Skip to main content
Top
Published in: Journal of Cachexia, Sarcopenia and Muscle 2/2012

Open Access 01-06-2012 | Review

Evolving concepts on the age-related changes in “muscle quality”

Authors: David W. Russ, Kimberly Gregg-Cornell, Matthew J. Conaway, Brian C. Clark

Published in: Journal of Cachexia, Sarcopenia and Muscle | Issue 2/2012

Login to get access

Abstract

The deterioration of skeletal muscle with advancing age has long been anecdotally recognized and has been of scientific interest for more than 150 years. Over the past several decades, the scientific and medical communities have recognized that skeletal muscle dysfunction (e.g., muscle weakness, poor muscle coordination, etc.) is a debilitating and life-threatening condition in the elderly. For example, the age-associated loss of muscle strength is highly associated with both mortality and physical disability. It is well-accepted that voluntary muscle force production is not solely dependent upon muscle size, but rather results from a combination of neurologic and skeletal muscle factors, and that biologic properties of both of these systems are altered with aging. Accordingly, numerous scientists and clinicians have used the term “muscle quality” to describe the relationship between voluntary muscle strength and muscle size. In this review article, we discuss the age-associated changes in the neuromuscular system—starting at the level of the brain and proceeding down to the subcellular level of individual muscle fibers—that are potentially influential in the etiology of dynapenia (age-related loss of muscle strength and power).
Literature
1.
go back to reference Federal Interagency Forum on Aging-Related Statistics. Older Americans 2008: key indicators of well-being. Washington: U.S. Government Printing Office; 2008. Federal Interagency Forum on Aging-Related Statistics. Older Americans 2008: key indicators of well-being. Washington: U.S. Government Printing Office; 2008.
2.
go back to reference Brooke JD, et al. Aging of human segmental oligosynaptic reflexes for control of leg movement. Neurobiol Aging. 1989;10:721–5.PubMedCrossRef Brooke JD, et al. Aging of human segmental oligosynaptic reflexes for control of leg movement. Neurobiol Aging. 1989;10:721–5.PubMedCrossRef
3.
go back to reference Cesari M, et al. Skeletal muscle and mortality results from the InCHIANTI Study. J Gerontol A Biol Sci Med Sci. 2009;64:377–84.PubMedCrossRef Cesari M, et al. Skeletal muscle and mortality results from the InCHIANTI Study. J Gerontol A Biol Sci Med Sci. 2009;64:377–84.PubMedCrossRef
4.
go back to reference Manini TM, et al. Knee extension strength cutpoints for maintaining mobility. J Am Geriatr Soc. 2007;55:451–7.PubMedCrossRef Manini TM, et al. Knee extension strength cutpoints for maintaining mobility. J Am Geriatr Soc. 2007;55:451–7.PubMedCrossRef
5.
go back to reference Newman AB, et al. Strength, but not muscle mass, is associated with mortality in the health, aging and body composition study cohort. J Gerontol A Biol Sci Med Sci. 2006;61:72–7.PubMedCrossRef Newman AB, et al. Strength, but not muscle mass, is associated with mortality in the health, aging and body composition study cohort. J Gerontol A Biol Sci Med Sci. 2006;61:72–7.PubMedCrossRef
6.
go back to reference Visser M, et al. Skeletal muscle mass and muscle strength in relation to lower-extremity performance in older men and women. J Am Geriatr Soc. 2000;48:381–6.PubMed Visser M, et al. Skeletal muscle mass and muscle strength in relation to lower-extremity performance in older men and women. J Am Geriatr Soc. 2000;48:381–6.PubMed
7.
go back to reference Visser M, et al. Change in muscle mass and muscle strength after a hip fracture: relationship to mobility recovery. J Gerontol A Biol Sci Med Sci. 2000;55:M434–40.PubMedCrossRef Visser M, et al. Change in muscle mass and muscle strength after a hip fracture: relationship to mobility recovery. J Gerontol A Biol Sci Med Sci. 2000;55:M434–40.PubMedCrossRef
8.
go back to reference von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachex Sarcopenia Muscle. 2010;1:129–33.CrossRef von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachex Sarcopenia Muscle. 2010;1:129–33.CrossRef
10.
go back to reference Delmonico MJ, et al. Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr. 2009;90:1579–85.PubMedCrossRef Delmonico MJ, et al. Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr. 2009;90:1579–85.PubMedCrossRef
11.
go back to reference Clark BC, et al. Adaptations in human neuromuscular function following prolonged unweighting: II. Neurological properties and motor imagery efficacy. J Appl Physiol. 2006;101:264–72.PubMedCrossRef Clark BC, et al. Adaptations in human neuromuscular function following prolonged unweighting: II. Neurological properties and motor imagery efficacy. J Appl Physiol. 2006;101:264–72.PubMedCrossRef
12.
go back to reference Clark BC, Fernhall B, Ploutz-Snyder LL. Adaptations in human neuromuscular function following prolonged unweighting: I. Skeletal muscle contractile properties and applied ischemia efficacy. J Appl Physiol. 2006;101:256–63.PubMedCrossRef Clark BC, Fernhall B, Ploutz-Snyder LL. Adaptations in human neuromuscular function following prolonged unweighting: I. Skeletal muscle contractile properties and applied ischemia efficacy. J Appl Physiol. 2006;101:256–63.PubMedCrossRef
13.
go back to reference Russ DW, et al. Ageing, but not yet senescent, rats exhibit reduced muscle quality and sarcoplasmic reticulum function. Acta Physiol. 2011;201:391–403.CrossRef Russ DW, et al. Ageing, but not yet senescent, rats exhibit reduced muscle quality and sarcoplasmic reticulum function. Acta Physiol. 2011;201:391–403.CrossRef
14.
15.
go back to reference Tomas MT, et al. Body composition, muscle strength, functional capacity, and physical disability risk in liver transplanted familial amyloidotic polyneuropathy patients. Clin Transpl. 2011;25:E406–14.CrossRef Tomas MT, et al. Body composition, muscle strength, functional capacity, and physical disability risk in liver transplanted familial amyloidotic polyneuropathy patients. Clin Transpl. 2011;25:E406–14.CrossRef
16.
go back to reference Scott D, et al. Statin therapy, muscle function and falls risk in community-dwelling older adults. QJM. 2009;102:625–33.PubMedCrossRef Scott D, et al. Statin therapy, muscle function and falls risk in community-dwelling older adults. QJM. 2009;102:625–33.PubMedCrossRef
17.
go back to reference Delmonico MJ, et al. Association of the ACTN3 genotype and physical functioning with age in older adults. J Gerontol A Biol Sci Med Sci. 2008;63:1227–34.PubMedCrossRef Delmonico MJ, et al. Association of the ACTN3 genotype and physical functioning with age in older adults. J Gerontol A Biol Sci Med Sci. 2008;63:1227–34.PubMedCrossRef
18.
go back to reference Reid KF, et al. Lower extremity power training in elderly subjects with mobility limitations: a randomized controlled trial. Aging Clin Exp Res. 2008;20:337–43.PubMed Reid KF, et al. Lower extremity power training in elderly subjects with mobility limitations: a randomized controlled trial. Aging Clin Exp Res. 2008;20:337–43.PubMed
19.
go back to reference Park SW, et al. Accelerated loss of skeletal muscle strength in older adults with type 2 diabetes: the health, aging, and body composition study. Diabetes Care. 2007;30:1507–12.PubMedCrossRef Park SW, et al. Accelerated loss of skeletal muscle strength in older adults with type 2 diabetes: the health, aging, and body composition study. Diabetes Care. 2007;30:1507–12.PubMedCrossRef
20.
go back to reference Brooks N, et al. Strength training improves muscle quality and insulin sensitivity in Hispanic older adults with type 2 diabetes. Int J Med Sci. 2007;4:19–27.CrossRef Brooks N, et al. Strength training improves muscle quality and insulin sensitivity in Hispanic older adults with type 2 diabetes. Int J Med Sci. 2007;4:19–27.CrossRef
21.
go back to reference Goodpaster BH, et al. The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 2006;61:1059–64.PubMedCrossRef Goodpaster BH, et al. The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. J Gerontol A Biol Sci Med Sci. 2006;61:1059–64.PubMedCrossRef
22.
go back to reference Kostek MC, et al. Muscle strength response to strength training is influenced by insulin-like growth factor 1 genotype in older adults. J Appl Physiol. 2005;98:2147–54.PubMedCrossRef Kostek MC, et al. Muscle strength response to strength training is influenced by insulin-like growth factor 1 genotype in older adults. J Appl Physiol. 2005;98:2147–54.PubMedCrossRef
23.
go back to reference Katsiaras A, et al. Skeletal muscle fatigue, strength, and quality in the elderly: the Health ABC Study. J Appl Physiol. 2005;99:210–6.PubMedCrossRef Katsiaras A, et al. Skeletal muscle fatigue, strength, and quality in the elderly: the Health ABC Study. J Appl Physiol. 2005;99:210–6.PubMedCrossRef
24.
go back to reference Newman AB, et al. Strength and muscle quality in a well-functioning cohort of older adults: the Health, Aging and Body Composition Study. J Am Geriatr Soc. 2003;51:323–30.PubMedCrossRef Newman AB, et al. Strength and muscle quality in a well-functioning cohort of older adults: the Health, Aging and Body Composition Study. J Am Geriatr Soc. 2003;51:323–30.PubMedCrossRef
25.
go back to reference Bigland-Ritchie B. Muscle fatigue and the influence of changing neural drive. Clin Chest Med. 1984;5:21–34.PubMed Bigland-Ritchie B. Muscle fatigue and the influence of changing neural drive. Clin Chest Med. 1984;5:21–34.PubMed
26.
