Skip to main content

Advertisement

Log in

Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness

  • Review Article
  • Published:
Biogerontology Aims and scope Submit manuscript

Abstract

Some of the most serious consequences of ageing are its effects on skeletal muscle. The term ‘sarcopenia’ describes the slow but progressive loss of muscle mass with advancing age and is characterised by a deterioration of muscle quantity and quality leading to a gradual slowing of movement and a decline in strength. The loss of muscle mass and strength is thought to be attributed to the progressive atrophy and loss of individual muscle fibres associated with the loss of motor units, and a concomitant reduction in muscle ‘quality’ due to the infiltration of fat and other non-contractile material. These age-related changes in skeletal muscle can be largely attributed to the complex interaction of factors affecting neuromuscular transmission, muscle architecture, fibre composition, excitation–contraction coupling, and metabolism. Given the magnitude of the growing public health problems associated with sarcopenia, there is considerable interest in the development and evaluation of therapeutic strategies to attenuate, prevent, or ultimately reverse age-related muscle wasting and weakness. The aim is to review our current understanding of some of the cellular and molecular mechanisms responsible for age-related changes in skeletal muscle.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  • Allen RE, Boxhorn LK (1989) Regulation of skeletal muscle satellite cell proliferation and differentiation by transforming growth factor-beta, insulin-like growth factor I, and fibroblast growth factor. J Cell Physiol 138:311–315

    PubMed  CAS  Google Scholar 

  • Altuwaijri S, Lee DK, Chuang KH, Ting HJ, Yang Z, Xu Q, Tsai MY, Yeh S, Hanchett LA, Chang HC, Chang C (2004) Androgen receptor regulates expression of skeletal muscle-specific proteins and muscle cell types. Endocrine 25:27–32

    PubMed  CAS  Google Scholar 

  • Alway SE, Degens H, Krishnamurthy G, Smith CA (2002a) Potential role for Id myogenic repressors in apoptosis and attenuation of hypertrophy in muscles of aged rats. Am J Physiol Cell Physiol 283:C66–C76

    PubMed  CAS  Google Scholar 

  • Alway SE, Degens H, Lowe DA, Krishnamurthy G (2002b) Increased myogenic repressor Id mRNA and protein levels in hindlimb muscles of aged rats. Am J Physiol Regul Integr Comp Physiol 282:R411–R422

    PubMed  CAS  Google Scholar 

  • Amin S, Riggs BL, Atkinson EJ, Oberg AL, Melton LJ 3rd, Khosla S (2004) A potentially deleterious role of IGFBP-2 on bone density in aging men and women. J Bone Miner Res 19:1075–1083

    PubMed  CAS  Google Scholar 

  • Barton ER, Morris L, Musaro A, Rosenthal N, Sweeney HL (2002) Muscle-specific expression of insulin-like growth factor I counters muscle decline in mdx mice. J Cell Biol 157:137–148

    PubMed  CAS  Google Scholar 

  • Baumann AP, Ibebunjo C, Grasser WA, Paralkar VM (2003) Myostatin expression in age and denervation-induced skeletal muscle atrophy. J Musculoskelet Neuronal Interact 3:8–16

    PubMed  CAS  Google Scholar 

  • Beitzel F, Gregorevic P, Ryall JG, Plant DR, Sillence MN, Lynch GS (2004) β2-adrenoceptor agonist fenoterol enhances functional repair of regenerating rat skeletal muscle after injury. J Appl Physiol 96:1385–1392

    PubMed  CAS  Google Scholar 

  • Beitzel F, Sillence MN, Lynch GS (2007) β-Adrenoceptor signaling in regenerating skeletal muscle after β-agonist administration. Am J Physiol Endocrinol Metab 293:E932–E940

    PubMed  CAS  Google Scholar 

  • Benbassat CA, Maki KC, Unterman TG (1997) Circulating levels of insulin-like growth factor (IGF) binding protein-1 and -3 in aging men: relationships to insulin, glucose, IGF, and dehydroepiandrosterone sulfate levels and anthropometric measures. J Clin Endocrinol Metab 82:1484–1491

    PubMed  CAS  Google Scholar 

  • Berchtold MW, Brinkmeier H, Muntener M (2000) Calcium ion in skeletal muscle: its crucial role for muscle function, plasticity, and disease. Physiol Rev 80:1215–1265

    PubMed  CAS  Google Scholar 

  • Berridge MJ, Bootman MD, Roderick HL (2003) Calcium signalling: dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol 4:517–529

    PubMed  CAS  Google Scholar 

  • Bigot A, Jacquemin V, Debacq-Chainiaux F, Butler-Browne GS, Toussaint O, Furling D, Mouly V (2008) Replicative aging down regulates the myogenic regulatory factors in human myoblasts. Biol Cell 100:189–199

    PubMed  CAS  Google Scholar 

  • Brack AS, Bildsoe H, Hughes SM (2005) Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J Cell Sci 118:4813–4821

    PubMed  CAS  Google Scholar 

  • Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA (2007) Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317:807–810

    PubMed  CAS  Google Scholar 

  • Brooks SV, Faulkner JA (1994) Skeletal muscle weakness in old age: underlying mechanisms. Med Sci Sports Exerc 26:432–439

