Skip to main content

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

  • Perspective
  • Published:

Aging through an epitranscriptomic lens

Abstract

The mechanistic causes of aging, the time-related decline in function and good health that leads to increased mortality, remain poorly understood. Here we propose that age-dependent alteration of the epitranscriptome, encompassing more than 150 chemically distinct post-transcriptional modifications or editing events, warrants exploration as an important modulator of aging. The epitranscriptome is a potent regulator of RNA function, diverse cellular processes and tissue regenerative capacity. To date, only a few studies link alterations in the epitranscriptome to molecular and physiological changes during aging; however, epitranscriptome dysfunction is associated with and underlies several age-associated pathologies, including cancer and neurodegenerative, cardiovascular and autoimmune diseases. For example, changes in RNA modifications (such as N6-methyladenosine and inosine) impact cardiac physiology and are linked to cardiac fibrosis. Although an uncharted research focus, mapping epitranscriptome alterations in the context of aging may elucidate novel predictors of both health and lifespan, and may identify therapeutic targets for attenuating aging and abrogating age-related diseases.

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: Main classes of RNA subject to post-transcriptional editing and modifications.
Fig. 2: Contribution of altered RNA modifications to the various hallmarks of aging.
Fig. 3: Molecular functions of RNA modifications.
Fig. 4: Role of RNA modifications in healthy and pathological tissue regeneration.

Similar content being viewed by others

References

  1. Niccoli, T. & Partridge, L. Ageing as a risk factor for disease. Curr. Biol. 22, R741–R752 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013). Landmark review article describing the hallmarks of aging—great introduction for newcomers to the aging field.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Wilkinson, G. S. & South, J. M. Life history, ecology and longevity in bats. Aging Cell 1, 124–131 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Buffenstein, R. et al. Probing pedomorphy and prolonged lifespan in naked mole-rats and dwarf mice. Physiology 35, 96–111 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Benayoun, B. A., Pollina, E. A. & Brunet, A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat. Rev. Mol. Cell Biol. 16, 593–610 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Gilbert, W. V., Bell, T. A. & Schaening, C. Messenger RNA modifications: form, distribution, and function. Science 352, 1408–1412 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. McCown, P. J. et al. Naturally occurring modified ribonucleosides. Wiley Interdiscip. Rev. RNA 11, e1595 (2020). Comprehensive review article on RNA modifications—great introduction for newcomers to the RNA modification field.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wiener, D. & Schwartz, S. The epitranscriptome beyond m6A. Nat. Rev. Genet. 22, 119–131 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dai, Q. et al. Nm-seq maps 2′-O-methylation sites in human mRNA with base precision. Nat. Methods 14, 695–698 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schwartz, S. et al. Transcriptome-wide mapping reveals widespread dynamic-regulated pseudouridylation of ncRNA and mRNA. Cell 159, 148–162 (2014). In the manuscript, pseudouridine modifications were profiled transcriptome-wide and found on human mRNA and ncRNAs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bazak, L. et al. A-to-I RNA editing occurs at over a hundred million genomic sites, located in a majority of human genes. Genome Res. 24, 365–376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Littlefield, J. W. & Dunn, D. B. Natural occurrence of thymine and three methylated adenine bases in several ribonucleic acids. Nature 181, 254–255 (1958).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. 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 

  19. Su, D. et al. Quantitative analysis of ribonucleoside modifications in tRNA by HPLC-coupled mass spectrometry. Nat. Protoc. 9, 828–841 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Boccaletto, P. et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 46, D303–D307 (2018). This article describes a database containing all known RNA modification pathways.

    Article  CAS  PubMed  Google Scholar 

  21. Nachtergaele, S. & He, C. Chemical modifications in the life of an mRNA transcript. Annu. Rev. Genet. 52, 349–372 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Shulman, Z. & Stern-Ginossar, N. The RNA modification N6-methyladenosine as a novel regulator of the immune system. Nat. Immunol. 21, 501–512 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Jonkhout, N. et al. The RNA modification landscape in human disease. RNA 23, 1754–1769 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Stojkovic, V. & Fujimori, D. G. Mutations in RNA methylating enzymes in disease. Curr. Opin. Chem. Biol. 41, 20–27 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  26. Abe, M. et al. Impact of age-associated increase in 2′-O-methylation of miRNAs on aging and neurodegeneration in Drosophila. Genes Dev. 28, 44–57 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Min, K. W. et al. Profiling of m6A RNA modifications identified an age-associated regulation of AGO2 mRNA stability. Aging Cell 17, e12753 (2018). This paper maps and quantifies m6A modifications in peripheral blood mononuclear cells from young and old individuals and reveals a global decrease in m6A RNA modifications with increasing age.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Nicholas, A. et al. Age-related gene-specific changes of A-to-I mRNA editing in the human brain. Mech. Ageing Dev. 131, 445–447 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dahlberg, A. E. The functional role of ribosomal RNA in protein synthesis. Cell 57, 525–529 (1989).

