239
Views
54
CrossRef citations to date
0
Altmetric
Review

Proteomic responses of skeletal and cardiac muscle to exercise

&
Pages 361-377 | Published online: 09 Jan 2014

References

  • Lakka TA, Venäläinen JM, Rauramaa R, Salonen R, Tuomilehto J, Salonen JT. Relation of leisure-time physical activity and cardiorespiratory fitness to the risk of acute myocardial infarction. N. Engl. J. Med.330, 1549–1554 (1994).
  • Warburton DE, Nicol CW, Bredin SS. Health benefits of physical activity: the evidence. CMAJ174, 801–809 (2006).
  • Myers J, Prakash M, Froelicher V, Do D, Partington S, Atwood JE. Exercise capacity and mortality among men referred for exercise testing. N. Engl. J. Med.346, 793–801 (2002).
  • Blair SN, Kampert JB, Kohl WH 3rd et al. Influences of cardiorespiratory fitness and other precursors on cardiovascular disease and all-cause mortality in men and women. JAMA276, 205–210 (1996).
  • Stepto NK, Coffey VG, Carey AL et al. Global gene expression in skeletal muscle from well-trained strength and endurance athletes. Med. Sci. Sports Exerc.41, 546–565 (2009).
  • Wittwer M, Billeter R, Hoppeler H, Flück M. Regulatory gene expression in skeletal muscle of highly endurance-trained humans. Acta Physiol. Scand.180, 217–227 (2004).
  • Chen YW, Hubal MJ, Hoffman EP, Thompson PD, Clarkson PM. Molecular responses of human muscle to eccentric exercise. J. Appl. Physiol.95, 2485–2494 (2003).
  • Liu D, Sartor MA, Nader GA et al. Skeletal muscle gene expression in response to resistance exercise: sex specific regulation. BMC Genomics11, 659 (2010).
  • 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.19, 1498–1500 (2005).
  • Gygi SP, Rochon Y, Franza BR, Aebersold R. Correlation between protein and mRNA abundance in yeast. Mol. Cell Biol.19, 1720–1730 (1999).
  • Goldspink DF. Exercise-related changes in protein turnover in mammalian striated muscle. J. Exp. Biol.160, 127–148 (1991).
  • 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 One4, e5610 (2009).
  • Chen YW, Nader GA, Baar KR, Fedele MJ, Hoffman EP, Esser KA. Response of rat muscle to acute resistance exercise defined by transcriptional and translational profiling. J. Physiol.545, 27–41 (2002).
  • Van Hall G, Saltin B, Wagenmakers AJ. Muscle protein degradation and amino acid metabolism during prolonged knee-extensor exercise in humans. Clin. Sci. (Lond.)97, 557–567 (1999).
  • Hittel DS, Hathout Y, Hoffman EP. Proteomics and systems biology in exercise and sport sciences research. Exerc. Sport Sci. Rev.35, 5–11 (2007).
  • Fu Q, Van Eyk JE. Proteomics and heart disease: identifying biomarkers of clinical utility. Expert Rev. Proteomics3, 237–249 (2006).
  • Ohlendieck K. Proteomics of skeletal muscle differentiation, neuromuscular disorders and fiber aging. Expert Rev. Proteomics7, 283–296 (2010).
  • Schiaffino S. Fibre types in skeletal muscle: a personal account. Acta Physiol.199, 451–463 (2010).
  • Schiaffino S, Reggiani C. Molecular diversity of myofibrillar proteins: gene regulation and functional significance. Physiol. Rev.76, 371–423 (1996).
  • Li XP, Pleissner KP, Scheler C, Regitz-Zagrosek V, Salnikow J, Jungblut PR. A two-dimensional electrophoresis database of rat heart proteins. Electrophoresis20, 891–897 (1999).
  • Sanchez J-C, Diego C, Converset V et al. The mouse SWISS-2D PAGE database: a tool for proteomics study of diabetes and obesity. Proteomics1, 136–163 (2001).
  • Kim NK, Joh JH, Park HR, Kim OH, Park BY, Lee CS. Differential expression profiling of the proteomes and their mRNAs in porcine white and red skeletal muscles. Proteomics4, 3422–3428 (2004).
