Skip to main content
Erschienen in: Sports Medicine 4/2019

19.02.2019 | Review Article

Physical Exercise and Epigenetic Modifications in Skeletal Muscle

verfasst von: Manuel Widmann, Andreas M. Nieß, Barbara Munz

Erschienen in: Sports Medicine | Ausgabe 4/2019

Einloggen, um Zugang zu erhalten

Abstract

Physical activity and sports play major roles in the overall health status of humans. It is well known that regular exercise helps to lower the risk for a broad variety of health problems, such as cardiovascular disease, type 2 diabetes, and cancer. Being physically active induces a wide variety of molecular adaptations, for example fiber type switches or other metabolic alterations, in skeletal muscle tissue. These adaptations are based on exercise-induced changes to the skeletal muscle transcriptome. Understanding their nature is crucial to improve the development of exercise-based therapeutic strategies. Recent research indicates that specifically epigenetic mechanisms, i.e., pathways that induce changes in gene expression patterns without altering the DNA base sequence, might play a major role in controlling skeletal muscle transcriptional patterns. Epigenetic mechanisms include DNA and histone modifications, as well as expression of specific microRNAs. They can be modulated by environmental factors or external stimuli, such as exercise, and eventually induce specific and fine-tuned changes to the transcriptional response. In this review, we highlight current knowledge on epigenetic changes induced in exercising skeletal muscle, their target genes, and resulting phenotypic changes. In addition, we raise the question of whether epigenetic modifications might serve as markers for the design and management of optimized and individualized training protocols, as prognostic tools to predict training adaptation, or even as targets for the design of “exercise mimics”.
Literatur
1.
Zurück zum Zitat Hechanova RL, Wegler JL, Forest CP. Exercise: a vitally important prescription. JAAPA. 2017;30:17–22.CrossRefPubMed Hechanova RL, Wegler JL, Forest CP. Exercise: a vitally important prescription. JAAPA. 2017;30:17–22.CrossRefPubMed
2.
Zurück zum Zitat Vina J, Sanchis-Gomar F, Martinez-Bello V, Gomez-Cabrera MC. Exercise acts as a drug; the pharmacological benefits of exercise. Br J Pharmacol. 2012;167:1–12.CrossRefPubMedPubMedCentral Vina J, Sanchis-Gomar F, Martinez-Bello V, Gomez-Cabrera MC. Exercise acts as a drug; the pharmacological benefits of exercise. Br J Pharmacol. 2012;167:1–12.CrossRefPubMedPubMedCentral
3.
4.
Zurück zum Zitat Sarzynski MA, Ghosh S, Bouchard C. Genomic and transcriptomic predictors of response levels to endurance exercise training. J Physiol. 2017;595:2931–9.CrossRefPubMed Sarzynski MA, Ghosh S, Bouchard C. Genomic and transcriptomic predictors of response levels to endurance exercise training. J Physiol. 2017;595:2931–9.CrossRefPubMed
5.
Zurück zum Zitat Ntanasis-Stathopoulos J, Tzanninis JG, Philippou A, Koutsilieris M. Epigenetic regulation on gene expression induced by physical exercise. J Musculoskelet Neuronal Interact. 2013;13:133–46.PubMed Ntanasis-Stathopoulos J, Tzanninis JG, Philippou A, Koutsilieris M. Epigenetic regulation on gene expression induced by physical exercise. J Musculoskelet Neuronal Interact. 2013;13:133–46.PubMed
6.
Zurück zum Zitat Ehlert T, Simon P, Moser DA. Epigenetics in sports. Sports Med. 2013;4:93–110.CrossRef Ehlert T, Simon P, Moser DA. Epigenetics in sports. Sports Med. 2013;4:93–110.CrossRef
7.
Zurück zum Zitat Pareja-Galeano H, Sanchis-Gomar F, García-Giménez JL. Physical exercise and epigenetic modulation: elucidating intricate mechanisms. Sports Med. 2014;44:429–36.CrossRefPubMed Pareja-Galeano H, Sanchis-Gomar F, García-Giménez JL. Physical exercise and epigenetic modulation: elucidating intricate mechanisms. Sports Med. 2014;44:429–36.CrossRefPubMed
8.
Zurück zum Zitat Soci UPR, Melo SFS, Gomes JLP, Silveira AC, Nóbrega C, de Oliveira EM. Exercise training and epigenetic regulation: multilevel modification and regulation of gene expression. Adv Exp Med Biol. 2017;1000:281–322.CrossRefPubMed Soci UPR, Melo SFS, Gomes JLP, Silveira AC, Nóbrega C, de Oliveira EM. Exercise training and epigenetic regulation: multilevel modification and regulation of gene expression. Adv Exp Med Biol. 2017;1000:281–322.CrossRefPubMed
9.
