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:

RNA in cancer

Abstract

While the processing of mRNA is essential for gene expression, recent findings have highlighted that RNA processing is systematically altered in cancer. Mutations in RNA splicing factor genes and the shortening of 3′ untranslated regions are widely observed. Moreover, evidence is accumulating that other types of RNAs, including circular RNAs, can contribute to tumorigenesis. In this Review, we highlight how altered processing or activity of coding and non-coding RNAs contributes to cancer. We introduce the regulation of gene expression by coding and non-coding RNA and discuss both established roles (microRNAs and long non-coding RNAs) and emerging roles (selective mRNA processing and circular RNAs) for RNAs, highlighting the potential mechanisms by which these RNA subtypes contribute to cancer. The widespread alteration of coding and non-coding RNA demonstrates that altered RNA biogenesis contributes to multiple hallmarks of cancer.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Processing and nuclear export of mRNA.
Fig. 2: Formation and functions of miRNAs.
Fig. 3: lncRNAs and their effects in cancers.
Fig. 4: Formation and functions of circRNAs.
Fig. 5: RNA splicing in cancer.
Fig. 6: Polyadenylation and mRNA export in cancer.

Similar content being viewed by others

References

  1. Bradner, J. E., Hnisz, D. & Young, R. A. Transcriptional addiction in cancer. Cell 168, 629–643 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Cooper, T. A., Wan, L. & Dreyfuss, G. RNA and disease. Cell 136, 777–793 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Matera, A. G. & Wang, Z. A day in the life of the spliceosome. Nat. Rev. Mol. Cell Biol. 15, 108–121 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Bracken, C. P., Scott, H. S. & Goodall, G. J. A network-biology perspective of microRNA function and dysfunction in cancer. Nat. Rev. Genet. 17, 719–732 (2016).

    CAS  PubMed  Google Scholar 

  5. Wickramasinghe, V. O. & Laskey, R. A. Control of mammalian gene expression by selective mRNA export. Nat. Rev. Mol. Cell Biol. 16, 431–442 (2015).

    CAS  PubMed  Google Scholar 

  6. Shi, Y. Mechanistic insights into precursor messenger RNA splicing by the spliceosome. Nat. Rev. Mol. Cell Biol. 18, 655–670 (2017).

    CAS  PubMed  Google Scholar 

  7. Wang, E. et al. Targeting an RNA-binding protein network in acute myeloid leukemia. Cancer Cell 35, 369–384 e367 (2019).

    PubMed  PubMed Central  Google Scholar 

  8. Vo, J. N. et al. The landscape of circular RNA in cancer. Cell 176, 869–881 e813 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Gruber, A. J. & Zavolan, M. Alternative cleavage and polyadenylation in health and disease. Nat. Rev. Genet. 20, 599–614 (2019).

    CAS  PubMed  Google Scholar 

  10. Dvinge, H., Guenthoer, J., Porter, P. L. & Bradley, R. K. RNA components of the spliceosome regulate tissue- and cancer-specific alternative splicing. Genome Res. 29, 1591–1604 (2019). Dvinge et al. (2019) and Shuai et al. (2019) demonstrate that RNA components of the spliceosome are also altered in cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, S. et al. Widespread and functional RNA circularization in localized prostate cancer. Cell 176, 831–843.e22 (2019).

    CAS  PubMed  Google Scholar 

  12. Lee, S. H. et al. Widespread intronic polyadenylation inactivates tumour suppressor genes in leukaemia. Nature 561, 127–131 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Lee, S. C. et al. Synthetic lethal and convergent biological effects of cancer-associated spliceosomal gene mutations. Cancer Cell 34, 225–241.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Climente-Gonzalez, H., Porta-Pardo, E., Godzik, A. & Eyras, E. The functional impact of alternative splicing in cancer. Cell Rep. 20, 2215–2226 (2017).

    CAS  PubMed  Google Scholar 

  15. Masamha, C. P. et al. CFIm25 links alternative polyadenylation to glioblastoma tumour suppression. Nature 510, 412–416 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Quesada, V. et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat. Genet. 44, 47–52 (2012). Quesada et al. (2012), Yoshida et al. (2011), Graubert et al. (2011), Papaemmanuil et al. (2011) and Wang et al. (2011) are the first to show that mutations in genes encoding splicing factors are widespread in haematological malignancies.

    CAS  Google Scholar 

  17. Yoshida, K. et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478, 64–69 (2011).

    CAS  PubMed  Google Scholar 

  18. Graubert, T. A. et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat. Genet. 44, 53–57 (2011).

    PubMed  PubMed Central  Google Scholar 

  19. Dvinge, H., Kim, E., Abdel-Wahab, O. & Bradley, R. K. RNA splicing factors as oncoproteins and tumour suppressors. Nat. Rev. Cancer 16, 413–430 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  21. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. 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).