    Article  CAS  PubMed  Google Scholar 

  31. Kirchner, S. & Ignatova, Z. Emerging roles of tRNA in adaptive translation, signalling dynamics and disease. Nat. Rev. Genet. 16, 98–112 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Fabian, M. R., Sonenberg, N. & Filipowicz, W. Regulation of mRNA translation and stability by microRNAs. Annu. Rev. Biochem. 79, 351–379 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Kim, N. K., Theimer, C. A., Mitchell, J. R., Collins, K. & Feigon, J. Effect of pseudouridylation on the structure and activity of the catalytically essential P6.1 hairpin in human telomerase RNA. Nucleic Acids Res. 38, 6746–6756 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tang, H. et al. HuR regulates telomerase activity through TERC methylation. Nat. Commun. 9, 2213 (2018). In the manuscript, the authors identify m5C modifications in TERC and demonstrate that m5C modifications impact telomerase activity.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Zaccara, S., Ries, R. J. & Jaffrey, S. R. Reading, writing and erasing mRNA methylation. Nat. Rev. Mol. Cell Biol. 20, 608–624 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Wilusz, C. J., Wormington, M. & Peltz, S. W. The cap-to-tail guide to mRNA turnover. Nat. Rev. Mol. Cell Biol. 2, 237–246 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  38. Sommer, S., Lavi, U. & Darnell, J. E. Jr The absolute frequency of labeled N-6-methyladenosine in HeLa cell messenger RNA decreases with label time. J. Mol. Biol. 124, 487–499 (1978).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  40. Zhang, C. et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature 549, 273–276 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Zaccara, S. & Jaffrey, S. R. A unified model for the function of YTHDF proteins in regulating m6A-modified mRNA. Cell https://doi.org/10.1016/j.cell.2020.05.012 (2020).

  42. Lasman, L. et al. Context-dependent functional compensation between Ythdf m6A reader proteins. Genes Dev. 34, 1373–1391 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 

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

    CAS  PubMed  Google Scholar 

  45. 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 

  46. Tuorto, F. et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat. Struct. Mol. Biol. 19, 900–905 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Blanco, S. et al. Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders. EMBO J. 33, 2020–2039 (2014). In the study, the authors demonstrate a role for NSUN2-mediated m5C tRNA modifications and tRNA-derived fragments in modulating cellular response to stress and in neurodevelopment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lee, B. P., Smith, M., Buffenstein, R. & Harries, L. W. Negligible senescence in naked mole rats may be a consequence of well-maintained splicing regulation. Geroscience 42, 633–651 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Deschenes, M. & Chabot, B. The emerging role of alternative splicing in senescence and aging. Aging Cell 16, 918–933 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Karijolich, J. & Yu, Y. T. Spliceosomal snRNA modifications and their function. RNA Biol. 7, 192–204 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Hasler, D. et al. The Alazami syndrome-associated protein LARP7 guides U6 small nuclear RNA modification and contributes to splicing robustness. Mol. Cell 77, 1014–1031 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Wang, X. et al. LARP7-mediated U6 snRNA modification ensures splicing fidelity and spermatogenesis in mice. Mol. Cell 77, 999–1013 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Rueter, S. M., Dawson, T. R. & Emeson, R. B. Regulation of alternative splicing by RNA editing. Nature 399, 75–80 (1999).

