Human Skeletal Muscle Possesses an Epigenetic Memory of Hypertrophy

Robert A Seaborne, Juliette Strauss, Matthew Cocks, Sam Shepherd, Thomas D O'Brien, Ken A van Someren, Phillip G Bell, Christopher Murgatroyd, James P Morton, Claire E Stewart, Adam P Sharples, Robert A Seaborne, Juliette Strauss, Matthew Cocks, Sam Shepherd, Thomas D O'Brien, Ken A van Someren, Phillip G Bell, Christopher Murgatroyd, James P Morton, Claire E Stewart, Adam P Sharples

Abstract

It is unknown if adult human skeletal muscle has an epigenetic memory of earlier encounters with growth. We report, for the first time in humans, genome-wide DNA methylation (850,000 CpGs) and gene expression analysis after muscle hypertrophy (loading), return of muscle mass to baseline (unloading), followed by later hypertrophy (reloading). We discovered increased frequency of hypomethylation across the genome after reloading (18,816 CpGs) versus earlier loading (9,153 CpG sites). We also identified AXIN1, GRIK2, CAMK4, TRAF1 as hypomethylated genes with enhanced expression after loading that maintained their hypomethylated status even during unloading where muscle mass returned to control levels, indicating a memory of these genes methylation signatures following earlier hypertrophy. Further, UBR5, RPL35a, HEG1, PLA2G16, SETD3 displayed hypomethylation and enhanced gene expression following loading, and demonstrated the largest increases in hypomethylation, gene expression and muscle mass after later reloading, indicating an epigenetic memory in these genes. Finally, genes; GRIK2, TRAF1, BICC1, STAG1 were epigenetically sensitive to acute exercise demonstrating hypomethylation after a single bout of resistance exercise that was maintained 22 weeks later with the largest increase in gene expression and muscle mass after reloading. Overall, we identify an important epigenetic role for a number of largely unstudied genes in muscle hypertrophy/memory.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
(A) Schematic representation of experimental conditions and types of analysis undertaken across the time-course. The image of a muscle represents the time point for analysis of muscle mass via (i) DEXA and strength via (ii) isometric quadriceps muscle torque using an isokinetic dynamometer. The images of muscle tissue also represent the time point of skeletal muscle biopsy of the Vastus Lateralis, muscle sample preparation for downstream analysis of (iii) Infinium MethylationEPIC BeadChip arrays (850 K CpG sites) methylome wide array (iv) and rt-qRT-PCR for gene expression analysis of important genes identified following methylome wide analysis. (B) Weekly total volume of resistance exercise undertaken by human participants (n = 7) during the first 7-week resistance exercise period (loading, weeks 1–7), followed by a 7 week cessation of resistance exercise (unloading, weeks 8–14) and the later second period of 7 weeks resistance exercise (reloading, weeks 15–21). Data represents volume load as calculated by ((load (Kg) x reps) x sets)) averaged across 3 resistance exercise sessions per week. Data presented mean ± SEM. (Ci) Lean lower limb mass changes in human subjects (n = 7) after a period of 7 weeks resistance exercise (loading), exercise cessation (unloading) and a subsequent second period of 7 weeks resistance exercise (reloading). Total limb lean mass normalised to baseline (percentage change). Significant change compared to baseline represented by * and significant difference to all other conditions represented by ** (Cii) Total lean mass percentage change when loading is normalised to baseline, and reloading normalised to unloading to account for starting lean mass in both conditions. Pairwise t-test of significance indicated by *. All data presented as mean ± SEM (n = 7).
Figure 2
Figure 2
(A) Infinium MethylationEPIC BeadChip arrays (850 K CpG sites) identified an enhanced frequency of hypomethylated CpG sites upon reloading (n = 7). (B) Gene ontology analysis using forest plot schematics confirmed an enhanced hypomethylated profile after reloading across various (i) molecular function, (ii) biological processes and (iii) cellular components. Functional groups with a fold enrichment >3 (as indicated via shaded blue region) represents statistically ‘over expressed’ (in this case epigenetically modified) KEGG pathways FDR < 0.05 (n = 8).