    PubMed  CAS  Google Scholar 

  • Buckingham M (2006) Myogenic progenitor cells and skeletal myogenesis in vertebrates. Curr Opin Genet Dev 16:525–532

    PubMed  CAS  Google Scholar 

  • Carlson BM, Faulkner JA (1989) Muscle transplantation between young and old rats: age of host determines recovery. Am J Physiol 256:C1262–C1266

    PubMed  CAS  Google Scholar 

  • Carlson BM, Faulkner JA (1996) The regeneration of noninnervated muscle grafts and marcaine-treated muscles in young and old rats. J Gerontol A Biol Sci Med Sci 51:B43–B49

    PubMed  CAS  Google Scholar 

  • Cederna PS, Asato H, Gu X, van der Meulen J, Kuzon WM Jr, Carlson BM, Faulkner JA (2001) Motor unit properties of nerve-intact extensor digitorum longus muscle grafts in young and old rats. J Gerontol A Biol Sci Med Sci 56:B254–B258

    PubMed  CAS  Google Scholar 

  • Chahal HS, Drake WM (2007) The endocrine system and ageing. J Pathol 211:173–180

    PubMed  CAS  Google Scholar 

  • Chargé SB, Rudnicki MA (2004) Cellular and molecular regulation of muscle regeneration. Physiol Rev 84:209–238

    PubMed  Google Scholar 

  • Chen Y, Zajac JD, MacLean HE (2005) Androgen regulation of satellite cell function. J Endocrinol 186:21–31

    PubMed  CAS  Google Scholar 

  • Chia DJ, Ono M, Woelfle J, Schlesinger-Massart M, Jiang H, Rotwein P (2006) Characterization of distinct Stat5b binding sites that mediate growth hormone-stimulated IGF-I gene transcription. J Biol Chem 281:3190–3197

    PubMed  CAS  Google Scholar 

  • Cohen TJ, Waddell DS, Barrientos T, Lu Z, Feng G, Cox GA, Bodine SC, Yao TP (2007) The histone deacetylase HDAC4 connects neural activity to muscle transcriptional reprogramming. J Biol Chem 282:33752–33759

    PubMed  CAS  Google Scholar 

  • Conboy IM, Rando TA (2002) The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell 3:397–409

    PubMed  CAS  Google Scholar 

  • Conboy IM, Conboy MJ, Smythe GM, Rando TA (2003) Notch-mediated restoration of regenerative potential to aged muscle. Science 302:1575–1577

    PubMed  CAS  Google Scholar 

  • Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA (2005) Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433:760–764

    PubMed  CAS  Google Scholar 

  • Conley KE, Cress ME, Jubrias SA, Esselman PC, Odderson IR (1995) From muscle properties to human performance, using magnetic resonance. J Gerontol A Biol Sci Med Sci 50(Spec No):35–40

    PubMed  Google Scholar 

  • Cree MG, Newcomer BR, Katsanos CS, Sheffield-Moore M, Chinkes D, Aarsland A, Urban R, Wolfe RR (2004) Intramuscular and liver triglycerides are increased in the elderly. J Clin Endocrinol Metab 89:3864–3871

    PubMed  CAS  Google Scholar 

  • Czifra G, Toth IB, Marincsak R, Juhasz I, Kovacs I, Acs P, Kovacs L, Blumberg PM, Biro T (2006) Insulin-like growth factor-I-coupled mitogenic signaling in primary cultured human skeletal muscle cells and in C2C12 myoblasts. A central role of protein kinase Cδ. Cell Signal 18:1461–1472

    PubMed  CAS  Google Scholar 

  • Dargelos E, Brule C, Combaret L, Hadj-Sassi A, Dulong S, Poussard S, Cottin P (2007) Involvement of the calcium-dependent proteolytic system in skeletal muscle aging. Exp Gerontol 42:1088–1098

    PubMed  CAS  Google Scholar 

  • Dedkov EI, Borisov AB, Carlson BM (2003a) Dynamics of postdenervation atrophy of young and old skeletal muscles: differential responses of fiber types and muscle types. J Gerontol A Biol Sci Med Sci 58:984–991

    PubMed  Google Scholar 

  • Dedkov EI, Kostrominova TY, Borisov AB, Carlson BM (2003b) MyoD and myogenin protein expression in skeletal muscles of senile rats. Cell Tissue Res 311:401–416

    PubMed  CAS  Google Scholar 

  • Dela F, Kjaer M (2006) Resistance training, insulin sensitivity and muscle function in the elderly. Essays Biochem 42:75–88

    PubMed  CAS  Google Scholar 

  • Delbono O (2003) Neural control of aging skeletal muscle. Aging Cell 2:21–29

    PubMed  CAS  Google Scholar 

  • Dirks AJ, Leeuwenburgh C (2006) Tumor necrosis factor alpha signaling in skeletal muscle: effects of age and caloric restriction. J Nutr Biochem 17:501–508

    PubMed  CAS  Google Scholar 

  • Dreyer HC, Blanco CE, Sattler FR, Schroeder ET, Wiswell RA (2006) Satellite cell numbers in young and older men 24 hours after eccentric exercise. Muscle Nerve 33:242–253

    PubMed  Google Scholar 

  • Edström L, Larsson L (1987) Effects of age on contractile and enzyme-histochemical properties of fast- and slow-twitch single motor units in the rat. J Physiol 392:129–145

