1,684
Views
24
CrossRef citations to date
0
Altmetric
Brief Report

The myonuclear DNA methylome in response to an acute hypertrophic stimulus

ORCID Icon, , , , , ORCID Icon & ORCID Icon show all

References

  • Murach KA, Fry CS, Kirby TJ, et al. Starring or supporting role? Satellite cells and skeletal muscle fiber size regulation. Physiology. 2018;33:26–38.
  • Tedesco FS, Moyle LA, Perdiguero E. Muscle interstitial cells: a brief field guide to non-satellite cell populations in skeletal muscle. Muscle Stem Cells: Methods Protoc. Methods Mol Biology. 2017;1556:129–147.
  • Lindholm ME, Marabita F, Gomez-Cabrero D, et al. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics. 2014;9:1557–1569.
  • Begue G, Raue U, Jemiolo B, et al. DNA methylation assessment from human slow-and fast-twitch skeletal muscle fibers. J Appl Physiol. 2017;122:952–967.
  • Murach KA, Raue U, Wilkerson BS, et al. Fiber type-specific gene expression with taper in competitive distance runners. PLoS One. 2014;9(9):e108547.
  • Murgia M, Toniolo L, Nagaraj N, et al. Single muscle fiber proteomics reveals fiber-type-specific features of human muscle aging. Cell Rep. 2017;19:2396–2409.
  • Raue U, Trappe TA, Estrem ST, et al. Transcriptome signature of resistance exercise adaptations: mixed muscle and fiber type specific profiles in young and old adults. J Appl Physiol. 2012;112:1625–1636.
  • Blackburn DM, Lazure F, Corchado AH, et al. High-resolution genome-wide expression analysis of single myofibers using SMART-Seq. J Biol Chem. 2019;294:20097–20108.
  • Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484.
  • Brenet F, Moh M, Funk P, et al. DNA methylation of the first exon is tightly linked to transcriptional silencing. PloS One. 2011;6:e14524.
  • Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301:89.
  • Bender CM, Gonzalgo ML, Gonzales FA, et al. Roles of cell division and gene transcription in the methylation of CpG islands. Mol Cell Biol. 1999;19:6690–6698.
  • Yang X, Han H, De Carvalho DD, et al. Gene body methylation can alter gene expression and is a therapeutic target in cancer. Cancer Cell. 2014;26:577–590.
  • Bagley JR, Burghardt KJ, McManus R, et al. Epigenetic responses to acute resistance exercise in trained vs. sedentary men. J Strength Cond Res. 2019. DOI:10.1519/JSC000000000000.3185,
  • Seaborne R, Strauss J, Cocks M, et al. Methylome of human skeletal muscle after acute & chronic resistance exercise training, detraining & retraining. Sci Data. 2018;5:180213.
  • Seaborne RA, Strauss J, Cocks M, et al. Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8:1898.
  • Iwata M, Englund DA, Wen Y, et al. A novel tetracycline-responsive transgenic mouse strain for skeletal muscle-specific gene expression. Skelet Muscle. 2018;8:33.
  • Kirby TJ, McCarthy JJ, Peterson CA, et al. Synergist ablation as a rodent model to study satellite cell dynamics in adult skeletal muscle. Skeletal muscle regeneration in the mouse: methods and protocols. 2016; 43–52.
  • Murach KA, White SH, Wen Y, et al. Differential requirement for satellite cells during overload-induced muscle hypertrophy in growing versus mature mice. Skelet Muscle. 2017;7:14.
  • von Walden F, Casagrande V, Östlund Farrants A-K, et al. Mechanical loading induces the expression of a Pol I regulon at the onset of skeletal muscle hypertrophy. Am J Physiol Cell Physiol. 2012;302:C1523–C1530.
  • Pawlikowski B, Pulliam C, Betta ND, et al. Pervasive satellite cell contribution to uninjured adult muscle fibers. Skelet Muscle. 2015;5:42.
  • Dungan CM, Murach KA, Frick KK, et al. Elevated myonuclear density during skeletal muscle hypertrophy in response to training is reversed during detraining. Am J Physiol Cell Physiol. 2019;316:C649–C654.
  • McCarthy JJ, Mula J, Miyazaki M, et al. Effective fiber hypertrophy in satellite cell-depleted skeletal muscle. Development. 2011;138:3657–3666.
  • Bodine SC, Stitt TN, Gonzalez M, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014.
  • McCarthy JJ, Murach KA. Anabolic and catabolic signaling pathways that regulate skeletal muscle mass. In: Debasis Bagchi, Sreejayan Nair, & Chandan K Sen, editors. Nutrition and enhanced sports performance.  London: Elsevier 2019. p. 275–290.