    PubMed  Google Scholar 

  24. Barbieri, I. & Kouzarides, T. Role of RNA modifications in cancer. Nat. Rev. Cancer 20, 303–322 (2020).

    CAS  PubMed  Google Scholar 

  25. Papaemmanuil, E. et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N. Engl. J. Med. 365, 1384–1395 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, L. et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N. Engl. J. Med. 365, 2497–2506 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Qiu, J. et al. Distinct splicing signatures affect converged pathways in myelodysplastic syndrome patients carrying mutations in different splicing regulators. RNA 22, 1535–1549 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chen, L. et al. The augmented R-loop is a unifying mechanism for myelodysplastic syndromes induced by high-risk splicing factor mutations. Mol. Cell 69, 412–425.e6 (2018). This study suggests that genomic instability caused by augmented R-loops induced by splicing factor gene mutations may directly contribute to compromised normal haematopoiesis in myelodysplastic syndromes.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  30. Eichhorn, S. W. et al. mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues. Mol. Cell 56, 104–115 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Engreitz, J. M., Ollikainen, N. & Guttman, M. Long non-coding RNAs: spatial amplifiers that control nuclear structure and gene expression. Nat. Rev. Mol. Cell Biol. 17, 756–770 (2016).

    CAS  PubMed  Google Scholar 

  32. Calin, G. A. et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl Acad. Sci. USA 99, 15524–15529 (2002). This study reports the first indication of involvement of a miRNA in cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Friedman, R. C., Farh, K. K., Burge, C. B. & Bartel, D. P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19, 92–105 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Gregory, P. A. et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 10, 593–601 (2008).

    CAS  PubMed  Google Scholar 

  35. Cursons, J. et al. Combinatorial targeting by microRNAs co-ordinates post-transcriptional control of EMT. Cell Syst. 7, 77–91.e7 (2018).

    CAS  PubMed  Google Scholar 

  36. Bartel, D. P. Metazoan microRNAs. Cell 173, 20–51 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hatley, M. E. et al. Modulation of K-Ras-dependent lung tumorigenesis by microRNA-21. Cancer Cell 18, 282–293 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Pfeffer, S. R., Yang, C. H. & Pfeffer, L. M. The role of miR-21 in cancer. Drug Dev. Res. 76, 270–277 (2015).

    CAS  PubMed  Google Scholar 

  39. O’Connell, R. M., Chaudhuri, A. A., Rao, D. S. & Baltimore, D. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc. Natl Acad. Sci. USA 106, 7113–7118 (2009).

    PubMed  PubMed Central  Google Scholar 

  40. Garofalo, M. et al. miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell 16, 498–509 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Kim, J. et al. Ablation of miR-10b suppresses oncogene-induced mammary tumorigenesis and metastasis and reactivates tumor-suppressive pathways. Cancer Res. 76, 6424–6435 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wei, Q., Lei, R. & Hu, G. Roles of miR-182 in sensory organ development and cancer. Thorac. Cancer 6, 2–9 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, Y., Choi, P. S., Casey, S. C., Dill, D. L. & Felsher, D. W. MYC through miR-17-92 suppresses specific target genes to maintain survival, autonomous proliferation, and a neoplastic state. Cancer Cell 26, 262–272 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Zindy, F. et al. Role of the miR-17 approximately 92 cluster family in cerebellar and medulloblastoma development. Biol. Open. 3, 597–605 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Lambo, S. et al. The molecular landscape of ETMR at diagnosis and relapse. Nature 576, 274–280 (2019).

    PubMed  PubMed Central  Google Scholar 

  46. Witten, L. W., Cheng, C. J. & Slack, F. J. miR-155 drives oncogenesis by promoting and cooperating with mutations in the c-Kit oncogene. Oncogene 38, 2151–2161 (2019).

    CAS  PubMed  Google Scholar 

  47. Chang, S. et al. Tumor suppressor BRCA1 epigenetically controls oncogenic microRNA-155. Nat. Med. 17, 1275–1282 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Jiang, S. et al. MicroRNA-155 functions as an OncomiR in breast cancer by targeting the suppressor of cytokine signaling 1 gene. Cancer Res. 70, 3119–3127 (2010).

    CAS  PubMed  Google Scholar 

  49. Nair, V. S., Maeda, L. S. & Ioannidis, J. P. Clinical outcome prediction by microRNAs in human cancer: a systematic review. J. Natl Cancer Inst. 104, 528–540 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  51. Gilles, M. E. & Slack, F. J. Let-7 microRNA as a potential therapeutic target with implications for immunotherapy. Expert Opin. Ther. Targets 22, 929–939 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Triboulet, R., Pirouz, M. & Gregory, R. I. A single Let-7 microRNA bypasses LIN28-mediated repression. Cell Rep. 13, 260–266 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Hagan, J. P., Piskounova, E. & Gregory, R. I. Lin28 recruits the TUTase Zcchc11 to inhibit let-7 maturation in mouse embryonic stem cells. Nat. Struct. Mol. Biol. 16, 1021–1025 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Adams, B. D. et al. miR-34a silences c-SRC to attenuate tumor growth in triple-negative breast cancer. Cancer Res. 76, 927–939 (2016).

    CAS  PubMed  Google Scholar 

  55. Jiang, L. & Hermeking, H. miR-34a and miR-34b/c suppress intestinal tumorigenesis. Cancer Res. 77, 2746–2758 (2017).

    CAS  PubMed  Google Scholar 

  56. Cheng, C. Y. et al. miR-34 cooperates with p53 in suppression of prostate cancer by joint regulation of stem cell compartment. Cell Rep. 6, 1000–1007 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Liu, C. et al. The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat. Med. 17, 211–215 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Okada, N. et al. A positive feedback between p53 and miR-34 miRNAs mediates tumor suppression. Genes Dev. 28, 438–450 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Pobezinsky, L. A. & Wells, A. C. Let’s fight cancer: let-7 is a tool to enhance antitumor immune responses. Future Oncol. 14, 1141–1145 (2018).