    PubMed  Google Scholar 

  • Edstrom E, Altun M, Bergman E, Johnson H, Kullberg S, Ramirez-Leon V, Ulfhake B (2007) Factors contributing to neuromuscular impairment and sarcopenia during aging. Physiol Behav 92:129–135

    PubMed  Google Scholar 

  • Einsiedel LJ, Luff AR (1992) Alterations in the contractile properties of motor units within the ageing rat medial gastrocnemius. J Neurol Sci 112:170–177

    PubMed  CAS  Google Scholar 

  • Esler M, Kaye D, Thompson J, Jennings G, Cox H, Turner A, Lambert G, Seals D (1995) Effects of aging on epinephrine secretion and regional release of epinephrine from the human heart. J Clin Endocrinol Metab 80:435–442

    PubMed  CAS  Google Scholar 

  • Faulkner JA, Larkin LM, Claflin DR, Brooks SV (2007) Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol 34:1091–1096

    Article  PubMed  CAS  Google Scholar 

  • Firth SM, Baxter RC (2002) Cellular actions of the insulin-like growth factor binding proteins. Endocr Rev 23:824–854

    PubMed  CAS  Google Scholar 

  • Fligger JM, Malven PV, Doumit ME, Merkel RA, Grant AL (1998) Increases in insulin-like growth factor binding protein-2 accompany decreases in proliferation and differentiation when porcine muscle satellite cells undergo multiple passages. J Anim Sci 76:2086–2093

    PubMed  CAS  Google Scholar 

  • Florini JR, Ewton DZ, Falen SL, Van Wyk JJ (1986) Biphasic concentration dependency of stimulation of myoblast differentiation by somatomedins. Am J Physiol 250:C771–C778

    PubMed  CAS  Google Scholar 

  • Foulstone EJ, Savage PB, Crown AL, Holly JM, Stewart CE (2003) Role of insulin-like growth factor binding protein-3 (IGFBP-3) in the differentiation of primary human adult skeletal myoblasts. J Cell Physiol 195:70–79

    PubMed  CAS  Google Scholar 

  • Fraysse B, Desaphy JF, Rolland JF, Pierno S, Liantonio A, Giannuzzi V, Camerino C, Didonna MP, Cocchi D, De Luca A, Conte Camerino D (2006) Fiber type-related changes in rat skeletal muscle calcium homeostasis during aging and restoration by growth hormone. Neurobiol Dis 21:372–380

    PubMed  CAS  Google Scholar 

  • Fulle S, Di Donna S, Puglielli C, Pietrangelo T, Beccafico S, Bellomo R, Protasi F, Fano G (2005) Age-dependent imbalance of the antioxidative system in human satellite cells. Exp Gerontol 40:189–197

    PubMed  CAS  Google Scholar 

  • Gallegly JC, Turesky NA, Strotman BA, Gurley CM, Peterson CA, Dupont-Versteegden EE (2004) Satellite cell regulation of muscle mass is altered at old age. J Appl Physiol 97:1082–1090

    PubMed  Google Scholar 

  • Giresi PG, Stevenson EJ, Theilhaber J, Koncarevic A, Parkington J, Fielding RA, Kandarian SC (2005) Identification of a molecular signature of sarcopenia. Physiol Genomics 21:253–263

    PubMed  CAS  Google Scholar 

  • Gissel H (2005) The role of Ca2+ in muscle cell damage. Ann NY Acad Sci 1066:166–180

    PubMed  CAS  Google Scholar 

  • Giustina A, Veldhuis JD (1998) Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev 19:717–797

    PubMed  CAS  Google Scholar 

  • Goldspink G, Harridge SD (2004) Growth factors and muscle ageing. Exp Gerontol 39:1433–1438

    PubMed  CAS  Google Scholar 

  • Goldspink G, Yang SY (2004) The splicing of the IGF-I gene to yield different muscle growth factors. Adv Genet 52:23–49

    Article  PubMed  CAS  Google Scholar 

  • Gregorevic P, Plant DR, Leeding KS, Bach LA, Lynch GS (2002) Improved contractile function of the mdx dystrophic mouse diaphragm muscle after insulin-like growth factor-I administration. Am J Pathol 161:2263–2272

    PubMed  CAS  Google Scholar 

  • Gregorevic P, Plant DR, Lynch GS (2004) Administration of insulin-like growth factor-I improves fatigue resistance of skeletal muscles from dystrophic mdx mice. Muscle Nerve 30:295–304

    PubMed  CAS  Google Scholar 

  • Hagen JL, Krause DJ, Baker DJ, Fu MH, Tarnopolsky MA, Hepple RT (2004) Skeletal muscle aging in F344BN F1-hybrid rats: I. Mitochondrial dysfunction contributes to the age-associated reduction in VO2max. J Gerontol A Biol Sci Med Sci. 59:1099–1110

    PubMed  Google Scholar 

  • Hamada K, Vannier E, Sacheck JM, Witsell AL, Roubenoff R (2005) Senescence of human skeletal muscle impairs the local inflammatory cytokine response to acute eccentric exercise. Faseb J 19:264–266

    PubMed  CAS  Google Scholar 

  • Hameed M, Orrell RW, Cobbold M, Goldspink G, Harridge SD (2003) Expression of IGF-I splice variants in young and old human skeletal muscle after high resistance exercise. J Physiol 547:247–254

