miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice

Adeel Safdar, Arkan Abadi, Mahmood Akhtar, Bart P Hettinga, Mark A Tarnopolsky, Adeel Safdar, Arkan Abadi, Mahmood Akhtar, Bart P Hettinga, Mark A Tarnopolsky

Abstract

MicroRNAs (miRNAs) are evolutionarily conserved small non-coding RNA species involved in post-transcriptional gene regulation. In vitro studies have identified a small number of skeletal muscle-specific miRNAs which play a crucial role in myoblast proliferation and differentiation. In skeletal muscle, an acute bout of endurance exercise results in the up-regulation of transcriptional networks that regulate mitochondrial biogenesis, glucose and fatty acid metabolism, and skeletal muscle remodelling. The purpose of this study was to assess the expressional profile of targeted miRNA species following an acute bout of endurance exercise and to determine relationships with previously established endurance exercise responsive transcriptional networks. C57Bl/6J wild-type male mice (N = 7/group) were randomly assigned to either sedentary or forced-endurance exercise (treadmill run @ 15 m/min for 90 min) group. The endurance exercise group was sacrificed three hours following a single bout of exercise. The expression of miR- 181, 1, 133, 23, and 107, all of which have been predicted to regulate transcription factors and co-activators involved in the adaptive response to exercise, was measured in quadriceps femoris muscle. Endurance exercise significantly increased the expression of miR-181, miR-1, and miR-107 by 37%, 40%, and 56%, respectively, and reduced miR-23 expression by 84% (P<or=0.05 for all), with no change in miR-133. Importantly, decreased expression of miRNA-23, a putative negative regulator of PGC-1alpha was consistent with increased expression of PGC-1alpha mRNA and protein along with several downstream targets of PGC-1alpha including ALAS, CS, and cytochrome c mRNA. PDK4 protein content remains unaltered despite an increase in its putative negative regulator, miR-107, and PDK4 mRNA expression. mRNA expression of miRNA processing machinery (Drosha, Dicer, and DGCR8) remained unchanged. We conclude that miRNA-mediated post-transcriptional regulation is potentially involved in the complex regulatory networks that govern skeletal muscle adaptation to endurance exercise in C57Bl/6J male mice.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. PGC-1α content and miR-23 expression…
Figure 1. PGC-1α content and miR-23 expression following exercise.
PGC-1α (A) mRNA expression and (B) protein content, and (C) miR-23 expression in the quadriceps of C57Bl/6J mice (N = 7/group) 3-hour following an acute bout of END exercise vs. SED group. (D) PGC-1α protein content negatively correlates (R = 0.62) with miR-23 content. PGC-1α mRNA expression, protein content and miR-23 expression are normalized to β-2 microglobulin, actin and Rnu6, respectively. Asterisks denote significant changes (P≤0.05).
Figure 2. Transcription of PGC-1α target genes…
Figure 2. Transcription of PGC-1α target genes following exercise.
Activation of ALAS, CS and cyt. c mRNA expression (fold-change) in the quadriceps of C57Bl/6J mice (N = 7/group) 3-hour following an acute bout of END exercise vs. SED group. ALAS, CS and cyt. c mRNA expression are normalized to β-2 microglobulin. Asterisks denote significant changes (P≤0.05).
Figure 3. PDK4 content and miR-107 expression…
Figure 3. PDK4 content and miR-107 expression following exercise.
(A) PDK4 mRNA expression and (B) protein content, and (C) miR-107 expression in the quadriceps of C57Bl/6J mice (N = 7/group) 3-hour following an acute bout of END exercise vs. SED group. PDK4 mRNA expression, protein content and miR-107 expression are normalized to β-2 microglobulin, actin and Rnu6, respectively. Asterisks denote significant changes (P≤0.05).
Figure 4. miR-1 and miR-181 expression following…
Figure 4. miR-1 and miR-181 expression following exercise.
miR-1 and miR-181 expression in the quadriceps of C57Bl/6J mice (N = 7/group) 3-hour following an acute bout of END exercise vs. SED group. miR-1 and miR-181 expression are normalized to Rnu6. Asterisks denote significant changes (P≤0.05).