go back to reference Klass M, Baudry S, Duchateau J. Voluntary activation during maximal contraction with advancing age: a brief review. Eur J Appl Physiol. 2007;100:543–51.PubMedCrossRef Klass M, Baudry S, Duchateau J. Voluntary activation during maximal contraction with advancing age: a brief review. Eur J Appl Physiol. 2007;100:543–51.PubMedCrossRef
28.
go back to reference Rekling JC, et al. Synaptic control of motoneuronal excitability. Physiol Rev. 2000;80:767–852.PubMed Rekling JC, et al. Synaptic control of motoneuronal excitability. Physiol Rev. 2000;80:767–852.PubMed
29.
go back to reference Behm D, Power K, Drinkwater E. Comparison of interpolation and central activation ratios as measures of muscle inactivation. Muscle Nerve. 2001;24:925–34.PubMedCrossRef Behm D, Power K, Drinkwater E. Comparison of interpolation and central activation ratios as measures of muscle inactivation. Muscle Nerve. 2001;24:925–34.PubMedCrossRef
30.
go back to reference Gandevia SC. Spinal and supraspinal factors in human muscle fatigue. Physiol Rev. 2001;81:1725–89.PubMed Gandevia SC. Spinal and supraspinal factors in human muscle fatigue. Physiol Rev. 2001;81:1725–89.PubMed
31.
go back to reference Kent-Braun JA. Noninvasive measures of central and peripheral activation in human muscle fatigue. Muscle Nerve Suppl. 1997;5:S98–S101.PubMedCrossRef Kent-Braun JA. Noninvasive measures of central and peripheral activation in human muscle fatigue. Muscle Nerve Suppl. 1997;5:S98–S101.PubMedCrossRef
32.
go back to reference Kent-Braun JA, Le Blanc R. Quantitation of central activation failure during maximal voluntary contractions in humans. Muscle Nerve. 1996;19:861–9.PubMedCrossRef Kent-Braun JA, Le Blanc R. Quantitation of central activation failure during maximal voluntary contractions in humans. Muscle Nerve. 1996;19:861–9.PubMedCrossRef
33.
go back to reference Merton PA. Voluntary strength and fatigue. J Physiol. 1954;123:553–64.PubMed Merton PA. Voluntary strength and fatigue. J Physiol. 1954;123:553–64.PubMed
34.
go back to reference Bilodeau M, et al. Fatigue of elbow flexor muscles in younger and older adults. Muscle Nerve. 2001;24:98–106.PubMedCrossRef Bilodeau M, et al. Fatigue of elbow flexor muscles in younger and older adults. Muscle Nerve. 2001;24:98–106.PubMedCrossRef
35.
go back to reference Callahan DM, Foulis SA, Kent-Braun JA. Age-related fatigue resistance in the knee extensor muscles is specific to contraction mode. Muscle Nerve. 2009;39:692–702.PubMedCrossRef Callahan DM, Foulis SA, Kent-Braun JA. Age-related fatigue resistance in the knee extensor muscles is specific to contraction mode. Muscle Nerve. 2009;39:692–702.PubMedCrossRef
36.
go back to reference Cannon J, et al. Comparative effects of resistance training on peak isometric torque, muscle hypertrophy, voluntary activation and surface EMG between young and elderly women. Clin Physiol Funct Imaging. 2007;27:91–100.PubMedCrossRef Cannon J, et al. Comparative effects of resistance training on peak isometric torque, muscle hypertrophy, voluntary activation and surface EMG between young and elderly women. Clin Physiol Funct Imaging. 2007;27:91–100.PubMedCrossRef
37.
go back to reference Chung LH, Callahan DM, Kent-Braun JA. Age-related resistance to skeletal muscle fatigue is preserved during ischemia. J Appl Physiol. 2007;103:1628–35.PubMedCrossRef Chung LH, Callahan DM, Kent-Braun JA. Age-related resistance to skeletal muscle fatigue is preserved during ischemia. J Appl Physiol. 2007;103:1628–35.PubMedCrossRef
38.
go back to reference Connelly DM, et al. Motor unit firing rates and contractile properties in tibialis anterior of young and old men. J Appl Physiol. 1999;87:843–52.PubMed Connelly DM, et al. Motor unit firing rates and contractile properties in tibialis anterior of young and old men. J Appl Physiol. 1999;87:843–52.PubMed
39.
go back to reference De Serres SJ, Enoka RM. Older adults can maximally activate the biceps brachii muscle by voluntary command. J Appl Physiol. 1998;84:284–91.PubMedCrossRef De Serres SJ, Enoka RM. Older adults can maximally activate the biceps brachii muscle by voluntary command. J Appl Physiol. 1998;84:284–91.PubMedCrossRef
40.
go back to reference Harridge SD, Kryger A, Stensgaard A. Knee extensor strength, activation, and size in very elderly people following strength training. Muscle Nerve. 1999;22:831–9.PubMedCrossRef Harridge SD, Kryger A, Stensgaard A. Knee extensor strength, activation, and size in very elderly people following strength training. Muscle Nerve. 1999;22:831–9.PubMedCrossRef
41.
go back to reference Hunter SK, et al. Recovery from supraspinal fatigue is slowed in old adults after fatiguing maximal isometric contractions. J Appl Physiol. 2008;105:1199–209.PubMedCrossRef Hunter SK, et al. Recovery from supraspinal fatigue is slowed in old adults after fatiguing maximal isometric contractions. J Appl Physiol. 2008;105:1199–209.PubMedCrossRef
42.
go back to reference Jakobi JM, Rice CL. Voluntary muscle activation varies with age and muscle group. J Appl Physiol. 2002;93:457–62.PubMed Jakobi JM, Rice CL. Voluntary muscle activation varies with age and muscle group. J Appl Physiol. 2002;93:457–62.PubMed
43.
go back to reference Kent-Braun JA, Ng AV. Specific strength and voluntary muscle activation in young and elderly women and men. J Appl Physiol. 1999;87:22–9.PubMed Kent-Braun JA, Ng AV. Specific strength and voluntary muscle activation in young and elderly women and men. J Appl Physiol. 1999;87:22–9.PubMed
44.
go back to reference Klass M, Baudry S, Duchateau J. Aging does not affect voluntary activation of the ankle dorsiflexors during isometric, concentric, and eccentric contractions. J Appl Physiol. 2005;99:31–8.PubMedCrossRef Klass M, Baudry S, Duchateau J. Aging does not affect voluntary activation of the ankle dorsiflexors during isometric, concentric, and eccentric contractions. J Appl Physiol. 2005;99:31–8.PubMedCrossRef
45.
go back to reference Klein CS, Rice CL, Marsh GD. Normalized force, activation, and coactivation in the arm muscles of young and old men. J Appl Physiol. 2001;91:1341–9.PubMed Klein CS, Rice CL, Marsh GD. Normalized force, activation, and coactivation in the arm muscles of young and old men. J Appl Physiol. 2001;91:1341–9.PubMed
46.
go back to reference Knight CA, Kamen G. Adaptations in muscular activation of the knee extensor muscles with strength training in young and older adults. J Electromyogr Kinesiol. 2001;11:405–12.PubMedCrossRef Knight CA, Kamen G. Adaptations in muscular activation of the knee extensor muscles with strength training in young and older adults. J Electromyogr Kinesiol. 2001;11:405–12.PubMedCrossRef
47.
go back to reference Lanza IR, Russ DW, Kent-Braun JA. Age-related enhancement of fatigue resistance is evident in men during both isometric and dynamic tasks. J Appl Physiol. 2004;97:967–75.PubMedCrossRef Lanza IR, Russ DW, Kent-Braun JA. Age-related enhancement of fatigue resistance is evident in men during both isometric and dynamic tasks. J Appl Physiol. 2004;97:967–75.PubMedCrossRef
48.
go back to reference Roos MR, et al. Quadriceps muscle strength, contractile properties, and motor unit firing rates in young and old men. Muscle Nerve. 1999;22:1094–103.PubMedCrossRef Roos MR, et al. Quadriceps muscle strength, contractile properties, and motor unit firing rates in young and old men. Muscle Nerve. 1999;22:1094–103.PubMedCrossRef
49.
go back to reference Simoneau E, Martin A, Van Hoecke J. Muscular performances at the ankle joint in young and elderly men. J Gerontol A Biol Sci Med Sci. 2005;60:439–47.PubMedCrossRef Simoneau E, Martin A, Van Hoecke J. Muscular performances at the ankle joint in young and elderly men. J Gerontol A Biol Sci Med Sci. 2005;60:439–47.PubMedCrossRef
50.
go back to reference Stevens JE, et al. Are voluntary muscle activation deficits in older adults meaningful? Muscle Nerve. 2003;27:99–101.PubMedCrossRef Stevens JE, et al. Are voluntary muscle activation deficits in older adults meaningful? Muscle Nerve. 2003;27:99–101.PubMedCrossRef
51.
go back to reference Wilder MR, Cannon J. Effect of age on muscle activation and twitch properties during static and dynamic actions. Muscle Nerve. 2009;39:683–91.PubMedCrossRef Wilder MR, Cannon J. Effect of age on muscle activation and twitch properties during static and dynamic actions. Muscle Nerve. 2009;39:683–91.PubMedCrossRef
52.
go back to reference Yue GH, et al. Older adults exhibit a reduced ability to fully activate their biceps brachii muscle. J Gerontol A Biol Sci Med Sci. 1999;54:M249–53.PubMedCrossRef Yue GH, et al. Older adults exhibit a reduced ability to fully activate their biceps brachii muscle. J Gerontol A Biol Sci Med Sci. 1999;54:M249–53.PubMedCrossRef
53.
go back to reference Yoon T, De-Lap BS, Griffith EE, Hunter SK. Age-related muscle fatigue after a low-force fatiguing contraction is explained by central fatigue. Muscle Nerve. 2007;37:457–66.CrossRef Yoon T, De-Lap BS, Griffith EE, Hunter SK. Age-related muscle fatigue after a low-force fatiguing contraction is explained by central fatigue. Muscle Nerve. 2007;37:457–66.CrossRef
54.
go back to reference Haug H, Eggers R. Morphometry of the human cortex cerebri and corpus striatum during aging. Neurobiol Aging. 1991;12:336–8.PubMedCrossRef Haug H, Eggers R. Morphometry of the human cortex cerebri and corpus striatum during aging. Neurobiol Aging. 1991;12:336–8.PubMedCrossRef
55.