    PubMed  CAS  Google Scholar 

  • Herbert J (1995) The age of dehydroepiandrosterone. Lancet 345:1193–1194

    PubMed  CAS  Google Scholar 

  • Herbst KL, Bhasin S (2004) Testosterone action on skeletal muscle. Curr Opin Clin Nutr Metab Care 7:271–277

    PubMed  CAS  Google Scholar 

  • Hepple RT, Baker DJ, McConkey M, Murynka T, Norris R (2006) Caloric restriction protects mitochondrial function with aging in skeletal and cardiac muscles. Rejuvenation Res 9:219–222

    PubMed  CAS  Google Scholar 

  • Hermann M, Berger P (2001) Hormonal changes in aging men: a therapeutic indication? Exp Gerontol 36:1075–1082

    PubMed  CAS  Google Scholar 

  • Iannuzzi-Sucich M, Prestwood KM, Kenny AM (2002) Prevalence of sarcopenia and predictors of skeletal muscle mass in healthy, older men and women. J Gerontol A Biol Sci Med Sci 57:M772–M777

    PubMed  Google Scholar 

  • Isgaard J, Carlsson L, Isaksson OG, Jansson JO (1988) Pulsatile intravenous growth hormone (GH) infusion to hypophysectomized rats increases insulin-like growth factor I messenger ribonucleic acid in skeletal tissues more effectively than continuous GH infusion. Endocrinology 123:2605–2610

    PubMed  CAS  Google Scholar 

  • Janssen JA, Stolk RP, Pols HA, Grobbee DE, Lamberts SW (1998) Serum free and total insulin-like growth factor-I, insulin-like growth factor binding protein-1 and insulin-like growth factor binding protein-3 Levels in healthy elderly individuals. Relation to self-reported quality of health and disability. Gerontology 44:277–280

    PubMed  CAS  Google Scholar 

  • Kadi F, Charifi N, Denis C, Lexell J (2004) Satellite cells and myonuclei in young and elderly women and men. Muscle Nerve 29:120–127

    PubMed  Google Scholar 

  • Kawada S, Tachi C, Ishii N (2001) Content and localization of myostatin in mouse skeletal muscles during aging, mechanical unloading and reloading. J Muscle Res Cell Motil 22:627–633

    PubMed  CAS  Google Scholar 

  • Kaye D, Esler M (2005) Sympathetic neuronal regulation of the heart in aging and heart failure. Cardiovasc Res 66:256–264

    PubMed  CAS  Google Scholar 

  • Kelly M, Keller C, Avilucea PR, Keller P, Luo Z, Xiang X, Giralt M, Hidalgo J, Saha AK, Pedersen BK, Ruderman NB (2004) AMPK activity is diminished in tissues of IL-6 knockout mice: the effect of exercise. Biochem Biophys Res Commun 320:449–454

    PubMed  CAS  Google Scholar 

  • Khosla S, Melton LJ 3rd, Atkinson EJ, O’Fallon WM, Klee GG, Riggs BL (1998) Relationship of serum sex steroid levels and bone turnover markers with bone mineral density in men and women: a key role for bioavailable estrogen. J Clin Endocrinol Metab 83:2266–2274

    PubMed  CAS  Google Scholar 

  • Kostrominova TY, Macpherson PC, Carlson BM, Goldman D (2000) Regulation of myogenin protein expression in denervated muscles from young and old rats. Am J Physiol Regul Integr Comp Physiol 279:R179–R188

    PubMed  CAS  Google Scholar 

  • Langley B, Thomas M, Bishop A, Sharma M, Gilmour S, Kambadur R (2002) Myostatin inhibits myoblast differentiation by down-regulating MyoD expression. J Biol Chem 277:49831–49840

    PubMed  CAS  Google Scholar 

  • Lanning NJ, Carter-Su C (2006) Recent advances in growth hormone signaling. Rev Endocr Metab Disord 7:225–235

    PubMed  CAS  Google Scholar 

  • Larkin LM, Halter JB, Supiano MA (1996) Effect of aging on rat skeletal muscle β-AR function in male Fischer 344 x brown Norway rats. Am J Physiol 270:R462–R468

    PubMed  CAS  Google Scholar 

  • Larsson L, Sjödin B, Karlsson J (1978) Histochemical and biochemical changes in human skeletal muscle with age in sedentary males, age 22–65 years. Acta Physiol Scand 103:31–39

    PubMed  CAS  Google Scholar 

  • Larsson L, Grimby G, Karlsson J (1979) Muscle strength and speed of movement in relation to age and muscle morphology. J Appl Physiol 46:451–456

    PubMed  CAS  Google Scholar 

  • Lee SJ (2004) Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol 20:61–86

    PubMed  CAS  Google Scholar 

  • Lexell J (1997) Evidence for nervous system degeneration with advancing age. J Nutr 127:1011S–1013S

    PubMed  CAS  Google Scholar 

  • Luff AR (1998) Age-associated changes in the innervation of muscle fibers and changes in the mechanical properties of motor units. Ann NY Acad Sci 854:92–101

    PubMed  CAS  Google Scholar 

  • Luo D, Renault VM, Rando TA (2005) The regulation of Notch signaling in muscle stem cell activation and postnatal myogenesis. Semin Cell Dev Biol 16:612–622