  • Bihan LC, Hou Y, Harris N, Tarelli E, Coulton GR. Proteomic analysis of fast and slow muscles from normal and kyphoscoliotic mice using protein arrays, 2-DE and MS. Proteomics6, 4646–4661 (2006).
  • Okumura N, Hashida-Okumura A, Kita K et al. Proteomic analysis of slow- and fast-twitch skeletal muscles. Proteomics5, 2896–2906 (2005).
  • Gelfi C, Vigano A, De Palma S et al. 2D protein maps of rat gastrocnemius and soleus muscles: a tool for muscle plasticity assessment. Proteomics6, 321–340 (2006).
  • Gelfi C, De Palma S, Cerretelli P, Begum S, Wait R. Two-dimensional protein map of human vastus lateralis muscle. Electrophoresis24, 286–295 (2003).
  • Pette D, Vrbova G. Adaptation of mammalian skeletal muscle fibers to chronic electrical stimulation. Rev. Physiol. Biochem. Pharmacol.120, 115–202 (1992).
  • Donoghue P, Doran P, Dowling P, Ohlendieck K. Differential expression of the fast skeletal muscle proteome following chronic low-frequency stimulation. Biochim. Biophys. Acta1752, 166–176 (2005).
  • Donoghue P, Doran P, Wynne K, Pedersen K, Dunn MJ, Ohlendieck K. Proteomic profiling of chronic low-frequency stimulated fast muscle. Proteomics7, 3417–3430 (2007).
  • Duchateau J, Semmler JG, Enoka RM. Training adaptations in the behavior of human motor units. J. Appl. Physiol.101, 1766–1775 (2006).
  • Lanza IR, Short DK, Short KR et al. Endurance exercise as a countermeasure for aging. Diabetes57, 2933–2942 (2008).
  • Capitanio D, Vigano A, Ricci E, Cerretelli P, Wait R, Gelfi C. Comparison of protein expression in human deltoideus and vastus lateralis muscles using two-dimensional gel electrophoresis. Proteomics5, 2577–2586 (2005).
  • Bouchard C, Rankinen T. Individual differences in response to regular physical activity. Med. Sci. Sports Exerc.33, S446–S451; discussion S452–S453 (2001).
  • Hubal MJ, Gordish-Dressman H, Thompson PD et al. Variability in muscle size and strength gain after unilateral resistance training. Med. Sci. Sports Exerc.37, 964–972 (2005).
  • Burniston JG. Changes in the rat skeletal muscle proteome induced by moderate-intensity endurance exercise. Biochim. Biophys. Acta1784, 1077–1086 (2008).
  • Burniston JG. Adaptation of the rat cardiac proteome in response to intensity-controlled endurance exercise. Proteomics9, 106–115 (2009).
  • Takahashi M, Kubota S. Exercise-related novel gene is involved in myoblast differentiation. Biomed. Res.26, 79–85 (2005).
  • Guelfi KJ, Casey TM, Giles JJ, Fournier PA, Arthur PG. A proteomic analysis of the acute effects of high-intensity exercise on skeletal muscle proteins in fasted rats. Clin. Exp. Pharmacol. Physiol.33, 952–957 (2006).
  • Gandra PG, Valente RH, Perales J, Pacheco AG, Macedo DV. Proteomic analysis of rat skeletal muscle submitted to one bout of incremental exercise. Scand. J. Med. Sci. Sports DOI: 10.1111/j.1600-0838.2010.01235.x (2010) (Epub ahead of print).
  • Shadel GS. Mitochondrial DNA, aconitase ‘wraps’ it up. Trends Biochem. Sci.30, 294–296 (2005).
  • Hojlund K, Wrzesinski K, Larsen PM et al. Proteome analysis reveals phosphorylation of ATP synthase β-subunit in human skeletal muscle and proteins with potential roles in Type 2 diabetes. J. Biol. Chem.278, 10436–10442 (2003).
  • Tjonna AE, Lee SJ, Rognmo O et al. Aerobic interval training versus continuous moderate exercise as a treatment for the metabolic syndrome: a pilot study. Circulation118, 346–354 (2008).