Zurück zum Zitat Ecker S, Pancaldi V, Valencia A, Beck S, Paul DS. Epigenetic and transcriptional variability shape phenotypic plasticity. Bioessays. 2018;40(2):1700148.CrossRef Ecker S, Pancaldi V, Valencia A, Beck S, Paul DS. Epigenetic and transcriptional variability shape phenotypic plasticity. Bioessays. 2018;40(2):1700148.CrossRef
11.
Zurück zum Zitat Meier K, Recillas-Targa F. New insights on the role of DNA methylation from a global view. Front Biosci (Landmark Ed). 2017;22:644–68.CrossRef Meier K, Recillas-Targa F. New insights on the role of DNA methylation from a global view. Front Biosci (Landmark Ed). 2017;22:644–68.CrossRef
13.
Zurück zum Zitat Delatte B, Deplus R, Fuks F. Playing TETris with DNA modifications. EMBO J. 2014;33:1198–211.CrossRef Delatte B, Deplus R, Fuks F. Playing TETris with DNA modifications. EMBO J. 2014;33:1198–211.CrossRef
14.
Zurück zum Zitat Ling C, Rönn T. Epigenetic adaptation to regular exercise in humans. Drug Discov Today. 2014;19:1015–8.CrossRefPubMed Ling C, Rönn T. Epigenetic adaptation to regular exercise in humans. Drug Discov Today. 2014;19:1015–8.CrossRefPubMed
15.
Zurück zum Zitat Barrès R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15:405–11.CrossRefPubMed Barrès R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15:405–11.CrossRefPubMed
16.
Zurück zum Zitat Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, 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;61:3322–32.CrossRefPubMedPubMedCentral Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, 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;61:3322–32.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Kanzleiter T, Jähnert M, Schulze G, Selbig J, Hallahan N, Schwenk RW, et al. Exercise training alters DNA methylation patterns in genes related to muscle growth and differentiation in mice. Am J Physiol Endocrinol Metab. 2015;308(10):E912–20.CrossRefPubMed Kanzleiter T, Jähnert M, Schulze G, Selbig J, Hallahan N, Schwenk RW, et al. Exercise training alters DNA methylation patterns in genes related to muscle growth and differentiation in mice. Am J Physiol Endocrinol Metab. 2015;308(10):E912–20.CrossRefPubMed
18.
Zurück zum Zitat Lindholm ME, Marabita F, Gomez-Cabrero D, Rundqvist H, Ekström TJ, Tegnér J, et al. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics. 2014;9:1557–69.CrossRefPubMedPubMedCentral Lindholm ME, Marabita F, Gomez-Cabrero D, Rundqvist H, Ekström TJ, Tegnér J, et al. An integrative analysis reveals coordinated reprogramming of the epigenome and the transcriptome in human skeletal muscle after training. Epigenetics. 2014;9:1557–69.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Rowlands DS, Page RA, Sukala WR, Giri M, Ghimbovschi SD, Hayat I, et al. Multi-omic integrated networks connect DNA methylation and miRNA with skeletal muscle plasticity to chronic exercise in type 2 diabetic obesity. Physiol Genom. 2014;46:747–65.CrossRef Rowlands DS, Page RA, Sukala WR, Giri M, Ghimbovschi SD, Hayat I, et al. Multi-omic integrated networks connect DNA methylation and miRNA with skeletal muscle plasticity to chronic exercise in type 2 diabetic obesity. Physiol Genom. 2014;46:747–65.CrossRef
21.
Zurück zum Zitat King-Himmelreich TS, Schramm S, Wolters MC, Schmetzer J, Möser CV, Knothe C, et al. The impact of endurance exercise on global and AMPK gene-specific DNA methylation. Biochem Biophys Res Commun. 2016;474:284–90.CrossRefPubMed King-Himmelreich TS, Schramm S, Wolters MC, Schmetzer J, Möser CV, Knothe C, et al. The impact of endurance exercise on global and AMPK gene-specific DNA methylation. Biochem Biophys Res Commun. 2016;474:284–90.CrossRefPubMed
22.
Zurück zum Zitat Lane SC, Camera DM, Lassiter DG, Areta JL, Bird SR, Yeo WK, et al. Effects of sleeping with reduced carbohydrate availability on acute training responses. J Appl Physiol (1985). 2015;119(6):643–55.CrossRef Lane SC, Camera DM, Lassiter DG, Areta JL, Bird SR, Yeo WK, et al. Effects of sleeping with reduced carbohydrate availability on acute training responses. J Appl Physiol (1985). 2015;119(6):643–55.CrossRef
23.
