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.

  • Article
  • Published:

m6A epitranscriptomic regulation of tissue homeostasis during primate aging

Abstract

How N6-methyladenosine (m6A), the most abundant mRNA modification, contributes to primate tissue homeostasis and physiological aging remains elusive. Here, we characterize the m6A epitranscriptome across the liver, heart and skeletal muscle in young and old nonhuman primates. Our data reveal a positive correlation between m6A modifications and gene expression homeostasis across tissues as well as tissue-type-specific aging-associated m6A dynamics. Among these tissues, skeletal muscle is the most susceptible to m6A loss in aging and shows a reduction in the m6A methyltransferase METTL3. We further show that METTL3 deficiency in human pluripotent stem cell-derived myotubes leads to senescence and apoptosis, and identify NPNT as a key element downstream of METTL3 involved in myotube homeostasis, whose expression and m6A levels are both decreased in senescent myotubes. Our study provides a resource for elucidating m6A-mediated mechanisms of tissue aging and reveals a METTL3–m6A–NPNT axis counteracting aging-associated skeletal muscle degeneration.

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: Characterization of aging-associated phenotypes of liver, skeletal muscle and heart from young and old primates.
Fig. 2: Profiling of m6A epitranscriptomes of liver, skeletal muscle and heart from primates.
Fig. 3: m6A modification dynamics in the liver, skeletal muscle and heart during primate aging.
Fig. 4: Downregulation of METTL3 and m6A contributes to skeletal muscle dysregulation during primate aging.
Fig. 5: NPNT is a key downstream effector of METTL3 in regulating skeletal muscle aging.
Fig. 6: METTL3 promotes NPNT expression and myotube homeostasis in an m6A-dependent manner.

Similar content being viewed by others

Data availability

High-throughput sequencing data generated in this study have been deposited in the Genome Sequence Archive (GSA) in the National Genomics Data Center, Beijing Institute of Genomics (China National Center for Bioinformation) of the Chinese Academy of Sciences under the accession numbers CRA005942 (monkey tissues) and HRA002143 (human myotubes) and are publicly accessible. In addition, the relevant processed data are also publicly accessible at the Aging Atlas database (https://bigd.big.ac.cn/aging/index)92. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

Code availability

All codes used for the analysis of m6A and RNA sequencing in this study are available at https://github.com/lumm11/M6a-Primate-Aging.

References

  1. Kennedy, B. K. et al. Geroscience: linking aging to chronic disease. Cell 159, 709–713 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kim, I. H., Kisseleva, T. & Brenner, D. A. Aging and liver disease. Curr. Opin. Gastroenterol. 31, 184–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Stern, S., Behar, S. & Gottlieb, S. Cardiology patient pages. Aging and diseases of the heart. Circulation 108, e99–e101 (2003).

    Article  PubMed  Google Scholar 

  4. Cai, Y. et al. Genetic enhancement: a new avenue to combat aging-related diseases. Life Med. 1, 307–318 (2022).

    Article  Google Scholar 

  5. Sun, Y., Li, Q. & Kirkland, J. L. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. Life Med. 1, 103–119 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Cai, Y. et al. The landscape of aging. Sci. China Life Sci. 65, 2354–2454 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kan, C., Ungelenk, L., Lupp, A., Dirsch, O. & Dahmen, U. Ischemia–reperfusion injury in aged livers–the energy metabolism, inflammatory response, and autophagy. Transplantation 102, 368–377 (2018).

    Article  PubMed  Google Scholar 

  8. Welch, A. A., Hayhoe, R. P. G. & Cameron, D. The relationships between sarcopenic skeletal muscle loss during ageing and macronutrient metabolism, obesity and onset of diabetes. Proc. Nutr. Soc. 79, 158–169 (2020).

