Nucleus Type-Specific DNA Methylomics Reveals Epigenetic "Memory" of Prior Adaptation in Skeletal Muscle

Yuan Wen, Cory M Dungan, C Brooks Mobley, Taylor Valentino, Ferdinand von Walden, Kevin A Murach, Yuan Wen, Cory M Dungan, C Brooks Mobley, Taylor Valentino, Ferdinand von Walden, Kevin A Murach

Abstract

Using a mouse model of conditional and inducible in vivo fluorescent myonuclear labeling (HSA-GFP), sorting purification of nuclei, low-input reduced representation bisulfite sequencing (RRBS), and a translatable and reversible model of exercise (progressive weighted wheel running, PoWeR), we provide the first nucleus type-specific epigenetic information on skeletal muscle adaptation and detraining. Adult (>4 mo) HSA-GFP mice performed PoWeR for 8 wk then detrained for 12 wk; age-matched untrained mice were used to control for the long duration of the study. Myonuclei and interstitial nuclei from plantaris muscles were isolated for RRBS. Relative to untrained, PoWeR caused similar myonuclear CpG hypo- and hyper-methylation of promoter regions and substantial hypomethylation in interstitial nuclear promoters. Over-representation analysis of promoters revealed a larger number of hyper- versus hypo-methylated pathways in both nuclear populations after training and evidence for reciprocal regulation of methylation between nucleus types, with hypomethylation of promoter regions in Wnt signaling-related genes in myonuclei and hypermethylation in interstitial nuclei. After 12 wk of detraining, promoter CpGs in documented muscle remodeling-associated genes and pathways that were differentially methylated immediately after PoWeR were persistently differentially methylated in myonuclei, along with long-term promoter hypomethylation in interstitial nuclei. No enduring gene expression changes in muscle tissue were observed using RNA-sequencing. Upon 4 wk of retraining, mice that trained previously grew more at the whole muscle and fiber type-specific cellular level than training naïve mice, with no difference in myonuclear number. Muscle nuclei have a methylation epi-memory of prior training that may augment muscle adaptability to retraining.

Keywords: epigenetics; exercise training; methylation; muscle memory; myonuclei; skeletal muscle.

© The Author(s) 2021. Published by Oxford University Press on behalf of American Physiological Society.

Figures

Graphical Abstract
Graphical Abstract
Figure 1.
Figure 1.
Myonuclear CpG promoter region DNA methylation changes in response to progressive weighted wheel running (PoWeR). (A) Study design schematic showing myonuclear labeling using the HSA-GFP mouse, PoWeR training (6M PoW) and detraining (9M PoW + DT), and age-matched untrained controls (6M UT and 9M UT), doxycycline treatment time points, and fluorescent activated cell sorting (FACS) of myonuclear and interstitial nuclear populations for downstream reduced representation bisulfite sequencing (RRBS) analysis. (B) Myonuclear and interstitial nuclear methylation in promoter regions after 8 wk of PoWeR. (C) Pathway analysis of hypomethylated promoters in myonuclei after PoWeR (relative to 6M UT). (D) Myonuclear genes with a hypomethylated promoter CpG in the NFκβ signaling pathway after PoWeR (FDR 

Figure 2.

Interstitial CpG promoter region DNA…

Figure 2.

Interstitial CpG promoter region DNA methylation changes in response to progressive weighted wheel…

Figure 2.
Interstitial CpG promoter region DNA methylation changes in response to progressive weighted wheel running (PoWeR). (A) Pathway analysis of hypomethylated promoters in interstitial nuclei after PoWeR (relative to 6M UT). (B) Interstitial nuclear genes with a hypomethylated promoter CpG in the fatty acid (FA) metabolism signaling pathway after PoWeR (FDR 

Figure 3.

Myonuclear promoter region methylation memory…

Figure 3.