  • Yamaguchi W, Fujimoto E, Higuchi M, Tabata I. A DIGE proteomic analysis for high-intensity exercise-trained rat skeletal muscle. J. Biochem.148, 327–333 (2010).
  • Holloway KV, O’Gorman M, Woods P et al. Proteomic investigation of changes in human vastus lateralis muscle in response to interval-exercise training. Proteomics9, 5155–5174 (2009).
  • Ogut O, Granzier H, Jin JP. Acidic and basic troponin T isoforms in mature fast-twitch skeletal muscle and effect on contractility. Am J. Physiol.276, C1162–C1170 (1999).
  • Yu ZB, Gao F, Feng HZ, Jin JP. Differential regulation of myofilament protein isoforms underlying the contractility changes in skeletal muscle unloading. Am. J. Physiol. Cell Physiol.292, C1192–C1203 (2007).
  • Ross A, Leveritt M. Long-term metabolic and skeletal muscle adaptations to short-sprint training: implications for sprint training and tapering. Sports Med.31, 1063–1082 (2001).
  • Korzeniewski B, Zoladz JA. Factors determining the oxygen consumption rate (VO2) on-kinetics in skeletal muscles. Biochem. J.379, 703–710 (2004).
  • Morton JP, Kayani AC, McArdle A, Drust B. The exercise-induced stress response of skeletal muscle, with specific emphasis on humans. Sports Med.39, 643–662 (2009).
  • Boluyt MO, Brevick JL, Rogers DS, Randall MJ, Scalia AF, Li ZB. Changes in the rat heart proteome induced by exercise training: increased abundance of heat-shock protein hsp20. Proteomics6, 3154–3169 (2006).
  • Fan GC, Chu G, Kranias EG. Hsp20 and its cardioprotection. Trends Cardiovasc. Med.15, 138–141 (2005).
  • Pipkin W, Johnson JA, Creazzo TL, Burch J, Komalavilas P, Brophy C. Localization, macromolecular associations, and function of the small heat shock-related protein HSP20 in rat heart. Circulation107, 469–476 (2003).
  • Fan GC, Chu G, Mitton B, Song Q, Yuan Q, Kranias EG. Small heat-shock protein Hsp20 phosphorylation inhibits β-agonist-induced cardiac apoptosis. Circ. Res.94, 1474–1482 (2004).
  • Fan GC, Ren X, Qian J et al. Novel cardioprotective role of a small heat-shock protein, Hsp20, against ischemia/reperfusion injury. Circulation111, 1792–1799 (2005).
  • Qian J, Ren X, Wang X et al. Blockade of Hsp20 phosphorylation exacerbates cardiac ischemia/reperfusion injury by suppressed autophagy and increased cell death. Circ. Res.105, 1223–1231 (2009).
  • Sun B, Wang JH, Lv YY, Zhu SS, Yang J, Ma JZ. Proteomic adaptation to chronic high intensity swimming training in the rat heart. Comparative Biochem. Physiol. D3, 108–117 (2008).
  • Hittel DS, Kraus WE, Tanner CJ, Houmard JA, Hoffman EP. Exercise training increases electron and substrate shuttling proteins in muscle of overweight men and women with the metabolic syndrome. J. Appl. Physiol.98, 168–179 (2005).
  • Roepstorff C, Helge JW, Vistisen B, Kiens B. Studies of plasma membrane fatty acid-binding protein and other lipid-binding proteins in human skeletal muscle. Proc. Nutr. Soc.63, 239–244 (2004).
  • Dai Q, Escobar GP, Hakala KW, Lambert JM, Weintraub ST, Lindsey ML. The left ventricle proteome differentiates middle-aged and old left ventricles in mice. J. Proteome Res.7, 756–765 (2008).
  • Grant JE, Bradshaw AD, Schwacke JH, Baicu CF, Zile MR, Schey KL. Quantification of protein expression changes in the aging left ventricle of Rattus norvegicus. J. Proteome Res.8, 4252–4263 (2009).