56.
57.
go back to reference Marner L, et al. Marked loss of myelinated nerve fibers in the human brain with age. J Comp Neurol. 2003;462:144–52.PubMedCrossRef Marner L, et al. Marked loss of myelinated nerve fibers in the human brain with age. J Comp Neurol. 2003;462:144–52.PubMedCrossRef
58.
go back to reference Madden DJ, et al. Diffusion tensor imaging of adult age differences in cerebral white matter: relation to response time. Neuroimage. 2004;21:1174–81.PubMedCrossRef Madden DJ, et al. Diffusion tensor imaging of adult age differences in cerebral white matter: relation to response time. Neuroimage. 2004;21:1174–81.PubMedCrossRef
59.
go back to reference Carlsson A. Treatment of Parkinson’s with L-DOPA. The early discovery phase, and a comment on current problems. J Neural Transm. 2002;109:777–87.PubMedCrossRef Carlsson A. Treatment of Parkinson’s with L-DOPA. The early discovery phase, and a comment on current problems. J Neural Transm. 2002;109:777–87.PubMedCrossRef
60.
go back to reference Morgan DG, May PC, Finch CE. Dopamine and serotonin systems in human and rodent brain: effects of age and neurodegenerative disease. J Am Geriatr Soc. 1987;35:334–45.PubMed Morgan DG, May PC, Finch CE. Dopamine and serotonin systems in human and rodent brain: effects of age and neurodegenerative disease. J Am Geriatr Soc. 1987;35:334–45.PubMed
61.
go back to reference Bigham MH, Lidow MS. Adrenergic and serotonergic receptors in aged monkey neocortex. Neurobiol Aging. 1995;16:91–104.PubMedCrossRef Bigham MH, Lidow MS. Adrenergic and serotonergic receptors in aged monkey neocortex. Neurobiol Aging. 1995;16:91–104.PubMedCrossRef
62.
go back to reference Bartus RT, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408–14.PubMedCrossRef Bartus RT, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408–14.PubMedCrossRef
63.
go back to reference Segovia G, et al. Glutamatergic neurotransmission in aging: a critical perspective. Mech Ageing Dev. 2001;122:1–29.PubMedCrossRef Segovia G, et al. Glutamatergic neurotransmission in aging: a critical perspective. Mech Ageing Dev. 2001;122:1–29.PubMedCrossRef
64.
65.
go back to reference Mora F, Segovia G, Del Arco A. Glutamate–dopamine–GABA interactions in the aging basal ganglia. Brain Res Rev. 2008;58:340–53.PubMedCrossRef Mora F, Segovia G, Del Arco A. Glutamate–dopamine–GABA interactions in the aging basal ganglia. Brain Res Rev. 2008;58:340–53.PubMedCrossRef
66.
go back to reference Roth GS, Joseph JA. Cellular and molecular mechanisms of impaired dopaminergic function during aging. Ann N Y Acad Sci. 1994;719:129–35.PubMedCrossRef Roth GS, Joseph JA. Cellular and molecular mechanisms of impaired dopaminergic function during aging. Ann N Y Acad Sci. 1994;719:129–35.PubMedCrossRef
67.
go back to reference Hayashi M, Yamashita A, Shimizu K. Somatostatin and brain-derived neurotrophic factor mRNA expression in the primate brain: decreased levels of mRNAs during aging. Brain Res. 1997;749:283–9.PubMedCrossRef Hayashi M, Yamashita A, Shimizu K. Somatostatin and brain-derived neurotrophic factor mRNA expression in the primate brain: decreased levels of mRNAs during aging. Brain Res. 1997;749:283–9.PubMedCrossRef
68.
go back to reference Volkow ND, et al. Dopamine transporters decrease with age. J Nucl Med. 1996;37:554–9.PubMed Volkow ND, et al. Dopamine transporters decrease with age. J Nucl Med. 1996;37:554–9.PubMed
69.
go back to reference Joseph JA, et al. Age-related changes in the nigrostriatum: a behavioral and biochemical analysis. J Gerontol. 1978;33:643–9.PubMedCrossRef Joseph JA, et al. Age-related changes in the nigrostriatum: a behavioral and biochemical analysis. J Gerontol. 1978;33:643–9.PubMedCrossRef
70.
go back to reference Kossev AR, et al. Increased intracortical inhibition in middle-aged humans; a study using paired-pulse transcranial magnetic stimulation. Neurosci Lett. 2002;333:83–6.PubMedCrossRef Kossev AR, et al. Increased intracortical inhibition in middle-aged humans; a study using paired-pulse transcranial magnetic stimulation. Neurosci Lett. 2002;333:83–6.PubMedCrossRef
71.
go back to reference Sale MV, Semmler JG. Age-related differences in corticospinal control during functional isometric contractions in left and right hands. J Appl Physiol. 2005;99:1483–93.PubMedCrossRef Sale MV, Semmler JG. Age-related differences in corticospinal control during functional isometric contractions in left and right hands. J Appl Physiol. 2005;99:1483–93.PubMedCrossRef
72.
go back to reference Smith AE, et al. Age-related changes in short-latency motor cortex inhibition. Exp Brain Res. 2009;198:489–500.PubMedCrossRef Smith AE, et al. Age-related changes in short-latency motor cortex inhibition. Exp Brain Res. 2009;198:489–500.PubMedCrossRef
73.
go back to reference McGinley M, et al. Older adults exhibit more intracortical inhibition and less intracortical facilitation than young adults. J Am Osteopathic Assoc. 2010;45:671–8. McGinley M, et al. Older adults exhibit more intracortical inhibition and less intracortical facilitation than young adults. J Am Osteopathic Assoc. 2010;45:671–8.
74.
go back to reference Heuninckx S, et al. Neural basis of aging: the penetration of cognition into action control. J Neurosci. 2005;25:6787–96.PubMedCrossRef Heuninckx S, et al. Neural basis of aging: the penetration of cognition into action control. J Neurosci. 2005;25:6787–96.PubMedCrossRef
75.
go back to reference Naccarato M, et al. Does healthy aging affect the hemispheric activation balance during paced index-to-thumb opposition task? An fMRI study. Neuroimage. 2006;32:1250–6.PubMedCrossRef Naccarato M, et al. Does healthy aging affect the hemispheric activation balance during paced index-to-thumb opposition task? An fMRI study. Neuroimage. 2006;32:1250–6.PubMedCrossRef
76.
go back to reference Rowe JB, et al. Aging is associated with contrasting changes in local and distant cortical connectivity in the human motor system. Neuroimage. 2006;32:747–60.PubMedCrossRef Rowe JB, et al. Aging is associated with contrasting changes in local and distant cortical connectivity in the human motor system. Neuroimage. 2006;32:747–60.PubMedCrossRef
77.
go back to reference Fathi D, et al. Effects of aging on the human motor cortical plasticity studied by paired associative stimulation. Clin Neurophysiol. 2010;121:90–3.PubMedCrossRef Fathi D, et al. Effects of aging on the human motor cortical plasticity studied by paired associative stimulation. Clin Neurophysiol. 2010;121:90–3.PubMedCrossRef
78.
go back to reference Sawaki L, et al. Age-dependent changes in the ability to encode a novel elementary motor memory. Ann Neurol. 2003;53:521–4.PubMedCrossRef Sawaki L, et al. Age-dependent changes in the ability to encode a novel elementary motor memory. Ann Neurol. 2003;53:521–4.PubMedCrossRef
79.
go back to reference Kobayashi M, Pascual-Leone A. Transcranial magnetic stimulation in neurology. Lancet Neurol. 2003;2:145–56.PubMedCrossRef Kobayashi M, Pascual-Leone A. Transcranial magnetic stimulation in neurology. Lancet Neurol. 2003;2:145–56.PubMedCrossRef
80.
go back to reference McGinley MP, Clark BC. Transcranial magnetic stimulation and the human neuromuscular system. In: Costa A, Villalba E, editors. Horizons in neuroscience research, vol. 4. Hauppauge: Nova Science; 2011. p. 69–88. McGinley MP, Clark BC. Transcranial magnetic stimulation and the human neuromuscular system. In: Costa A, Villalba E, editors. Horizons in neuroscience research, vol. 4. Hauppauge: Nova Science; 2011. p. 69–88.