    Article  PubMed  Google Scholar 

  9. Hunt, N. J., Kang, S. W. S., Lockwood, G. P., Le Couteur, D. G. & Cogger, V. C. Hallmarks of aging in the liver. Comput. Struct. Biotechnol. J. 17, 1151–1161 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tian, X., Seluanov, A. & Gorbunova, V. Beyond making ends meet: DNA-PK, metabolism, and aging. Cell Metab. 25, 991–992 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Yang, Y., Li, X., Zhang, T. & Xu, D. RIP kinases and necroptosis in aging and aging-related diseases. Life Med. 1, 2–20 (2022).

    Article  Google Scholar 

  12. Sithara, T. & Drosatos, K. Metabolic complications in cardiac aging. Front. Physiol. 12, 669497 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zhang, W. et al. SIRT6 deficiency results in developmental retardation in cynomolgus monkeys. Nature 560, 661–665 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Wang, S. et al. Single-cell transcriptomic atlas of primate ovarian aging. Cell 180, 585–600 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Ma, S. et al. Single-cell transcriptomic atlas of primate cardiopulmonary aging. Cell Res. 31, 415–432 (2020).

    Article  PubMed  Google Scholar 

  16. Zhang, W. et al. A single-cell transcriptomic landscape of primate arterial aging. Nat. Commun. 11, 2202 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zou, X. et al. From monkey single-cell atlases into a broader biomedical perspective. Life Med. 1, 254–257 (2022).

    Article  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Roignant, J. Y. & Soller, M. m6A in mRNA: an ancient mechanism for fine-tuning gene expression. Trends Genet. 33, 380–390 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Niu, Y. et al. N6-methyl-adenosine (m6A) in RNA: an old modification with a novel epigenetic function. Genomics Proteomics Bioinformatics 11, 8–17 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Deng, X. et al. RNA N6-methyladenosine modification in cancers: current status and perspectives. Cell Res. 28, 507–517 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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  Google Scholar 

  24. Yao, Y. et al. METTL3-dependent m6A modification programs T follicular helper cell differentiation. Nat. Commun. 12, 1333 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Meyer, K. D. & Jaffrey, S. R. Rethinking m6A readers, writers and erasers. Annu. Rev. Cell Dev. Biol. 33, 319–342 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hu, L. et al. m6A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat. Biotechnol. 40, 1210–1219 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Liu, J. et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Han, M. et al. Abnormality of m6A mRNA methylation is involved in Alzheimer’s disease. Front. Neurosci. 14, 98 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Chen, X. et al. Down-regulation of m6A mRNA methylation is involved in dopaminergic neuronal death. ACS Chem. Neurosci. 10, 2355–2363 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Ma, S. et al. Caloric restriction reprograms the single-cell transcriptional landscape of Rattus norvegicus aging. Cell 180, 984–1001 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Kuk, J. L., Saunders, T. J., Davidson, L. E. & Ross, R. Age-related changes in total and regional fat distribution. Ageing Res. Rev. 8, 339–348 (2009).

    Article  PubMed  Google Scholar 

  33. Geng, L. et al. A comparative study of metformin and nicotinamide riboside in alleviating tissue aging in rats. Life Med. https://doi.org/10.1093/lifemedi/lnac045 (2022).

  34. Freund, A., Laberge, R. M., Demaria, M. & Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 23, 2066–2075 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Siparsky, P. N., Kirkendall, D. T. & Garrett, W. E. Jr. Muscle changes in aging: understanding sarcopenia. Sports Health 6, 36–40 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Olivetti, G., Melissari, M., Capasso, J. M. & Anversa, P. Cardiomyopathy of the aging human heart. Myocyte loss and reactive cellular hypertrophy. Circ. Res. 68, 1560–1568 (1991).

    Article  CAS  PubMed  Google Scholar 

  37. Jing, Y. et al. Single-nucleus profiling unveils a geroprotective role of the FOXO3 in primate skeletal muscle aging. Protein Cell https://doi.org/10.1093/procel/pwac061 (2022).

  38. Zhang, Y. et al. Single-nucleus transcriptomics reveals a gatekeeper role for FOXP1 in primate cardiac aging. Protein Cell https://doi.org/10.1093/procel/pwac038 (2022).