    PubMed  CAS  Google Scholar 

  • Lynch GS (2004a) Tackling Australia’s future health problems: developing strategies to combat sarcopenia–age-related muscle wasting and weakness. Intern Med J 34:294–296

    PubMed  CAS  Google Scholar 

  • Lynch GS (2004b) Emerging drugs for sarcopenia: age-related muscle wasting. Expert Opin Emerg Drugs 9:345–361

    PubMed  CAS  Google Scholar 

  • Lynch GS, Ryall JG (2008) Role of β-adrenoceptor signaling in skeletal muscle—implications for muscle wasting and disease. Physiol Rev. doi:10.1152/physrev.00028.2007

    PubMed  Google Scholar 

  • Marcell TJ, Harman SM, Urban RJ, Metz DD, Rodgers BD, Blackman MR (2001) Comparison of GH, IGF-I, and testosterone with mRNA of receptors and myostatin in skeletal muscle in older men. Am J Physiol Endocrinol Metab 281:E1159–E1164

    PubMed  CAS  Google Scholar 

  • Mariotti S, Franceschi C, Cossarizza A, Pinchera A (1995) The aging thyroid. Endocr Rev 16:686–715

    PubMed  CAS  Google Scholar 

  • McCroskery S, Thomas M, Maxwell L, Sharma M, Kambadur R (2003) Myostatin negatively regulates satellite cell activation and self-renewal. J Cell Biol 162:1135–1147

    PubMed  CAS  Google Scholar 

  • McCroskery S, Thomas M, Platt L, Hennebry A, Nishimura T, McLeay L, Sharma M, Kambadur R (2005) Improved muscle healing through enhanced regeneration and reduced fibrosis in myostatin-null mice. J Cell Sci 118:3531–3541

    PubMed  CAS  Google Scholar 

  • McNeil CJ, Doherty TJ, Stashuk DW, Rice CL (2005) Motor unit number estimates in the tibialis anterior muscle of young, old, and very old men. Muscle Nerve 31:461–467

    PubMed  Google Scholar 

  • McPherron AC, Lawler AM, Lee SJ (1997) Regulation of skeletal muscle mass in mice by a new TGF-β superfamily member. Nature 387:83–90

    PubMed  CAS  Google Scholar 

  • Messi ML, Delbono O (2003) Target-derived trophic effect on skeletal muscle innervation in senescent mice. J Neurosci 23:1351–1359

    PubMed  CAS  Google Scholar 

  • Morley JE, Kaiser F, Raum WJ, Perry HM 3rd, Flood JF, Jensen J, Silver AJ, Roberts E (1997) Potentially predictive and manipulable blood serum correlates of aging in the healthy human male: progressive decreases in bioavailable testosterone, dehydroepiandrosterone sulfate, and the ratio of insulin-like growth factor 1 to growth hormone. Proc Natl Acad Sci U S A 94:7537–7542

    PubMed  CAS  Google Scholar 

  • Musarò A, Cusella De Angelis MG, Germani A, Ciccarelli C, Molinaro M, Zani BM (1995) Enhanced expression of myogenic regulatory genes in aging skeletal muscle. Exp Cell Res 221:241–248

    PubMed  Google Scholar 

  • Musarò A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, Rosenthal N (2001) Localized IGF-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet 27:195–200

    PubMed  Google Scholar 

  • Nagley P, Mackay IR, Baumer A, Maxwell RJ, Vaillant F, Wang ZX, Zhang C, Linnane AW (1992) Mitochondrial DNA mutation associated with aging and degenerative disease. Ann NY Acad Sci 673:92–102

    PubMed  CAS  Google Scholar 

  • Nair KS, Rizza RA, O’Brien P, Dhatariya K, Short KR, Nehra A, Vittone JL, Klee GG, Basu A, Basu R, Cobelli C, Toffolo G, Dalla Man C, Tindall DJ, Melton LJ 3rd, Smith GE, Khosla S, Jensen MD (2006) DHEA in elderly women and DHEA or testosterone in elderly men. N Engl J Med 355:1647–1659

    PubMed  CAS  Google Scholar 

  • Narayanan N, Jones DL, Xu A, Yu JC (1996) Effects of aging on sarcoplasmic reticulum function and contraction duration in skeletal muscles of the rat. Am J Physiol 271:C1032–C1040

    PubMed  CAS  Google Scholar 

  • O’Connor KG, Tobin JD, Harman SM, Plato CC, Roy TA, Sherman SS, Blackman MR (1998) Serum levels of insulin-like growth factor-I are related to age and not to body composition in healthy women and men. J Gerontol A Biol Sci Med Sci 53:M176–M182

    PubMed  CAS  Google Scholar 

  • Owino V, Yang SY, Goldspink G (2001) Age-related loss of skeletal muscle function and the inability to express the autocrine form of insulin-like growth factor-1 (MGF) in response to mechanical overload. FEBS Lett 505:259–263

    PubMed  CAS  Google Scholar 

  • Pampusch MS, Kamanga-Sollo E, White ME, Hathaway MR, Dayton WR (2003) Effect of recombinant porcine IGF-binding protein-3 on proliferation of embryonic porcine myogenic cell cultures in the presence and absence of IGF-I. J Endocrinol 176:227–235