Zurück zum Zitat Laker RC, Lillard TS, Okutsu M, Zhang M, Hoehn KL, Connelly JJ, et al. Exercise prevents maternal high-fat diet-induced hypermethylation of the Pgc-1α gene and age-dependent metabolic dysfunction in the offspring. Diabetes. 2014;63:1605–11.CrossRefPubMedPubMedCentral Laker RC, Lillard TS, Okutsu M, Zhang M, Hoehn KL, Connelly JJ, et al. Exercise prevents maternal high-fat diet-induced hypermethylation of the Pgc-1α gene and age-dependent metabolic dysfunction in the offspring. Diabetes. 2014;63:1605–11.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Lochmann TL, Thomas RR, Bennett JP Jr, Taylor SM. Epigenetic modifications of the PGC-1α promoter during exercise induced expression in mice. PLoS One. 2015;10(6):e0129647.CrossRefPubMedPubMedCentral Lochmann TL, Thomas RR, Bennett JP Jr, Taylor SM. Epigenetic modifications of the PGC-1α promoter during exercise induced expression in mice. PLoS One. 2015;10(6):e0129647.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Kasch J, Kanzleiter I, Saussenthaler S, Schürmann A, Keijer J, van Schothorst E, et al. Insulin sensitivity linked skeletal muscle Nr4a1 DNA methylation is programmed by the maternal diet and modulated by voluntary exercise in mice. J Nutr Biochem. 2018;57:86–92.CrossRefPubMed Kasch J, Kanzleiter I, Saussenthaler S, Schürmann A, Keijer J, van Schothorst E, et al. Insulin sensitivity linked skeletal muscle Nr4a1 DNA methylation is programmed by the maternal diet and modulated by voluntary exercise in mice. J Nutr Biochem. 2018;57:86–92.CrossRefPubMed
26.
Zurück zum Zitat Nguyen A, Duquette N, Mamarbachi M, Thorin E. Epigenetic regulatory effect of exercise on glutathione peroxidase 1 expression in the skeletal muscle of severely dyslipidemic mice. PLoS One. 2016;11(3):e0151526.CrossRefPubMedPubMedCentral Nguyen A, Duquette N, Mamarbachi M, Thorin E. Epigenetic regulatory effect of exercise on glutathione peroxidase 1 expression in the skeletal muscle of severely dyslipidemic mice. PLoS One. 2016;11(3):e0151526.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Seaborne RA, Strauss J, Cocks M, Shepherd S, O’Brien TD, Someren KAV, et al. Methylome of human skeletal muscle after acute and chronic resistance exercise training, detraining and retraining. Sci Data. 2018;5:180213.CrossRefPubMedPubMedCentral Seaborne RA, Strauss J, Cocks M, Shepherd S, O’Brien TD, Someren KAV, et al. Methylome of human skeletal muscle after acute and chronic resistance exercise training, detraining and retraining. Sci Data. 2018;5:180213.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Seaborne RA, Strauss J, Cocks M, Shepherd S, O’Brien TD, van Someren KA, et al. Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8:1898.CrossRefPubMedPubMedCentral Seaborne RA, Strauss J, Cocks M, Shepherd S, O’Brien TD, van Someren KA, et al. Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8:1898.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Laker RC, Garde C, Camera DM, Smiles WJ, Zierath JR, Hawley JA, et al. Transcriptomic and epigenetic responses to short-term nutrient-exercise stress in humans. Sci Rep. 2017;7:15134.CrossRefPubMedPubMedCentral Laker RC, Garde C, Camera DM, Smiles WJ, Zierath JR, Hawley JA, et al. Transcriptomic and epigenetic responses to short-term nutrient-exercise stress in humans. Sci Rep. 2017;7:15134.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Brown WM. Exercise-associated DNA methylation change in skeletal muscle and the importance of imprinted genes: a bioinformatics meta-analysis. Br J Sports Med. 2015;49:1567–78.CrossRefPubMed Brown WM. Exercise-associated DNA methylation change in skeletal muscle and the importance of imprinted genes: a bioinformatics meta-analysis. Br J Sports Med. 2015;49:1567–78.CrossRefPubMed
31.
Zurück zum Zitat Carter HN, Pauly M, Tryon LD, Hood DA. Effect of contractile activity on PGC-1α transcription in young and aged skeletal muscle. J Appl Physiol. 1985;2018(124):1605–15. Carter HN, Pauly M, Tryon LD, Hood DA. Effect of contractile activity on PGC-1α transcription in young and aged skeletal muscle. J Appl Physiol. 1985;2018(124):1605–15.
32.
Zurück zum Zitat Fisher AG, Seaborne RA, Hughes TM, Gutteridge A, Stewart C, Coulson JM, et al. Transcriptomic and epigenetic regulation of disuse atrophy and the return to activity in skeletal muscle. FASEB J. 2017;31:5268–82.CrossRefPubMed Fisher AG, Seaborne RA, Hughes TM, Gutteridge A, Stewart C, Coulson JM, et al. Transcriptomic and epigenetic regulation of disuse atrophy and the return to activity in skeletal muscle. FASEB J. 2017;31:5268–82.CrossRefPubMed
33.