Figure 3
Figure 3
(A) Representation of the DNA methylation modifications that occurred within the PI3K/AKT KEGG pathway following 7 weeks of reloading in human subjects. Signalling analysis performed on statistically differentially regulated CpG sites compared to baseline, with green indicating a hypomethylated fold change and red indicating a hypermethylated change, with strength of colour representing the intensity of fold change–. Figure 3b. Venn diagram analysis of the statistically differentially regulated CpG sites attributed to the PI3K/AKT pathway following loading, unloading and reloading, compared to be baseline. Ellipsis reports number of commonly statistically differentially regulated CpG sites across each condition. Analysis confirms an enhanced number of differentially regulated CpG sites upon reloading condition.
Figure 4
Figure 4
(A) Heat map depicting unsupervised hierarchical clustering of the top 500 statistically differentially regulated CpG loci (columns) and conditions (baseline, loading, unloading and reloading) in previously untrained male participants (n = 8). The heat-map colours correspond to standardised expression normalised β-values, with green representing hypomethylation, red hypermethylation and unchanged sites are represented in black. (4B and C) Relative gene expression (i) and schematic representation of CpG DNA methylation and gene expression relationship (ii) in two identified gene clusters from genome wide methylation analysis after a period of 7 weeks resistance exercise (loading), exercise cessation (unloading) and a subsequent secondary period of 7 weeks resistance exercise (reloading). (Bi) Expression of genes that displayed a significant increase compared to baseline (represented by *) upon earlier loading, that returned to baseline during unloading, and displayed enhanced expression after reloading (significantly different to all other conditions **). MANOVA analysis reported a significant effect over the entire time course of the experiment (P < 0.0001). (Bii) Representative schematic displaying the inverse relationship between mean gene expression (solid black lines) and CpG DNA methylation (dashed black lines) of grouped transcripts (RPL35a, C12orf50, BICC1, ZFP2, UBR5, HEG1, PLA2G16, SETD3 and ODF2). Data represented as fold change for DNA methylation (left y axis) and gene/mRNA expression (right y axis). (Ci) Clustering of genes that portrayed an accumulative increase in gene expression after loading, unloading and reloading. With the largest increase in gene expression after reloading. Culminating in significance in the unloading (baseline vs. unloading*), and reloading (reloading vs. baseline**). (Cii) Representative schematic displaying the inverse relationship between mean gene expression (solid black lines) and CpG DNA methylation (dashed black lines) of grouped transcripts (AXIN1, TRAF1, GRIK2, CAMK4). Data represented as fold change for methylation (left y axis) and mRNA expression (right y axis). All data represented as mean ± SEM for gene expression (n = 7 for UBR5, PLA2G16, AXIN1, GRIK2; n = 8 for all others) and CpG DNA methylation (n = 8 for baseline, loading and unloading; n = 7 for reloading).
Figure 5
Figure 5
Relative fold changes in: (A) gene expression; (B) correlation between gene expression and lower limb lean mass across experimental conditions, and; (C) schematic representation of relationship between fold changes in CpG DNA methylation (dashed black line; left y axis) and fold change in gene/mRNA expression (solid black line; right y axis) for identified genes: RPL35a (I), UBR5 (II), SETD3 (III), PLA2G16 (IV) and HEG1 (V). Statistical significance compared to baseline and unloading represented by* and** respectively. All significance taken as p less than or equal to 0.05 unless otherwise state on graph. All data presented as mean ± SEM (n = 7/8).
Figure 6
Figure 6
Representation and characterisation of the DNA methylation modifications that occurred within the ubiquitin mediated proteolysis pathway across all conditions of loading, unloading and reloading compared to baseline (ANOVA). Signalling analysis performed on statistically differentially regulated CpG sites compared to baseline, with green indicating a hypomethylated fold change and red indicating a hypermethylated change, with strength of colour representing the intensity of fold change–. Importantly, the novel HECT-type E3 ubiquitin ligase, UBR5, displays a significantly hypomethylated state within this pathway.