    PubMed  CAS  Google Scholar 

  • Pampusch MS, Xi G, Kamanga-Sollo E, Loseth KJ, Hathaway MR, Dayton WR, White ME (2005) Production of recombinant porcine IGF-binding protein-5 and its effect on proliferation of porcine embryonic myoblast cultures in the presence and absence of IGF-I and Long-R3-IGF-I. J Endocrinol 185:197–206

    PubMed  CAS  Google Scholar 

  • Pedersen BK, Fischer CP (2007) Physiological roles of muscle-derived interleukin-6 in response to exercise. Curr Opin Clin Nutr Metab Care 10:265–271

    Article  PubMed  CAS  Google Scholar 

  • Pedersen M, Steensberg A, Keller C, Osada T, Zacho M, Saltin B, Febbraio MA, Pedersen BK (2004) Does the aging skeletal muscle maintain its endocrine function? Exerc Immunol Rev 10:42–55

    PubMed  Google Scholar 

  • Petrella JK, Kim JS, Cross JM, Kosek DJ, Bamman MM (2006) Efficacy of myonuclear addition may explain differential myofiber growth among resistance-trained young and older men and women. Am J Physiol Endocrinol Metab 291:E937–E946

    PubMed  CAS  Google Scholar 

  • Plant DR, Lynch GS (2002) Excitation–contraction coupling and sarcoplasmic reticulum function in mechanically skinned fibres from fast skeletal muscles of aged mice. J Physiol 543:169–176

    PubMed  CAS  Google Scholar 

  • Plomgaard P, Penkowa M, Pedersen BK (2005) Fiber type specific expression of TNF-alpha, IL-6 and IL-18 in human skeletal muscles. Exerc Immunol Rev 11:53–63

    PubMed  Google Scholar 

  • Prakash YS, Sieck GC (1998) Age-related remodeling of neuromuscular junctions on type-identified diaphragm fibers. Muscle Nerve 21:887–895

    PubMed  CAS  Google Scholar 

  • Przybyla B, Gurley C, Harvey JF, Bearden E, Kortebein P, Evans WJ, Sullivan DH, Peterson CA, Dennis RA (2006) Aging alters macrophage properties in human skeletal muscle both at rest and in response to acute resistance exercise. Exp Gerontol 41:320–327

    PubMed  CAS  Google Scholar 

  • Raue U, Slivka D, Jemiolo B, Hollon C, Trappe S (2006) Myogenic gene expression at rest and after a bout of resistance exercise in young (18–30 yr) and old (80–89 yr) women. J Appl Physiol 101:53–59

    PubMed  CAS  Google Scholar 

  • Renault V, Thornell LE, Eriksson PO, Butler-Browne G, Mouly V (2002) Regenerative potential of human skeletal muscle during aging. Aging Cell 1:132–139

    PubMed  CAS  Google Scholar 

  • Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, Yancopoulos GD, Glass DJ (2001) Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol 3:1009–1013

    PubMed  CAS  Google Scholar 

  • Rosenheimer JL, Smith DO (1985) Differential changes in the end-plate architecture of functionally diverse muscles during aging. J Neurophysiol 53:1567–1581

    PubMed  CAS  Google Scholar 

  • Roth SM, Martel GF, Ivey FM, Lemmer JT, Metter EJ, Hurley BF, Rogers MA (2000) Skeletal muscle satellite cell populations in healthy young and older men and women. Anat Rec 260:351–358

    PubMed  CAS  Google Scholar 

  • Roubenoff R (2003) Catabolism of aging: is it an inflammatory process? Curr Opin Clin Nutr Metab Care 6:295–299

    PubMed  Google Scholar 

  • Roubenoff R, Harris TB, Abad LW, Wilson PW, Dallal GE, Dinarello CA (1998) Monocyte cytokine production in an elderly population: effect of age and inflammation. J Gerontol A Biol Sci Med Sci 53:M20–M26

    PubMed  CAS  Google Scholar 

  • Ryall JG, Schertzer JD, Lynch GS (2007) Attenuation of age-related muscle wasting and weakness in rats after formoterol treatment: therapeutic implications for sarcopenia. J Gerontol A Biol Sci Med Sci 62:813–823

    PubMed  Google Scholar 

  • Sacheck JM, Cannon JG, Hamada K, Vannier E, Blumberg JB, Roubenoff R (2006) Age-related loss of associations between acute exercise-induced IL-6 and oxidative stress. Am J Physiol Endocrinol Metab 291:E340–E349

    PubMed  CAS  Google Scholar 

  • Salviati G, Zeviani M, Betto R, Nacamulli D, Busnardo B (1985) Effects of thyroid hormones on the biochemical specialization of human muscle fibers. Muscle Nerve 8:363–371

    PubMed  CAS  Google Scholar 

  • Schaap LA, Pluijm SM, Smit JH, van Schoor NM, Visser M, Gooren LJ, Lips P (2005) The association of sex hormone levels with poor mobility, low muscle strength and incidence of falls among older men and women. Clin Endocrinol (Oxf) 63:152–160

    CAS  Google Scholar 

  • Schertzer JD, Plant DR, Ryall JG, Beitzel F, Stupka N, Lynch GS (2005) β2-agonist administration increases sarcoplasmic reticulum Ca2+-ATPase activity in aged rat skeletal muscle. Am J Physiol Endocrinol Metab 288:E526–E533

