7,516
Views
67
CrossRef citations to date
0
Altmetric
Review

Epigenetic changes in healthy human skeletal muscle following exercise– a systematic review

ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon & ORCID Icon
Pages 633-648 | Received 18 Feb 2019, Accepted 26 Apr 2019, Published online: 13 May 2019

References

  • Coffey VG, Hawley JA. The molecular basis of training adaptation. Sport Med. 2007;37:737–763.
  • Hargreaves M. Exercise and gene expression. Prog Mol Biol Transl Sci. 2015. p. 457–469.
  • Bishop DJ, Granata C, Eynon N. Can we optimise the exercise training prescription to maximise improvements in mitochondria function and content? Biochim Biophys Acta Gen Subj. 2014;1840:1266–1275.
  • Hawley JA, Hargreaves M, Joyner MJ, et al. Review integrative biology of exercise. Cell. 2014;159:738–749.
  • Soci UPR, Melo SFS, Gomes JLP, et al. Exercise training and epigenetic regulation: multilevel modification and regulation of gene expression. Adv Exp Med Biol.. 2017. p. 281–322.
  • Voisin S, Eynon N, Yan X, et al. Exercise training and DNA methylation in humans. Acta Physiol. 2015;213:39–59.
  • Landen S, Voisin S, Craig JM, et al. Genetic and epigenetic sex-specific adaptations to endurance exercise. Epigenetics. 2019; DOI:10.1080/15592294.2019.1603961.
  • Lappalainen T, Greally JM. Associating cellular epigenetic models with human phenotypes. Nat Rev Genet. 2017;18:441–451.
  • Weinhold B. Epigenetics: the science of change. Environ Heal Perspect. 2006;114:160–167.
  • Huang B, Zhang R. Regulatory non-coding RNAs: revolutionizing the RNA world. Mol Biol Rep. 2014;41:3915–3923.
  • Tao H, Yang -J-J, Shi K-H. Non-coding RNAs as direct and indirect modulators of epigenetic mechanism regulation of cardiac fibrosis. Expert Opin Ther Targets. 2015;19:707–716.
  • Peschansky VJ, Wahlestedt C. Non-coding RNAs as direct and indirect modulators of epigenetic regulation. Epigenetics. 2014;9:3–12.
  • Rooney J Further thoughts on mercury, epigenetics, genetics and amyotrophic lateral sclerosis. 2011;523–524.
  • van Dijk SJ, Molloy PL, Varinli H, et al. Epigenetics and human obesity. Int J Obes (Lond). 2015;39:85–97.
  • Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33:245–254.
  • 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.
  • Li E, Zhang Y. DNA methylation in mammals. Cold Spring Harb Perspect Biol. 2014;1;6:a019133.
  • Nan X, Ng HH, Johnson CA, et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature. 1998;393:386–389.
  • Ohm JE, McGarvey KM, Yu X, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39:237–242.
  • Schlesinger Y, Straussman R, Keshet I, et al. Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet. 2007;39:232–236.
  • Gal-Yam EN, Egger G, Iniguez L, et al. Frequent switching of polycomb repressive marks and DNA hypermethylation in the PC3 prostate cancer cell line. Proc Natl Acad Sci U S A. 2008;105:12979–12984.
  • Strunnikova M, Schagdarsurengin U, Kehlen A, et al. Chromatin inactivation precedes de novo DNA methylation during the progressive epigenetic silencing of the RASSF1A promoter. Mol Cell Biol. 2005;25:3923–3933.
  • Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484–492.
  • Pacis A, Mailhot-Leonard F, Tailleux L, et al. Gene activation precedes DNA demethylation in response to infection in human dendritic cells. Proc Natl Acad Sci U S A. 2019;116:6938–6943.
  • Guibert S, Weber M. Functions of DNA methylation and hydroxymethylation in mammalian development. Curr Top Dev Biol. 2013;104:47–83.
  • Bentley GA, Lewit-Bentley A, Finch JT, et al. Crystal structure of the nucleosome core particle at 16A resolution. J Mol Biol. 1984;176:55–75.
  • McGee SL, Hargreaves M. Histone modifications and exercise adaptations. J Appl Physiol. 2011;110:258–263.
  • Zhang T, Cooper S, Brockdorff N. The interplay of histone modifications - writers that read. EMBO Rep. 2015;16:1467–1481.
  • Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21:381–395.
  • Hussain MU. Micro-RNAs (miRNAs): genomic organisation, biogenesis and mode of action. Cell Tissue Res. 2012;349:405–413.
  • Widmann M, Nieß AM, Munz B. Physical exercise and epigenetic modifications in skeletal muscle. Sport Med. 2019.