    CAS  PubMed  Google Scholar 

  60. Baer, C. et al. Suppression of microRNA activity amplifies IFN-gamma-induced macrophage activation and promotes anti-tumour immunity. Nat. Cell Biol. 18, 790–802 (2016).

    CAS  PubMed  Google Scholar 

  61. Rodriguez, A., Griffiths-Jones, S., Ashurst, J. L. & Bradley, A. Identification of mammalian microRNA host genes and transcription units. Genome Res. 14, 1902–1910 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Moutinho, C. & Esteller, M. MicroRNAs and epigenetics. Adv. Cancer Res. 135, 189–220 (2017).

    CAS  PubMed  Google Scholar 

  63. Lim, Y. Y. et al. Epigenetic modulation of the miR-200 family is associated with transition to a breast cancer stem-cell-like state. J. Cell Sci. 126, 2256–2266 (2013).

    CAS  PubMed  Google Scholar 

  64. Davalos, V. et al. Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 31, 2062–2074 (2012).

    CAS  PubMed  Google Scholar 

  65. Toyota, M. et al. Epigenetic silencing of microRNA-34b/c and B-cell translocation gene 4 is associated with CpG island methylation in colorectal cancer. Cancer Res. 68, 4123–4132 (2008).

    CAS  PubMed  Google Scholar 

  66. Lodygin, D. et al. Inactivation of miR-34a by aberrant CpG methylation in multiple types of cancer. Cell Cycle 7, 2591–2600 (2008).

    CAS  PubMed  Google Scholar 

  67. Rupaimoole, R. et al. Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression. Nat. Commun. 5, 5202 (2014).

    CAS  PubMed  Google Scholar 

  68. Golden, R. J. et al. An Argonaute phosphorylation cycle promotes microRNA-mediated silencing. Nature 542, 197–202 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Shen, J. et al. EGFR modulates microRNA maturation in response to hypoxia through phosphorylation of AGO2. Nature 497, 383–387 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Bracken, C. P. et al. A double-negative feedback loop between ZEB1-SIP1 and the microRNA-200 family regulates epithelial-mesenchymal transition. Cancer Res. 68, 7846–7854 (2008).

    CAS  PubMed  Google Scholar 

  71. van den Beucken, T. et al. Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of DICER. Nat. Commun. 5, 5203 (2014).

    PubMed  Google Scholar 

  72. Huang, X. et al. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol. Cell 35, 856–867 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Mathew, L. K. & Simon, M. C. mir-210: a sensor for hypoxic stress during tumorigenesis. Mol. Cell 35, 737–738 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Rupaimoole, R. et al. Hypoxia-upregulated microRNA-630 targets Dicer, leading to increased tumor progression. Oncogene 35, 4312–4320 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Melo, S. A. et al. A genetic defect in exportin-5 traps precursor microRNAs in the nucleus of cancer cells. Cancer Cell 18, 303–315 (2010). Melo et al. (2010) and Sun et al. (2016) demonstrate the oncogenic potential of defective miRNA export from the nucleus.

    CAS  PubMed  Google Scholar 

  76. Sun, H. L. et al. ERK activation globally downregulates miRNAs through phosphorylating exportin-5. Cancer Cell 30, 723–736 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, L. et al. microRNAs exhibit high frequency genomic alterations in human cancer. Proc. Natl Acad. Sci. USA 103, 9136–9141 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Tuck, A. C. & Tollervey, D. A transcriptome-wide atlas of RNP composition reveals diverse classes of mRNAs and lncRNAs. Cell 154, 996–1009 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Gupta, R. A. et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464, 1071–1076 (2010). This is one of the seminal reports demonstrating that lncRNAs can modulate the cancer epigenome.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Huarte, M. et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142, 409–419 (2010). This is a seminal report linking a lncRNA function of the important tumour suppressor p53.

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Dimitrova, N. et al. LincRNA-p21 activates p21 in cis to promote polycomb target gene expression and to enforce the G1/S checkpoint. Mol. Cell 54, 777–790 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Yoon, J. H. et al. LincRNA-p21 suppresses target mRNA translation. Mol. Cell 47, 648–655 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Yang, F., Zhang, H., Mei, Y. & Wu, M. Reciprocal regulation of HIF-1alpha and lincRNA-p21 modulates the Warburg effect. Mol. Cell 53, 88–100 (2014).

    CAS  PubMed  Google Scholar 

  84. Kopp, F. & Mendell, J. T. Functional classification and experimental dissection of long noncoding RNAs. Cell 172, 393–407 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Li, X. & Fu, X. D. Chromatin-associated RNAs as facilitators of functional genomic interactions. Nat. Rev. Genet. 20, 503–519 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Yari, H. et al. LncRNA REG1CP promotes tumorigenesis through an enhancer complex to recruit FANCJ helicase for REG3A transcription. Nat. Commun. 10, 5334 (2019).