81.
go back to reference Florian J, et al. Inhibitory circuits and the nature of their interactions in the human motor cortex—a pharmacological TMS study. J Physiol. 2008;586:495–514.PubMedCrossRef Florian J, et al. Inhibitory circuits and the nature of their interactions in the human motor cortex—a pharmacological TMS study. J Physiol. 2008;586:495–514.PubMedCrossRef
83.
go back to reference McDonnell MN, Orekhov Y, Ziemann U. The role of GABA(B) receptors in intracortical inhibition in the human motor cortex. Exp Brain Res. 2006;173:86–93.PubMedCrossRef McDonnell MN, Orekhov Y, Ziemann U. The role of GABA(B) receptors in intracortical inhibition in the human motor cortex. Exp Brain Res. 2006;173:86–93.PubMedCrossRef
84.
go back to reference Reis J, et al. Contribution of transcranial magnetic stimulation to the understanding of cortical mechanisms involved in motor control. J Physiol. 2008;586:325–51.PubMedCrossRef Reis J, et al. Contribution of transcranial magnetic stimulation to the understanding of cortical mechanisms involved in motor control. J Physiol. 2008;586:325–51.PubMedCrossRef
86.
go back to reference Powers RK, Binder MD. Input–output functions of mammalian motoneurons. Rev Physiol Biochem Pharmacol. 2001;143:137–263.PubMedCrossRef Powers RK, Binder MD. Input–output functions of mammalian motoneurons. Rev Physiol Biochem Pharmacol. 2001;143:137–263.PubMedCrossRef
87.
go back to reference Henneman E, Somjen G, Carpenter DO. Excitability and inhibitability of motoneurons of different sizes. J Neurophysiol. 1965;28:599–620.PubMed Henneman E, Somjen G, Carpenter DO. Excitability and inhibitability of motoneurons of different sizes. J Neurophysiol. 1965;28:599–620.PubMed
88.
go back to reference Powers RK, Turker KS, Binder MD. What can be learned about motoneurone properties from studying firing patterns? Adv Exp Med Biol. 2002;508:199–205.PubMedCrossRef Powers RK, Turker KS, Binder MD. What can be learned about motoneurone properties from studying firing patterns? Adv Exp Med Biol. 2002;508:199–205.PubMedCrossRef
89.
go back to reference Heckman CJ. Active conductances in motoneuron dendrites enhance movement capabilities. Exerc Sport Sci Rev. 2003;31:96–101.PubMedCrossRef Heckman CJ. Active conductances in motoneuron dendrites enhance movement capabilities. Exerc Sport Sci Rev. 2003;31:96–101.PubMedCrossRef
90.
go back to reference Heckman CJ, Lee RH, Brownstone RM. Hyperexcitable dendrites in motoneurons and their neuromodulatory control during motor behavior. Trends Neurosci. 2003;26:688–95.PubMedCrossRef Heckman CJ, Lee RH, Brownstone RM. Hyperexcitable dendrites in motoneurons and their neuromodulatory control during motor behavior. Trends Neurosci. 2003;26:688–95.PubMedCrossRef
91.
go back to reference Tomlinson BE, Irving D. The numbers of limb motor neurons in the human lumbosacral cord throughout life. J Neurol Sci. 1977;34:213–9.PubMedCrossRef Tomlinson BE, Irving D. The numbers of limb motor neurons in the human lumbosacral cord throughout life. J Neurol Sci. 1977;34:213–9.PubMedCrossRef
92.
go back to reference Siu PM, Alway SE. Response and adaptation of skeletal muscle to denervation stress: the role of apoptosis in muscle loss. Frontiers Biosci: J Virtual Libr. 2009;14:432–52.CrossRef Siu PM, Alway SE. Response and adaptation of skeletal muscle to denervation stress: the role of apoptosis in muscle loss. Frontiers Biosci: J Virtual Libr. 2009;14:432–52.CrossRef
93.
94.
go back to reference Deschenes MR, et al. Remodeling of the neuromuscular junction precedes sarcopenia related alterations in myofibers. Exp Gerontol. 2010;45:389–93.PubMedCrossRef Deschenes MR, et al. Remodeling of the neuromuscular junction precedes sarcopenia related alterations in myofibers. Exp Gerontol. 2010;45:389–93.PubMedCrossRef
95.
go back to reference Andonian MH, Fahim MA. Effects of endurance exercise on the morphology of mouse neuromuscular junctions during ageing. J Neurocytol. 1987;16:589–99.PubMedCrossRef Andonian MH, Fahim MA. Effects of endurance exercise on the morphology of mouse neuromuscular junctions during ageing. J Neurocytol. 1987;16:589–99.PubMedCrossRef
96.
go back to reference Courtney J, Steinbach JH. Age changes in neuromuscular junction morphology and acetylcholine receptor distribution on rat skeletal muscle fibres. J Physiol. 1981;320:435–47.PubMed Courtney J, Steinbach JH. Age changes in neuromuscular junction morphology and acetylcholine receptor distribution on rat skeletal muscle fibres. J Physiol. 1981;320:435–47.PubMed
97.
go back to reference Oda K. Age changes of motor innervation and acetylcholine receptor distribution on human skeletal muscle fibres. J Neurol Sci. 1984;66:327–38.PubMedCrossRef Oda K. Age changes of motor innervation and acetylcholine receptor distribution on human skeletal muscle fibres. J Neurol Sci. 1984;66:327–38.PubMedCrossRef
98.
go back to reference Robbins N, Fahim MA. Progression of age changes in mature mouse motor nerve terminals and its relation to locomotor activity. J Neurocytol. 1985;14:1019–36.PubMedCrossRef Robbins N, Fahim MA. Progression of age changes in mature mouse motor nerve terminals and its relation to locomotor activity. J Neurocytol. 1985;14:1019–36.PubMedCrossRef
99.
go back to reference Deschenes MR, Wilson MH. Age-related differences in synaptic plasticity following muscle unloading. J Neurobiol. 2003;57:246–56.PubMedCrossRef Deschenes MR, Wilson MH. Age-related differences in synaptic plasticity following muscle unloading. J Neurobiol. 2003;57:246–56.PubMedCrossRef
100.
go back to reference Fahim MA, Robbins N. Ultrastructural studies of young and old mouse neuromuscular junctions. J Neurocytol. 1982;11:641–56.PubMedCrossRef Fahim MA, Robbins N. Ultrastructural studies of young and old mouse neuromuscular junctions. J Neurocytol. 1982;11:641–56.PubMedCrossRef
101.
go back to reference Deschenes MR. Motor unit and neuromuscular junction remodeling with aging. Curr Aging Sci. 2011;4:209–20.PubMedCrossRef Deschenes MR. Motor unit and neuromuscular junction remodeling with aging. Curr Aging Sci. 2011;4:209–20.PubMedCrossRef
102.
go back to reference Morita H, et al. Progressive decrease in heteronymous monosynaptic Ia facilitation with human ageing. Exp Brain Res Experimentelle Hirnforschung Experiment Cerebrale. 1995;104:167–70. Morita H, et al. Progressive decrease in heteronymous monosynaptic Ia facilitation with human ageing. Exp Brain Res Experimentelle Hirnforschung Experiment Cerebrale. 1995;104:167–70.
103.
go back to reference Kamen G, et al. Motor unit discharge behavior in older adults during maximal-effort contractions. J Appl Physiol. 1995;79:1908–13.PubMed Kamen G, et al. Motor unit discharge behavior in older adults during maximal-effort contractions. J Appl Physiol. 1995;79:1908–13.PubMed
104.
go back to reference Tracy BL, et al. Variability of motor unit discharge and force fluctuations across a range of muscle forces in older adults. Muscle Nerve. 2005;32:533–40.PubMedCrossRef Tracy BL, et al. Variability of motor unit discharge and force fluctuations across a range of muscle forces in older adults. Muscle Nerve. 2005;32:533–40.PubMedCrossRef
105.
go back to reference Christie A, Kamen G. Doublet discharges in motoneurons of young and older adults. J Neurophysiol. 2006;95:2787–95.PubMedCrossRef Christie A, Kamen G. Doublet discharges in motoneurons of young and older adults. J Neurophysiol. 2006;95:2787–95.PubMedCrossRef
106.
go back to reference Lexell J, Taylor CC, Sjostrom M. What is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15- to 83-year-old men. J Neurol Sci. 1988;84:275–94.PubMedCrossRef Lexell J, Taylor CC, Sjostrom M. What is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15- to 83-year-old men. J Neurol Sci. 1988;84:275–94.PubMedCrossRef
107.
go back to reference Lee WS, et al. Age-associated decrease of type IIA/B human skeletal muscle fibers. Clin Orthop Relat Res. 2006;450:231–7.PubMedCrossRef Lee WS, et al. Age-associated decrease of type IIA/B human skeletal muscle fibers. Clin Orthop Relat Res. 2006;450:231–7.PubMedCrossRef
108.
go back to reference Jakobsson F, Borg K, Edstrom L. Fibre-type composition, structure and cytoskeletal protein location of fibres in anterior tibial muscle. Comparison between young adults and physically active aged humans. Acta Neuropathol. 1990;80:459–68.PubMedCrossRef Jakobsson F, Borg K, Edstrom L. Fibre-type composition, structure and cytoskeletal protein location of fibres in anterior tibial muscle. Comparison between young adults and physically active aged humans. Acta Neuropathol. 1990;80:459–68.PubMedCrossRef
109.
go back to reference Lenk K, Schuler G, Adams V. Skeletal muscle wasting in cachexia and sarcopenia: molecular pathophysiology and impact of exercise training. J Cachex Sarcopenia Muscle. 2010;1:9–21.CrossRef Lenk K, Schuler G, Adams V. Skeletal muscle wasting in cachexia and sarcopenia: molecular pathophysiology and impact of exercise training. J Cachex Sarcopenia Muscle. 2010;1:9–21.CrossRef
110.
go back to reference Fry CS, Rasmussen BB. Skeletal muscle protein balance and metabolism in the elderly. Curr Aging Sci. 2011;4:260–8.PubMedCrossRef Fry CS, Rasmussen BB. Skeletal muscle protein balance and metabolism in the elderly. Curr Aging Sci. 2011;4:260–8.PubMedCrossRef
111.
go back to reference Cuthbertson D, et al. Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. FASEB J. 2005;19:422–4.PubMed Cuthbertson D, et al. Anabolic signaling deficits underlie amino acid resistance of wasting, aging muscle. FASEB J. 2005;19:422–4.PubMed
112.