  • Faber MJ, Dalinghaus M, Lankhuizen IM et al. Proteomic changes in the pressure overloaded right ventricle after 6 weeks in young rats: correlations with the degree of hypertrophy. Proteomics5, 2519–2530 (2005).
  • Bugger H, Schwarzer M, Chen D et al. Proteomic remodelling of mitochondrial oxidative pathways in pressure overload-induced heart failure. Cardiovasc. Res.85, 376–384 (2010).
  • Kavazis AN, Alvarez S, Talbert E, Lee Y, Powers SK. Exercise training induces a cardioprotective phenotype and alterations in cardiac subsarcolemmal and intermyofibrillar mitochondrial proteins. Am. J. Physiol. Heart Circ. Physiol.297, H144–H152 (2009).
  • Kaludercic N, Takimoto E, Nagayama T et al. Monoamine oxidase A-mediated enhanced catabolism of norepinephrine contributes to adverse remodeling and pump failure in hearts with pressure overload. Circ. Res.106, 193–202 (2010).
  • MacDonnell SM, Kubo H, Crabbe DL et al. Improved myocardial β-adrenergic responsiveness and signaling with exercise training in hypertension. Circulation111, 3420–3428 (2005).
  • Kemi OJ, Hoydal MA, Haram PM et al. Exercise training restores aerobic capacity and energy transfer systems in heart failure treated with losartan. Cardiovasc. Res.76, 91–99 (2007).
  • Bansal A, Dai Q, Chiao YA et al. Proteomic analysis reveals late exercise effects on cardiac remodeling following myocardial infarction. Proteomics73(10), 2041–2049 (2010).
  • Ascensao A, Ferreira R, Magalhaes J. Exercise-induced cardioprotection – biochemical, morphological and functional evidence in whole tissue and isolated mitochondria. Int. J. Cardiol.117, 16–30 (2007).
  • Koch LG, Britton SL. Artificial selection for intrinsic aerobic endurance running capacity in rats. Physiol. Genomics5, 45–52 (2001).
  • Wisloff U, Najjar SM, Ellingsen O et al. Cardiovascular risk factors emerge after artificial selection for low aerobic capacity. Science307, 418–420 (2005).
  • Lujan HL, Britton SL, Koch LG, DiCarlo SE. Reduced susceptibility to ventricular tachyarrhythmias in rats selectively bred for high aerobic capacity. Am. J. Physiol. Heart Circ. Physiol.291, H2933–H2941 (2006).
  • Noland RC, Thyfault JP, Henes ST et al. Artificial selection for high-capacity endurance running is protective against high-fat diet-induced insulin resistance. Am. J. Physiol. Endocrinol. Metab.293, E31–E41 (2007).
  • Palpant NJ, Szatkowski ML, Wang W et al. Artificial selection for whole animal low intrinsic aerobic capacity co-segregates with hypoxia-induced cardiac pump failure. PLoS One4, e6117 (2009).
  • Bye A, Langaas M, Hoydal MA et al. Aerobic capacity-dependent differences in cardiac gene expression. Physiol. Genomics33, 100–109 (2008).
  • Strom CC, Aplin M, Ploug T et al. Expression profiling reveals differences in metabolic gene expression between exercise-induced cardiac effects and maladaptive cardiac hypertrophy. FEBS J.272, 2684–2695 (2005).
  • Baldwin KM. Research in the exercise sciences: where do we go from here? J. Appl. Physiol.88, 332–336 (2000).
  • Boja ES, Phillips D, French SA, Harris RA, Balaban RS. Quantitative mitochondrial phosphoproteomics using iTRAQ on an LTQ-Orbitrap with high energy collision dissociation. J. Proteome Res.8, 4665–4675 (2009).
  • Gannon J, Staunton L, O’Connell K, Doran P, Ohlendieck K. Phosphoproteomic analysis of aged skeletal muscle. Int. J. Mol. Med.22, 33–42 (2008).
  • Lefort N, Yi Z, Bowen B et al. Proteome profile of functional mitochondria from human skeletal muscle using one-dimensional gel electrophoresis and HPLC–ESI–MS/MS. J. Proteomics72, 1046–1060 (2009).