    Article  CAS  PubMed  Google Scholar 

  54. Solomon, O. et al. Global regulation of alternative splicing by adenosine deaminase acting on RNA (ADAR). RNA 19, 591–604 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhao, X. et al. FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res. 24, 1403–1419 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Liu, N. et al. N6-methyladenosine-dependent RNA structural switches regulate RNA–protein interactions. Nature 518, 560–564 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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 

  58. Marcel, V. et al. p53 acts as a safeguard of translational control by regulating fibrillarin and rRNA methylation in cancer. Cancer Cell 24, 318–330 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yoon, A. et al. Impaired control of IRES-mediated translation in X-linked dyskeratosis congenita. Science 312, 902–906 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Nachmani, D. et al. Germline NPM1 mutations lead to altered rRNA 2′-O-methylation and cause dyskeratosis congenita. Nat. Genet. 51, 1518–1529 (2019). This study describes how novel mutations in NPM1 cause dyskeratosis congenita and decrease site-specific ribosomal RNA Nm modifications.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Taoka, M. et al. Landscape of the complete RNA chemical modifications in the human 80S ribosome. Nucleic Acids Res. 46, 9289–9298 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ignatova, V. V. et al. The rRNA m6A methyltransferase METTL5 is involved in pluripotency and developmental programs. Genes Dev. 34, 715–729 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Begley, U. et al. Trm9-catalyzed tRNA modifications link translation to the DNA damage response. Mol. Cell 28, 860–870 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Laxman, S. et al. Sulfur amino acids regulate translational capacity and metabolic homeostasis through modulation of tRNA thiolation. Cell 154, 416–429 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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 

  66. Rapino, F. et al. Codon-specific translation reprogramming promotes resistance to targeted therapy. Nature 558, 605–609 (2018). In this manuscript, inhibition of RNA-modifying enzymes involved in tRNA U34 modification restores sensitivity to BRAF inhibition in drug-resistant melanomas.

    Article  CAS  PubMed  Google Scholar 

  67. Thiaville, P. C. et al. Global translational impacts of the loss of the tRNA modification t6A in yeast. Microb. Cell 3, 29–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Pollo-Oliveira, L. et al. Loss of elongator- and KEOPS-dependent tRNA modifications leads to severe growth phenotypes and protein aggregation in yeast. Biomolecules https://doi.org/10.3390/biom10020322 (2020).

  69. Chan, C. T. et al. Reprogramming of tRNA modifications controls the oxidative stress response by codon-biased translation of proteins. Nat. Commun. 3, 937 (2012).

    Article  PubMed  CAS  Google Scholar 

  70. Fu, D. et al. Human AlkB homolog ABH8 is a tRNA methyltransferase required for wobble uridine modification and DNA damage survival. Mol. Cell. Biol. 30, 2449–2459 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Endres, L. et al. Alkbh8 regulates selenocysteine-protein expression to protect against reactive oxygen species damage. PLoS ONE 10, e0131335 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mao, Y. et al. m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat. Commun. 10, 5332 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Zhou, J. et al. Dynamic m6A mRNA methylation directs translational control of heat shock response. Nature 526, 591–594 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. 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 

  77. North, B. J. & Sinclair, D. A. The intersection between aging and cardiovascular disease. Circ. Res. 110, 1097–1108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Mathiyalagan, P. et al. FTO-dependent N6-methyladenosine regulates cardiac function during remodeling and repair. Circulation 139, 518–532 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kmietczyk, V. et al. m6A-mRNA methylation regulates cardiac gene expression and cellular growth. Life Sci. Alliance https://doi.org/10.26508/lsa.201800233 (2019).

  80. Dorn, L. E. et al. The N6-methyladenosine mRNA methylase METTL3 controls cardiac homeostasis and hypertrophy. Circulation 139, 533–545 (2019).

    Article  CAS  PubMed  Google Scholar 

  81. Frayling, T. M. et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316, 889–894 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Liu, C., Mou, S. & Pan, C. The FTO gene rs9939609 polymorphism predicts risk of cardiovascular disease: a systematic review and meta-analysis. PLoS ONE 8, e71901 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. 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 

  85. Wei, J. et al. Differential m6A, m6Am, and m1A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol. Cell 71, 973–985 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Moore, J. B. T. et al. The A-to-I RNA editing enzyme Adar1 is essential for normal embryonic cardiac growth and development. Circ. Res. 127, 550–552 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. El Azzouzi, H. et al. Cardiomyocyte specific deletion of ADAR1 causes severe cardiac dysfunction and increased lethality. Front. Cardiovasc. Med. 7, 30 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Fei, J., Cui, X. B., Wang, J. N., Dong, K. & Chen, S. Y. ADAR1-mediated RNA editing, a novel mechanism controlling phenotypic modulation of vascular smooth muscle cells. Circ. Res. 119, 463–469 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Stellos, K. et al. Adenosine-to-inosine RNA editing controls cathepsin S expression in atherosclerosis by enabling HuR-mediated post-transcriptional regulation. Nat. Med. 22, 1140–1150 (2016). This article implicates elevated A-to-I editing of the mRNA encoding cathepsin S in the pathogenesis of atherosclerosis.