  • Kravic B, Harbauer AB, Romanello V, et al. In mammalian skeletal muscle, phosphorylation of TOMM22 by protein kinase CSNK2/CK2 controls mitophagy. Autophagy. 2018;14:311–335.
  • Mammucari C, Milan G, Romanello V, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471.
  • Moresi V, Carrer M, Grueter CE, et al. Histone deacetylases 1 and 2 regulate autophagy flux and skeletal muscle homeostasis in mice. Proc Nat Acad Sci. 2012;109:1649–1654.
  • Mukhopadhyay S, Panda PK, Sinha N, et al. Autophagy and apoptosis: where do they meet? Apoptosis. 2014;19:555–566.
  • Figueiredo VC, McCarthy JJ. Regulation of ribosome biogenesis in skeletal muscle hypertrophy. Physiology. 2018;34:30–42.
  • Nakada S, Ogasawara R, Kawada S, et al. Correlation between ribosome biogenesis and the magnitude of hypertrophy in overloaded skeletal muscle. PloS One. 2016;11:e0147284.
  • von Walden F. Ribosome biogenesis in skeletal muscle: coordination of transcription and translation. J Appl Physiol. 2019;127(2):591–598.
  • Wen Y, Alimov AP, McCarthy JJ. Ribosome biogenesis is necessary for skeletal muscle hypertrophy. Exerc Sport Sci Rev. 2016;44:110.
  • Lorincz MC, Dickerson DR, Schmitt M, et al. Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol. 2004;11:1068.
  • Maunakea AK, Chepelev I, Cui K, et al. Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Res. 2013;23:1256.
  • Maunakea AK, Nagarajan RP, Bilenky M, et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010;466:253.
  • Neri F, Rapelli S, Krepelova A, et al. Intragenic DNA methylation prevents spurious transcription initiation. Nature. 2017;543:72.
  • Rea M, Eckstein M, Eleazer R, et al. Genome-wide DNA methylation reprogramming in response to inorganic arsenic links inhibition of CTCF binding, DNMT expression and cellular transformation. Sci Rep. 2017;7:1–14.
  • Chaillou T, Lee JD, England JH, et al. Time course of gene expression during mouse skeletal muscle hypertrophy. J Appl Physiol. 2013;115:1065–1074.
  • Altıntaş A, Laker RC, Garde C, et al. Transcriptomic and epigenomics atlas of myotubes reveals insight into the circadian control of metabolism and development. Epigenomics. 2020. DOI:10.2217/epi-2019-0391,
  • Santoro R, Grummt I. Molecular mechanisms mediating methylation-dependent silencing of ribosomal gene transcription. Mol Cell. 2001;8:719–725.
  • Laker R, Garde C, Camera D, et al. Transcriptomic and epigenetic responses to short-term nutrient-exercise stress in humans. Sci Rep. 2017;7:1–12.
  • Rowlands DS, Page RA, Sukala WR, et al. Multi-omic integrated networks connect DNA methylation and miRNA with skeletal muscle plasticity to chronic exercise in Type 2 diabetic obesity. Physiol Genomics. 2014;46:747–765.
  • Sailani MR, Halling JF, Møller HD, et al. Lifelong physical activity is associated with promoter hypomethylation of genes involved in metabolism, myogenesis, contractile properties and oxidative stress resistance in aged human skeletal muscle. Sci Rep. 2019;9:1–11.
  • Turner DC, Seaborne RA, Sharples AP. Comparative transcriptome and methylome analysis in human skeletal muscle anabolism, hypertrophy and epigenetic memory. Sci Rep. 2019;9:1–12.
  • Barres R, Yan J, Egan B, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15:405–411.
  • Hodges E, Molaro A, Dos Santos CO, et al. Directional DNA methylation changes and complex intermediate states accompany lineage specificity in the adult hematopoietic compartment. Mol Cell. 2011;44:17–28.
  • Xie W, Ling T, Zhou Y, et al. The chromatin remodeling complex NuRD establishes the poised state of rRNA genes characterized by bivalent histone modifications and altered nucleosome positions. Proc Nat Acad Sci. 2012;109:8161–8166.
  • Cedernaes J, Schönke M, Westholm JO, et al. Acute sleep loss results in tissue-specific alterations in genome-wide DNA methylation state and metabolic fuel utilization in humans. Sci Adv. 2018;4:eaar8590.
  • Harfmann BD, Schroder EA, Esser KA. Circadian rhythms, the molecular clock, and skeletal muscle. J Biol Rhythms. 2015;30:84–94.