  • Yin X, Cuello F, Mayr U et al. Proteomic analysis of the cardiac myofilament subproteome reveals dynamic alterations in phosphatase subunit distribution. Mol. Cell Proteomics9(3), 497–509 (2009).
  • Yuan C, Sheng Q, Tang H, Li Y, Zeng R, Solaro RJ. Quantitative comparison of sarcomeric phosphoproteomes of neonatal and adult rat hearts. Am. J. Physiol. Heart Circ. Physiol.295, H647–H656 (2008).
  • Deng N, Zhang J, Zong C et al. Phosphoproteome analysis reveals regulatory sites in major pathways of cardiac mitochondria. Mol. Cell Proteomics10(2), M110.000117 (2011).
  • Boersema PJ, Mohammed S, Heck AJ. Phosphopeptide fragmentation and analysis by mass spectrometry. J. Mass Spectrom.44, 861–878 (2009).
  • Scruggs SB, Hinken AC, Thawornkaiwong A et al. Ablation of ventricular myosin regulatory light chain phosphorylation in mice causes cardiac dysfunction in situ and affects neighboring myofilament protein phosphorylation. J. Biol. Chem.284, 5097–5106 (2009).
  • Olsson MC, Patel JR, Fitzsimons DP, Walker JW, Moss RL. Basal myosin light chain phosphorylation is a determinant of Ca2+ sensitivity of force and activation dependence of the kinetics of myocardial force development. Am. J. Physiol. Heart Circ. Physiol.287, H2712–H2718 (2004).
  • Kemi OJ, Ellingsen O, Smith GL, Wisloff U. Exercise-induced changes in calcium handling in left ventricular cardiomyocytes. Front Biosci.13, 356–368 (2008).
  • Moriggi M, Cassano P, Vasso M et al. A DIGE approach for the assessment of rat soleus muscle changes during unloading: effect of acetyl-L-carnitine supplementation. Proteomics8(17), 3588–3604 (2008).
  • Mintz M, Vanderver A, Brown KJ et al. Time series proteome profiling to study endoplasmic reticulum stress response. J. Proteome Res.7, 2435–2444 (2008).
  • Burniston JG, Connolly JB. Characterisation of isoform-specific tryptic peptides of rat cardiac myosin heavy chains using automated liquid chromatography-matrix assisted laser desorption ionisation (LC–MALDI) mass spectrometry. Int. J. Cardiol.140, 363–366 (2010).
  • Hojlund K, Yi Z, Hwang H et al. Characterization of the human skeletal muscle proteome by one-dimensional gel electrophoresis and HPLC–ESI–MS/MS. Mol. Cell Proteomics7, 257–267 (2007).
  • Parker KC, Walsh R, Salajegheh M et al. Characterization of human skeletal muscle biopsy samples using shotgun proteomics. J. Proteome Res.8(7), 3265–3277 (2009).
  • Bousette N, Kislinger T, Fong V et al. Large-scale characterization and analysis of the murine cardiac proteome. J. Proteome Res.8, 1887–1901 (2009).
  • Hwang H, Bowen BP, Lefort N et al. Proteomics analysis of human skeletal muscle reveals novel abnormalities in obesity and Type 2 diabetes. Diabetes59, 33–42 (2010).
  • Koch LG, Britton SL. Evolution, atmospheric oxygen, and complex disease. Physiol. Genomics30, 205–208 (2007).
  • Das AM. Regulation of the mitochondrial ATP-synthase in health and disease. Mol. Genet. Metab.79, 71–82 (2003).
  • Rose AJ, Frosig C, Kiens B, Wojtaszewski JF, Richter EA. Effect of endurance exercise training on Ca2+ calmodulin-dependent protein kinase II expression and signalling in skeletal muscle of humans. J. Physiol.583, 785–795 (2007).
  • Arrell DK, Elliott ST, Kane LA et al Proteomic analysis of pharmacological preconditioning: novel protein targets converge to mitochondrial metabolism pathways. Circ. Res.99, 706–714 (2006).
  • Kane LA, Van Eyk JE. Post-translational modifications of ATP synthase in the heart: biology and function. J. Bioenerg. Biomembr.41, 145–150 (2009).