    Article  CAS  PubMed  Google Scholar 

  90. Suzuki, T., Nagao, A. & Suzuki, T. Human mitochondrial tRNAs: biogenesis, function, structural aspects, and diseases. Annu. Rev. Genet. 45, 299–329 (2011).

    Article  CAS  PubMed  Google Scholar 

  91. Fulop, T. et al. Immunosenescence and inflamm-aging as two sides of the same coin: friends or foes? Front. Immunol. 8, 1960 (2017).

    Article  PubMed  CAS  Google Scholar 

  92. O’Connell, M. A., Mannion, N. M. & Keegan, L. P. The epitranscriptome and innate immunity. PLoS Genet. 11, e1005687 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Kariko, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005). Key study demonstrating that delivery of modified RNA suppresses RNA recognition by Toll-like receptors.

    Article  CAS  PubMed  Google Scholar 

  94. Nelson, J. et al. Impact of mRNA chemistry and manufacturing process on innate immune activation. Sci. Adv. 6, eaaz6893 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Lu, M. et al. N6-methyladenosine modification enables viral RNA to escape recognition by RNA sensor RIG-I. Nat. Microbiol. 5, 584–598 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Agrawal, A. Mechanisms and implications of age-associated impaired innate interferon secretion by dendritic cells: a mini-review. Gerontology 59, 421–426 (2013).

    Article  PubMed  Google Scholar 

  97. Hartner, J. C., Walkley, C. R., Lu, J. & Orkin, S. H. ADAR1 is essential for the maintenance of hematopoiesis and suppression of interferon signaling. Nat. Immunol. 10, 109–115 (2009).

    Article  CAS  PubMed  Google Scholar 

  98. George, C. X., Ramaswami, G., Li, J. B. & Samuel, C. E. Editing of cellular self-RNAs by adenosine deaminase ADAR1 suppresses innate immune stress responses. J. Biol. Chem. 291, 6158–6168 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Rice, G. I. et al. Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a type I interferon signature. Nat. Genet. 44, 1243–1248 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Liu, E. Y., Cali, C. P. & Lee, E. B. RNA metabolism in neurodegenerative disease. Dis. Models Mech. 10, 509–518 (2017).

    Article  CAS  Google Scholar 

  101. Reitz, C. et al. Genetic variants in the fat and obesity associated (FTO) gene and risk of Alzheimer’s disease. PLoS ONE 7, e50354 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Yamashita, T. et al. Rescue of amyotrophic lateral sclerosis phenotype in a mouse model by intravenous AAV9-ADAR2 delivery to motor neurons. EMBO Mol. Med. 5, 1710–1719 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Hideyama, T. et al. Profound downregulation of the RNA editing enzyme ADAR2 in ALS spinal motor neurons. Neurobiol. Dis. 45, 1121–1128 (2012).

    Article  CAS  PubMed  Google Scholar 

  104. Brusa, R. et al. Early-onset epilepsy and postnatal lethality associated with an editing-deficient GluR-B allele in mice. Science 270, 1677–1680 (1995).

    Article  CAS  PubMed  Google Scholar 

  105. Higuchi, M. et al. Point mutation in an AMPA receptor gene rescues lethality in mice deficient in the RNA-editing enzyme ADAR2. Nature 406, 78–81 (2000).

    Article  CAS  PubMed  Google Scholar 

  106. Yamashita, T. et al. A role for calpain-dependent cleavage of TDP-43 in amyotrophic lateral sclerosis pathology. Nat. Commun. 3, 1307 (2012).

    Article  PubMed  CAS  Google Scholar 

  107. Moore, S. et al. ADAR2 mislocalization and widespread RNA editing aberrations in C9orf72-mediated ALS/FTD. Acta Neuropathol. 138, 49–65 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Parikshak, N. N., Gandal, M. J. & Geschwind, D. H. Systems biology and gene networks in neurodevelopmental and neurodegenerative disorders. Nat. Rev. Genet. 16, 441–458 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Flores, J. V. et al. Cytosine-5 RNA methylation regulates neural stem cell differentiation and motility. Stem Cell Rep. 8, 112–124 (2017).