go back to reference Katsanos CS, et al. Aging is associated with diminished accretion of muscle proteins after the ingestion of a small bolus of essential amino acids. Am J Clin Nutr. 2005;82:1065–73.PubMed Katsanos CS, et al. Aging is associated with diminished accretion of muscle proteins after the ingestion of a small bolus of essential amino acids. Am J Clin Nutr. 2005;82:1065–73.PubMed
113.
go back to reference Katsanos CS, et al. A high proportion of leucine is required for optimal stimulation of the rate of muscle protein synthesis by essential amino acids in the elderly. Am J Physiol Endocrinol Metab. 2006;291:E381–7.PubMedCrossRef Katsanos CS, et al. A high proportion of leucine is required for optimal stimulation of the rate of muscle protein synthesis by essential amino acids in the elderly. Am J Physiol Endocrinol Metab. 2006;291:E381–7.PubMedCrossRef
114.
go back to reference Volpi E, et al. Oral amino acids stimulate muscle protein anabolism in the elderly despite higher first-pass splanchnic extraction. Am J Physiol. 1999;277:E513–20.PubMed Volpi E, et al. Oral amino acids stimulate muscle protein anabolism in the elderly despite higher first-pass splanchnic extraction. Am J Physiol. 1999;277:E513–20.PubMed
115.
go back to reference Volpi E, et al. The response of muscle protein anabolism to combined hyperaminoacidemia and glucose-induced hyperinsulinemia is impaired in the elderly. J Clin Endocrinol Metabol. 2000;85:4481–90.CrossRef Volpi E, et al. The response of muscle protein anabolism to combined hyperaminoacidemia and glucose-induced hyperinsulinemia is impaired in the elderly. J Clin Endocrinol Metabol. 2000;85:4481–90.CrossRef
116.
go back to reference Volpi E, et al. Basal muscle amino acid kinetics and protein synthesis in healthy young and older men. JAMA. 2001;286:1206–12.PubMedCrossRef Volpi E, et al. Basal muscle amino acid kinetics and protein synthesis in healthy young and older men. JAMA. 2001;286:1206–12.PubMedCrossRef
117.
go back to reference Rasmussen BB, Wolfe RR, Volpi E. Oral and intravenously administered amino acids produce similar effects on muscle protein synthesis in the elderly. J Nutr Health Aging. 2002;6:358–62.PubMed Rasmussen BB, Wolfe RR, Volpi E. Oral and intravenously administered amino acids produce similar effects on muscle protein synthesis in the elderly. J Nutr Health Aging. 2002;6:358–62.PubMed
118.
go back to reference Paddon-Jones D, et al. Amino acid ingestion improves muscle protein synthesis in the young and elderly. Am J Physiol Endocrinol Metab. 2004;286:E321–8.PubMedCrossRef Paddon-Jones D, et al. Amino acid ingestion improves muscle protein synthesis in the young and elderly. Am J Physiol Endocrinol Metab. 2004;286:E321–8.PubMedCrossRef
119.
go back to reference Paddon-Jones D, et al. Differential stimulation of muscle protein synthesis in elderly humans following isocaloric ingestion of amino acids or whey protein. Exp Gerontol. 2006;41:215–9.PubMedCrossRef Paddon-Jones D, et al. Differential stimulation of muscle protein synthesis in elderly humans following isocaloric ingestion of amino acids or whey protein. Exp Gerontol. 2006;41:215–9.PubMedCrossRef
120.
go back to reference Rennie MJ, et al. Muscle protein synthesis measured by stable isotope techniques in man: the effects of feeding and fasting. Clin Sci. 1982;63:519–23.PubMed Rennie MJ, et al. Muscle protein synthesis measured by stable isotope techniques in man: the effects of feeding and fasting. Clin Sci. 1982;63:519–23.PubMed
121.
go back to reference Volpi E, et al. Exogenous amino acids stimulate net muscle protein synthesis in the elderly. J Clin Invest. 1998;101:2000–7.PubMedCrossRef Volpi E, et al. Exogenous amino acids stimulate net muscle protein synthesis in the elderly. J Clin Invest. 1998;101:2000–7.PubMedCrossRef
122.
go back to reference Volpi E, et al. Essential amino acids are primarily responsible for the amino acid stimulation of muscle protein anabolism in healthy elderly adults. Am J Clin Nutr. 2003;78:250–8.PubMed Volpi E, et al. Essential amino acids are primarily responsible for the amino acid stimulation of muscle protein anabolism in healthy elderly adults. Am J Clin Nutr. 2003;78:250–8.PubMed
123.
go back to reference Sheffield-Moore M, et al. Mixed muscle and hepatic derived plasma protein metabolism is differentially regulated in older and younger men following resistance exercise. Am J Physiol Endocrinol Metab. 2005;288:E922–9.PubMedCrossRef Sheffield-Moore M, et al. Mixed muscle and hepatic derived plasma protein metabolism is differentially regulated in older and younger men following resistance exercise. Am J Physiol Endocrinol Metab. 2005;288:E922–9.PubMedCrossRef
124.
go back to reference Kumar V, et al. Age-related differences in the dose–response relationship of muscle protein synthesis to resistance exercise in young and old men. J Physiol. 2009;587:211–7.PubMedCrossRef Kumar V, et al. Age-related differences in the dose–response relationship of muscle protein synthesis to resistance exercise in young and old men. J Physiol. 2009;587:211–7.PubMedCrossRef
125.
go back to reference Mayhew DL, et al. Translational signaling responses preceding resistance training-mediated myofiber hypertrophy in young and old humans. J Appl Physiol. 2009;107:1655–62.PubMedCrossRef Mayhew DL, et al. Translational signaling responses preceding resistance training-mediated myofiber hypertrophy in young and old humans. J Appl Physiol. 2009;107:1655–62.PubMedCrossRef
126.
go back to reference Guillet C, et al. Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans. FASEB J. 2004;18:1586–7.PubMed Guillet C, et al. Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans. FASEB J. 2004;18:1586–7.PubMed
127.
go back to reference Drummond MJ, et al. Skeletal muscle protein anabolic response to resistance exercise and essential amino acids is delayed with aging. J Appl Physiol. 2008;104:1452–61.PubMedCrossRef Drummond MJ, et al. Skeletal muscle protein anabolic response to resistance exercise and essential amino acids is delayed with aging. J Appl Physiol. 2008;104:1452–61.PubMedCrossRef
128.
go back to reference Altun M, et al. Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. J Biol Chem. 2010;285:39597–608.PubMedCrossRef Altun M, et al. Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. J Biol Chem. 2010;285:39597–608.PubMedCrossRef
129.
go back to reference Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009;1793:664–73.PubMedCrossRef Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009;1793:664–73.PubMedCrossRef
130.
go back to reference Rajawat YS, Hilioti Z, Bossis I. Aging: central role for autophagy and the lysosomal degradative system. Ageing Res Rev. 2009;8:199–213.PubMedCrossRef Rajawat YS, Hilioti Z, Bossis I. Aging: central role for autophagy and the lysosomal degradative system. Ageing Res Rev. 2009;8:199–213.PubMedCrossRef
131.
go back to reference McMullen CA, et al. Age-related changes of cell death pathways in rat extraocular muscle. Exp Gerontol. 2009;44:420–5.PubMedCrossRef McMullen CA, et al. Age-related changes of cell death pathways in rat extraocular muscle. Exp Gerontol. 2009;44:420–5.PubMedCrossRef
132.
go back to reference Wohlgemuth SE, et al. Skeletal muscle autophagy and apoptosis during aging: effects of calorie restriction and life-long exercise. Exp Gerontol. 2010;45:138–48.PubMedCrossRef Wohlgemuth SE, et al. Skeletal muscle autophagy and apoptosis during aging: effects of calorie restriction and life-long exercise. Exp Gerontol. 2010;45:138–48.PubMedCrossRef
133.
go back to reference Lee CM, et al. Influences of caloric restriction on age-associated skeletal muscle fiber characteristics and mitochondrial changes in rats and mice. Ann N Y Acad Sci. 1998;854:182–91.PubMedCrossRef Lee CM, et al. Influences of caloric restriction on age-associated skeletal muscle fiber characteristics and mitochondrial changes in rats and mice. Ann N Y Acad Sci. 1998;854:182–91.PubMedCrossRef
134.
go back to reference Marzetti E, et al. Sarcopenia of aging: underlying cellular mechanisms and protection by calorie restriction. Biofactors. 2009;35:28–35.PubMedCrossRef Marzetti E, et al. Sarcopenia of aging: underlying cellular mechanisms and protection by calorie restriction. Biofactors. 2009;35:28–35.PubMedCrossRef
135.
go back to reference Goodpaster BH, et al. Skeletal muscle attenuation determined by computed tomography is associated with skeletal muscle lipid content. J Appl Physiol. 2000;89:104–10.PubMed Goodpaster BH, et al. Skeletal muscle attenuation determined by computed tomography is associated with skeletal muscle lipid content. J Appl Physiol. 2000;89:104–10.PubMed
136.
go back to reference Kurz MJ, Stergiou N. The aging human neuromuscular system expresses less certainty for selecting joint kinematics during gait. Neurosci Lett. 2003;348:155–8.PubMedCrossRef Kurz MJ, Stergiou N. The aging human neuromuscular system expresses less certainty for selecting joint kinematics during gait. Neurosci Lett. 2003;348:155–8.PubMedCrossRef
137.
go back to reference Christie A, Kamen G. Short-term training adaptations in maximal motor unit firing rates and afterhyperpolarization duration. Muscle Nerve. 2010;41:651–60.PubMed Christie A, Kamen G. Short-term training adaptations in maximal motor unit firing rates and afterhyperpolarization duration. Muscle Nerve. 2010;41:651–60.PubMed
138.