  39. Tzahor, E. & Poss, K. D. Cardiac regeneration strategies: staying young at heart. Science 356, 1035–1039 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xiao, S. et al. The RNA N6-methyladenosine modification landscape of human fetal tissues. Nat. Cell Biol. 21, 651–661 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Zhang, S. et al. FTO stabilizes MIS12 and counteracts senescence. Protein Cell 13, 954–960 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Sunadome, K. et al. ERK5 regulates muscle cell fusion through Klf transcription factors. Dev. Cell 20, 192–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Liu, P. et al. m6A-independent genome-wide METTL3 and METTL14 redistribution drives the senescence-associated secretory phenotype. Nat. Cell Biol. 23, 355–365 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yankova, E. et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 593, 597–601 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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 

  47. Ma, L. et al. Evolution of transcript modification by N6-methyladenosine in primates. Genome Res. 27, 385–392 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. McMahon, M., Forester, C. & Buffenstein, R. Aging through an epitranscriptomic lens. Nat. Aging 1, 335–346 (2021).

    Article  PubMed  Google Scholar 

  49. Wu, Z. et al. Emerging role of RNA m6A modification in aging regulation. Curr. Med. 1, 8 (2022).

    Article  Google Scholar 

  50. Wu, Z. et al. METTL3 counteracts premature aging via m6A-dependent stabilization of MIS12 mRNA. Nucleic Acids Res. 48, 11083–11096 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Min, K. W. et al. Profiling of m6A RNA modifications identified an age-associated regulation of AGO2 mRNA stability. Aging Cell 17, e12753 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zhang, J. et al. Lamin A safeguards the m6A methylase METTL14 nuclear speckle reservoir to prevent cellular senescence. Aging Cell 19, e13215 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Lovric, J. et al. Terminal differentiation of cardiac and skeletal myocytes induces permissivity to AAV transduction by relieving inhibition imposed by DNA damage response proteins. Mol. Ther. 20, 2087–2097 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liang, Y. et al. METTL3-mediated m6A methylation regulates muscle stem cells and muscle regeneration by Notch signaling pathway. Stem Cells Int. 2021, 9955691 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Petrosino, J. M. et al. The m6A methyltransferase METTL3 regulates muscle maintenance and growth in mice. Nat. Commun. 13, 168 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wu, W. et al. AMPK regulates lipid accumulation in skeletal muscle cells through FTO-dependent demethylation of N6-methyladenosine. Sci Rep. 7, 41606 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Morales, P. E., Bucarey, J. L. & Espinosa, A. Muscle lipid metabolism: role of lipid droplets and perilipins. J. Diabetes Res. 2017, 1789395 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Kitada, M. & Koya, D. Autophagy in metabolic disease and ageing. Nat. Rev. Endocrinol. 17, 647–661 (2021).

    Article  PubMed  Google Scholar 

  60. Wang, Y., Welc, S. S., Wehling-Henricks, M. & Tidball, J. G. Myeloid cell-derived tumor necrosis factor-alpha promotes sarcopenia and regulates muscle cell fusion with aging muscle fibers. Aging Cell 17, e12828 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Girardi, F. et al. TGFβ signaling curbs cell fusion and muscle regeneration. Nat. Commun. 12, 750 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Tidball, J. G., Flores, I., Welc, S. S., Wehling-Henricks, M. & Ochi, E. Aging of the immune system and impaired muscle regeneration: a failure of immunomodulation of adult myogenesis. Exp. Gerontol. 145, 111200 (2021).