    PubMed  CAS  Google Scholar 

  • Schertzer JD, Ryall JG, Lynch GS (2006) Systemic administration of IGF-I enhances oxidative status and reduces contraction-induced injury in skeletal muscles of mdx dystrophic mice. Am J Physiol Endocrinol Metab 291:E499–E505

    PubMed  CAS  Google Scholar 

  • Schertzer JD, van der Poel C, Shavlakadze T, Grounds MD, Lynch GS (2008) Muscle specific overexpression of IGF-I improves E–C coupling in skeletal muscle fibers from dystrophic mdx mice. Am J Physiol Cell Physiol 294:C161–C168

    PubMed  CAS  Google Scholar 

  • Serrano AL, Baeza-Raja B, Perdiguero E, Jardi M, Munoz-Canoves P (2008) Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle hypertrophy. Cell Metab 7:33–44

    PubMed  CAS  Google Scholar 

  • Sherlock M, Toogood AA (2007) Aging and the growth hormone/insulin like growth factor-I axis. Pituitary 10:189–203

    PubMed  CAS  Google Scholar 

  • Short KR, Moller N, Bigelow ML, Coenen-Schimke J, Nair KS (2008) Enhancement of muscle mitochondrial function by growth hormone. J Clin Endocrinol Metab 93:597–604

    PubMed  CAS  Google Scholar 

  • Simonides WS, Brent GA, Thelen MH, van der Linden CG, Larsen PR, van Hardeveld C (1996) Characterization of the promoter of the rat sarcoplasmic endoplasmic reticulum Ca2+-ATPase 1 gene and analysis of thyroid hormone responsiveness. J Biol Chem 271:32048–32056

    PubMed  CAS  Google Scholar 

  • Sinha-Hikim I, Taylor WE, Gonzalez-Cadavid NF, Zheng W, Bhasin S (2004) Androgen receptor in human skeletal muscle and cultured muscle satellite cells: up-regulation by androgen treatment. J Clin Endocrinol Metab 89:5245–5255

    PubMed  CAS  Google Scholar 

  • Sinha-Hikim I, Cornford M, Gaytan H, Lee ML, Bhasin S (2006) Effects of testosterone supplementation on skeletal muscle fiber hypertrophy and satellite cells in community-dwelling older men. J Clin Endocrinol Metab 91:3024–3033

    PubMed  CAS  Google Scholar 

  • Siriett V, Platt L, Salerno MS, Ling N, Kambadur R, Sharma M (2006) Prolonged absence of myostatin reduces sarcopenia. J Cell Physiol 209:866–873

    PubMed  CAS  Google Scholar 

  • Siriett V, Salerno MS, Berry C, Nicholas G, Bower R, Kambadur R, Sharma M (2007) Antagonism of myostatin enhances muscle regeneration during sarcopenia. Mol Ther 15:1463–1470

    PubMed  CAS  Google Scholar 

  • Snow MH (1977) The effects of aging on satellite cells in skeletal muscles of mice and rats. Cell Tissue Res 185:399–408

    PubMed  CAS  Google Scholar 

  • Sohal RS, Weindruch R (1996) Oxidative stress, caloric restriction, and aging. Science 273:59–63

    PubMed  CAS  Google Scholar 

  • Solomon AM, Bouloux PM (2006) Modifying muscle mass - the endocrine perspective. J Endocrinol 191:349–360

    PubMed  CAS  Google Scholar 

  • Sotiropoulos A, Ohanna M, Kedzia C, Menon RK, Kopchick JJ, Kelly PA, Pende M (2006) Growth hormone promotes skeletal muscle cell fusion independent of insulin-like growth factor 1 up-regulation. Proc Natl Acad Sci U S A 103:7315–7320

    PubMed  CAS  Google Scholar 

  • Spangenburg EE, Abraha T, Childs TE, Pattison JS, Booth FW (2003) Skeletal muscle IGF-binding protein-3 and -5 expressions are age, muscle, and load dependent. Am J Physiol Endocrinol Metab 284:E340–E350

    PubMed  CAS  Google Scholar 

  • Stewart CE, Newcomb PV, Holly JM (2004) Multifaceted roles of TNF-alpha in myoblast destruction: a multitude of signal transduction pathways. J Cell Physiol 198:237–247

    PubMed  CAS  Google Scholar 

  • Szulc P, Beck TJ, Marchand F, Delmas PD (2005) Low skeletal muscle mass is associated with poor structural parameters of bone and impaired balance in elderly men–the MINOS study. J Bone Miner Res 20:721–729

    PubMed  Google Scholar 

  • Thompson RW, McClung JM, Baltgalvis KA, Davis JM, Carson JA (2006) Modulation of overload-induced inflammation by aging and anabolic steroid administration. Exp Gerontol 41:1136–1148

    PubMed  CAS  Google Scholar 

  • Tsujinaka T, Fujita J, Ebisui C, Yano M, Kominami E, Suzuki K, Tanaka K, Katsume A, Ohsugi Y, Shiozaki H, Monden M (1996) Interleukin 6 receptor antibody inhibits muscle atrophy and modulates proteolytic systems in interleukin 6 transgenic mice. J Clin Invest 97:244–249