go back to reference Han YS, et al. ATP consumption rate per cross bridge depends on myosin heavy chain isoform. J Appl Physiol. 2003;94:2188–96.PubMedCrossRef Han YS, et al. ATP consumption rate per cross bridge depends on myosin heavy chain isoform. J Appl Physiol. 2003;94:2188–96.PubMedCrossRef
139.
go back to reference He ZH, et al. ATP consumption and efficiency of human single muscle fibers with different myosin isoform composition. Biophys J. 2000;79:945–61.PubMedCrossRef He ZH, et al. ATP consumption and efficiency of human single muscle fibers with different myosin isoform composition. Biophys J. 2000;79:945–61.PubMedCrossRef
140.
go back to reference Rivero JL, Talmadge RJ, Edgerton VR. Interrelationships of myofibrillar ATPase activity and metabolic properties of myosin heavy chain-based fibre types in rat skeletal muscle. Histochem Cell Biol. 1999;111:277–87.PubMedCrossRef Rivero JL, Talmadge RJ, Edgerton VR. Interrelationships of myofibrillar ATPase activity and metabolic properties of myosin heavy chain-based fibre types in rat skeletal muscle. Histochem Cell Biol. 1999;111:277–87.PubMedCrossRef
141.
go back to reference Stienen GJ, et al. Myofibrillar ATPase activity in skinned human skeletal muscle fibres: fibre type and temperature dependence. J Physiol. 1996;493:299–307.PubMed Stienen GJ, et al. Myofibrillar ATPase activity in skinned human skeletal muscle fibres: fibre type and temperature dependence. J Physiol. 1996;493:299–307.PubMed
142.
go back to reference Szentesi P, et al. ATP utilization for calcium uptake and force production in different types of human skeletal muscle fibres. J Physiol. 2001;531:393–403.PubMedCrossRef Szentesi P, et al. ATP utilization for calcium uptake and force production in different types of human skeletal muscle fibres. J Physiol. 2001;531:393–403.PubMedCrossRef
143.
go back to reference D’Antona G, et al. The effect of ageing and immobilization on structure and function of human skeletal muscle fibres. J Physiol. 2003;552:499–511.PubMedCrossRef D’Antona G, et al. The effect of ageing and immobilization on structure and function of human skeletal muscle fibres. J Physiol. 2003;552:499–511.PubMedCrossRef
144.
go back to reference Haddad F, Adams GR. Aging-sensitive cellular and molecular mechanisms associated with skeletal muscle hypertrophy. J Appl Physiol. 2006;100:1188–203.PubMedCrossRef Haddad F, Adams GR. Aging-sensitive cellular and molecular mechanisms associated with skeletal muscle hypertrophy. J Appl Physiol. 2006;100:1188–203.PubMedCrossRef
145.
go back to reference Piec I, et al. Differential proteome analysis of aging in rat skeletal muscle. FASEB J. 2005;19:1143–5.PubMed Piec I, et al. Differential proteome analysis of aging in rat skeletal muscle. FASEB J. 2005;19:1143–5.PubMed
146.
go back to reference Thompson LV, et al. Myosin and actin expression and oxidation in aging muscle. J Appl Physiol. 2006;101:1581–7.PubMedCrossRef Thompson LV, et al. Myosin and actin expression and oxidation in aging muscle. J Appl Physiol. 2006;101:1581–7.PubMedCrossRef
147.
go back to reference Clark BC. In vivo alterations in skeletal muscle form and function after disuse atrophy. Med Sci Sports Exerc. 2009;41:1869–75.PubMedCrossRef Clark BC. In vivo alterations in skeletal muscle form and function after disuse atrophy. Med Sci Sports Exerc. 2009;41:1869–75.PubMedCrossRef
148.
go back to reference Riley DA, et al. Decreased thin filament density and length in human atrophic soleus muscle fibers after spaceflight. J Appl Physiol. 2000;88:567–72.PubMed Riley DA, et al. Decreased thin filament density and length in human atrophic soleus muscle fibers after spaceflight. J Appl Physiol. 2000;88:567–72.PubMed
149.
go back to reference Klitgaard H, et al. Ageing alters the myosin heavy chain composition of single fibres from human skeletal muscle. Acta Physiol Scand. 1990;140:55–62.PubMedCrossRef Klitgaard H, et al. Ageing alters the myosin heavy chain composition of single fibres from human skeletal muscle. Acta Physiol Scand. 1990;140:55–62.PubMedCrossRef
150.
go back to reference Sullivan VK, et al. Myosin heavy chain composition in young and old rat skeletal muscle: effects of endurance exercise. J Appl Physiol. 1995;78:2115–20.PubMed Sullivan VK, et al. Myosin heavy chain composition in young and old rat skeletal muscle: effects of endurance exercise. J Appl Physiol. 1995;78:2115–20.PubMed
151.
go back to reference Carlsen RC, Walsh DA. Decrease in force potentiation and appearance of alpha-adrenergic mediated contracture in aging rat skeletal muscle. Pflugers Archiv: Eur J Physiol. 1987;408:224–30.CrossRef Carlsen RC, Walsh DA. Decrease in force potentiation and appearance of alpha-adrenergic mediated contracture in aging rat skeletal muscle. Pflugers Archiv: Eur J Physiol. 1987;408:224–30.CrossRef
152.
go back to reference Petrella RJ, et al. Comparison of twitch potentiation in the gastrocnemius of young and elderly men. Eur J Appl Physiol Occup Physiol. 1989;58:395–9.PubMedCrossRef Petrella RJ, et al. Comparison of twitch potentiation in the gastrocnemius of young and elderly men. Eur J Appl Physiol Occup Physiol. 1989;58:395–9.PubMedCrossRef
153.
go back to reference Russ DW, et al. Contrasting influences of age and sex on muscle fatigue. Med Sci Sports Exerc. 2008;40:234–41.PubMedCrossRef Russ DW, et al. Contrasting influences of age and sex on muscle fatigue. Med Sci Sports Exerc. 2008;40:234–41.PubMedCrossRef
154.
go back to reference Larsson L. Morphological and functional characteristics of the ageing skeletal muscle in man. A cross-sectional study. Acta Physiol Scand Suppl. 1978;457:1–36.PubMedCrossRef Larsson L. Morphological and functional characteristics of the ageing skeletal muscle in man. A cross-sectional study. Acta Physiol Scand Suppl. 1978;457:1–36.PubMedCrossRef
155.
go back to reference Lexell J, et al. Distribution of different fiber types in human skeletal muscles: effects of aging studied in whole muscle cross sections. Muscle Nerve. 1983;6:588–95.PubMedCrossRef Lexell J, et al. Distribution of different fiber types in human skeletal muscles: effects of aging studied in whole muscle cross sections. Muscle Nerve. 1983;6:588–95.PubMedCrossRef
156.
go back to reference Geiger PC, et al. Maximum specific force depends on myosin heavy chain content in rat diaphragm muscle fibers. J Appl Physiol. 2000;89:695–703.PubMed Geiger PC, et al. Maximum specific force depends on myosin heavy chain content in rat diaphragm muscle fibers. J Appl Physiol. 2000;89:695–703.PubMed
157.
go back to reference Bodine SC, et al. Maximal force as a function of anatomical features of motor units in the cat tibialis anterior. J Neurophysiol. 1987;57:1730–45.PubMed Bodine SC, et al. Maximal force as a function of anatomical features of motor units in the cat tibialis anterior. J Neurophysiol. 1987;57:1730–45.PubMed
158.
159.
go back to reference Ulfhake B, et al. Regulation of neurotrophin signaling in aging sensory and motoneurons: dissipation of target support? Mol Neurobiol. 2000;21:109–35.PubMedCrossRef Ulfhake B, et al. Regulation of neurotrophin signaling in aging sensory and motoneurons: dissipation of target support? Mol Neurobiol. 2000;21:109–35.PubMedCrossRef
160.
go back to reference Larsson L, Li X, Frontera WR. Effects of aging on shortening velocity and myosin isoform composition in single human skeletal muscle cells. Am J Physiol. 1997;272:C638–49.PubMed Larsson L, Li X, Frontera WR. Effects of aging on shortening velocity and myosin isoform composition in single human skeletal muscle cells. Am J Physiol. 1997;272:C638–49.PubMed
161.
go back to reference Hook P, Larsson L. Actomyosin interactions in a novel single muscle fiber in vitro motility assay. J Muscle Res Cell Motil. 2000;21:357–65.PubMedCrossRef Hook P, Larsson L. Actomyosin interactions in a novel single muscle fiber in vitro motility assay. J Muscle Res Cell Motil. 2000;21:357–65.PubMedCrossRef
162.
go back to reference Hook P, et al. In vitro motility speed of slow myosin extracted from single soleus fibres from young and old rats. J Physiol. 1999;520:463–71.PubMedCrossRef Hook P, et al. In vitro motility speed of slow myosin extracted from single soleus fibres from young and old rats. J Physiol. 1999;520:463–71.PubMedCrossRef
163.
go back to reference Lowe DA, et al. Electron paramagnetic resonance reveals age-related myosin structural changes in rat skeletal muscle fibers. Am J Physiol Cell Physiol. 2001;280:C540–7.PubMed Lowe DA, et al. Electron paramagnetic resonance reveals age-related myosin structural changes in rat skeletal muscle fibers. Am J Physiol Cell Physiol. 2001;280:C540–7.PubMed
164.
go back to reference Lowe DA, et al. Muscle activity and aging affect myosin structural distribution and force generation in rat fibers. J Appl Physiol. 2004;96:498–506.PubMedCrossRef Lowe DA, et al. Muscle activity and aging affect myosin structural distribution and force generation in rat fibers. J Appl Physiol. 2004;96:498–506.PubMedCrossRef
165.