Zurück zum Zitat Stephens NA, Brouwers B, Eroshkin AM, Yi F, Cornnell HH, Meyer C, et al. Exercise response variations in skeletal muscle PCr recovery rate and insulin sensitivity relate to muscle epigenomic profiles in individuals with type 2 diabetes. Diabetes Care. 2018;41:2245–54.CrossRefPubMed Stephens NA, Brouwers B, Eroshkin AM, Yi F, Cornnell HH, Meyer C, et al. Exercise response variations in skeletal muscle PCr recovery rate and insulin sensitivity relate to muscle epigenomic profiles in individuals with type 2 diabetes. Diabetes Care. 2018;41:2245–54.CrossRefPubMed
34.
Zurück zum Zitat Bajpeyi S, Covington JD, Taylor EM, Stewart LK, Galgani JE, Henagan TM. Skeletal muscle PGC1α -1 nucleosome position and -260 nt DNA methylation determine exercise response and prevent ectopic lipid accumulation in men. Endocrinology. 2017;158(7):2190–9.CrossRefPubMedPubMedCentral Bajpeyi S, Covington JD, Taylor EM, Stewart LK, Galgani JE, Henagan TM. Skeletal muscle PGC1α -1 nucleosome position and -260 nt DNA methylation determine exercise response and prevent ectopic lipid accumulation in men. Endocrinology. 2017;158(7):2190–9.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Terruzzi I, Senesi P, Montesano A, La Torre A, Alberti G, Benedini S, et al. Genetic polymorphisms of the enzymes involved in DNA methylation and synthesis in elite athletes. Physiol Genom. 2011;43:965–73.CrossRef Terruzzi I, Senesi P, Montesano A, La Torre A, Alberti G, Benedini S, et al. Genetic polymorphisms of the enzymes involved in DNA methylation and synthesis in elite athletes. Physiol Genom. 2011;43:965–73.CrossRef
36.
Zurück zum Zitat Gates LA, Foulds CE, O’Malley BW. Histone marks in the ‘driver’s seat’: functional roles in steering the transcription cycle. Trends Biochem Sci. 2017;42:977–89.CrossRefPubMedPubMedCentral Gates LA, Foulds CE, O’Malley BW. Histone marks in the ‘driver’s seat’: functional roles in steering the transcription cycle. Trends Biochem Sci. 2017;42:977–89.CrossRefPubMedPubMedCentral
37.
38.
Zurück zum Zitat McGee SL, Hargreaves M. Histone modifications and exercise adaptations. J Appl Physiol. 1985;2011(110):258–63. McGee SL, Hargreaves M. Histone modifications and exercise adaptations. J Appl Physiol. 1985;2011(110):258–63.
39.
Zurück zum Zitat Potthoff MJ, Wu H, Arnold MA, Shelton JM, Backs J, McAnally J, et al. Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. J Clin Investig. 2007;117:2459–67.CrossRefPubMed Potthoff MJ, Wu H, Arnold MA, Shelton JM, Backs J, McAnally J, et al. Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. J Clin Investig. 2007;117:2459–67.CrossRefPubMed
40.
Zurück zum Zitat Smith JA, Kohn TA, Chetty AK, Ojuka EO. CaMK activation during exercise is required for histone hyperacetylation and MEF2A binding at the MEF2 site on the Glut4 gene. Am J Physiol Endocrinol Metab. 2008;295:E698–704.CrossRefPubMed Smith JA, Kohn TA, Chetty AK, Ojuka EO. CaMK activation during exercise is required for histone hyperacetylation and MEF2A binding at the MEF2 site on the Glut4 gene. Am J Physiol Endocrinol Metab. 2008;295:E698–704.CrossRefPubMed
41.
Zurück zum Zitat Joseph JS, Ayeleso AO, Mukwevho E. Exercise increases hyper-acetylation of histones on the Cis-element of NRF-1 binding to the Mef2a promoter: implications on type 2 diabetes. Biochem Biophys Res Commun. 2017;486:83–7.CrossRefPubMed Joseph JS, Ayeleso AO, Mukwevho E. Exercise increases hyper-acetylation of histones on the Cis-element of NRF-1 binding to the Mef2a promoter: implications on type 2 diabetes. Biochem Biophys Res Commun. 2017;486:83–7.CrossRefPubMed
42.
Zurück zum Zitat Hong S, Zhou W, Fang B, Lu W, Loro E, Damle M, et al. Dissociation of muscle insulin sensitivity from exercise endurance in mice by HDAC3 depletion. Nat Med. 2017;23:223–34.CrossRefPubMed Hong S, Zhou W, Fang B, Lu W, Loro E, Damle M, et al. Dissociation of muscle insulin sensitivity from exercise endurance in mice by HDAC3 depletion. Nat Med. 2017;23:223–34.CrossRefPubMed
43.