Myonuclear promoter region methylation memory of prior PoWeR training may facilitate retraining adaptations.…

Figure 3.
Myonuclear promoter region methylation memory of prior PoWeR training may facilitate retraining adaptations. (A) Memory of promoter site-specific CpG methylation in myonuclear Pkd2l1 (FDR < 0.05). (B) Memory of promoter region-specific hypomethylation in myonuclear Gdf10. (C) Memory of promoter region-specific hypermethylation in myonuclear Eif1a, Pitx1, Sun2, Usp43, and Ski (FDR < 0.05). (D) Evidence for promoter region CpG methylation memory of previous PoWeR at the pathway level in myonuclei. (E) Study design schematic showing how mice were subjected to PoWeR for 8 wk, detrained for 3 mo, and retrained for 4 wk (PoW + DT + RT); age-matched mice that only trained for 4 wk served as controls (4 wk PoW). (F) Average nightly running volume during 4 wk of retraining. (G) Body weight at the time of being euthanized. (H) Absolute plantaris muscle weight in milligrams (mg; *P<0.05, directional t-test). (I) Plantaris muscle weight (mg) normalized to body weight in grams (g; *P<0.05, directional t-test). (J) Myosin heavy chain (MyHC) 2A cross sectional area (CSA) of gastrocnemius muscle fibers (*P<0.05, directional t-test). (K) MyHC 2A proportion. (L) MyHC 2A myonuclear number measured using dystrophin and DAPI. (M) Representative image of dystrophin, MyHC 2A fibers, and nuclei in 4 wk PoW and PoW + DT + RT muscles. Scale bar = 50 µm.
Comment in
Similar articles
Cited by
References
    1. Seaborne RA, Strauss J, Cocks Met al. . Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8(1): 1898. - PMC - PubMed
    1. 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):1–12. - PMC - PubMed
    1. Fisher AG, Seaborne RA, Hughes TMet al. . Transcriptomic and epigenetic regulation of disuse atrophy and the return to activity in skeletal muscle. FASEB J. 2017;31(12): 5268–5282. - PubMed
    1. von Walden F, Rea M, Mobley CBet al. . The myonuclear DNA methylome in response to an acute hypertrophic stimulus. Epigenetics. 2020;15(11): 1151–1162. - PMC - PubMed
    1. Iwata M, Englund DA, Wen Yet al. . A novel tetracycline-responsive transgenic mouse strain for skeletal muscle-specific gene expression. Skelet Musc. 2018;8(1): 33. - PMC - PubMed
Show all 84 references
Publication types
LinkOut - more resources
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 2.
Figure 2.
Interstitial CpG promoter region DNA methylation changes in response to progressive weighted wheel running (PoWeR). (A) Pathway analysis of hypomethylated promoters in interstitial nuclei after PoWeR (relative to 6M UT). (B) Interstitial nuclear genes with a hypomethylated promoter CpG in the fatty acid (FA) metabolism signaling pathway after PoWeR (FDR 

Figure 3.

Myonuclear promoter region methylation memory…

Figure 3.

Myonuclear promoter region methylation memory of prior PoWeR training may facilitate retraining adaptations.…