go back to reference Balagopal P, et al. Effects of aging on in vivo synthesis of skeletal muscle myosin heavy-chain and sarcoplasmic protein in humans. Am J Physiol. 1997;273:E790–800.PubMed Balagopal P, et al. Effects of aging on in vivo synthesis of skeletal muscle myosin heavy-chain and sarcoplasmic protein in humans. Am J Physiol. 1997;273:E790–800.PubMed
166.
go back to reference Koopman R, et al. Co-ingestion of protein and leucine stimulates muscle protein synthesis rates to the same extent in young and elderly lean men. Am J Clin Nutr. 2006;84:623–32.PubMed Koopman R, et al. Co-ingestion of protein and leucine stimulates muscle protein synthesis rates to the same extent in young and elderly lean men. Am J Clin Nutr. 2006;84:623–32.PubMed
167.
go back to reference Short KR, et al. Age and aerobic exercise training effects on whole body and muscle protein metabolism. Am J Physiol Endocrinol Metab. 2004;286:E92–E101.PubMedCrossRef Short KR, et al. Age and aerobic exercise training effects on whole body and muscle protein metabolism. Am J Physiol Endocrinol Metab. 2004;286:E92–E101.PubMedCrossRef
168.
go back to reference Welle S, Thornton C, Statt M. Myofibrillar protein synthesis in young and old human subjects after three months of resistance training. Am J Physiol. 1995;268:E422–7.PubMed Welle S, Thornton C, Statt M. Myofibrillar protein synthesis in young and old human subjects after three months of resistance training. Am J Physiol. 1995;268:E422–7.PubMed
169.
go back to reference Kawai M, Ishiwata S. Use of thin filament reconstituted muscle fibres to probe the mechanism of force generation. J Muscle Res Cell Motil. 2006;27:455–68.PubMedCrossRef Kawai M, Ishiwata S. Use of thin filament reconstituted muscle fibres to probe the mechanism of force generation. J Muscle Res Cell Motil. 2006;27:455–68.PubMedCrossRef
170.
go back to reference Kanski J, Hong SJ, Schoneich C. Proteomic analysis of protein nitration in aging skeletal muscle and identification of nitrotyrosine-containing sequences in vivo by nanoelectrospray ionization tandem mass spectrometry. J Biol Chem. 2005;280:24261–6.PubMedCrossRef Kanski J, Hong SJ, Schoneich C. Proteomic analysis of protein nitration in aging skeletal muscle and identification of nitrotyrosine-containing sequences in vivo by nanoelectrospray ionization tandem mass spectrometry. J Biol Chem. 2005;280:24261–6.PubMedCrossRef
171.
go back to reference Capitanio D, et al. Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE. Proteomics. 2009;9:2004–20.PubMedCrossRef Capitanio D, et al. Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE. Proteomics. 2009;9:2004–20.PubMedCrossRef
172.
go back to reference Donoghue P, et al. DIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult versus aged gastrocnemius tissue. J Proteomics. 2010;73:1441–53.PubMedCrossRef Donoghue P, et al. DIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult versus aged gastrocnemius tissue. J Proteomics. 2010;73:1441–53.PubMedCrossRef
173.
go back to reference Balogh J, et al. Lower active force generation and improved fatigue resistance in skeletal muscle from desmin deficient mice. J Muscle Res Cell Motil. 2003;24:453–9.PubMedCrossRef Balogh J, et al. Lower active force generation and improved fatigue resistance in skeletal muscle from desmin deficient mice. J Muscle Res Cell Motil. 2003;24:453–9.PubMedCrossRef
174.
go back to reference Li Z, et al. Cardiovascular lesions and skeletal myopathy in mice lacking desmin. Dev Biol. 1996;175:362–6.PubMedCrossRef Li Z, et al. Cardiovascular lesions and skeletal myopathy in mice lacking desmin. Dev Biol. 1996;175:362–6.PubMedCrossRef
175.
go back to reference Sam M, et al. Desmin knockout muscles generate lower stress and are less vulnerable to injury compared with wild-type muscles. Am J Physiol Cell Physiol. 2000;279:C1116–22.PubMed Sam M, et al. Desmin knockout muscles generate lower stress and are less vulnerable to injury compared with wild-type muscles. Am J Physiol Cell Physiol. 2000;279:C1116–22.PubMed
176.
go back to reference Wieneke S, et al. Generation of tension by skinned fibers and intact skeletal muscles from desmin-deficient mice. Biochem Biophys Res Commun. 2000;278:419–25.PubMedCrossRef Wieneke S, et al. Generation of tension by skinned fibers and intact skeletal muscles from desmin-deficient mice. Biochem Biophys Res Commun. 2000;278:419–25.PubMedCrossRef
177.
go back to reference Ansved T, Wallner P, Larsson L. Spatial distribution of motor unit fibres in fast- and slow-twitch rat muscles with special reference to age. Acta Physiol Scand. 1991;143:345–54.PubMedCrossRef Ansved T, Wallner P, Larsson L. Spatial distribution of motor unit fibres in fast- and slow-twitch rat muscles with special reference to age. Acta Physiol Scand. 1991;143:345–54.PubMedCrossRef
178.
go back to reference Russ DW, Grandy JS. Increased desmin expression in hindlimb muscles of aging rats. J Cachex Sarcopenia Muscle. 2011;2:175–80.CrossRef Russ DW, Grandy JS. Increased desmin expression in hindlimb muscles of aging rats. J Cachex Sarcopenia Muscle. 2011;2:175–80.CrossRef
179.
go back to reference Boriek AM, et al. Desmin integrates the three-dimensional mechanical properties of muscles. Am J Physiol Cell Physiol. 2001;280:C46–52.PubMed Boriek AM, et al. Desmin integrates the three-dimensional mechanical properties of muscles. Am J Physiol Cell Physiol. 2001;280:C46–52.PubMed
180.
go back to reference Di Biase V, Franzini-Armstrong C. Evolution of skeletal type e–c coupling: a novel means of controlling calcium delivery. J Cell Biol. 2005;171:695–704.PubMedCrossRef Di Biase V, Franzini-Armstrong C. Evolution of skeletal type e–c coupling: a novel means of controlling calcium delivery. J Cell Biol. 2005;171:695–704.PubMedCrossRef
181.
go back to reference Dargelos E, et al. Calcium-dependent proteolytic system and muscle dysfunctions: a possible role of calpains in sarcopenia. Biochimie. 2008;90:359–68.PubMedCrossRef Dargelos E, et al. Calcium-dependent proteolytic system and muscle dysfunctions: a possible role of calpains in sarcopenia. Biochimie. 2008;90:359–68.PubMedCrossRef
182.
go back to reference Goll DE, et al. Myofibrillar protein turnover: the proteasome and the calpains. J Anim Sci. 2008;86:E19–35.PubMedCrossRef Goll DE, et al. Myofibrillar protein turnover: the proteasome and the calpains. J Anim Sci. 2008;86:E19–35.PubMedCrossRef
183.
go back to reference Hunter SK, et al. Human skeletal sarcoplasmic reticulum Ca2+ uptake and muscle function with aging and strength training. J Appl Physiol. 1999;86:1858–65.PubMed Hunter SK, et al. Human skeletal sarcoplasmic reticulum Ca2+ uptake and muscle function with aging and strength training. J Appl Physiol. 1999;86:1858–65.PubMed
184.
go back to reference Thomas MM, et al. Initiating treadmill training in late middle age offers modest adaptations in Ca2+ handling but enhances oxidative damage in senescent rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1269–78.PubMedCrossRef Thomas MM, et al. Initiating treadmill training in late middle age offers modest adaptations in Ca2+ handling but enhances oxidative damage in senescent rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1269–78.PubMedCrossRef
185.
go back to reference Ma J, Pan Z. Junctional membrane structure and store operated calcium entry in muscle cells. Front Biosci: J Virtual Libr. 2003;8:d242–55.CrossRef Ma J, Pan Z. Junctional membrane structure and store operated calcium entry in muscle cells. Front Biosci: J Virtual Libr. 2003;8:d242–55.CrossRef
186.
188.
go back to reference Pozzan T, et al. Molecular and cellular physiology of intracellular calcium stores. Physiol Rev. 1994;74:595–636.PubMedCrossRef Pozzan T, et al. Molecular and cellular physiology of intracellular calcium stores. Physiol Rev. 1994;74:595–636.PubMedCrossRef
189.
go back to reference Rios E, Pizarro G, Stefani E. Charge movement and the nature of signal transduction in skeletal muscle excitation–contraction coupling. Annu Rev Physiol. 1992;54:109–33.PubMedCrossRef Rios E, Pizarro G, Stefani E. Charge movement and the nature of signal transduction in skeletal muscle excitation–contraction coupling. Annu Rev Physiol. 1992;54:109–33.PubMedCrossRef
190.
go back to reference Putney Jr JW. The integration of receptor-regulated intracellular calcium release and calcium entry across the plasma membrane. Curr Top Cell Regul. 1990;31:111–27.PubMed Putney Jr JW. The integration of receptor-regulated intracellular calcium release and calcium entry across the plasma membrane. Curr Top Cell Regul. 1990;31:111–27.PubMed
191.
go back to reference Zhao X, et al. Compromised store-operated Ca2+ entry in aged skeletal muscle. Aging Cell. 2008;7:561–8.PubMedCrossRef Zhao X, et al. Compromised store-operated Ca2+ entry in aged skeletal muscle. Aging Cell. 2008;7:561–8.PubMedCrossRef
192.
go back to reference Boncompagni S, et al. Progressive disorganization of the excitation–contraction coupling apparatus in aging human skeletal muscle as revealed by electron microscopy: a possible role in the decline of muscle performance. J Gerontol A Biol Sci Med Sci. 2006;61:995–1008.PubMedCrossRef Boncompagni S, et al. Progressive disorganization of the excitation–contraction coupling apparatus in aging human skeletal muscle as revealed by electron microscopy: a possible role in the decline of muscle performance. J Gerontol A Biol Sci Med Sci. 2006;61:995–1008.PubMedCrossRef
193.