  • Richardson MR, Lai X, Dixon JL, Sturek M, Witzmann FA. Diabetic dyslipidemia and exercise alter the plasma low-density lipoproteome in Yucatan pigs. Proteomics9, 2468–2483 (2009).
  • Sietsema KE, Meng F, Yates NA et al. Potential biomarkers of muscle injury after eccentric exercise. Biomarkers15, 249–258 (2010).
  • Hu S, Xie Y, Ramachandran P et al. Large-scale identification of proteins in human salivary proteome by liquid chromatography/mass spectrometry and two-dimensional gel electrophoresis–mass spectrometry. Proteomics5, 1714–1728 (2005).
  • Kohler M, Franz S, Regeniter A et al. Comparison of the urinary protein patterns of athletes by 2D-gel electrophoresis and mass spectrometry – a pilot study. Drug Test Anal.1, 382–386 (2009).
  • Maiorana A, O’Driscoll G, Cheetham C et al. Combined aerobic and resistance exercise training improves functional capacity and strength in CHF. J. Appl. Physiol.88, 1565–1570 (2000).
  • Goldspink DF. Ageing and activity: their effects on the functional reserve capacities of the heart and vascular smooth and skeletal muscles. Ergonomics48, 1334–1351 (2005).
  • Doherty MK, McLean L, Hayter JR et al. The proteome of chicken skeletal muscle: changes in soluble protein expression during growth in a layer strain. Proteomics4, 2082–2093 (2004).
  • Burniston JG, McLean L, Beynon RJ, Goldspink DF. Anabolic effects of a non-myotoxic dose of the β2-adrenergic receptor agonist clenbuterol on the rat plantaris muscle. Muscle Nerve35, 217–223 (2007).
  • Isfort RJ, Wang F, Greis KD et al. Proteomic analysis of rat soleus muscle undergoing hindlimb suspension-induced atrophy and reweighting hypertrophy. Proteomics2, 543–550 (2002).
  • Nader GA. Concurrent strength and endurance training: from molecules to man. Med. Sci. Sports Exerc.38, 1965–1970 (2006).
  • Ding Q, Vaynman S, Souda P, Whitelegge JP, Gomez-Pinilla F. Exercise affects energy metabolism and neural plasticity-related proteins in the hippocampus as revealed by proteomic analysis. Eur. J. Neurosci.24, 1265–1276 (2006).
  • Kirchner L, Chen WQ, Afjehi-Sadat L et al. Hippocampal metabolic proteins are modulated in voluntary and treadmill exercise rats. Exp. Neurol.212, 145–151 (2008).
  • Mizutani K, Sonoda S, Hayashi N et al. Analysis of protein expression profile in the cerebellum of cerebral infarction rats after treadmill training. Am. J. Phys. Med. Rehabil.89, 107–114 (2010).
  • Moriggi M, Vasso M, Fania C et al. Long term bed rest with and without vibration exercise countermeasures: effects on human muscle protein dysregulation. Proteomics10, 3756–3774 (2010).
  • Yang KD, Chang WC, Chuang H et al. Increased complement factor H with decreased factor B determined by proteomic differential displays as a biomarker of tai chi chuan exercise. Clin. Chem.56, 127–131 (2010).
  • Ichibangase T, Imai K. Application of fluorogenic derivatization-liquid chromatography-tandem mass spectrometric proteome method to skeletal muscle proteins in fast thoroughbred horses. J. Proteome Res.8(4), 2129–2134 (2009).
  • Bouwman FG, van Ginneken MM, Noben JP et al. Differential expression of equine muscle biopsy proteins during normal training and intensified training in young standardbred horses using proteomics technology. Comp. Biochem. Physiol. Part D Genomics Proteomics5, 55–64 (2010).
  • de Moraes R, Valente RH, León IR et al. Chronic dynamic exercise increases apolipoprotein A-I expression in rabbit renal cortex as determined by proteomic technology. Br. J. Sports Med.42, 386–388 (2008).
  • Gerecke KM, Jiao Y, Pani A, Pagala V, Smeyne RJ. Exercise protects against MPTP-induced neurotoxicity in mice. Brain Res.1341, 72–83 (2010).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.