    Article  CAS  Google Scholar 

  110. Braun, D. A. et al. Mutations in KEOPS-complex genes cause nephrotic syndrome with primary microcephaly. Nat. Genet. 49, 1529–1538 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Arrondel, C. et al. Defects in t6A tRNA modification due to GON7 and YRDC mutations lead to Galloway-Mowat syndrome. Nat. Commun. 10, 3967 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Livneh, I., Moshitch-Moshkovitz, S., Amariglio, N., Rechavi, G. & Dominissini, D. The m6A epitranscriptome: transcriptome plasticity in brain development and function. Nat. Rev. Neurosci. 21, 36–51 (2020).

    Article  CAS  PubMed  Google Scholar 

  113. White, M. C. et al. Age and cancer risk: a potentially modifiable relationship. Am. J. Prev. Med. 46, S7–S15 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Shen, S. et al. An epitranscriptomic mechanism underlies selective mRNA translation remodelling in melanoma persister cells. Nat. Commun. 10, 5713 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. 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). This and the study below identify a role for m6A RNA modifications in normal hematopoiesis and in promoting leukemia.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Weng, H. et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A modification. Cell Stem Cell 22, 191–205 (2018). This and the study above identify a role for m6A RNA modifications in normal hematopoiesis and in promoting leukemia.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ishizuka, J. J. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 565, 43–48 (2019).

    Article  CAS  PubMed  Google Scholar 

  119. Ruggero, D. et al. Dyskeratosis congenita and cancer in mice deficient in ribosomal RNA modification. Science 299, 259–262 (2003).

    Article  CAS  PubMed  Google Scholar 

  120. Alter, B. P., Giri, N., Savage, S. A. & Rosenberg, P. S. Cancer in dyskeratosis congenita. Blood 113, 6549–6557 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Ronchetti, D. et al. The expression pattern of small nucleolar and small Cajal body-specific RNAs characterizes distinct molecular subtypes of multiple myeloma. Blood Cancer J. 2, e96 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Valleron, W. et al. Small nucleolar RNA expression profiling identifies potential prognostic markers in peripheral T-cell lymphoma. Blood 120, 3997–4005 (2012).

    Article  CAS  PubMed  Google Scholar 

  123. McMahon, M. et al. A single H/ACA small nucleolar RNA mediates tumor suppression downstream of oncogenic RAS. eLife https://doi.org/10.7554/eLife.48847 (2019).

  124. Babaian, A. et al. Loss of m1acp3Ψ ribosomal RNA modification is a major feature of cancer. Cell Rep. 31, 107611 (2020).

    Article  CAS  PubMed  Google Scholar 

  125. Lee, M. Y., Leonardi, A., Begley, T. J. & Melendez, J. A. Loss of epitranscriptomic control of selenocysteine utilization engages senescence and mitochondrial reprogramming. Redox Biol. 28, 101375 (2020).

    Article  CAS  PubMed  Google Scholar 

  126. Li, Q. et al. NSUN2-mediated m5C methylation and METTL3/METTL14-mediated m6A methylation cooperatively enhance p21 translation. J. Cell. Biochem. 118, 2587–2598 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Bellodi, C., Kopmar, N. & Ruggero, D. Deregulation of oncogene-induced senescence and p53 translational control in X-linked dyskeratosis congenita. EMBO J. 29, 1865–1876 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Wang, Y. et al. N6-methyladenosine RNA modification regulates embryonic neural stem cell self-renewal through histone modifications. Nat. Neurosci. 21, 195–206 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Zhang, C. et al. METTL3 and N6-methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA–RNA hybrid accumulation. Mol. Cell 79, 425–442 (2020).

    Article  CAS  PubMed  Google Scholar 

  131. De Jesus, D. F. et al. m6A mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes. Nat. Metab. 1, 765–774 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Fakruddin, M. et al. Defective mitochondrial tRNA taurine modification activates global proteostress and leads to mitochondrial disease. Cell Rep. 22, 482–496 (2018).