Figure 3.
Myonuclear promoter region methylation memory of prior PoWeR training may facilitate retraining adaptations. (A) Memory of promoter site-specific CpG methylation in myonuclear Pkd2l1 (FDR < 0.05). (B) Memory of promoter region-specific hypomethylation in myonuclear Gdf10. (C) Memory of promoter region-specific hypermethylation in myonuclear Eif1a, Pitx1, Sun2, Usp43, and Ski (FDR < 0.05). (D) Evidence for promoter region CpG methylation memory of previous PoWeR at the pathway level in myonuclei. (E) Study design schematic showing how mice were subjected to PoWeR for 8 wk, detrained for 3 mo, and retrained for 4 wk (PoW + DT + RT); age-matched mice that only trained for 4 wk served as controls (4 wk PoW). (F) Average nightly running volume during 4 wk of retraining. (G) Body weight at the time of being euthanized. (H) Absolute plantaris muscle weight in milligrams (mg; *P<0.05, directional t-test). (I) Plantaris muscle weight (mg) normalized to body weight in grams (g; *P<0.05, directional t-test). (J) Myosin heavy chain (MyHC) 2A cross sectional area (CSA) of gastrocnemius muscle fibers (*P<0.05, directional t-test). (K) MyHC 2A proportion. (L) MyHC 2A myonuclear number measured using dystrophin and DAPI. (M) Representative image of dystrophin, MyHC 2A fibers, and nuclei in 4 wk PoW and PoW + DT + RT muscles. Scale bar = 50 µm.
Figure 3.
Figure 3.
Myonuclear promoter region methylation memory of prior PoWeR training may facilitate retraining adaptations. (A) Memory of promoter site-specific CpG methylation in myonuclear Pkd2l1 (FDR < 0.05). (B) Memory of promoter region-specific hypomethylation in myonuclear Gdf10. (C) Memory of promoter region-specific hypermethylation in myonuclear Eif1a, Pitx1, Sun2, Usp43, and Ski (FDR < 0.05). (D) Evidence for promoter region CpG methylation memory of previous PoWeR at the pathway level in myonuclei. (E) Study design schematic showing how mice were subjected to PoWeR for 8 wk, detrained for 3 mo, and retrained for 4 wk (PoW + DT + RT); age-matched mice that only trained for 4 wk served as controls (4 wk PoW). (F) Average nightly running volume during 4 wk of retraining. (G) Body weight at the time of being euthanized. (H) Absolute plantaris muscle weight in milligrams (mg; *P<0.05, directional t-test). (I) Plantaris muscle weight (mg) normalized to body weight in grams (g; *P<0.05, directional t-test). (J) Myosin heavy chain (MyHC) 2A cross sectional area (CSA) of gastrocnemius muscle fibers (*P<0.05, directional t-test). (K) MyHC 2A proportion. (L) MyHC 2A myonuclear number measured using dystrophin and DAPI. (M) Representative image of dystrophin, MyHC 2A fibers, and nuclei in 4 wk PoW and PoW + DT + RT muscles. Scale bar = 50 µm.