Figure 7
Figure 7
Response of the methylome after acute resistance loading stimulus compared to baseline, 7 weeks loading and 7 weeks reloading: (A) Heat map depicting unsupervised hierarchical clustering of statistically differentially regulated (P = 0.05) CpG loci following exposure to acute RE compared to baseline; (B) a Venn diagram depicting the number of CpG sites that were significantly differentially regulated in both methylome analysis experiments (base, loading, unloading and reloading, blue circle; baseline and acute resistance stimulus, red circle), and the amount of genes analysed for gene expression across acute, 7 weeks loading and 7 weeks reloading, respectively; (C) temporal pattern of fold change in DNA CpG methylation of the identified overlapping CpG sites that mapped to relevant gene transcripts; (D) correlation of CpG DNA methylation of acute RE vs. 7 weeks loading and reloading conditions, and (E) Heat map depicting unsupervised hierarchical clustering of statistically differentially regulated (P = 0.05) CpG loci following exposure to acute RE compared to baseline, loading and reloading (F) representative schematic displaying the inverse relationship between mean gene expression (solid black lines) and CpG DNA methylation (dashed black lines) of identified transcripts. Significance indicated in gene expression (*) and in CpG DNA methylation (§) when compared to baseline.

References

    1. 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. doi: 10.1111/acel.12486.
    1. Patel HP, et al. Developmental influences, muscle morphology, and sarcopenia in community-dwelling older men. J Gerontol A Biol Sci Med Sci. 2012;67:82–87. doi: 10.1093/gerona/glr020.
    1. Patel HP, et al. Lean mass, muscle strength and gene expression in community dwelling older men: findings from the Hertfordshire Sarcopenia Study (HSS) Calcified tissue international. 2014;95:308–316. doi: 10.1007/s00223-014-9894-z.
    1. Laker RC, et al. Exercise prevents maternal high-fat diet-induced hypermethylation of the Pgc-1alpha gene and age-dependent metabolic dysfunction in the offspring. Diabetes. 2014;63:1605–1611. doi: 10.2337/db13-1614.
    1. Zeng Y, Gu P, Liu K, Huang P. Maternal protein restriction in rats leads to reduced PGC-1alpha expression via altered DNA methylation in skeletal muscle. Molecular medicine reports. 2013;7:306–312. doi: 10.3892/mmr.2012.1134.
    1. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nature genetics. 2003;33:Suppl, 245–254. doi: 10.1038/ng1089.
    1. Green CJ, Bunprajun T, Pedersen BK, Scheele C. Physical activity is associated with retained muscle metabolism in human myotubes challenged with palmitate. J Physiol. 2013;591:4621–4635. doi: 10.1113/jphysiol.2013.251421.
    1. Aguer C, et al. Intramyocellular lipid accumulation is associated with permanent relocation ex vivo and in vitro of fatty acid translocase (FAT)/CD36 in obese patients. Diabetologia. 2010;53:1151–1163. doi: 10.1007/s00125-010-1708-x.
    1. Maples JM, et al. Lipid exposure elicits differential responses in gene expression and DNA methylation in primary human skeletal muscle cells from severely obese women. Physiological genomics. 2015;47:139–146. doi: 10.1152/physiolgenomics.00065.2014.
    1. Foulstone EJ, Savage PB, Crown AL, Holly JM, Stewart CE. Adaptations of the IGF system during malignancy: human skeletal muscle versus the systemic environment. Horm Metab Res. 2003;35:667–674. doi: 10.1055/s-2004-814159.
    1. Sharples AP, et al. Skeletal muscle cells possess a ‘memory’ of acute early life TNF-alpha exposure: role of epigenetic adaptation. Biogerontology. 2016;17:603–617. doi: 10.1007/s10522-015-9604-x.
    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. The Journal of Physiology. 2013;591:6221–6230. doi: 10.1113/jphysiol.2013.264457.
    1. Bruusgaard JC, Johansen IB, Egner IM, Rana ZA, Gundersen K. Myonuclei acquired by overload exercise precede hypertrophy and are not lost on detraining. Proc Natl Acad Sci USA. 2010;107:15111–15116. doi: 10.1073/pnas.0913935107.
    1. Jacobsen SC, et al. Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia. 2012;55:3341–3349. doi: 10.1007/s00125-012-2717-8.
    1. Barres R, et al. Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density. Cell Metab. 2009;10:189–198. doi: 10.1016/j.cmet.2009.07.011.
    1. Barres R, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 2012;15:405–411. doi: 10.1016/j.cmet.2012.01.001.
    1. Fisher A, et al. Transcriptomic and Epigenetic Regulation of Disuse Atrophy and the Return to Activity in Skeletal Muscle. Faseb J. 2017;31:5268–5282. doi: 10.1096/fj.201700089RR.