    Article  CAS  PubMed  Google Scholar 

  63. Moustogiannis, A. et al. The effects of muscle cell aging on myogenesis. Int. J. Mol. Sci. 22, 3721 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Nehlin, J. O., Just, M., Rustan, A. C. & Gaster, M. Human myotubes from myoblast cultures undergoing senescence exhibit defects in glucose and lipid metabolism. Biogerontology 12, 349–365 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Kuek, V. et al. NPNT is expressed by osteoblasts and mediates angiogenesis via the activation of extracellular signal-regulated kinase. Sci Rep. 6, 36210 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Gache, V., Gomes, E. R. & Cadot, B. Microtubule motors involved in nuclear movement during skeletal muscle differentiation. Mol. Biol. Cell 28, 865–874 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kapitansky, O. & Gozes, I. ADNP differentially interact with genes/proteins in correlation with aging: a novel marker for muscle aging. Geroscience 41, 321–340 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kapitansky, O. et al. Age and sex-dependent ADNP regulation of muscle gene expression is correlated with motor behavior: possible feedback mechanism with PACAP. Int. J. Mol. Sci. 21, 6715 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ottenheijm, C. A. & Granzier, H. Role of titin in skeletal muscle function and disease. Adv. Exp. Med. Biol. 682, 105–122 (2010).

    Article  CAS  PubMed  Google Scholar 

  70. Swist, S. et al. Maintenance of sarcomeric integrity in adult muscle cells crucially depends on Z-disc anchored titin. Nat. Commun. 11, 4479 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Maffioletti, S. M. et al. Efficient derivation and inducible differentiation of expandable skeletal myogenic cells from human ES and patient-specific iPS cells. Nat. Protoc. 10, 941–958 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Deshmukh, D. V. et al. Screening method to identify hydrogel formulations that facilitate myotube formation from encapsulated primary myoblasts. Bioeng. Transl. Med. 5, e10181 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. He, Y. et al. 4E-BP1 counteracts human mesenchymal stem cell senescence via maintaining mitochondrial homeostasis. Protein Cell https://doi.org/10.1093/procel/pwac037 (2022).

  74. Ham, D. J. et al. The neuromuscular junction is a focal point of mTORC1 signaling in sarcopenia. Nat. Commun. 11, 4510 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Emde, B., Heinen, A., Gödecke, A. & Bottermann, K. Wheat germ agglutinin staining as a suitable method for detection and quantification of fibrosis in cardiac tissue after myocardial infarction. Eur. J. Histochem. 58, 2448 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  77. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  Google Scholar 

  78. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Liu, J. et al. Landscape and regulation of m6A and m6Am methylome across human and mouse tissues. Mol. Cell 77, 426–440 (2020).

    Article  CAS  PubMed  Google Scholar 

  82. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Xiong, X. et al. Genetic drivers of m6A methylation in human brain, lung, heart and muscle. Nat. Genet. 53, 1156–1165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lu, Y. et al. Gene expression variation and parental allele inheritance in a Xiphophorus interspecies hybridization model. PLoS Genet. 14, e1007875 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Shen, S. et al. rMATS: robust and flexible detection of differential alternative splicing from replicate RNA-seq data. Proc. Natl Acad. Sci. USA 111, E5593–E5601 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Liu, X. et al. Resurrection of endogenous retroviruses during aging reinforces senescence. Cell 186, 287–304 (2023).

    Article  CAS  PubMed  Google Scholar 

  91. Zhao, H. et al. Destabilizing heterochromatin by APOE mediates senescence. Nat. Aging 2, 303–316 (2022).

    Article  CAS  PubMed  Google Scholar 

  92. Aging Atlas Consortium. Aging Atlas: a multi-omics database for aging biology. Nucleic Acids Res. 49, D825–D830 (2021).