  • McClay JL, Shabalin AA, Dozmorov MG, et al. High density methylation QTL analysis in human blood via next-generation sequencing of the methylated genomic DNA fraction. Genome Biol. 2015;16:291.
  • Hannon E, Spiers H, Viana J, et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat Neurosci. 2015;19:48.
  • Banovich NE, Lan X, McVicker G, et al. Methylation QTLs are associated with coordinated changes in transcription factor binding, histone modifications, and gene expression levels. PLoS Genet. 2014;10:e1004663.
  • Fraga MF, Ballestar E, Paz MF, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A. 2005;102:10–15.
  • van Dongen J, Nivard MG, Willemsen G, et al. Genetic and environmental influences interact with age and sex in shaping the human methylome. Nat Commun. 2016;7:11115.
  • Zimmer P, Bloch W. Physical exercise and epigenetic adaptations of the cardiovascular system. Herz. 2015;40:353–360.
  • Barrès R, Yan J, Egan B, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15:405–411.
  • Fabre O, Ingerslev LR, Garde C, et al. Exercise training alters the genomic response to acute exercise in human adipose tissue. Epigenomics. 2018;10:1033–1050.
  • Russell AP, Lamon S, Boon H, et al. Regulation of miRNAs in human skeletal muscle following acute endurance exercise and short-term endurance training. J Physiol. 2013;591:4637–4653.
  • Moher D, Shamseer L, Clarke M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev. 2015;4:1–19.
  • Alibegovic AC, Sonne MP, Højbjerre L, et al. Insulin resistance induced by physical inactivity is associated with multiple transcriptional changes in skeletal muscle in young men. Am J Physiol - Endocrinol Metab. 2010;299:752–763.
  • Bajpeyi S, Covington JD, Taylor EM, et al. Skeletal muscle PGC1alpha −1 nucleosome position and −260nt DNA methylation determine exercise response and prevent ectopic lipid accumulation in men. Endocrinology. 2017;158:2190–2199.
  • 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.
  • Nitert MD, Dayeh T, Volkov P, et al. Impact of an exercise intervention on DNA methylation in skeletal muscle from first-degree relatives of patients with type 2 diabetes. Diabetes. 2012;61:3322–3332.
  • Seaborne RA, Strauss J, Cocks M, et al. Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8:1898.
  • Robinson MM, Dasari S, Konopka AR, et al. Enhanced protein translation underlies improved metabolic and physical adaptations to different exercise training modes in young and old humans. Cell Metab. 2017;25:581–592.
  • Lindholm ME, Giacomello S, Werne Solnestam B, et al. The impact of endurance training on human skeletal muscle memory, global isoform expression and novel transcripts. PLOS Genet. 2016;12:e1006294.
  • McGee SL, Fairlie E, Garnham AP, et al. Exercise-induced histone modifications in human skeletal muscle. J Physiol. 2009;587:5951–5958.
  • Nielsen S, Scheele C, Yfanti C, et al. Muscle specific microRNAs are regulated by endurance exercise in human skeletal muscle. J Physiol. 2010;588:4029–4037.
  • Ringholm S, Biensø RS, Kiilerich K, et al. Bed rest reduces metabolic protein content and abolishes exercise-induced mRNA responses in human skeletal muscle. Am J Physiol Endocrinol Metab. 2011;301:E649–58.
  • Rivas DA, Lessard SJ, Rice NP, et al. Diminished skeletal muscle microRNA expression with aging is associated with attenuated muscle plasticity and inhibition of IGF-1 signaling. Faseb J. 2014;28:4133–4147.
  • D’Souza RF, Markworth JF, Aasen KMM, et al. Acute resistance exercise modulates microRNA expression profiles: combined tissue and circulatory targeted analyses. PLoS One. 2017;12e0181594.
  • Fyfe JJ, Bishop DJ, Zacharewicz E, et al. Concurrent exercise incorporating high-intensity interval or continuous training modulates mTORC1 signaling and microRNA expression in human skeletal muscle. Am J Physiol - Regul Integr Comp Physiol. 2016;310:R1297–311.
  • Keller P, Vollaard NBJ, Gustafsson T, et al. A transcriptional map of the impact of endurance exercise training on skeletal muscle phenotype. J Appl Physiol. 2011;110:46–59.