Zurück zum Zitat Ohsawa I, Konno R, Masuzawa R, Kawano F. Amount of daily exercise is an essential stimulation to alter the epigenome of skeletal muscle in rats. J Appl Physiol (1985). 2018;125(4):1097–104.CrossRef Ohsawa I, Konno R, Masuzawa R, Kawano F. Amount of daily exercise is an essential stimulation to alter the epigenome of skeletal muscle in rats. J Appl Physiol (1985). 2018;125(4):1097–104.CrossRef
44.
Zurück zum Zitat Barreiro E, Sznajder JI. Epigenetic regulation of muscle phenotype and adaptation: a potential role in COPD muscle dysfunction. J Appl Physiol. 1985;2013(114):1263–72. Barreiro E, Sznajder JI. Epigenetic regulation of muscle phenotype and adaptation: a potential role in COPD muscle dysfunction. J Appl Physiol. 1985;2013(114):1263–72.
45.
Zurück zum Zitat Thalacker-Mercer A, Stec M, Cui X, Cross J, Windham S, Bamman M. Cluster analysis reveals differential transcript profiles associated with resistance training-induced human skeletal muscle hypertrophy. Physiol Genom. 2013;45:499–507.CrossRef Thalacker-Mercer A, Stec M, Cui X, Cross J, Windham S, Bamman M. Cluster analysis reveals differential transcript profiles associated with resistance training-induced human skeletal muscle hypertrophy. Physiol Genom. 2013;45:499–507.CrossRef
47.
Zurück zum Zitat Pandorf CE, Haddad F, Wright C, Bodell PW, Baldwin KM. Differential epigenetic modifications of histones at the myosin heavy chain genes in fast and slow skeletal muscle fibers and in response to muscle unloading. Am J Physiol Cell Physiol. 2009;297:C6–16.CrossRefPubMedPubMedCentral Pandorf CE, Haddad F, Wright C, Bodell PW, Baldwin KM. Differential epigenetic modifications of histones at the myosin heavy chain genes in fast and slow skeletal muscle fibers and in response to muscle unloading. Am J Physiol Cell Physiol. 2009;297:C6–16.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Kawano F, Nimura K, Ishino S, Nakai N, Nakata K, Ohira Y. Differences in histone modifications between slow- and fast-twitch muscle of adult rats and following overload, denervation, or valproic acid administration. J Appl Physiol. 1985;2015(119):1042–52. Kawano F, Nimura K, Ishino S, Nakai N, Nakata K, Ohira Y. Differences in histone modifications between slow- and fast-twitch muscle of adult rats and following overload, denervation, or valproic acid administration. J Appl Physiol. 1985;2015(119):1042–52.
49.
Zurück zum Zitat Willkomm L, Gehlert S, Jacko D, Schiffer T, Bloch W. p38 MAPK activation and H3K4 trimethylation is decreased by lactate in vitro and high intensity resistance training in human skeletal muscle. PLoS One. 2017;12(5):e0176609.CrossRefPubMedPubMedCentral Willkomm L, Gehlert S, Jacko D, Schiffer T, Bloch W. p38 MAPK activation and H3K4 trimethylation is decreased by lactate in vitro and high intensity resistance training in human skeletal muscle. PLoS One. 2017;12(5):e0176609.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Ribich S, Harvey D, Copeland RA. Drug discovery and chemical biology of cancer epigenetics. Cell Chem Biol. 2017;24:1120–47.CrossRefPubMed Ribich S, Harvey D, Copeland RA. Drug discovery and chemical biology of cancer epigenetics. Cell Chem Biol. 2017;24:1120–47.CrossRefPubMed
52.
Zurück zum Zitat Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science. 2007;318(5858):1931–4.CrossRefPubMed Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation. Science. 2007;318(5858):1931–4.CrossRefPubMed
53.
Zurück zum Zitat Polakovičová M, Musil P, Laczo E, Hamar D, Kyselovič J. Circulating microRNAs as potential biomarkers of exercise response. Int J Mol Sci. 2016;17(10):E1553.CrossRefPubMed Polakovičová M, Musil P, Laczo E, Hamar D, Kyselovič J. Circulating microRNAs as potential biomarkers of exercise response. Int J Mol Sci. 2016;17(10):E1553.CrossRefPubMed
54.
Zurück zum Zitat Silva GJJ, Bye A, El Azzouzi H, Wisløff U. MicroRNAs as important regulators of exercise adaptation. Prog Cardiovasc Dis. 2017;2017(60):130–51.CrossRef Silva GJJ, Bye A, El Azzouzi H, Wisløff U. MicroRNAs as important regulators of exercise adaptation. Prog Cardiovasc Dis. 2017;2017(60):130–51.CrossRef
55.