References

    1. Seaborne RA, Strauss J, Cocks Met al. . Human skeletal muscle possesses an epigenetic memory of hypertrophy. Sci Rep. 2018;8(1): 1898.
    1. 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):1–12.
    1. Fisher AG, Seaborne RA, Hughes TMet al. . Transcriptomic and epigenetic regulation of disuse atrophy and the return to activity in skeletal muscle. FASEB J. 2017;31(12): 5268–5282.
    1. von Walden F, Rea M, Mobley CBet al. . The myonuclear DNA methylome in response to an acute hypertrophic stimulus. Epigenetics. 2020;15(11): 1151–1162.
    1. Iwata M, Englund DA, Wen Yet al. . A novel tetracycline-responsive transgenic mouse strain for skeletal muscle-specific gene expression. Skelet Musc. 2018;8(1): 33.
    1. Figueiredo VC, Wen Y, Alkner Bet al. . Genetic and epigenetic regulation of skeletal muscle ribosome biogenesis with exercise. J Physiol. 2021;599(13): 3363–3384.
    1. Dungan CM, Murach KA, Frick KKet al. . Elevated myonuclear density during skeletal muscle hypertrophy in response to training is reversed during detraining. Am J Physiol Cell Physiol. 2019;316(5): C649–C654.
    1. Murach KA, Mobley CB, Zdunek CJet al. . Muscle memory: myonuclear accretion, maintenance, morphology, and miRNA levels with training and detraining in adult mice. J Cachexia Sarcopenia Musc. 2020;11(6): 1705–1722.
    1. Englund D, Figueiredo VC, Dungan CMet al. . Satellite cell depletion disrupts transcriptional coordination and muscle adaptation to exercise. Function. 2020;2(1): zqaa033.
    1. Murach KA, McCarthy JJ, Peterson CA, Dungan CM. Making mice mighty: recent advances in translational models of load-induced muscle hypertrophy. J Appl Physiol. 2020;129(3): 516–521.
    1. Wen Y, Englund DA, Peck BDet al. . Myonuclear transcriptional dynamics in response to exercise following satellite cell depletion. iScience. 2021;24(8):102838. doi: 10.1016/j.isci.2021.102838.
    1. Englund DA, Murach KA, Dungan CMet al. . Depletion of resident muscle stem cells negatively impacts running volume, physical function and muscle hypertrophy in response to lifelong physical activity. Am J Physiol Cell Physiol. 2020;318(6): C1178–C1188.
    1. Siegfried Z, Eden S, Mendelsohn Met al. . DNA methylation represses transcription in vivo. Nat Genet. 1999;22(2): 203–206.
    1. Kang JG, Park JS, Ko J-H, Kim Y-S. Regulation of gene expression by altered promoter methylation using a CRISPR/Cas9-mediated epigenetic editing system. Sci Rep. 2019;9(1): 1–12.
    1. Busslinger M, Hurst J, Flavell R. DNA methylation and the regulation of globin gene expression. Cell. 1983;34(1): 197–206.
    1. Walsh CP, Chaillet JR, Bestor TH. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat Genet. 1998;20(2): 116–117.
    1. Cooper SJ, Trinklein ND, Anton ED, Nguyen L, Myers RM. Comprehensive analysis of transcriptional promoter structure and function in 1% of the human genome. Genome Res. 2006;16(1): 1–10.
    1. Chen C-H, Zheng R, Tokheim Cet al. . Determinants of transcription factor regulatory range. Nat Commun. 2020;11(1): 1–15.
    1. Lee C, Wang K, Qin T, Sartor MA. Testing proximity of genomic regions to transcription start sites and enhancers complements gene set enrichment testing. Front Genet. 2020;11: 199. doi: 10.3389/fgene.2020.00199.
    1. Saxonov S, Berg P, Brutlag DL. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc Natl Acad Sci. 2006;103(5): 1412–1417.
    1. Rowlands DS, Page RA, Sukala WRet 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(20): 747–765.
    1. Nitert MD, Dayeh T, Volkov Pet 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(12): 3322–3332.
    1. Jacques M, Hiam D, Craig Jet al. . Epigenetic changes in healthy human skeletal muscle following exercise–a systematic review. Epigenetics. 2019;14(7): 633–648.