    PubMed  PubMed Central  Google Scholar 

  87. Hua, J. T. et al. Risk SNP-mediated promoter-enhancer switching drives prostate cancer through lncRNA PCAT19. Cell 174, 564–575 e518 (2018).

    CAS  PubMed  Google Scholar 

  88. Gao, P. et al. Biology and clinical implications of the 19q13 aggressive prostate cancer susceptibility locus. Cell 174, 576–589.e18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Liu, P. Y. et al. The long noncoding RNA lncNB1 promotes tumorigenesis by interacting with ribosomal protein RPL35. Nat. Commun. 10, 5026 (2019).

    PubMed  PubMed Central  Google Scholar 

  90. Hu, Q. et al. Oncogenic lncRNA downregulates cancer cell antigen presentation and intrinsic tumor suppression. Nat. Immunol. 20, 835–851 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Coe, E. A. et al. The MITF-SOX10 regulated long non-coding RNA DIRC3 is a melanoma tumour suppressor. PLoS Genet. 15, e1008501 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Wang, Y. Q. et al. SATB2-AS1 suppresses colorectal carcinoma aggressiveness by inhibiting SATB2-dependent snail transcription and epithelial-mesenchymal transition. Cancer Res. 79, 3542–3556 (2019).

    CAS  PubMed  Google Scholar 

  93. Shahabi, S. et al. LINC00261 is an epigenetically regulated tumor suppressor essential for activation of the DNA damage response. Cancer Res. 79, 3050–3062 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Saha, S. et al. Long noncoding RNA DRAIC inhibits prostate cancer progression by interacting with IKK to inhibit NF-kappaB activation. Cancer Res. 80, 950–963 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Wang, Y. et al. LncRNA-encoded polypeptide ASRPS inhibits triple-negative breast cancer angiogenesis. J. Exp. Med. 217, e20190950 (2020).

    PubMed  Google Scholar 

  96. Tseng, Y. Y. et al. PVT1 dependence in cancer with MYC copy-number increase. Nature 512, 82–86 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Olivero, C. E. et al. p53 activates the long noncoding RNA Pvt1b to inhibit Myc and suppress tumorigenesis. Mol. Cell 77, 761–774.e8 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Cho, S. W. et al. Promoter of lncRNA gene PVT1 is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Jiang, Y. et al. Co-activation of super-enhancer-driven CCAT1 by TP63 and SOX2 promotes squamous cancer progression. Nat. Commun. 9, 3619 (2018).

    PubMed  PubMed Central  Google Scholar 

  100. Xiang, J. F. et al. Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 24, 513–531 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Cai, Z. et al. RIC-seq for global in situ profiling of RNA-RNA spatial interactions. Nature 582, 432–437 (2020).

    CAS  PubMed  Google Scholar 

  102. Sun, Y. & Ma, L. New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers 11, 216 (2019).

    CAS  PubMed Central  Google Scholar 

  103. Kim, J. et al. Long noncoding RNA MALAT1 suppresses breast cancer metastasis. Nat. Genet. 50, 1705–1715 (2018). This works warns that a widely recognized protumorigenic RNA may have a suppressive role in breast cancers.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Salzman, J., Gawad, C., Wang, P. L., Lacayo, N. & Brown, P. O. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS ONE 7, e30733 (2012). This is a seminal report of the widespread occurrence of circRNAs in many cell types.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Chen, L. L. The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat. Rev. Mol. Cell Biol. 21, 475–490 (2020).

    CAS  PubMed  Google Scholar 

  106. Xiao, M. S., Ai, Y. & Wilusz, J. E. Biogenesis and functions of circular RNAs come into focus. Trends Cell Biol. 30, 226–240 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Li, Z. et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat. Struct. Mol. Biol. 22, 256–264 (2015).

    PubMed  Google Scholar 

  108. Guarnerio, J. et al. Intragenic antagonistic roles of protein and circRNA in tumorigenesis. Cell Res. 29, 628–640 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Conn, S. J. et al. The RNA binding protein quaking regulates formation of circRNAs. Cell 160, 1125–1134 (2015).

    CAS  PubMed  Google Scholar 

  110. Memczak, S. et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 495, 333–338 (2013). Memczak et al. (2013) and Hansen et al.(2013) are two reports that highlight the remarkable titration of miR-7 by an abundant circRNA.

    CAS  PubMed  Google Scholar 

  111. Hansen, T. B. et al. Natural RNA circles function as efficient microRNA sponges. Nature 495, 384–388 (2013).

    CAS  PubMed  Google Scholar 

  112. Barrett, S. P., Parker, K. R., Horn, C., Mata, M. & Salzman, J. ciRS-7 exonic sequence is embedded in a long non-coding RNA locus. PLoS Genet. 13, e1007114 (2017).

    PubMed  PubMed Central  Google Scholar 

  113. Piwecka, M. et al. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 57, eaam8526 (2017).

    Google Scholar 

  114. Hanniford, D. et al. Epigenetic silencing of CDR1as drives IGF2BP3-mediated melanoma invasion and metastasis. Cancer Cell 37, 55–70.e15 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Bosson, A. D., Zamudio, J. R. & Sharp, P. A. Endogenous miRNA and target concentrations determine susceptibility to potential ceRNA competition. Mol. Cell 56, 347–359 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Jens, M. & Rajewsky, N. Competition between target sites of regulators shapes post-transcriptional gene regulation. Nat. Rev. Genet. 16, 113–126 (2015).

    CAS  PubMed  Google Scholar 

  117. Denzler, R. et al. Impact of microRNA levels, target-site complementarity, and cooperativity on competing endogenous RNA-regulated gene expression. Mol. Cell 64, 565–579 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Panda, A. C. et al. Identification of senescence-associated circular RNAs (SAC-RNAs) reveals senescence suppressor CircPVT1. Nucleic Acids Res. 45, 4021–4035 (2017).

    CAS  PubMed  Google Scholar 

  119. Verduci, L. et al. The oncogenic role of circPVT1 in head and neck squamous cell carcinoma is mediated through the mutant p53/YAP/TEAD transcription-competent complex. Genome Biol. 18, 237 (2017).

    PubMed  PubMed Central  Google Scholar 

  120. Li, Q. et al. CircACC1 regulates assembly and activation of AMPK complex under metabolic stress. Cell Metab. 30, 157–173.e7 (2019).

    PubMed  Google Scholar 

  121. Zhang, Y. et al. The biogenesis of nascent circular RNAs. Cell Rep. 15, 611–624 (2016).

    CAS  PubMed  Google Scholar 

  122. Enuka, Y. et al. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res. 44, 1370–1383 (2016).

    CAS  PubMed  Google Scholar 

  123. Bachmayr-Heyda, A. et al. Correlation of circular RNA abundance with proliferation–exemplified with colorectal and ovarian cancer, idiopathic lung fibrosis, and normal human tissues. Sci. Rep. 5, 8057 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Pillman, K. A. et al. miR-200/375 control epithelial plasticity-associated alternative splicing by repressing the RNA-binding protein Quaking. EMBO J 37, e99016 (2018).

    PubMed  PubMed Central  Google Scholar 

  125. Kornblihtt, A. R. et al. Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat. Rev. Mol. Cell Biol. 14, 153–165 (2013).

    CAS  PubMed  Google Scholar 

  126. Nilsen, T. W. & Graveley, B. R. Expansion of the eukaryotic proteome by alternative splicing. Nature 463, 457–463 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Weatheritt, R. J., Sterne-Weiler, T. & Blencowe, B. J. The ribosome-engaged landscape of alternative splicing. Nat. Struct. Mol. Biol. 23, 1117–1123 (2016). Weatheritt et al. (2016) and Liu et al. (2017) are among the first to examine the impact of alternative splicing at the protein level using ribosome profiling and quantitative proteomics.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Liu, Y. et al. Impact of alternative splicing on the human proteome. Cell Rep. 20, 1229–1241 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Wahl, M. C., Will, C. L. & Luhrmann, R. The spliceosome: design principles of a dynamic RNP machine. Cell 136, 701–718 (2009).

    CAS  PubMed  Google Scholar 

  130. Wang, E. T. et al. Alternative isoform regulation in human tissue transcriptomes. Nature 456, 470–476 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Pan, Q., Shai, O., Lee, L. J., Frey, B. J. & Blencowe, B. J. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat. Genet. 40, 1413–1415 (2008).

    CAS  PubMed  Google Scholar 

  132. Reyes, A. & Huber, W. Alternative start and termination sites of transcription drive most transcript isoform differences across human tissues. Nucleic Acids Res. 46, 582–592 (2018).

    CAS  PubMed  Google Scholar 

  133. Viphakone, N. et al. Co-transcriptional loading of RNA export factors shapes the human transcriptome. Mol. Cell 75, 310–323.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Chen, S. et al. The mRNA export receptor NXF1 coordinates transcriptional dynamics, alternative polyadenylation, and mRNA export. Mol. Cell 74, 118–131.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Carey, K. T. & Wickramasinghe, V. O. Regulatory potential of the RNA processing machinery: implications for human disease. Trends Genet. 34, 279–290 (2018).

    CAS  PubMed  Google Scholar 

  136. Wickramasinghe, V. O. et al. Regulation of constitutive and alternative mRNA splicing across the human transcriptome by PRPF8 is determined by 5’ splice site strength. Genome Biol. 16, 201 (2015).

    PubMed  PubMed Central  Google Scholar 

  137. Papasaikas, P., Tejedor, J. R., Vigevani, L. & Valcarcel, J. Functional splicing network reveals extensive regulatory potential of the core spliceosomal machinery. Mol. Cell 57, 7–22 (2015).

    CAS  PubMed  Google Scholar 

  138. Saltzman, A. L., Pan, Q. & Blencowe, B. J. Regulation of alternative splicing by the core spliceosomal machinery. Genes Dev. 25, 373–384 (2011).

    PubMed  PubMed Central  Google Scholar 

  139. Kahles, A. et al. Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer Cell 34, 211–224.e6 (2018).

    CAS  PubMed  Google Scholar 

  140. Harbour, J. W. et al. Recurrent mutations at codon 625 of the splicing factor SF3B1 in uveal melanoma. Nat. Genet. 45, 133–135 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Martin, M. et al. Exome sequencing identifies recurrent somatic mutations in EIF1AX and SF3B1 in uveal melanoma with disomy 3. Nat. Genet. 45, 933–936 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Biankin, A. V. et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491, 399–405 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Stephens, P. J. et al. The landscape of cancer genes and mutational processes in breast cancer. Nature 486, 400–404 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Maguire, S. L. et al. SF3B1 mutations constitute a novel therapeutic target in breast cancer. J. Pathol. 235, 571–580 (2015).

    CAS  PubMed  Google Scholar 

  145. Mian, S. A. et al. SF3B1 mutant MDS-initiating cells may arise from the haematopoietic stem cell compartment. Nat. Commun. 6, 10004 (2015).

    CAS  PubMed  Google Scholar 

  146. Lindsley, R. C. et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 125, 1367–1376 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Shuai, S. et al. The U1 spliceosomal RNA is recurrently mutated in multiple cancers. Nature 574, 712–716 (2019).

    CAS  PubMed  Google Scholar 

  148. Obeng, E. A. et al. Physiologic expression of Sf3b1(K700E) causes impaired erythropoiesis, aberrant splicing, and sensitivity to therapeutic spliceosome modulation. Cancer Cell 30, 404–417 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Shirai, C. L. et al. Mutant U2AF1 expression alters hematopoiesis and pre-mRNA splicing in vivo. Cancer Cell 27, 631–643 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Kim, E. et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell 27, 617–630 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Ernst, T. et al. Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat. Genet. 42, 722–726 (2010).

    CAS  PubMed  Google Scholar 

  152. Nikoloski, G. et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat. Genet. 42, 665–667 (2010).

    CAS  PubMed  Google Scholar 

  153. Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Smith, M. A. et al. U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nat. Cell Biol. 21, 640–650 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Adler, A. S. et al. An integrative analysis of colon cancer identifies an essential function for PRPF6 in tumor growth. Genes Dev. 28, 1068–1084 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Venkitaraman, A. R. Cancer suppression by the chromosome custodians, BRCA1 and BRCA2. Science 343, 1470–1475 (2014).

    CAS  PubMed  Google Scholar 

  157. Wang, L. et al. Transcriptomic characterization of SF3B1 mutation reveals its pleiotropic effects in chronic lymphocytic leukemia. Cancer Cell 30, 750–763 (2016).

    PubMed  PubMed Central  Google Scholar 

  158. Park, S. M. et al. U2AF35(S34F) promotes transformation by directing aberrant ATG7 Pre-mRNA 3’ end formation. Mol. Cell 62, 479–490 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Nguyen, H. D. et al. Spliceosome mutations induce R loop-associated sensitivity to ATR inhibition in myelodysplastic syndromes. Cancer Res. 78, 5363–5374 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Wickramasinghe, V. O. & Venkitaraman, A. R. RNA processing and genome stability: cause and consequence. Mol. Cell 61, 496–505 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Sollier, J. & Cimprich, K. A. Breaking bad: R-loops and genome integrity. Trends Cell Biol. 25, 514–522 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Gruber, A. J. et al. Discovery of physiological and cancer-related regulators of 3’ UTR processing with KAPAC. Genome Biol. 19, 44 (2018).

    PubMed  PubMed Central  Google Scholar 

  163. Mayr, C. & Bartel, D. P. Widespread shortening of 3’UTRs by alternative cleavage and polyadenylation activates oncogenes in cancer cells. Cell 138, 673–684 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Singh, P. et al. Global changes in processing of mRNA 3’ untranslated regions characterize clinically distinct cancer subtypes. Cancer Res. 69, 9422–9430 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Xia, Z. et al. Dynamic analyses of alternative polyadenylation from RNA-seq reveal a 3’-UTR landscape across seven tumour types. Nat. Commun. 5, 5274 (2014).

    CAS  PubMed  Google Scholar 

  166. Mapendano, C. K., Lykke-Andersen, S., Kjems, J., Bertrand, E. & Jensen, T. H. Crosstalk between mRNA 3’ end processing and transcription initiation. Mol. Cell 40, 410–422 (2010).

    CAS  PubMed  Google Scholar 

  167. Millevoi, S. et al. A physical and functional link between splicing factors promotes pre-mRNA 3’ end processing. Nucleic Acids Res. 37, 4672–4683 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Siddiqui, N. & Borden, K. L. mRNA export and cancer. Wiley Interdiscip. Rev. RNA 3, 13–25 (2012).

    CAS  PubMed  Google Scholar 

  169. Wickramasinghe, V. O. et al. Human inositol polyphosphate multikinase regulates transcript-selective nuclear mRNA export to preserve genome integrity. Mol. Cell 51, 737–750 (2013).

    CAS  PubMed  Google Scholar 

  170. Culjkovic-Kraljacic, B., Baguet, A., Volpon, L., Amri, A. & Borden, K. L. The oncogene eIF4E reprograms the nuclear pore complex to promote mRNA export and oncogenic transformation. Cell Rep. 2, 207–215 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Assouline, S. et al. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin. Blood 114, 257–260 (2009).

    CAS  PubMed  Google Scholar 

  172. Culjkovic, B. et al. The eIF4E RNA regulon promotes the Akt signaling pathway. J. Cell Biol. 181, 51–63 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Dominguez-Sanchez, M. S., Saez, C., Japon, M. A., Aguilera, A. & Luna, R. Differential expression of THOC1 and ALY mRNP biogenesis/export factors in human cancers. BMC Cancer 11, 77 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Guo, S. et al. Linking transcriptional elongation and messenger RNA export to metastatic breast cancers. Cancer Res. 65, 3011–3016 (2005).

    CAS  PubMed  Google Scholar 

  175. Saito, Y. et al. ALY as a potential contributor to metastasis in human oral squamous cell carcinoma. J. Cancer Res. Clin. Oncol. 139, 585–594 (2013).

    CAS  PubMed  Google Scholar 

  176. Viphakone, N. et al. Luzp4 defines a new mRNA export pathway in cancer cells. Nucleic Acids Res. 43, 2353–2366 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Wang, K. et al. A U2-snRNP-independent role of SF3b in promoting mRNA export. Proc. Natl Acad. Sci. USA 116, 7837–7846 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Agrawal, A. A., Yu, L., Smith, P. G. & Buonamici, S. Targeting splicing abnormalities in cancer. Curr. Opin. Genet. Dev. 48, 67–74 (2018).

    CAS  PubMed  Google Scholar 

  179. Lee, S. C. & Abdel-Wahab, O. Therapeutic targeting of splicing in cancer. Nat. Med. 22, 976–986 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Folco, E. G., Coil, K. E. & Reed, R. The anti-tumor drug E7107 reveals an essential role for SF3b in remodeling U2 snRNP to expose the branch point-binding region. Genes Dev. 25, 440–444 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Eskens, F. A. et al. Phase I pharmacokinetic and pharmacodynamic study of the first-in-class spliceosome inhibitor E7107 in patients with advanced solid tumors. Clin. Cancer Res. 19, 6296–6304 (2013).

    CAS  PubMed  Google Scholar 

  182. Seiler, M. et al. H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers. Nat. Med. 24, 497–504 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Uehara, T. et al. Selective degradation of splicing factor CAPERalpha by anticancer sulfonamides. Nat. Chem. Biol. 13, 675–680 (2017).

    CAS  PubMed  Google Scholar 

  184. Han, T. et al. Anticancer sulfonamides target splicing by inducing RBM39 degradation via recruitment to DCAF15. Science 356, eaal3755 (2017).

    PubMed  Google Scholar 

  185. Hanna, J., Hossain, G. S. & Kocerha, J. The potential for microRNA therapeutics and clinical research. Front. Genet. 10, 478 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Rupaimoole, R. & Slack, F. J. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 16, 203–222 (2017).

    CAS  PubMed  Google Scholar 

  187. van Zandwijk, N. et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 18, 1386–1396 (2017). This reports a promising initial phase I trial of a delivery system for a miRNA as therapy for mesothelioma.

    PubMed  Google Scholar 

  188. Will, C. L. & Luhrmann, R. Spliceosome structure and function. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a003707 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  189. Shi, Y. et al. Molecular architecture of the human pre-mRNA 3’ processing complex. Mol. Cell 33, 365–376 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Christofori, G. & Keller, W. 3’ cleavage and polyadenylation of mRNA precursors in vitro requires a poly(A) polymerase, a cleavage factor, and a snRNP. Cell 54, 875–889 (1988).

    CAS  PubMed  Google Scholar 

  191. Cheng, H. et al. Human mRNA export machinery recruited to the 5’ end of mRNA. Cell 127, 1389–1400 (2006).

    CAS  PubMed  Google Scholar 

  192. Strasser, K. et al. TREX is a conserved complex coupling transcription with messenger RNA export. Nature 417, 304–308 (2002).

    PubMed  Google Scholar 

  193. Wickramasinghe, V. O. et al. Selective nuclear export of specific classes of mRNA from mammalian nuclei is promoted by GANP. Nucleic Acids Res. 42, 5059–5071 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Jani, D. et al. Functional and structural characterization of the mammalian TREX-2 complex that links transcription with nuclear messenger RNA export. Nucleic Acids Res. 40, 4562–4573 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Ellisdon, A. M., Dimitrova, L., Hurt, E. & Stewart, M. Structural basis for the assembly and nucleic acid binding of the TREX-2 transcription-export complex. Nat. Struct. Mol. Biol. 19, 328–336 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Vihandha, O. et al. mRNA export from mammalian cell nuclei is dependent on GANP. Curr. Biol. 20, 25–31 (2010).

    Google Scholar 

  197. Bjork, P., Persson, J. O. & Wieslander, L. Intranuclear binding in space and time of exon junction complex and NXF1 to premRNPs/mRNPs in vivo. J. Cell Biol. 211, 63–75 (2015).

    PubMed  PubMed Central  Google Scholar 

  198. Viphakone, N. et al. TREX exposes the RNA-binding domain of Nxf1 to enable mRNA export. Nat. Commun. 3, 1006 (2012).

    PubMed  Google Scholar 

  199. Zhang, X. O. et al. Complementary sequence-mediated exon circularization. Cell 159, 134–147 (2014).

    CAS  PubMed  Google Scholar 

  200. Liang, D. & Wilusz, J. E. Short intronic repeat sequences facilitate circular RNA production. Genes Dev. 28, 2233–2247 (2014).

    PubMed  PubMed Central  Google Scholar 

  201. Jeck, W. R. et al. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA 19, 141–157 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Thomson, D. W. et al. Assessing the gene regulatory properties of Argonaute-bound small RNAs of diverse genomic origin. Nucleic Acids Res. 43, 470–481 (2015).

    CAS  PubMed  Google Scholar 

  203. Boskovic, A., Bing, X. Y., Kaymak, E. & Rando, O. J. Control of noncoding RNA production and histone levels by a 5’ tRNA fragment. Genes Dev. 34, 118–131 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Sobala, A. & Hutvagner, G. Small RNAs derived from the 5’ end of tRNA can inhibit protein translation in human cells. RNA Biol. 10, 553–563 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Balatti, V., Pekarsky, Y. & Croce, C. M. Role of the tRNA-derived small RNAs in cancer: new potential biomarkers and target for therapy. Adv. Cancer Res. 135, 173–187 (2017).

    CAS  PubMed  Google Scholar 

  206. Maute, R. L. et al. tRNA-derived microRNA modulates proliferation and the DNA damage response and is down-regulated in B cell lymphoma. Proc. Natl Acad. Sci. USA 110, 1404–1409 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Pekarsky, Y. et al. Dysregulation of a family of short noncoding RNAs, tsRNAs, in human cancer. Proc. Natl Acad. Sci. USA 113, 5071–5076 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Goodarzi, H. et al. Endogenous tRNA-derived fragments suppress breast cancer progression via YBX1 displacement. Cell 161, 790–802 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Santos, M., Fidalgo, A., Varanda, A. S., Oliveira, C. & Santos, M. A. S. tRNA deregulation and its consequences in cancer. Trends Mol. Med. 25, 853–865 (2019).

    CAS  PubMed  Google Scholar 

  210. Goodarzi, H. et al. Modulated expression of specific tRNAs drives gene expression and cancer progression. Cell 165, 1416–1427 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  212. 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).

    CAS  PubMed  Google Scholar 

  213. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Wang, X. et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  217. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Roundtree, I. A. et al. YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. eLife 6, e31311 (2017).

    PubMed  PubMed Central  Google Scholar 

  219. Xiao, W. et al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol. Cell 61, 507–519 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  221. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Weng, H. et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m(6)A modification. Cell Stem Cell 22, 191–205.e9 (2018).

    CAS  PubMed  Google Scholar 

  224. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank W. Filipowicz and P. Gregory for critically reading the manuscript. They gratefully acknowledge funding from the Australian National Health and Medical Research Council (GNT1127745 to V.O.W, and GNT1126711 and research fellowship GNT1118170 to GJG). V.O.W. is supported by an innovation fellowship from veski and a mid-career fellowship from the Victorian Cancer Agency.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Gregory J. Goodall or Vihandha O. Wickramasinghe.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks G. Calin, L.-L. Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Related links

PubMed: https://pubmed.ncbi.nlm.nih.gov/

Glossary

Spliceosome

Executes RNA splicing reactions and is composed of a number of distinct small nuclear ribonucleoprotein complexes containing proteins that are stably bound to uridine-rich small nuclear RNA molecules. It is dynamically remodelled during the splicing reaction.

Genomic instability

An underlying hallmark of cancers is their genomic instability, which is associated with a greater tendency to accumulate DNA damage. Genomic instability and mutation are an enabling characteristic driving tumour progression.

TREX and TREX2

Export of mRNA from the nucleus to the cytoplasm requires the transcription–export complexes TREX and TREX2, which recognize mRNAs during their biogenesis. Cargo mRNAs are transferred to the transport factors NXF1 and p15 for transit through nuclear pores. Expression of TREX components such as ALYREF and THOC1 and the TREX2 component GANP is altered in cancer.

Epithelial-to-mesenchymal transition

(EMT). A change in cell phenotype that normally occurs during embryogenesis as part of tissue remodelling, and during wound healing, but can also be recapitulated by cancers. EMT confers on cancer cells the ability to invade and metastasize, and also contributes to chemoresistance.

M1 macrophage

A proinflammatory macrophage that secretes inflammatory cytokines such as IL-1, IL-6, IL-12 and tumour necrosis factor (TNF).

Genotoxic stress

Cellular exposure to DNA-damaging agents, which elicits biochemical responses to repair DNA damage, enhancing cell survival or leading to cell death.

Nucleators

Molecules that nucleate complex formation by binding to multiple binding partners.

3′ end processing complex

Interacts with sequence motifs in nascent RNAs to determine the site of cleavage and polyadenylation and comprising a number of subcomplexes (cleavage and polyadenylation factor, cleavage factors 1 and 2 and cleavage stimulation factor). Following endonucleolytic cleavage, the poly(A) tail is synthesized on the 3′ end of the cleaved product by poly(A) polymerase.

Nuclear pore complexes

Proteins and RNA molecules are transported between the nucleus and cytoplasm through nuclear pore complexes. They are ~110-MDa macromolecular complexes with an overall octagonal symmetry and consist of several copies of ~30 proteins called ‘nucleoporins’. mRNA export factors such as NXF1 directly interact with nucleoporins, which have regions rich in phenylalanine–glycine repeats (FG nucleoporins) that line the inner part of the channel and help to move the cargo through the pore.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goodall, G.J., Wickramasinghe, V.O. RNA in cancer. Nat Rev Cancer 21, 22–36 (2021). https://doi.org/10.1038/s41568-020-00306-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-00306-0

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