Zurück zum Zitat Wei JW, Huang K, Yang C, Kang CS. Non-coding RNAs as regulators in epigenetics. Oncol Rep. 2017;37:3–9.CrossRefPubMed Wei JW, Huang K, Yang C, Kang CS. Non-coding RNAs as regulators in epigenetics. Oncol Rep. 2017;37:3–9.CrossRefPubMed
56.
Zurück zum Zitat Paul P, Chakraborty A, Sarkar D, Langthasa M, Rahman M, Bari M, et al. Interplay between miRNAs and human diseases. J Cell Physiol. 2018;233:2007–18.CrossRefPubMed Paul P, Chakraborty A, Sarkar D, Langthasa M, Rahman M, Bari M, et al. Interplay between miRNAs and human diseases. J Cell Physiol. 2018;233:2007–18.CrossRefPubMed
57.
Zurück zum Zitat Daskalakis NP, Provost AC, Hunter RG, Guffanti G. Noncoding RNAs: stress, glucocorticoids, and posttraumatic stress disorder. Biol Psychol. 2018;83:849–65.CrossRef Daskalakis NP, Provost AC, Hunter RG, Guffanti G. Noncoding RNAs: stress, glucocorticoids, and posttraumatic stress disorder. Biol Psychol. 2018;83:849–65.CrossRef
59.
Zurück zum Zitat Luo W, Nie Q, Zhang X. MicroRNAs involved in skeletal muscle differentiation. J Genet Genom. 2013;40:107–16.CrossRef Luo W, Nie Q, Zhang X. MicroRNAs involved in skeletal muscle differentiation. J Genet Genom. 2013;40:107–16.CrossRef
60.
Zurück zum Zitat Horak M, Novak J, Bienertova-Vasku J. Muscle-specific microRNAs in skeletal muscle development. Dev Biol. 2016;410(1):1–13.CrossRefPubMed Horak M, Novak J, Bienertova-Vasku J. Muscle-specific microRNAs in skeletal muscle development. Dev Biol. 2016;410(1):1–13.CrossRefPubMed
61.
Zurück zum Zitat Safdar A, Abadi A, Akhtar M, Hettinga BP, Tarnopolsky MA. miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice. PLoS One. 2009;4(5):e5610.CrossRefPubMedPubMedCentral Safdar A, Abadi A, Akhtar M, Hettinga BP, Tarnopolsky MA. miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice. PLoS One. 2009;4(5):e5610.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Aoi W, Naito Y, Mizushima K, Takanami Y, Kawai Y, Ichikawa H, et al. The microRNA miR-696 regulates PGC-1 alpha in mouse skeletal muscle in response to physical activity. Am J Physiol Endocrinol Metab. 2010;298:E799–806.CrossRefPubMed Aoi W, Naito Y, Mizushima K, Takanami Y, Kawai Y, Ichikawa H, et al. The microRNA miR-696 regulates PGC-1 alpha in mouse skeletal muscle in response to physical activity. Am J Physiol Endocrinol Metab. 2010;298:E799–806.CrossRefPubMed
63.
Zurück zum Zitat Yamamoto H, Morino K, Nishio Y, Ugi S, Yoshizaki T, Kashiwagi A, et al. MicroRNA-494 regulates mitochondrial biogenesis in skeletal muscle through mitochondrial transcription factor A and Forkhead box j3. Am J Physiol Endocrinol Metab. 2012;303:E1419–27.CrossRefPubMed Yamamoto H, Morino K, Nishio Y, Ugi S, Yoshizaki T, Kashiwagi A, et al. MicroRNA-494 regulates mitochondrial biogenesis in skeletal muscle through mitochondrial transcription factor A and Forkhead box j3. Am J Physiol Endocrinol Metab. 2012;303:E1419–27.CrossRefPubMed
64.
Zurück zum Zitat Keller P, Vollaard NB, Gustafsson T, Gallagher IJ, Sundberg CJ, Rankinen T, et al. A transcriptional map of the impact of endurance exercise training on skeletal muscle phenotype. J Appl Physiol. 1985;110:46–59.CrossRef Keller P, Vollaard NB, Gustafsson T, Gallagher IJ, Sundberg CJ, Rankinen T, et al. A transcriptional map of the impact of endurance exercise training on skeletal muscle phenotype. J Appl Physiol. 1985;110:46–59.CrossRef
65.