References

    1. Hamilton MT, Booth FW. Skeletal muscle adaptation to exercise: a century of progress. J Appl Physiol. 2000;88:327–331.
    1. Mahoney DJ, Parise G, Melov S, Safdar A, Tarnopolsky MA. Analysis of global mRNA expression in human skeletal muscle during recovery from endurance exercise. Faseb J. 2005;19:1498–1500.
    1. Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, et al. Endurance exercise as a countermeasure for aging. Diabetes. 2008;57:2933–2942.
    1. Wisloff U, Najjar SM, Ellingsen O, Haram PM, Swoap S, et al. Cardiovascular risk factors emerge after artificial selection for low aerobic capacity. Science. 2005;307:418–420.
    1. Chakravarty EF, Hubert HB, Lingala VB, Fries JF. Reduced disability and mortality among aging runners: a 21-year longitudinal study. Arch Intern Med. 2008;168:1638–1646.
    1. Bonen A, Dyck DJ, Ibrahimi A, Abumrad NA. Muscle contractile activity increases fatty acid metabolism and transport and FAT/CD36. Am J Physiol. 1999;276:E642–649.
    1. Carter SL, Rennie C, Tarnopolsky MA. Substrate utilization during endurance exercise in men and women after endurance training. Am J Physiol Endocrinol Metab. 2001;280:E898–907.
    1. Gollnick PD, Saltin B. Significance of skeletal muscle oxidative enzyme enhancement with endurance training. Clin Physiol. 1982;2:1–12.
    1. Ren JM, Semenkovich CF, Gulve EA, Gao J, Holloszy JO. Exercise induces rapid increases in GLUT4 expression, glucose transport capacity, and insulin-stimulated glycogen storage in muscle. J Biol Chem. 1994;269:14396–14401.
    1. Baar K, Song Z, Semenkovich CF, Jones TE, Han DH, et al. Skeletal muscle overexpression of nuclear respiratory factor 1 increases glucose transport capacity. Faseb J. 2003;17:1666–1673.
    1. Baar K, Wende AR, Jones TE, Marison M, Nolte LA, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC-1. Faseb J. 2002;16:1879–1886.
    1. Hawley JA, Hargreaves M, Zierath JR. Signalling mechanisms in skeletal muscle: role in substrate selection and muscle adaptation. Essays Biochem. 2006;42:1–12.
    1. Mahoney DJ, Carey K, Fu MH, Snow R, Cameron-Smith D, et al. Real-time RT-PCR analysis of housekeeping genes in human skeletal muscle following acute exercise. Physiol Genomics. 2004;18:226–231.
    1. Widegren U, Jiang XJ, Krook A, Chibalin AV, Bjornholm M, et al. Divergent effects of exercise on metabolic and mitogenic signaling pathways in human skeletal muscle. Faseb J. 1998;12:1379–1389.
    1. Hoppeler H, Klossner S, Fluck M. Gene expression in working skeletal muscle. Adv Exp Med Biol. 2007;618:245–254.
    1. Joseph AM, Pilegaard H, Litvintsev A, Leick L, Hood DA. Control of gene expression and mitochondrial biogenesis in the muscular adaptation to endurance exercise. Essays Biochem. 2006;42:13–29.
    1. Coffey VG, Hawley JA. The molecular bases of training adaptation. Sports Med. 2007;37:737–763.
    1. Rose AJ, Broholm C, Kiillerich K, Finn SG, Proud CG, et al. Exercise rapidly increases eukaryotic elongation factor 2 phosphorylation in skeletal muscle of men. J Physiol. 2005;569:223–228.
    1. Keller P, Vollaard N, Babraj J, Ball D, Sewell DA, et al. Using systems biology to define the essential biological networks responsible for adaptation to endurance exercise training. Biochem Soc Trans. 2007;35:1306–1309.
    1. Asli NS, Pitulescu ME, Kessel M. MicroRNAs in organogenesis and disease. Curr Mol Med. 2008;8:698–710.
    1. Papagiannakopoulos T, Kosik KS. MicroRNAs: regulators of oncogenesis and stemness. BMC Med. 2008;6:15.
    1. Callis TE, Deng Z, Chen JF, Wang DZ. Muscling through the microRNA world. Exp Biol Med (Maywood) 2008;233:131–138.
    1. Walden TB, Timmons JA, Keller P, Nedergaard J, Cannon B. Distinct expression of muscle-specific microRNAs (myomirs) in brown adipocytes. J Cell Physiol. 2009;218:444–449.
    1. Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91:209–217.
    1. Poy MN, Spranger M, Stoffel M. microRNAs and the regulation of glucose and lipid metabolism. Diabetes Obes Metab. 2007;9(Suppl 2):67–73.
    1. van Rooij E, Liu N, Olson EN. MicroRNAs flex their muscles. Trends Genet. 2008;24:159–166.
    1. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20.
    1. Cannell IG, Kong YW, Bushell M. How do microRNAs regulate gene expression? Biochem Soc Trans. 2008;36:1224–1231.
    1. Sokol NS, Ambros V. Mesodermally expressed Drosophila microRNA-1 is regulated by Twist and is required in muscles during larval growth. Genes Dev. 2005;19:2343–2354.
    1. Zhao Y, Samal E, Srivastava D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature. 2005;436:214–220.
    1. Eisenberg I, Eran A, Nishino I, Moggio M, Lamperti C, et al. Distinctive patterns of microRNA expression in primary muscular disorders. Proc Natl Acad Sci U S A. 2007;104:17016–17021.
    1. Jackson RJ, Standart N. How do microRNAs regulate gene expression? Sci STKE. 2007;2007:re1.