    Article  Google Scholar 

Download references

Acknowledgements

We are grateful to L. Bai, Q. Chu, R. Bai, S. Ma, J. Lu, L. Tian, Y. Yang, J. Chen and X. Jin for their administrative assistance. We thank Q. Ji, Y. Zhang, S. Yang, L. Li, P. Yan, X. Jiang, J. Li, S. Li, H. Zhao, C. Liang, Y. He, H. Hu, Z. Wang, L. Fu, S. Ma, H. Yan and Z. Diao for their technical support. We also thank the Data Center for Stem Cell and Regeneration (DCSCR) for supporting the data analysis. This work was supported by the National Key Research and Development Program of China (2022YFA1103700 to W.Z. and J.Q.), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16000000 to G.-H.L., J.Q., W.Z., W.C., J.R., S.W. and Y.-G.Y.), the National Key Research and Development Program of China (2020YFA0804000 to G.-H.L. and S.W., 2018YFC2000100 to J.Q., 2018YFA0107203 to J.Q., 2020YFA0112200 to G.-H.L., 2021YFF1201005 to W.Z., 2019YFA0110900 to W.C., 2022YFA1103800 to Z.W. and T.W., 2019YFA0802202 to J.R., 2020YFA0803401 to J.R.), the National Natural Science Foundation of China (32100937 to Z.W., 81921006 to G.-H.L. and J.Q., 92149301 to G.-H.L. and S.W., 92168201 to G.-H.L., 82125011 to J.Q., 92049304 to J.Q., 91949209 to J.Q., 92049116 to W.Z., 32121001 to W.Z. and J.R., 82192863 to W.Z., 82122024 to S.W., 82071588 to S.W., 31970597 to J.R., 82173061 to W.C., 82001477 to Q.Z.), the STI2030-Major Projects (2021ZD0202400 to S.W.), CAS Project for Young Scientists in Basic Research (YSBR-076 to G.-H.L., J.Q. and J.R., YSBR-012 to W.Z.), CAS Special Research Assistant Program (to Z.W.), the Program of the Beijing Natural Science Foundation (Z190019 to G.-H.L. and W.Z.), K. C. Wong Education Foundation (GJTD-2019-06 to J.Q., GJTD-2019-08 to W.Z.), the Pilot Project for Public Welfare Development and Reform of Beijing-affiliated Medical Research Institutes (11000022T000000461062 to S.W.), Youth Innovation Promotion Association of CAS (E1CAZW0401 to W.Z.), Young Elite Scientists Sponsorship Program by CAST (YESS20200012 to S.W.), the Informatization Plan of Chinese Academy of Sciences (CAS-WX2021SF-0301 to G.-H.L., CAS-WX2022SDC-XK14 to G.-H.L., CAS-WX2021SF-0101 to J.Q.) and the Tencent Foundation (2021-1045 to G.-H.L.).

Author information

Authors and Affiliations

Authors

Contributions

G.-H.L., W.C., W.Z. and J.Q. conceived the project and supervised overall experiments. Z.W. performed the phenotypic and mechanistic analyses with assistance from D.L., J.R. and S.W. M.L. and Y.S. performed bioinformatic analyses with assistance from J.R., S.W., Z.W. and Z.L. Y.S. performed the m6A-seq and RNA-seq library construction with the help from Z.Y., D.L., S.Z. and S.B. Y.J., Q.Z. and H.L. performed the plasmid construction and lentivirus packaging. G.-H.L., W.C., W.Z., J.Q., Z.W., M.L., D.L., Y.S., J.R. and S.W. performed the data analysis. G.-H.L., W.C., W.Z., J.Q., Z.W., M.L., D.L., Y.S., J.R., S.W., T.W., Y.-G.Y., J.X. and J.C.I.B. wrote the initial draft of this manuscript. All the authors participated in editing both the form and content of this manuscript and approved the final version.

Corresponding authors

Correspondence to Jing Qu, Weiqi Zhang, Weimin Ci or Guang-Hui Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Mary McMahon and the other, anonymous, reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Characterization of mRNA m6A modification in liver, skeletal muscle, and heart from young and old primates.

a. Heatmaps showing the mRNA levels of IFN-I and SASP-related genes (IL1A, IL1B, IFNB1, and IL6) in liver, skeletal muscle, and heart from young and old primates. The color keys from blue to brown represent the mRNA levels from low to high. Eight animals each group were used and experiments were repeated three times independently. b. Motif analysis for m6A peaks in liver, skeletal muscle, and heart from young and old primates. c. Distribution of m6As along the 5′UTR, CDS, and 3′UTR regions of the total mRNAs across all tissues after normalization to transcript length. d. Heatmap showing Pearson’s correlation of m6A modification levels across all tissue samples from old primates. e. Heatmap showing the Pearson’s correlation of the RNA expression levels across all tissue samples from young primates. f. Heatmap showing the Pearson’s correlation of the RNA expression levels across all tissue samples from old tissues. g. Venn diagram showing the overlap of m6A peaks identified in liver, skeletal muscle, and heart from old primates. h. Functional pathway enrichment analysis of genes encoding mRNAs with common m6As shared by liver, skeletal muscle, and heart from old primates. i. Functional pathway enrichment analysis of genes encoding mRNAs with tissue-specific m6As in liver, skeletal muscle, and heart from old primates. The number of genes for each term is indicated in grey round shadow. j. Violin plots showing the distribution of expression level of mRNAs with common m6As and tissue-specific m6As in liver, skeletal muscle, and heart from old primates. k. Violin plots showing the distribution of expression divergence of mRNAs with common m6As and tissue-specific m6As in liver, skeletal muscle, and heart from old primates. The one-sided binomial test was used for statistical analysis in b. The accumulative hypergeometric test was used for statistical analysis in h and i. The two-tailed Mann-Whitney U-test was used for statistical analysis in j and k. Bioinformatic analyses: n = 8 (liver and skeletal muscle) or 5 (heart) biological replicates. P values are indicated in the figure. The color keys from light to dark represent the similarities (Pearson’s correlation) from low to high in d-f.

Source data

Extended Data Fig. 2 The correlation between m6A modification and gene expression dynamics during primate aging.

a. Venn diagram showing the overlap of m6A peaks or genes encoding mRNAs with m6A modification between young and old tissues. b. Venn diagram showing the overlap of genes encoding mRNAs with stable, old-gain or old-loss m6As in liver, skeletal muscle, and heart. c. Boxplots showing the expression level of genes encoding mRNAs with stable, old-loss and old-gain m6As at the 5′ UTR, CDS, and 3′ UTR of their transcripts. d. Boxplots showing the expression divergence of genes encoding mRNAs with stable, old-loss and old-gain m6As at the 5′ UTR, CDS, and 3′ UTR of their transcripts. The two-tailed Mann-Whitney U-test was used for statistical analysis in c and d. Box plots indicate median values and interquartile ranges; whiskers indicate 1.5x interquartile range. n = 8 (for liver and skeletal muscle) or 5 (for heart) biological replicates (see Methods and Supplementary Table 1 for details). P values are indicated in the figure.

Extended Data Fig. 3 Expression alterations in genes encoding mRNAs with m6A loss or gain during aging in the liver, skeletal muscle, and heart.

a-c, Cumulative plots showing the expression alterations in genes encoding mRNAs with m6A loss (left) or gain (right) between young and old livers (a), skeletal muscles (b), and hearts (c). The two-tailed Mann-Whitney U-test was used for statistical analysis. n = 8 (for liver and skeletal muscle) or 5 (for heart) biological replicates (see Methods and Supplementary Table 1 for details). P values are indicated in the figure.

Extended Data Fig. 4 Analysis of m6A modification and expression levels of m6A-related enzymes in young and old tissues.

a. Cumulative plot showing the m6A peak intensity in young and old livers. b. Cumulative plot showing the m6A peak intensity in young and old hearts. c. Dot blot analysis of m6A in mRNAs of young and old livers.d. Dot blot analysis of m6A in mRNAs of young and old hearts. e. Statistical results of the protein levels of METTL14, FTO and ALKBH5 in young and old skeletal muscles (corresponding to Fig. 4c). Triangles indicate female samples, and filled circles indicate male ones. f. Western blot analysis of the m6A-related enzymes in young and old livers. Triangles indicate female samples, and filled circles indicate male ones. g. Western blot analysis of the m6A-related enzymes in young and old hearts. Triangles indicate female samples, and filled circles indicate male ones. The two-tailed Mann-Whitney U-test was used for statistical analysis in a and b. Two-tailed Student’s t-test was used for statistical analysis in c-g. Data are presented as the mean ± s.e.m. n = 8 biological replicates in c-g. P values are indicated in the figure. For dot blot analysis in c and d, MB staining was used as the loading control. For western blot analysis in f and g, GAPDH was used as the loading control.