Zurück zum Zitat Nielsen S, Scheele C, Yfanti C, Akerström T, Nielsen AR, Pedersen BK, et al. Muscle specific microRNAs are regulated by endurance exercise in human skeletal muscle. J Physiol. 2010;588:4029–37.CrossRefPubMedPubMedCentral Nielsen S, Scheele C, Yfanti C, Akerström T, Nielsen AR, Pedersen BK, et al. Muscle specific microRNAs are regulated by endurance exercise in human skeletal muscle. J Physiol. 2010;588:4029–37.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Russell AP, Lamon S, Boon H, Wada S, Güller I, Brown EL, et al. Regulation of miRNAs in human skeletal muscle following acute endurance exercise and short-term endurance training. J Physiol. 2013;591:4637–53.CrossRefPubMedPubMedCentral Russell AP, Lamon S, Boon H, Wada S, Güller I, Brown EL, et al. Regulation of miRNAs in human skeletal muscle following acute endurance exercise and short-term endurance training. J Physiol. 2013;591:4637–53.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. J Appl Physiol. 1985;2007(102):306–13. McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. J Appl Physiol. 1985;2007(102):306–13.
69.
Zurück zum Zitat Mueller M, Breil FA, Lurman G, Klossner S, Flück M, Billeter R, et al. Different molecular and structural adaptations with eccentric and conventional strength training in elderly men and women. Gerontology. 2011;57:528–38.CrossRefPubMed Mueller M, Breil FA, Lurman G, Klossner S, Flück M, Billeter R, et al. Different molecular and structural adaptations with eccentric and conventional strength training in elderly men and women. Gerontology. 2011;57:528–38.CrossRefPubMed
70.
Zurück zum Zitat Davidsen PK, Gallagher IJ, Hartman JW, Tarnopolsky MA, Dela F, Helge JW, et al. High responders to resistance exercise training demonstrate differential regulation of skeletal muscle microRNA expression. J Appl Physiol. 1985;2011(110):309–17. Davidsen PK, Gallagher IJ, Hartman JW, Tarnopolsky MA, Dela F, Helge JW, et al. High responders to resistance exercise training demonstrate differential regulation of skeletal muscle microRNA expression. J Appl Physiol. 1985;2011(110):309–17.
71.
Zurück zum Zitat Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. Aging differentially affects human skeletal muscle microRNA expression at rest and after an anabolic stimulus of resistance exercise and essential amino acids. Am J Physiol Endocrinol Metab. 2008;295:E1333–40.CrossRefPubMedPubMedCentral Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. Aging differentially affects human skeletal muscle microRNA expression at rest and after an anabolic stimulus of resistance exercise and essential amino acids. Am J Physiol Endocrinol Metab. 2008;295:E1333–40.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Ringholm S, Biensø RS, Kiilerich K, Guadalupe-Grau A, Aachmann-Andersen NJ, Saltin B, 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.CrossRefPubMed Ringholm S, Biensø RS, Kiilerich K, Guadalupe-Grau A, Aachmann-Andersen NJ, Saltin B, 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.CrossRefPubMed
73.
Zurück zum Zitat Fyfe JJ, Bishop DJ, Zacharewicz E, Russell AP, Stepto NK. 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;31:R1297–311.CrossRef Fyfe JJ, Bishop DJ, Zacharewicz E, Russell AP, Stepto NK. 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;31:R1297–311.CrossRef
74.
Zurück zum Zitat Makarova JA, Maltseva DV, Galatenko VV, Abbasi A, Maximenko DG, Grigoriev AI, et al. Exercise immunology meets miRNAs. Exerc Immunol Rev. 2014;20:135–64.PubMed Makarova JA, Maltseva DV, Galatenko VV, Abbasi A, Maximenko DG, Grigoriev AI, et al. Exercise immunology meets miRNAs. Exerc Immunol Rev. 2014;20:135–64.PubMed
75.
Zurück zum Zitat Mooren FC, Viereck J, Krüger K, Thum T. Circulating microRNAs as potential biomarkers of aerobic exercise capacity. Am J Physiol Heart Circ Physiol. 2014;306(4):H557–63.CrossRefPubMed Mooren FC, Viereck J, Krüger K, Thum T. Circulating microRNAs as potential biomarkers of aerobic exercise capacity. Am J Physiol Heart Circ Physiol. 2014;306(4):H557–63.CrossRefPubMed
76.
Zurück zum Zitat Baggish AL, Hale A, Weiner RB, Lewis GD, Systrom D, Wang F, et al. Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J Physiol. 2011;589:3983–94.CrossRefPubMedPubMedCentral Baggish AL, Hale A, Weiner RB, Lewis GD, Systrom D, Wang F, et al. Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J Physiol. 2011;589:3983–94.CrossRefPubMedPubMedCentral
77.
Zurück zum Zitat Wardle SL, Bailey ME, Kilikevicius A, Malkova D, Wilson RH, Venckunas T, et al. Plasma microRNA levels differ between endurance and strength athletes. PLoS One. 2015;10(4):e0122107.CrossRefPubMedPubMedCentral Wardle SL, Bailey ME, Kilikevicius A, Malkova D, Wilson RH, Venckunas T, et al. Plasma microRNA levels differ between endurance and strength athletes. PLoS One. 2015;10(4):e0122107.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Sawada S, Kon M, Wada S, Ushida T, Suzuki K, Akimoto T. Profiling of circulating microRNAs after a bout of acute resistance exercise in humans. PLoS One. 2013;8(7):e70823.CrossRefPubMedPubMedCentral Sawada S, Kon M, Wada S, Ushida T, Suzuki K, Akimoto T. Profiling of circulating microRNAs after a bout of acute resistance exercise in humans. PLoS One. 2013;8(7):e70823.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Bye A, Røsjø H, Aspenes ST, Condorelli G, Omland T, Wisløff U. Circulating microRNAs and aerobic fitness—the HUNT-Study. PLoS One. 2013;8(2):e57496.CrossRefPubMedPubMedCentral Bye A, Røsjø H, Aspenes ST, Condorelli G, Omland T, Wisløff U. Circulating microRNAs and aerobic fitness—the HUNT-Study. PLoS One. 2013;8(2):e57496.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Nielsen S, Åkerström T, Rinnov A, Yfanti C, Scheele C, Pedersen BK, et al. The miRNA plasma signature in response to acute aerobic exercise and endurance training. PLoS One. 2014;9(2):e87308.CrossRefPubMedPubMedCentral Nielsen S, Åkerström T, Rinnov A, Yfanti C, Scheele C, Pedersen BK, et al. The miRNA plasma signature in response to acute aerobic exercise and endurance training. PLoS One. 2014;9(2):e87308.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Aoi W, Ichikawa H, Mune K, Tanimura Y, Mizushima K, Naito Y, et al. Muscle-enriched microRNA miR-486 decreases in circulation in response to exercise in young men. Front Physiol. 2013;4:80.CrossRefPubMedPubMedCentral Aoi W, Ichikawa H, Mune K, Tanimura Y, Mizushima K, Naito Y, et al. Muscle-enriched microRNA miR-486 decreases in circulation in response to exercise in young men. Front Physiol. 2013;4:80.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat Uhlemann M, Möbius-Winkler S, Fikenzer S, Adam J, Redlich M, Möhlenkamp S, et al. Circulating microRNA-126 increases after different forms of endurance exercise in healthy adults. Eur J Prev Cardiol. 2014;21:484–91.CrossRefPubMed Uhlemann M, Möbius-Winkler S, Fikenzer S, Adam J, Redlich M, Möhlenkamp S, et al. Circulating microRNA-126 increases after different forms of endurance exercise in healthy adults. Eur J Prev Cardiol. 2014;21:484–91.CrossRefPubMed
84.
Zurück zum Zitat Håkansson KEJ, Sollie O, Simons KH, Quax PHA, Jensen J, Nossent AY. Circulating Small non-coding RNAs as biomarkers for recovery after exhaustive or repetitive exercise. Front Physiol. 2018;9:1136.CrossRefPubMedPubMedCentral Håkansson KEJ, Sollie O, Simons KH, Quax PHA, Jensen J, Nossent AY. Circulating Small non-coding RNAs as biomarkers for recovery after exhaustive or repetitive exercise. Front Physiol. 2018;9:1136.CrossRefPubMedPubMedCentral
85.
Zurück zum Zitat Ramos AE, Lo C, Estephan LE, Tai YY, Tang Y, Zhao J, 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(2):H273–83.CrossRefPubMedPubMedCentral Ramos AE, Lo C, Estephan LE, Tai YY, Tang Y, Zhao J, 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(2):H273–83.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Denham J, Marques FZ, O’Brien BJ, Charchar FJ. Exercise: putting action into our epigenome. Sports Med. 2014;44:189–209.CrossRefPubMed Denham J, Marques FZ, O’Brien BJ, Charchar FJ. Exercise: putting action into our epigenome. Sports Med. 2014;44:189–209.CrossRefPubMed
87.
Zurück zum Zitat 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;15:603–16.CrossRefPubMedPubMedCentral 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;15:603–16.CrossRefPubMedPubMedCentral
Metadaten
Titel
Physical Exercise and Epigenetic Modifications in Skeletal Muscle
verfasst von
Manuel Widmann
Andreas M. Nieß
Barbara Munz
Publikationsdatum
19.02.2019
Verlag
Springer International Publishing
Erschienen in
Sports Medicine / Ausgabe 4/2019
Print ISSN: 0112-1642
Elektronische ISSN: 1179-2035
DOI
https://doi.org/10.1007/s40279-019-01070-4

Weitere Artikel der Ausgabe 4/2019

Sports Medicine 4/2019 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.