    Article  CAS  PubMed  Google Scholar 

  133. Lin, H. et al. CO2-sensitive tRNA modification associated with human mitochondrial disease. Nat. Commun. 9, 1875 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Pang, W. W. et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc. Natl Acad. Sci. USA 108, 20012–20017 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Mejia-Ramirez, E. & Florian, M. C. Understanding intrinsic hematopoietic stem cell aging. Haematologica 105, 22–37 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Li, Z. et al. Suppression of m6A reader Ythdf2 promotes hematopoietic stem cell expansion. Cell Res. 28, 904–917 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Guzzi, N. et al. Pseudouridylation of tRNA-derived fragments steers translational control in stem cells. Cell 173, 1204–1216 (2018).

    Article  CAS  PubMed  Google Scholar 

  138. Gu, B. W., Fan, J. M., Bessler, M. & Mason, P. J. Accelerated hematopoietic stem cell aging in a mouse model of dyskeratosis congenita responds to antioxidant treatment. Aging Cell 10, 338–348 (2011).

    Article  CAS  PubMed  Google Scholar 

  139. Kirwan, M. & Dokal, I. Dyskeratosis congenita: a genetic disorder of many faces. Clin. Genet. 73, 103–112 (2008).

    Article  CAS  PubMed  Google Scholar 

  140. Verdu, E., Ceballos, D., Vilches, J. J. & Navarro, X. Influence of aging on peripheral nerve function and regeneration. J. Peripher. Nerv. Syst. 5, 191–208 (2000).

    Article  CAS  PubMed  Google Scholar 

  141. Weng, Y. L. et al. Epitranscriptomic m6A regulation of axon regeneration in the adult mammalian nervous system. Neuron 97, 313–325 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Murtha, L. A. et al. The role of pathological aging in cardiac and pulmonary fibrosis. Aging Dis. 10, 419–428 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Kropski, J. A. et al. A novel dyskerin (DKC1) mutation is associated with familial interstitial pneumonia. Chest 146, e1–e7 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Ben-Shoshan, S. O. et al. ADAR1 deletion induces NFκB and interferon signaling dependent liver inflammation and fibrosis. RNA Biol. 14, 587–602 (2017). In this manuscript it was shown that ADAR1 depletion in vivo induces inflammation and liver fibrosis.

    Article  PubMed  Google Scholar 

  145. Hebras, J., Krogh, N., Marty, V., Nielsen, H. & Cavaille, J. Developmental changes of rRNA ribose methylations in the mouse. RNA Biol. 17, 150–164 (2020).

    Article  CAS  PubMed  Google Scholar 

  146. Shafik, A. M. et al. N6-methyladenosine dynamics in neurodevelopment and aging, and its potential role in Alzheimer’s disease. Genome Biol. 22, 17 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Asadi Shahmirzadi, A. et al. Alpha-ketoglutarate, an endogenous metabolite, extends lifespan and compresses morbidity in aging mice. Cell Metab. 32, 447–456 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Thomas, J. M., Batista, P. J. & Meier, J. L. Metabolic regulation of the epitranscriptome. ACS Chem. Biol. 14, 316–324 (2019).

    Article  CAS  PubMed  Google Scholar 

  149. Fedeles, B. I., Singh, V., Delaney, J. C., Li, D. & Essigmann, J. M. The AlkB family of Fe(II)/alpha-ketoglutarate-dependent dioxygenases: repairing nucleic acid alkylation damage and beyond. J. Biol. Chem. 290, 20734–20742 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Wu, Z. et al. METTL3 counteracts premature aging via m6A-dependent stabilization of MIS12 mRNA. Nucleic Acids Res. 48, 11083–11096 (2020). In this study, it was found that m6A is decreased in stem cell models of the progeroid syndromes Hutchinson–Gilford progeria syndrome and Werner syndrome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Sebastiani, P. et al. RNA editing genes associated with extreme old age in humans and with lifespan in C. elegans. PLoS ONE 4, e8210 (2009). This article and the one below describe SNPs in factors involved in A-to-I editing and m6A modifications and their association with human longevity.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Cardelli, M. et al. A polymorphism of the YTHDF2 gene (1p35) located in an Alu-rich genomic domain is associated with human longevity. J. Gerontol. A 61, 547–556 (2006). This article and the one above describe SNPs in factors involved in A-to-I editing and m6A modifications and their association with human longevity.