    1. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–854.
    1. Holloszy JO, Booth FW. Biochemical adaptations to endurance exercise in muscle. Annu Rev Physiol. 1976;38:273–291.
    1. Kelly DP, Scarpulla RC. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 2004;18:357–368.
    1. Puigserver P, Adelmant G, Wu Z, Fan M, Xu J, et al. Activation of PPARgamma coactivator-1 through transcription factor docking. Science. 1999;286:1368–1371.
    1. Puigserver P, Spiegelman BM. Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator. Endocr Rev. 2003;24:78–90.
    1. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 1999;98:115–124.
    1. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, et al. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature. 2002;418:797–801.
    1. Wright DC, Han DH, Garcia-Roves PM, Geiger PC, Jones TE, et al. Exercise-induced mitochondrial biogenesis begins before the increase in muscle PGC-1alpha expression. J Biol Chem. 2007;282:194–199.
    1. Sandri M, Lin J, Handschin C, Yang W, Arany ZP, et al. PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci U S A. 2006;103:16260–16265.
    1. Handschin C, Kobayashi YM, Chin S, Seale P, Campbell KP, et al. PGC-1alpha regulates the neuromuscular junction program and ameliorates Duchenne muscular dystrophy. Genes Dev. 2007;21:770–783.
    1. Hanai J, Cao P, Tanksale P, Imamura S, Koshimizu E, et al. The muscle-specific ubiquitin ligase atrogin-1/MAFbx mediates statin-induced muscle toxicity. J Clin Invest. 2007;117:3940–3951.
    1. Hildebrandt AL, Pilegaard H, Neufer PD. Differential transcriptional activation of select metabolic genes in response to variations in exercise intensity and duration. Am J Physiol Endocrinol Metab. 2003;285:E1021–1027.
    1. Pilegaard H, Keller C, Steensberg A, Helge JW, Pedersen BK, et al. Influence of pre-exercise muscle glycogen content on exercise-induced transcriptional regulation of metabolic genes. J Physiol. 2002;541:261–271.
    1. Pilegaard H, Ordway GA, Saltin B, Neufer PD. Transcriptional regulation of gene expression in human skeletal muscle during recovery from exercise. Am J Physiol Endocrinol Metab. 2000;279:E806–814.
    1. Holness MJ, Sugden MC. Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation. Biochem Soc Trans. 2003;31:1143–1151.
    1. Sugden MC, Holness MJ. Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs. Am J Physiol Endocrinol Metab. 2003;284:E855–862.
    1. Holness MJ, Kraus A, Harris RA, Sugden MC. Targeted upregulation of pyruvate dehydrogenase kinase (PDK)-4 in slow-twitch skeletal muscle underlies the stable modification of the regulatory characteristics of PDK induced by high-fat feeding. Diabetes. 2000;49:775–781.
    1. Peters SJ, Harris RA, Heigenhauser GJ, Spriet LL. Muscle fiber type comparison of PDH kinase activity and isoform expression in fed and fasted rats. Am J Physiol Regul Integr Comp Physiol. 2001;280:R661–668.
    1. Ramaswamy G, Karim MA, Murti KG, Jackowski S. PPARalpha controls the intracellular coenzyme A concentration via regulation of PANK1alpha gene expression. J Lipid Res. 2004;45:17–31.
    1. Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest. 2006;116:571–580.
    1. Skrede S, Sorensen HN, Larsen LN, Steineger HH, Hovik K, et al. Thia fatty acids, metabolism and metabolic effects. Biochim Biophys Acta. 1997;1344:115–131.
    1. Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang G, et al. The expression of microRNA miR-107 decreases early in Alzheimer's disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28:1213–1223.
    1. Larson EB, Wang L, Bowen JD, McCormick WC, Teri L, et al. Exercise is associated with reduced risk for incident dementia among persons 65 years of age and older. Ann Intern Med. 2006;144:73–81.
    1. Teri L, Gibbons LE, McCurry SM, Logsdon RG, Buchner DM, et al. Exercise plus behavioral management in patients with Alzheimer disease: a randomized controlled trial. Jama. 2003;290:2015–2022.
    1. Fluck M. Functional, structural and molecular plasticity of mammalian skeletal muscle in response to exercise stimuli. J Exp Biol. 2006;209:2239–2248.
    1. Vissing K, McGee SL, Roepstorff C, Schjerling P, Hargreaves M, et al. Effect of sex differences on human MEF2 regulation during endurance exercise. Am J Physiol Endocrinol Metab. 2008;294:E408–415.
    1. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet. 2006;38:228–233.
    1. Naguibneva I, Ameyar-Zazoua M, Polesskaya A, Ait-Si-Ali S, Groisman R, et al. The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol. 2006;8:278–284.
    1. McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. J Appl Physiol. 2007;102:306–313.
    1. Xu C, Lu Y, Pan Z, Chu W, Luo X, et al. The muscle-specific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. J Cell Sci. 2007;120:3045–3052.

Source: PubMed

3
Abonnere