    1. 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(24): 1567–1578.
    1. Barres R, Yan J, Egan Bet al. . Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15(3): 405–411.
    1. Herwig R, Hardt C, Lienhard M, Kamburov A. Analyzing and interpreting genome data at the network level with ConsensusPathDB. Nat Protoc. 2016;11(10): 1889.
    1. Okazaki Y, Ohshima N, Yoshizawa Iet al. . A novel glycerophosphodiester phosphodiesterase, GDE5, controls skeletal muscle development via a non-enzymatic mechanism. J Biol Chem. 2010;285(36): 27652–27663.
    1. Shan T, Liu J, Xu Z, Wang Y. Roles of phosphatase and tensin homolog in skeletal muscle. J Cell Physiol. 2019;234(4): 3192–3196.
    1. Wijesekara N, Konrad D, Eweida Met al. . Muscle-specific Pten deletion protects against insulin resistance and diabetes. Mol Cell Biol. 2005;25(3): 1135–1145.
    1. Lu Z, Cui Y, Wei Xet al. . Deficiency of PKD2L1 (TRPP3) exacerbates pathological cardiac hypertrophy by augmenting NCX1-mediated mitochondrial calcium overload. Cell Rep. 2018;24(6): 1639–1652.
    1. Camps J, Breuls N, Sifrim Aet al. . Interstitial cell remodeling promotes aberrant adipogenesis in dystrophic muscles. Cell Rep. 2020;31(5): 107597.
    1. Pandey SN, Cabotage J, Shi Ret al. . Conditional over-expression of PITX1 causes skeletal muscle dystrophy in mice. Biol Open. 2012;1(7): 629–639.
    1. Stewart RM, Rodriguez EC, King MC. Ablation of SUN2-containing LINC complexes drives cardiac hypertrophy without interstitial fibrosis. Mol Biol Cell. 2019;30(14): 1664–1675.
    1. He L, Liu X, Yang Jet al. . Imbalance of the reciprocally inhibitory loop between the ubiquitin-specific protease USP43 and EGFR/PI3K/AKT drives breast carcinogenesis. Cell Res. 2018;28(9): 934–951.
    1. Löw P, Talián GC, Sass M. Up-and downregulated genes in muscles that undergo developmentally programmed cell death in the insect Manduca sexta. FEBS Lett. 2005;579(22): 4943–4948.
    1. Chargé SB, Brack AS, Hughes SM. Aging-related satellite cell differentiation defect occurs prematurely after Ski-induced muscle hypertrophy. Am J Physiol Cell Physiol. 2002;283(4): C1228–C1241.
    1. Bruusgaard J, Brack A, Hughes S, Gundersen K. Muscle hypertrophy induced by the Ski protein: cyto-architecture and ultrastructure. Acta Physiol Scand. 2005;185(2): 141–149.
    1. Sutrave P, Kelly A, Hughes S. Ski can cause selective growth of skeletal muscle in transgenic mice. Genes Dev. 1990;4(9): 1462–1472.
    1. Sutrave P, Leferovich JM, Kelly AM, Hughes SH. The induction of skeletal muscle hypertrophy by a ski transgene is promoter-dependent. Gene. 2000;241(1): 107–116.
    1. Costelli P, Carbó N, Busquets Set al. . Reduced protein degradation rates and low expression of proteolytic systems support skeletal muscle hypertrophy in transgenic mice overexpressing the c-ski oncogene. Cancer Lett. 2003;200(2): 153–160.
    1. Diaz M, Martel N, Fitzsimmons RLet al. . Ski overexpression in skeletal muscle modulates genetic programs that control susceptibility to diet-induced obesity and insulin signaling. Obesity. 2012;20(11): 2157–2167.
    1. Knopp P, Figeac N, Fortier M, Moyle L, Zammit PS. Pitx genes are redeployed in adult myogenesis where they can act to promote myogenic differentiation in muscle satellite cells. Dev Biol. 2013;377(1): 293–304.
    1. Wen Y, Alimov AP, McCarthy JJ. Ribosome biogenesis is necessary for skeletal muscle hypertrophy. Exerc Sport Sci Rev. 2016;44(3): 110.
    1. von Walden F. Ribosome biogenesis in skeletal muscle: coordination of transcription and translation. J Appl Physiol. 2019;127(2): 591–598.
    1. Figueiredo VC, McCarthy JJ. Regulation of ribosome biogenesis in skeletal muscle hypertrophy. Physiology. 2019;34(1): 30–42.
    1. McCarthy JJ, Murach KA. in Nutrition and Enhanced Sports Performance. The Netherlands: Elsevier, 2019;275–290.
    1. Timmons JA, Jansson E, Fischer Het al. . Modulation of extracellular matrix genes reflects the magnitude of physiological adaptation to aerobic exercise training in humans. BMC Biol. 2005;3(1): 1–10.
    1. Hjorth M, Norheim F, Meen AJet al. . The effect of acute and long-term physical activity on extracellular matrix and serglycin in human skeletal muscle. Physiol Rep. 2015;3(8): e12473.
    1. Mendias CL, Schwartz AJ, Grekin JA, Gumucio JP, Sugg KB. Changes in muscle fiber contractility and extracellular matrix production during skeletal muscle hypertrophy. J Appl Physiol. 2017;122(3): 571–579.
    1. Guzzoni V, Ribeiro MBT, Lopes GNet al. . Effect of resistance training on extracellular matrix adaptations in skeletal muscle of older rats. Front Physiol. 2018;9:374. doi: 10.3389/fphys.2018.00374.
    1. Pillon NJ, Gabriel BM, Dollet Let al. . Transcriptomic profiling of skeletal muscle adaptations to exercise and inactivity. Nat Commun. 2020;11(1): 1–15.
    1. Stantzou A, Relizani K, Morales-Gonzalez Set al. . Extracellular matrix remodelling is associated with muscle force increase in overloaded mouse plantaris muscle. Neuropathol Appl Neurobiol. 2021;47(2): 218–235.
    1. Williams K, Carrasquilla GD, Ingerslev LRet al. . Epigenetic rewiring of skeletal muscle enhancers after exercise training supports arole in whole-body function and human health. Mol Metabol. 2021;53: 101290. doi: 10.1016/j.molmet.2021.101290.
    1. Nikooie R, Jafari-Sardoie S, Sheibani V, Nejadvaziri Chatroudi A. Resistance training-induced muscle hypertrophy is mediated by TGF-β1-Smad signaling pathway in male Wistar rats. J Cell Physiol. 2020;235(7-8): 5649–5665.
    1. Fuentes EN, Pino K, Navarro Cet al. . Transient inactivation of myostatin induces muscle hypertrophy and overcompensatory growth in zebrafish via inactivation of the SMAD signaling pathway. J Biotechnol. 2013;168(4): 295–302.
    1. Goodman CA, Hornberger TA. New roles for Smad signaling and phosphatidic acid in the regulation of skeletal muscle mass. F1000prime Rep. 2014;6(20). doi: 10.12703/P6-20.
    1. Leal ML, Lamas L, Aoki MSet al. . Effect of different resistance-training regimens on the WNT-signaling pathway. Eur J Appl Physiol. 2011;111(10): 2535–2545.
    1. Armstrong DD, Esser KA. Wnt/β-catenin signaling activates growth-control genes during overload-induced skeletal muscle hypertrophy. Am J Physiol Cell Physiol. 2005;289(4): C853–C859.
    1. Schmutz S, Däpp C, Wittwer Met al. . Endurance training modulates the muscular transcriptome response to acute exercise. Pflügers Archiv Eur J Physiol. 2006;451(5): 678–687.
    1. Perry CG, Lally J, Holloway GPet al. . Repeated transient mRNA bursts precede increases in transcriptional and mitochondrial proteins during training in human skeletal muscle. J Physiol. 2010;588(23): 4795–4810.
    1. Raue U, Trappe TA, Estrem STet al. . Transcriptome signature of resistance exercise adaptations: mixed muscle and fiber type specific profiles in young and old adults. J Appl Physiol. 2012;112(10): 1625–1636.
    1. Murach KA, Walton RG, Fry CSet al. . Cycle training modulates satellite cell and transcriptional responses to a bout of resistance exercise. Physiol Rep. 2016;4(18): e12973.
    1. Nader GA, von Walden F, Liu Cet al. . Resistance exercise training modulates acute gene expression during human skeletal muscle hypertrophy. J Appl Physiol. 2014;116(6): 693–702.
    1. Egan B, O'connor PL, Zierath JR, O'gorman DJ. Time course analysis reveals gene-specific transcript and protein kinetics of adaptation to short-term aerobic exercise training in human skeletal muscle. PLoS ONE. 2013;8(9): e74098.