  • McCarthy JJ, Andrews JL, McDearmon EL, et al. Identification of the circadian transcriptome in adult mouse skeletal muscle. Physiol Genomics. 2007;31:86–95.
  • Gabriel BM, Zierath JR. Circadian rhythms and exercise—re-setting the clock in metabolic disease. Nat Rev Endocrinol. 2019;15:197–206.
  • Schroder EA, Esser KA. Circadian rhythms, skeletal muscle molecular clocks and exercise. Exerc Sport Sci Rev. 2013;41:224–229.
  • Wolff CA, Esser KA. Exercise timing and circadian rhythms. Curr Opin Physiol. 2019;10:64–69.
  • Goodman CA, Frey JW, Mabrey DM, et al. The role of skeletal muscle mTOR in the regulation of mechanical load‐induced growth. J Physiol. 2011;589:5485–5501.
  • Bai X, Ma D, Liu A, et al. Rheb activates mTOR by antagonizing its endogenous inhibitor, FKBP38. Science. 2007;318:977–980.
  • Goodman CA. Role of mTORC1 in mechanically induced increases in translation and skeletal muscle mass. J Appl Physiol. 2019;127:581–590.
  • Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307:1098–1101.
  • Yu T, Li J, Yan M, et al. MicroRNA-193a-3p and-5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene. 2015;34:413–423.
  • Mayer C, Grummt I. Ribosome biogenesis and cell growth: mTOR coordinates transcription by all three classes of nuclear RNA polymerases. Oncogene. 2006;25:6384–6391.
  • Nader GA, McLoughlin TJ, Esser KA. mTOR function in skeletal muscle hypertrophy: increased ribosomal RNA via cell cycle regulators. Am J Physiol Cell Physiol. 2005;289:C1457–C1465.
  • von Walden F, Liu C, Aurigemma N, et al. mTOR signaling regulates myotube hypertrophy by modulating protein synthesis, rDNA transcription, and chromatin remodeling. Am J Physiol Cell Physiol. 2016;311:C663–C672.
  • Arabi A, Wu S, Ridderstråle K, et al. c-Myc associates with ribosomal DNA and activates RNA polymerase I transcription. Nat Cell Biol. 2005;7:303–310.
  • Van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer. 2010;10:301.
  • Maiuri MC, Galluzzi L, Morselli E, et al. Autophagy regulation by p53. Curr Opin Cell Biol. 2010;22:181–185.
  • Beharry AW, Judge AR. Differential expression of HDAC and HAT genes in atrophying skeletal muscle. Muscle Nerve. 2015;52:1098–1101.
  • Beharry AW, Sandesara PB, Roberts BM, et al. HDAC1 activates FoxO and is both sufficient and required for skeletal muscle atrophy. J Cell Sci. 2014;127:1441–1453.
  • Bell RA, Al-Khalaf M, Megeney LA. The beneficial role of proteolysis in skeletal muscle growth and stress adaptation. Skelet Muscle. 2016;6:16.
  • Masiero E, Agatea L, Mammucari C, et al. Autophagy is required to maintain muscle mass. Cell Metab. 2009;10:507–515.
  • von Walden F, Jakobsson F, Edström L, et al. Altered autophagy gene expression and persistent atrophy suggest impaired remodeling in chronic hemiplegic human skeletal muscle. Muscle Nerve. 2012;46:785–792.
  • Sanchez AM, Bernardi H, Py G, et al. Autophagy is essential to support skeletal muscle plasticity in response to endurance exercise. Am J Physiol Regul Integr Comp Physiol. 2014;307:R956–R969.
  • He C, Bassik MC, Moresi V, et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012;481:511.
  • Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168:960–976.
  • Suzuki MM, Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet. 2008;9:465.
  • Mendoza MC, Er EE, Blenis J. The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends Biochem Sci. 2011;36:320–328.
  • Murach KA, Dungan CM, Dupont-Versteegden EE, et al. “Muscle memory” not mediated by myonuclear number?: secondary analysis of human detraining data. J Appl Physiol. 2019;127:1814–1816.
  • Sharples AP, Stewart CE, Seaborne RA. Does skeletal muscle have an ‘epi’‐memory? The role of epigenetics in nutritional programming, metabolic disease, aging and exercise. Aging Cell. 2016;15:603–616.
  • Tumbar T, Guasch G, Greco V, et al. Defining the epithelial stem cell niche in skin. Science. 2004;303:359–363.
  • Okada S, Saiwai H, Kumamaru H, et al. Flow cytometric sorting of neuronal and glial nuclei from central nervous system tissue. J Cell Physiol. 2011;226:552–558.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.