    1. Bigot A, et al. Age-Associated Methylation Suppresses SPRY1, Leading to a Failure of Re-quiescence and Loss of the Reserve Stem Cell Pool in Elderly Muscle. Cell reports. 2015;13:1172–1182. doi: 10.1016/j.celrep.2015.09.067.
    1. Bogdanovic O, Veenstra GJ. DNA methylation and methyl-CpG binding proteins: developmental requirements and function. Chromosoma. 2009;118:549–565. doi: 10.1007/s00412-009-0221-9.
    1. Peterson MD, Pistilli E, Haff GG, Hoffman EP, Gordon PM. Progression of volume load and muscular adaptation during resistance exercise. European journal of applied physiology. 2011;111:1063–1071. doi: 10.1007/s00421-010-1735-9.
    1. Maksimovic J, Gordon L, Oshlack A. SWAN: Subset-quantile within array normalization for illumina infinium HumanMethylation450 BeadChips. Genome biology. 2012;13:R44. doi: 10.1186/gb-2012-13-6-r44.
    1. Pidsley R, et al. Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome biology. 2016;17:208. doi: 10.1186/s13059-016-1066-1.
    1. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic acids research. 2000;28:27–30. doi: 10.1093/nar/28.1.27.
    1. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic acids research. 2017;45:D353–d361. doi: 10.1093/nar/gkw1092.
    1. Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic acids research. 2016;44:D457–462. doi: 10.1093/nar/gkv1070.
    1. Egerman MA, Glass DJ. Signaling pathways controlling skeletal muscle mass. Critical Reviews in Biochemistry and Molecular Biology. 2014;49:59–68. doi: 10.3109/10409238.2013.857291.
    1. Schiaffino S, Mammucari C. Regulation of skeletal muscle growth by the IGF1-Akt/PKB pathway: insights from genetic models. Skeletal Muscle. 2011;1:4. doi: 10.1186/2044-5040-1-4.
    1. Fuks F, et al. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. The Journal of biological chemistry. 2003;278:4035–4040. doi: 10.1074/jbc.M210256200.
    1. Lunyak VV, et al. Corepressor-dependent silencing of chromosomal regions encoding neuronal genes. Science (New York, N.Y.) 2002;298:1747–1752. doi: 10.1126/science.1076469.
    1. Rountree MR, Selker EU. DNA methylation inhibits elongation but not initiation of transcription in Neurospora crassa. Genes & development. 1997;11:2383–2395. doi: 10.1101/gad.11.18.2383.
    1. Figeac N, Zammit PS. Coordinated action of Axin1 and Axin2 suppresses beta-catenin to regulate muscle stem cell function. Cellular signalling. 2015;27:1652–1665. doi: 10.1016/j.cellsig.2015.03.025.
    1. Huraskin D, et al. Wnt/beta-catenin signaling via Axin2 is required for myogenesis and, together with YAP/Taz and Tead1, active in IIa/IIx muscle fibers. Development. 2016;143:3128–3142. doi: 10.1242/dev.139907.
    1. Han Y, Wang C, Park JS, Niu L. Channel-opening kinetic mechanism for human wild-type GluK2 and the M867I mutant kainate receptor. Biochemistry. 2010;49:9207–9216. doi: 10.1021/bi100819v.
    1. Blaeser F, Ho N, Prywes R, Chatila TA. Ca2+-dependent Gene Expression Mediated by MEF2 Transcription Factors. Journal of Biological Chemistry. 2000;275:197–209. doi: 10.1074/jbc.275.1.197.
    1. Wu H, et al. MEF2 responds to multiple calcium-regulated signals in the control of skeletal muscle fiber type. Embo j. 2000;19:1963–1973. doi: 10.1093/emboj/19.9.1963.
    1. Nitert MD, 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. doi: 10.2337/db11-1653.
    1. Fry AC. The Role of Resistance Exercise Intensity on Muscle Fibre Adaptations. Sports Medicine. 2004;34:663–679. doi: 10.2165/00007256-200434100-00004.
    1. Pomerantz JL, Baltimore D. NF-kappaB activation by a signaling complex containing TRAF2, TANK and TBK1, a novel IKK-related kinase. Embo j. 1999;18:6694–6704. doi: 10.1093/emboj/18.23.6694.