    Article  Google Scholar 

  153. 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 

  154. Liberman, N. et al. N6-adenosine methylation of ribosomal RNA affects lipid oxidation and stress resistance. Sci. Adv. 6, eaaz4370 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Heissenberger, C. et al. Loss of the ribosomal RNA methyltransferase NSUN5 impairs global protein synthesis and normal growth. Nucleic Acids Res. 47, 11807–11825 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Marchand, V. et al. AlkAniline-Seq: profiling of m7G and m3C RNA modifications at single nucleotide resolution. Angew. Chem. Int. Ed. 57, 16785–16790 (2018).

    Article  CAS  Google Scholar 

  158. Liu, H. et al. Accurate detection of m6A RNA modifications in native RNA sequences. Nat. Commun. 10, 4079 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Heiss, M., Reichle, V. F. & Kellner, S. Observing the fate of tRNA and its modifications by nucleic acid isotope labeling mass spectrometry: NAIL-MS. RNA Biol. 14, 1260–1268 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Liu, X. M., Zhou, J., Mao, Y., Ji, Q. & Qian, S. B. Programmable RNA N6-methyladenosine editing by CRISPR-Cas9 conjugates. Nat. Chem. Biol. 15, 865–871 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Richard, E. M. et al. Bi-allelic variants in METTL5 cause autosomal-recessive intellectual disability and microcephaly. Am. J. Hum. Genet. 105, 869–878 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Braun, D. A. et al. Mutations in WDR4 as a new cause of Galloway–Mowat syndrome. Am. J. Med. Genet. A 176, 2460–2465 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Monies, D., Vagbo, C. B., Al-Owain, M., Alhomaidi, S. & Alkuraya, F. S. Recessive truncating mutations in ALKBH8 cause intellectual disability and severe impairment of wobble uridine modification. Am. J. Hum. Genet. 104, 1202–1209 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. de Brouwer, A. P. M. et al. Variants in PUS7 cause intellectual disability with speech delay, microcephaly, short stature, and aggressive behavior. Am. J. Hum. Genet. 103, 1045–1052 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. de Paiva, A. R. B. et al. PUS3 mutations are associated with intellectual disability, leukoencephalopathy, and nephropathy. Neurol. Genet. 5, e306 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Shen, F. et al. Decreased N6-methyladenosine in peripheral blood RNA from diabetic patients is associated with FTO expression rather than ALKBH5. J. Clin. Endocrinol. Metab. 100, E148–E154 (2015).

    Article  CAS  PubMed  Google Scholar 

  167. Vlachogiannis, N. I. et al. Increased adenosine-to-inosine RNA editing in rheumatoid arthritis. J. Autoimmun. 106, 102329 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. 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 

  169. Ghezzi, D. et al. Mutations of the mitochondrial-tRNA modifier MTO1 cause hypertrophic cardiomyopathy and lactic acidosis. Am. J. Hum. Genet. 90, 1079–1087 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Kopajtich, R. et al. Mutations in GTPBP3 cause a mitochondrial translation defect associated with hypertrophic cardiomyopathy, lactic acidosis, and encephalopathy. Am. J. Hum. Genet. 95, 708–720 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Zhang, X. et al. Small RNA modifications in Alzheimer’s disease. Neurobiol. Dis. 145, 105058 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Liddicoat, B. J. et al. RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself. Science 349, 1115–1120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Kimmel and R. Cohen at Calico Life Sciences LLC as well as D. Fujimori (UCSF) and C. Stumpf (eFFECTOR Therapeutics) for their helpful comments on this article. Figures were created using BioRender.com. M.M. and R.B. have been funded by Calico Life Sciences LLC. C.F. was funded by the NIH/NIDDK (1K08DK119561-01) and the American Society of Hematology (A132428).

Author information

Authors and Affiliations

Authors

Contributions

M.M. and R.B. led article conceptualization and writing. M.M., C.F. and R.B. researched data for the article. M.M., C.F. and R.B. discussed the content and contributed to writing and editing of the manuscript before submission.

Corresponding authors

Correspondence to Mary McMahon or Rochelle Buffenstein.

Ethics declarations

Competing interests

M.M. and R.B. are employees of Calico Life Sciences LLC. C.F. declares no competing interests.

Additional information

Peer review information Nature Aging thanks the anonymous reviewers 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.

Supplementary information

Supplementary Information

List of abbreviations used.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McMahon, M., Forester, C. & Buffenstein, R. Aging through an epitranscriptomic lens. Nat Aging 1, 335–346 (2021). https://doi.org/10.1038/s43587-021-00058-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00058-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing