

Corresponding author

Correspondence to Tony Kouzarides.

Ethics declarations

Competing interests

T.K. is a co-founder of Abcam and Storm Therapeutics, Cambridge, UK. I.B. declares no competing interests.

Additional information

Publisher’s note

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

Glossary

Watson–Crick edge

The side of purine and pyrimidine bases involved in the canonical Watson–Crick base pairing.

Cap

A characteristic structure of all eukaryotic mRNAs that consists of a N7-methylated guanine nucleotide bound to the first nucleotide of the mRNA via a 5′–5′ triphosphate bond.

Primary miRNA

(pri-miRNA). Primary transcript from which one or several microRNAs originate. In many cases the same pri-miRNA can be a primary transcript for mRNA and other non-coding RNAs.

Small nuclear RNA

(snRNA). A class of small non-coding RNA localized in the nucleus of eukaryotic cells as part of ribonucleoprotein complexes mediating mRNA maturation and splicing. They are functionally different from small nucleolar RNAs, which are involved in ribosome biogenesis.

Hypomorphic mutations

Mutations partially impairing the function of the encoded protein.

Enhancer RNA

(eRNA). Short (50–200 nucleotides) non-coding RNAs transcribed from enhancer regions and involved in transcriptional regulation both in cis (adjacent loci) and in trans (distant loci).

Bisulfite sequencing

Technique making possible the mapping of 5-methylcytosine (m5C)-modified nucleotides within DNA or RNA. Cytosine nucleotides are converted into uracil on treatment with bisulfite, while methylated nucleotides are protected from this conversion. Methylation sites can then be detected by standard sequencing methods.

Vault RNA

Vaults, large ribonucleoprotein particles in the cytoplasm isolated from higher eukaryotes, contain a small portion of small untranslated RNAs called vault RNAs. Vault RNA is transcribed by RNA polymerase II and, despite being part of the vault complex, does not have a structural role.

Hoogsteen base pairing

A variation of base pairing in nucleic acids, formed by the interaction of the N7 position (as a hydrogen bond acceptor) and C6 amino group (as a donor) of a purine base, with the Watson–Crick (N3–C4) surface of a pyrimidine base.

G-quadruplex structures

Formed in DNA and RNA molecules rich in guanines where four guanine bases can associate by Hoogsteen pairings to form a guanine tetrad, which in turn can stack on top of each other.

Exonucleases

Enzymes removing single nucleotides from the ends of a DNA or RNA polynucleotide chain.

Small nucleolar RNA

(snoRNA). A non-coding RNA localized in the nucleolus and part of ribonucleoprotein complexes involved in the modification of ribosomal RNA precursors and transfer RNAs.

Precursor miRNA

(pre-miRNA). Hairpin intermediate precursors of microRNAs (miRNAs) produced by the cleavage of primary miRNAs by Drosha ribonuclease III (DROSHA) and microprocessor complex subunit DGCR8.

Exoribonuclease

Enzyme that removes single nucleotides from the ends of an RNA polynucleotide chain.

Wilms tumours

Rare kidney cancers affecting children.

Transposable elements

Genomic elements capable of changing their position and copy numbers within the genome. DNA transposons do not go through an RNA intermediate, while retrotransposons are transcribed into an RNA intermediate and successively retrotranscribed to double-stranded DNA.

Alu repeats

A type of genomic short interspersed element (SINE) representing the most common transposable element in the human genome with more than one million copies. They can influence gene expression and are involved in disease, including cancer.

Seed sequence

Conserved heptameric region of a microRNA, typically positioned at the 5′ end determining target specificity (the binding of the microRNA to its target mRNA).

Wobble position

The third base of a codon (or anticodon) characterized by promiscuous pairings. Modifications of this position on transfer RNAs allow non-Watson–Crick base pairing to occur and allow different codons to encode the same amino acid.

Elongator complex

Multisubunit protein complex associated with RNA polymerase II and involved in 5-methoxycarbonylmethyl (mcm5) and 5-carbamoylmethyl (ncm5) modifications on uridines at wobble positions in transfer RNA and histone acetylation.

Internal ribosome entry site

(IRES). RNA elements that allow internal cap-independent translation initiation. They are common in viral RNAs but are also found in stress-induced transcripts in eukaryotic cells.

Rossmann fold

Structural motif commonly found in proteins that bind to nucleotides.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barbieri, I., Kouzarides, T. Role of RNA modifications in cancer. Nat Rev Cancer 20, 303–322 (2020). https://doi.org/10.1038/s41568-020-0253-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-0253-2

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