    1. Foulstone EJ, Huser C, Crown AL, Holly JM, Stewart CE. Differential signalling mechanisms predisposing primary human skeletal muscle cells to altered proliferation and differentiation: roles of IGF-I and TNFalpha. Exp Cell Res. 2004;294:223–235. doi: 10.1016/j.yexcr.2003.10.034.
    1. Girven M, et al. l-glutamine Improves Skeletal Muscle Cell Differentiation and Prevents Myotube Atrophy After Cytokine (TNF-alpha) Stress Via Reduced p38 MAPK Signal Transduction. Journal of cellular physiology. 2016;231:2720–2732. doi: 10.1002/jcp.25380.
    1. Li YP. TNF-alpha is a mitogen in skeletal muscle. Am J Physiol Cell Physiol. 2003;285:C370–376. doi: 10.1152/ajpcell.00453.2002.
    1. Mackey AL, et al. The influence of anti-inflammatory medication on exercise-induced myogenic precursor cell responses in humans. J Appl Physiol. 2007;103:425–431. doi: 10.1152/japplphysiol.00157.2007.
    1. van de Vyver M, Myburgh KH. Cytokine and satellite cell responses to muscle damage: interpretation and possible confounding factors in human studies. J Muscle Res Cell Motil. 2012;33:177–185. doi: 10.1007/s10974-012-9303-z.
    1. Li YP, et al. TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle. Faseb J. 2005;19:362–370. doi: 10.1096/fj.04-2364com.
    1. Eom GH, et al. Histone Methyltransferase SETD3 Regulates Muscle Differentiation. Journal of Biological Chemistry. 2011;286:34733–34742. doi: 10.1074/jbc.M110.203307.
    1. Jaworski K, et al. AdPLA ablation increases lipolysis and prevents obesity induced by high-fat feeding or leptin deficiency. Nature medicine. 2009;15:159–168. doi: 10.1038/nm.1904.
    1. Kleaveland B, et al. Regulation of cardiovascular development and integrity by the heart of glass-cerebral cavernous malformation protein pathway. Nature medicine. 2009;15:169–176. doi: 10.1038/nm.1918.
    1. Tsuji S, et al. HEG1 is a novel mucin-like membrane protein that serves as a diagnostic and therapeutic target for malignant mesothelioma. Scientific reports. 2017;7:45768. doi: 10.1038/srep45768.
    1. Callaghan MJ, et al. Identification of a human HECT family protein with homology to the Drosophila tumor suppressor gene hyperplastic discs. Oncogene. 1998;17:3479–3491. doi: 10.1038/sj.onc.1202249.
    1. Buetow L, Huang DT. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nature reviews. Molecular cell biology. 2016;17:626–642. doi: 10.1038/nrm.2016.91.
    1. Bodine SC, et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. 2001;294:1704–1708. doi: 10.1126/science.1065874.
    1. Sartori R, et al. BMP signaling controls muscle mass. Nature genetics. 2013;45:1309–1318. doi: 10.1038/ng.2772.
    1. Hu G, et al. Modulation of myocardin function by the ubiquitin E3 ligase UBR5. The Journal of biological chemistry. 2010;285:11800–11809. doi: 10.1074/jbc.M109.079384.
    1. Wang Z, Wang DZ, Pipes GC, Olson EN. Myocardin is a master regulator of smooth muscle gene expression. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:7129–7134. doi: 10.1073/pnas.1232341100.
    1. Meadows SM, Warkman AS, Salanga MC, Small EM, Krieg PA. The myocardin-related transcription factor, MASTR, cooperates with MyoD to activate skeletal muscle gene expression. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:1545–1550. doi: 10.1073/pnas.0703918105.
    1. Long X, Creemers EE, Wang DZ, Olson EN, Miano JM. Myocardin is a bifunctional switch for smooth versus skeletal muscle differentiation. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:16570–16575. doi: 10.1073/pnas.0708253104.
    1. Muráni E, Murániová M, Ponsuksili S, Schellander K, Wimmers K. Identification of genes differentially expressed during prenatal development of skeletal muscle in two pig breeds differing in muscularity. BMC Developmental Biology. 2007;7:109. doi: 10.1186/1471-213X-7-109.
    1. Kong X, et al. Distinct functions of human cohesin-SA1 and cohesin-SA2 in double-strand break repair. Mol Cell Biol. 2014;34:685–698. doi: 10.1128/MCB.01503-13.

Source: PubMed

3
S'abonner