  • Zhang T, Birbrair A, Wang ZM, et al. Improved knee extensor strength with resistance training associates with muscle specific miRNAs in older adults. Exp Gerontol. 2015;62:7–13.
  • Mueller M, Breil FA, Lurman G, et al. Different molecular and structural adaptations with eccentric and conventional strength training in elderly men and women. Gerontology. 2011;57:528–538.
  • D’Souza RF, Bjørnsen T, Zeng N, et al. MicroRNAs in muscle: characterizing the powerlifter phenotype. Front Physiol. 2017;8:1–12.
  • Chen J-F, Mandel EM, Thomson JM, et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet. 2006;38:228–233.
  • Sun X, Lin J, Zhang Y, et al. MicroRNA-181b improves glucose homeostasis and insulin sensitivity by regulating endothelial function in white adipose tissue. Circ Res. 2016;118:810–821.
  • Mercatelli N, Fittipaldi S, De Paola E, et al. MiR-23-TrxR1 as a novel molecular axis in skeletal muscle differentiation. Sci Rep. 2017;7:7219.
  • Brunet-Vega A, Pericay C, Quílez ME, et al. Variability in microRNA recovery from plasma: comparison of five commercial kits. Anal Biochem. 20154;88:28–35.
  • Whiley PJ, De La Hoya M, Thomassen M, et al. Comparison of mRNA splicing assay protocols across multiple laboratories: recommendations for best practice in standardized clinical testing. Clin Chem. 2014.
  • Russell AP, Lamon S. Exercise, skeletal muscle and circulating microRNAs. Prog Mol Biol Transl Sci. 2015;135:471–496.
  • Ogasawara R, Akimoto T, Umeno T, et al. MicroRNA expression profiling in skeletal muscle reveals different regulatory patterns in high and low responders to resistance training. Physiol Genomics. 2016;48:320–324.
  • Zacharewicz E, Della Gatta P, Reynolds J, et al. Identification of microRNAs linked to regulators of muscle protein synthesis and regeneration in young and old skeletal muscle. PLoS One. 2014;9(12):e114009.
  • McLean CS, Mielke C, Cordova JM, et al. Gene and microRNA expression responses to exercise; relationship with insulin sensitivity. PLoS One. 2015;10:1–14.
  • Russell AP, Wallace MA, Kalanon M, et al. Striated muscle activator of Rho signalling (STARS) is reduced in ageing human skeletal muscle and targeted by miR-628-5p. Acta Physiol. 2017;220:263–274.
  • Hodson N, Philp A. The importance of mTOR trafficking for human skeletal muscle translational control. Exerc Sport Sci Rev. 2019;47(1):46–53.
  • Krist B, Florczyk U, Pietraszek-Gremplewicz K, et al. The role of miR-378a in metabolism, angiogenesis, and muscle biology. Int J Endocrinol. 2015;2015:281756.
  • Horak M, Novak J, Bienertova-Vasku J. Muscle-specific microRNAs in skeletal muscle development. Dev Biol. 2016;410:1–13.
  • Siracusa J, Koulmann N, Banzet S. Circulating myomiRs: a new class of biomarkers to monitor skeletal muscle in physiology and medicine. J Cachexia Sarcopenia Muscle. 2018;9:20–27.
  • Ramos AE, Lo C, Estephan LE, et al. Specific circulating microRNAs display dose-dependent responses to variable intensity and duration of endurance exercise. Am J Physiol Heart Circ Physiol. 2018;315:H273–83.
  • Nohata N, Hanazawa T, Enokida H, et al. microRNA-1/133a and microRNA-206/133b clusters: dysregulation and functional roles in human cancers. Oncotarget. 2012;3:9–21.
  • Lu L, Zhou L, Chen EZ, et al. A novel YY1-miR-1 regulatory circuit in skeletal myogenesis revealed by genome-wide prediction of YY1-miRNA network. PLoS One. 2012;7: e27596.
  • Tam S, Tsao MS, McPherson JD. Optimization of miRNA-seq data preprocessing. Brief Bioinform. 2015;16:950–963.
  • Chazaud B. Inflammation during skeletal muscle regeneration and tissue remodeling: application to exercise-induced muscle damage management. Immunol Cell Biol. 2016;94:140–145.
  • Malm C, Nyberg P, Engstr M, et al. Muscular adaptation to physical stress is of significant importance for normal muscular development and function. without stimulation from physical activity. 2000;243–262.
  • Bheda P, Schneider R. Epigenetics reloaded: the single-cell revolution. Trends Cell Biol. 2014;24:712–723.
  • Riley RD, Higgins JPT, Deeks JJ. Interpretation of random effects meta-analyses. Bmj. 2011;342:964–967.

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.