Source data

Extended Data Fig. 5 Characterization of mRNA m6A modification in control and METTL3-deficient myotubes.

a. Motif analysis for m6A peaks in control (sgNTC) and METTL3-deficient (sgMETTL3) myotubes. b. Distribution of m6As along the 5′UTR, CDS, and 3′UTR regions of the total mRNAs after normalization to transcript length in sgNTC and sgMETTL3 myotubes. c. Pie chart showing the fraction of m6As in different transcript segments in sgNTC and sgMETTL3 myotubes. d. Functional pathway enrichment analysis of genes encoding mRNAs with m6A loss in their transcripts in sgMETTL3 myotubes. Gene numbers are indicated on the right. The color key from light to dark indicates the -log10(P value) from low to high, respectively. e. Heatmap showing the clusters of differentially expressed genes (DEGs) encoding mRNAs with m6A loss in their transcripts in sgMETTL3 myotubes relative to sgNTC myotubes. f. Functional pathway enrichment analysis of differentially expressed genes encoding mRNAs with m6A loss in their transcripts in sgMETTL3 myotubes relative to sgNTC myotubes. The number of genes for each term is indicated in grey round shadow. g. Heatmap showing the differential expression of SASP-related genes in sgNTC and sgMETTL3 myotubes. h. Heatmap showing the differential expression of adipogenesis-related genes in sgNTC and sgMETTL3 myotubes. i. Heatmaps showing the m6A and mRNA levels of NPNT in young and old skeletal muscles. Eight animals each group were used and experiments were repeated three times independently. j. IGV views showing the m6A signals in NPNT in sgNTC and sgMETTL3 myotubes and in young and old skeletal muscles. The color from light to dark represents the m6A signal from low to high, respectively. The one-sided binomial test was used for statistical analysis in a. The accumulative hypergeometric test was used for statistical analysis in d and f. For bioinformatic analyses in sgNTC and sgMETTL3 myotubes, two biological replicates were used. The color keys from blue to brown indicate the normalized RPKM (e, g and h) or the m6A and mRNA levels of NPNT (i) from low to high.

Source data

Extended Data Fig. 6 NPNT is a key downstream effector of METTL3 in counteracting myotube senescence.

a. Heatmap showing the Pearson’s correlation of RNA-seq data in replicates of control (siNC) and NPNT-knockdown (siNPNT) myotubes. The color key from blue to red indicates the Pearson’s correlation from low to high, respectively. b. Heatmap showing the expression level of SASP-related genes in siNC and siNPNT myotubes. The color key from blue to red indicates the expression level (normalized RPKM) from low to high, respectively. c. Heatmap showing the differential expression of adipogenesis-related genes in siNC and siNPNT myotubes. The color key from blue to red indicates the normalized RPKM from low to high, respectively. d. Functional pathway enrichment analysis of downregulated genes in siNPNT myotubes relative to siNC myotubes. e. Heatmaps showing the expression level of genes related to terms including “regulation of muscle contraction” (left), “regulation of muscle system process” (middle), and “muscle cell differentiation” (right) in siNC and siNPNT myotubes. The color key from blue to red indicates the expression level (normalized RPKM) from low to high, respectively. The gene symbol of NPNT is highlighted in red. f. Venn diagram showing the overlap of downregulated genes in METTL3-deficient myotubes and NPNT-knockdown myotubes. g. Functional pathway enrichment analysis of genes showing downregulation in both METTL3-deficient myotubes and NPNT-knockdown myotubes. h. Western blot analysis of NPNT and P16 in METTL3-deficient myotubes transduced with lentiviruses expressing Flag-Luc or Flag-NPNT. i. RT-qPCR analysis of CDKN2A (P16), IL6 and CXCL8 in METTL3-deficient myotubes transduced with lentiviruses expressing Flag-Luc or Flag-NPNT. j. SA-β-gal staining in METTL3-deficient myotubes transduced with lentiviruses expressing Flag-Luc or Flag-NPNT. The accumulative hypergeometric test was used for statistical analysis in d and g. For bioinformatic analyses in siNC and siNPNT myotubes, three biological replicates were used. Two-tailed Student’s t-test was used for statistical analysis in h-j. Data are presented as the mean ± s.e.m. n = 3 biological replicates. P values are indicated in the figure. GAPDH was used as the loading control in h. Scale bars in j, 50 μm.