    1. Murach K, Raue U, Wilkerson Bet al. . Single muscle fiber gene expression with run taper. PLoS ONE. 2014;9(9): e108547.
    1. Lindholm ME, Giacomello S, Solnestam BWet al. . The impact of endurance training on human skeletal muscle memory, global isoform expression and novel transcripts. PLos Genet. 2016;12(9): e1006294.
    1. Moberg M, Lindholm ME, Reitzner SMet al. . Exercise induces different molecular responses in trained and untrained human muscle. Med Sci Sports Exercise. 2020;52(8): 1679.
    1. Kubo K, Ikebukuro T, Yata H, Tsunoda N, Kanehisa H. Time course of changes in muscle and tendon properties during strength training and detraining. J Strength Condition Res. 2010;24(2): 322–331.
    1. Murach KA, Dungan CM, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. “Muscle memory” not mediated by myonuclear number?: secondary analysis of human detraining data. J Appl Physiol. 2019;127(6): 1814–1816.
    1. Psilander N, Eftestøl E, Cumming KTet al. . Effects of training, detraining, and retraining on strength, hypertrophy, and myonuclear number in human skeletal muscle. J Appl Physiol. 2019;126(6): 1636–1645.
    1. Blocquiaux S, Gorski T, Van Roie Eet al. . The effect of resistance training, detraining and retraining on muscle strength and power, myofibre size, satellite cells and myonuclei in older men. Exp Gerontol. 2020;133: 110860. doi: 10.1016/j.exger.2020.110897.
    1. Snijders T, Aussieker T, Holwerda Aet al. . The concept of skeletal muscle memory: evidence from animal and human studies. Acta Physiol Scand. 2020;229(3): e13465.
    1. Snijders T, Leenders M, de Groot L, van Loon LJ, Verdijk LB. Muscle mass and strength gains following 6 months of resistance type exercise training are only partly preserved within one year with autonomous exercise continuation in older adults. Exp Gerontol. 2019;121: 71–78.. doi: 10.1016/j.exger.2019.04.002.
    1. Egner IM, Bruusgaard JC, Eftestøl E, Gundersen K. A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids. J Physiol. 2013;591(24): 6221–6230.
    1. Lee H, Kim K, Kim Bet al. . A cellular mechanism of muscle memory facilitates mitochondrial remodelling following resistance training. J Physiol. 2018;596(18): 4413–4426.
    1. Seaborne R, Strauss J, Cocks Met al. . Methylome of human skeletal muscle after acute, chronic resistance exercise training, detraining, retraining. Sci Data. 2018;5(1): 180213.
    1. Small L, Ingerslev LR, Manitta Eet al. . Ablation of DNA-methyltransferase 3A in skeletal muscle does not affect energy metabolism or exercise capacity. PLos Genet. 2021;17(1): e1009325.
    1. Villivalam SD, Ebert SM, Lim HWet al. . A necessary role of DNMT3A in endurance exercise by suppressing ALDH1L1-mediated oxidative stress. EMBO J. 2021;40(9): e106491.
    1. Tumbar T, Guasch G, Greco Vet al. . Defining the epithelial stem cell niche in skin. Science. 2004;303(5656): 359–363.
    1. Murach KA, Vechetti IJ Jr., Van Pelt DWet al. . Fusion-independent satellite cell communication to muscle fibers during load-induced hypertrophy. Function. 2020;1(1): zqaa009.
    1. Grozdanov P, Georgiev O, Karagyozov L. Complete sequence of the 45-kb mouse ribosomal DNA repeat: analysis of the intergenic spacer. Genomics. 2003;82(6): 637–643.
    1. Park Y, Figueroa ME, Rozek LS, Sartor MA. MethylSig: a whole genome DNA methylation analysis pipeline. Bioinformatics. 2014;30(17): 2414–2422.
    1. Wen Y, Murach KA, Vechetti IJ Jret al. . MyoVision: software for automated high-content analysis of skeletal muscle immunohistochemistry. J Appl Physiol. 2018;124(1): 40–51.
    1. Anders S, Huber W. Differential expression analysis for sequence count data. Nat Prec. 2010;11(10): 1–1.

Source: PubMed

3
Abonnere