    Article  PubMed  CAS  Google Scholar 

  • Turner JD, Rotwein P, Novakofski J, Bechtel PJ (1988) Induction of mRNA for IGF-I and -II during growth hormone-stimulated muscle hypertrophy. Am J Physiol 255:E513–E517

    PubMed  CAS  Google Scholar 

  • van der Linden GC, Simonides WS, van Hardeveld C (1992) Thyroid hormone regulates Ca2+-ATPase mRNA levels of sarcoplasmic reticulum during neonatal development of fast skeletal muscle. Mol Cell Endocrinol 90:125–131

    PubMed  Google Scholar 

  • van den Beld AW, de Jong FH, Grobbee DE, Pols HA, Lamberts SW (2000) Measures of bioavailable serum testosterone and estradiol and their relationships with muscle strength, bone density, and body composition in elderly men. J Clin Endocrinol Metab 85:3276–3282

    PubMed  Google Scholar 

  • van den Beld AW, Lamberts SW (2002) Endocrine aspects of healthy ageing in men. In: Novartis Foundation (eds) Endocrine facets of ageing. Wiley, New York, pp 3–25

    Google Scholar 

  • van den Beld AW, Blum WF, Pols HA, Grobbee DE, Lamberts SW (2003) Serum insulin-like growth factor binding protein-2 levels as an indicator of functional ability in elderly men. Eur J Endocrinol 148:627–634

    PubMed  Google Scholar 

  • Veldhuis JD, Iranmanesh A, Weltman A (1997) Elements in the pathophysiology of diminished growth hormone (GH) secretion in aging humans. Endocrine 7:41–48

    PubMed  CAS  Google Scholar 

  • Verdijk LB, Koopman R, Schaart G, Meijer K, Savelberg HH, van Loon LJ (2007) Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly. Am J Physiol Endocrinol Metab 292:E151–E157

    PubMed  CAS  Google Scholar 

  • Vermeulen A (1991) Clinical review 24: Androgens in the aging male. J Clin Endocrinol Metab 73:221–224

    PubMed  CAS  Google Scholar 

  • Viguerie N, Langin D (2003) Effect of thyroid hormone on gene expression. Curr Opin Clin Nutr Metab Care 6:377–381

    PubMed  CAS  Google Scholar 

  • Visser M, Pahor M, Taaffe DR, Goodpaster BH, Simonsick EM, Newman AB, Nevitt M, Harris TB (2002) Relationship of interleukin-6 and tumor necrosis factor-alpha with muscle mass and muscle strength in elderly men and women: the Health ABC Study. J Gerontol A Biol Sci Med Sci 57:M326–M332

    PubMed  Google Scholar 

  • Wagner KR, Liu X, Chang X, Allen RE (2005) Muscle regeneration in the prolonged absence of myostatin. Proc Natl Acad Sci U S A 102:2519–2524

    PubMed  CAS  Google Scholar 

  • Waters DL, Yau CL, Montoya GD, Baumgartner RN (2003) Serum Sex Hormones, IGF-1, and IGFBP3 Exert a Sexually Dimorphic Effect on Lean Body Mass in Aging. J Gerontol A Biol Sci Med Sci 58:648–652

    PubMed  Google Scholar 

  • Welle S, Bhatt K, Shah B, Thornton C (2002) Insulin-like growth factor-1 and myostatin mRNA expression in muscle: comparison between 62–77 and 21–31 yr old men. Exp Gerontol 37:833–839

    PubMed  CAS  Google Scholar 

  • Yarasheski KE (2003) Exercise, aging, and muscle protein metabolism. J Gerontol A Biol Sci Med Sci 58:M918–M922

    PubMed  Google Scholar 

  • Yu F, Hedstrom M, Cristea A, Dalen N, Larsson L (2007) Effects of ageing and gender on contractile properties in human skeletal muscle and single fibres. Acta Physiol (Oxf) 190:229–241

    CAS  Google Scholar 

  • Zadik Z, Chalew SA, McCarter RJ Jr, Meistas M, Kowarski AA (1985) The influence of age on the 24-hour integrated concentration of growth hormone in normal individuals. J Clin Endocrinol Metab 60:513–516

    Article  PubMed  CAS  Google Scholar 

  • Zebedee Z, Hara E (2001) Id proteins in cell cycle control and cellular senescence. Oncogene 20:8317–8325

    PubMed  CAS  Google Scholar 

  • Zheng Z, Wang ZM, Delbono O (2002) Insulin-like growth factor-1 increases skeletal muscle dihydropyridine receptor α1S transcriptional activity by acting on the cAMP-response element-binding protein element of the promoter region. J Biol Chem 277:50535–50542

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Supported by research grant funding from the Australian Research Council Discovery-Project funding scheme (DP0665071, DP0772781), the National Health and Medical Research Council of Australia (350439, 454561, 509313), the Muscular Dystrophy Association (USA, 3595, 4167), the Rebecca L. Cooper Medical Research Foundation, and Pfizer Inc. (USA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gordon S. Lynch.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ryall, J.G., Schertzer, J.D. & Lynch, G.S. Cellular and molecular mechanisms underlying age-related skeletal muscle wasting and weakness. Biogerontology 9, 213–228 (2008). https://doi.org/10.1007/s10522-008-9131-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10522-008-9131-0

Keywords

Navigation