Source data

Extended Data Fig. 7 METTL3 regulates NPNT expression and myotube homeostasis in a catalytic activity-dependent manner.

a. Immunostaining of m6A in wild-type myotubes treated with vehicle or STM2457. b. RT-qPCR analysis of NPNT in wild-type myotubes treated with vehicle or STM2457. c. SA-β-gal staining in wild-type myotubes treated with vehicle or STM2457. d. Immunostaining of MYHC in wild-type myotubes treated with vehicle or STM2457. The yellow line segments indicate the representative diameters of indicated myotubes. Statistical analysis of myotube diameter is shown on the right. e. Western blot analysis of METTL3 and NPNT in wild-type myotubes cultured for indicated times. f. SA-β-gal staining in wild-type myotubes cultured for 8 and 14 days. Two-tailed Student’s t-test was used for statistical analysis. Data are presented as the mean ± s.e.m. n = 15 random fields from three biological replicates in a. n = 4 biological replicates in b. n = 3 biological replicates in c-f. P values are indicated in the figure. Scale bars in a, c, d and f, 50 μm. GAPDH was used as the loading control in e.

Source data

Extended Data Fig. 8 METTL3 promotes the stability of NPNT mRNA via IGF2BP1.

a. Schematic diagram showing the potential transcripts of NPNT with different skipping exons (indicated by dashed black box). b. Sashimi plots showing the major splicing events of the indicated skipping exons of NPNT in control (sgNTC) and METTL3-deficient (sgMETTL3) myotubes (left), as well as in young and old skeletal muscles (right). c. Heatmaps showing the IncLevel values of differential alternative splicing skipping-exon events between sgNTC and sgMETTL3 myotubes (left), as well as between young and old skeletal muscles (right). The color keys from blue to red indicate the IncLevel from low to high. d. mRNA stability analysis of NPNT in sgNTC and sgMETTL3 myotubes upon treatment with ACTD for the indicated times. e. RIP-qPCR analysis showing the enrichment of IgG or IGF2BP1 on NPNT mRNA. f. Western blot analysis of IGF2BP1 in control (siNC) and IGF2BP1-knockdown (siIGF2BP1) myotubes. GAPDH was used as the loading control. g. mRNA stability analysis of NPNT in siNC and siIGF2BP1 myotubes upon treatment with ACTD for the indicated times. Two-tailed Student’s t-test was used for statistical analysis in d-g. Data are presented as the mean ± s.e.m. n = 3 biological replicates in d-f. n = 4 biological replicates in g. P values are indicated in the figure. For alternative splicing analyses with RNA-seq data in b and c, two biological replicates were used in sgNTC and sgMETTL3 myotubes and eight biological replicates were used in young and old skeletal muscles.

Source data

Supplementary information

Supplementary Information

Legends for Supplementary Tables 1–5.

Reporting Summary

Supplementary Tables 1–5

Supplementary Table 1. Information of cynomolgus monkeys used in this study. Table 2. Differential m6A peaks during primate aging in liver, skeletal muscle and heart. Table 3. Functional annotation of DEGs with m6A loss in METTL3-deficient myotubes. Table 4. Functional annotation of genes with m6A loss in both aged skeletal muscles and METTL3-deficient myotubes. Table 5. Sequences of sgRNA, siRNA and primers for construction of lentiviral expression vector and RT–qPCR analysis.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 4

Unprocessed dot and western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed dot and western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, Z., Lu, M., Liu, D. et al. m6A epitranscriptomic regulation of tissue homeostasis during primate aging. Nat Aging 3, 705–721 (2023). https://doi.org/10.1038/s43587-023-00393-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00393-2

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