go back to reference Gonzalez E, Messi ML, Delbono O. The specific force of single intact extensor digitorum longus and soleus mouse muscle fibers declines with aging. J Membr Biol. 2000;178:175–83.PubMedCrossRef Gonzalez E, Messi ML, Delbono O. The specific force of single intact extensor digitorum longus and soleus mouse muscle fibers declines with aging. J Membr Biol. 2000;178:175–83.PubMedCrossRef
194.
go back to reference Renganathan M, Delbono O. Caloric restriction prevents age-related decline in skeletal muscle dihydropyridine receptor and ryanodine receptor expression. FEBS Lett. 1998;434:346–50.PubMedCrossRef Renganathan M, Delbono O. Caloric restriction prevents age-related decline in skeletal muscle dihydropyridine receptor and ryanodine receptor expression. FEBS Lett. 1998;434:346–50.PubMedCrossRef
195.
go back to reference Wieslander A, et al. Biological significance of reducing glucose degradation products in peritoneal dialysis fluids. Perit Dial Int. 2000;20:S23–7.PubMed Wieslander A, et al. Biological significance of reducing glucose degradation products in peritoneal dialysis fluids. Perit Dial Int. 2000;20:S23–7.PubMed
196.
go back to reference Delbono O, Renganathan M, Messi ML. Excitation–Ca2+ release–contraction coupling in single aged human skeletal muscle fiber. Muscle Nerve Suppl. 1997;5:S88–92.PubMedCrossRef Delbono O, Renganathan M, Messi ML. Excitation–Ca2+ release–contraction coupling in single aged human skeletal muscle fiber. Muscle Nerve Suppl. 1997;5:S88–92.PubMedCrossRef
197.
go back to reference Margreth A, Damiani E, Bortoloso E. Sarcoplasmic reticulum in aged skeletal muscle. Acta Physiol Scand. 1999;167:331–8.PubMedCrossRef Margreth A, Damiani E, Bortoloso E. Sarcoplasmic reticulum in aged skeletal muscle. Acta Physiol Scand. 1999;167:331–8.PubMedCrossRef
198.
go back to reference Narayanan N, et al. Effects of aging on sarcoplasmic reticulum function and contraction duration in skeletal muscles of the rat. Am J Physiol. 1996;271:C1032–40.PubMed Narayanan N, et al. Effects of aging on sarcoplasmic reticulum function and contraction duration in skeletal muscles of the rat. Am J Physiol. 1996;271:C1032–40.PubMed
199.
go back to reference Wang ZM, Messi ML, Delbono O. L-type Ca(2+) channel charge movement and intracellular Ca(2+) in skeletal muscle fibers from aging mice. Biophys J. 2000;78:1947–54.PubMedCrossRef Wang ZM, Messi ML, Delbono O. L-type Ca(2+) channel charge movement and intracellular Ca(2+) in skeletal muscle fibers from aging mice. Biophys J. 2000;78:1947–54.PubMedCrossRef
200.
go back to reference Delbono O. Molecular mechanisms and therapeutics of the deficit in specific force in ageing skeletal muscle. Biogerontology. 2002;3:265–70.PubMedCrossRef Delbono O. Molecular mechanisms and therapeutics of the deficit in specific force in ageing skeletal muscle. Biogerontology. 2002;3:265–70.PubMedCrossRef
201.
go back to reference Moreno RJ, et al. Role of sustained overexpression of central nervous system IGF-I in the age-dependent decline of mouse excitation–contraction coupling. J Membr Biol. 2006;212:147–61.PubMedCrossRef Moreno RJ, et al. Role of sustained overexpression of central nervous system IGF-I in the age-dependent decline of mouse excitation–contraction coupling. J Membr Biol. 2006;212:147–61.PubMedCrossRef
202.
go back to reference Neuhuber B, et al. Association of calcium channel alpha1S and beta1a subunits is required for the targeting of beta1a but not of alpha1S into skeletal muscle triads. Proc Natl Acad Sci USA. 1998;95:5015–20.PubMedCrossRef Neuhuber B, et al. Association of calcium channel alpha1S and beta1a subunits is required for the targeting of beta1a but not of alpha1S into skeletal muscle triads. Proc Natl Acad Sci USA. 1998;95:5015–20.PubMedCrossRef
203.
go back to reference Strube C, et al. Reduced Ca2+ current, charge movement, and absence of Ca2+ transients in skeletal muscle deficient in dihydropyridine receptor beta 1 subunit. Biophys J. 1996;71:2531–43.PubMedCrossRef Strube C, et al. Reduced Ca2+ current, charge movement, and absence of Ca2+ transients in skeletal muscle deficient in dihydropyridine receptor beta 1 subunit. Biophys J. 1996;71:2531–43.PubMedCrossRef
204.
go back to reference Gregg RG, et al. Absence of the beta subunit (cchb1) of the skeletal muscle dihydropyridine receptor alters expression of the alpha 1 subunit and eliminates excitation–contraction coupling. Proc Natl Acad Sci USA. 1996;93:13961–6.PubMedCrossRef Gregg RG, et al. Absence of the beta subunit (cchb1) of the skeletal muscle dihydropyridine receptor alters expression of the alpha 1 subunit and eliminates excitation–contraction coupling. Proc Natl Acad Sci USA. 1996;93:13961–6.PubMedCrossRef
205.
go back to reference Cheng W, et al. Interaction between the dihydropyridine receptor Ca2+ channel beta-subunit and ryanodine receptor type 1 strengthens excitation–contraction coupling. Proc Natl Acad Sci USA. 2005;102:19225–30.PubMedCrossRef Cheng W, et al. Interaction between the dihydropyridine receptor Ca2+ channel beta-subunit and ryanodine receptor type 1 strengthens excitation–contraction coupling. Proc Natl Acad Sci USA. 2005;102:19225–30.PubMedCrossRef
206.
go back to reference Schredelseker J, et al. The beta 1a subunit is essential for the assembly of dihydropyridine-receptor arrays in skeletal muscle. Proc Natl Acad Sci USA. 2005;102:17219–24.PubMedCrossRef Schredelseker J, et al. The beta 1a subunit is essential for the assembly of dihydropyridine-receptor arrays in skeletal muscle. Proc Natl Acad Sci USA. 2005;102:17219–24.PubMedCrossRef
207.
go back to reference Taylor JR, et al. Increased Ca(V)beta(1a) expression with aging contributes skeletal muscle weakness. Aging Cell. 2009;8:584–94.PubMedCrossRef Taylor JR, et al. Increased Ca(V)beta(1a) expression with aging contributes skeletal muscle weakness. Aging Cell. 2009;8:584–94.PubMedCrossRef
208.
go back to reference Payne AM, et al. Role of Ca2+, membrane excitability, and Ca2+ stores in failing muscle contraction with aging. Exp Gerontol. 2009;44:261–73.PubMedCrossRef Payne AM, et al. Role of Ca2+, membrane excitability, and Ca2+ stores in failing muscle contraction with aging. Exp Gerontol. 2009;44:261–73.PubMedCrossRef
209.
go back to reference Andersson DC, et al. Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging. Cell Metab. 2011;14:196–207.PubMedCrossRef Andersson DC, et al. Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging. Cell Metab. 2011;14:196–207.PubMedCrossRef
210.
go back to reference Bellinger AM, et al. Remodeling of ryanodine receptor complex causes “leaky” channels: a molecular mechanism for decreased exercise capacity. Proc Natl Acad Sci USA. 2008;105:2198–202.PubMedCrossRef Bellinger AM, et al. Remodeling of ryanodine receptor complex causes “leaky” channels: a molecular mechanism for decreased exercise capacity. Proc Natl Acad Sci USA. 2008;105:2198–202.PubMedCrossRef
211.
go back to reference Ward CW, et al. Defects in ryanodine receptor calcium release in skeletal muscle from post-myocardial infarct rats. FASEB J. 2003;17:1517–9.PubMed Ward CW, et al. Defects in ryanodine receptor calcium release in skeletal muscle from post-myocardial infarct rats. FASEB J. 2003;17:1517–9.PubMed
212.
go back to reference Ferrington DA, Krainev AG, Bigelow DJ. Altered turnover of calcium regulatory proteins of the sarcoplasmic reticulum in aged skeletal muscle. J Biol Chem. 1998;273:5885–91.PubMedCrossRef Ferrington DA, Krainev AG, Bigelow DJ. Altered turnover of calcium regulatory proteins of the sarcoplasmic reticulum in aged skeletal muscle. J Biol Chem. 1998;273:5885–91.PubMedCrossRef
213.
go back to reference Papageorgopoulos C, et al. Measuring synthesis rates of muscle creatine kinase and myosin with stable isotopes and mass spectrometry. Anal Biochem. 2002;309:1–10.PubMedCrossRef Papageorgopoulos C, et al. Measuring synthesis rates of muscle creatine kinase and myosin with stable isotopes and mass spectrometry. Anal Biochem. 2002;309:1–10.PubMedCrossRef
214.
go back to reference Brandt NR, et al. The role of mitsugumin 29 in transverse tubules of rabbit skeletal muscle. Arch Biochem Biophys. 2001;385:406–9.PubMedCrossRef Brandt NR, et al. The role of mitsugumin 29 in transverse tubules of rabbit skeletal muscle. Arch Biochem Biophys. 2001;385:406–9.PubMedCrossRef
215.
go back to reference Corona BT, et al. Junctophilin damage contributes to early strength deficits and EC coupling failure after eccentric contractions. Am J Physiol Cell Physiol. 2010;298:C365–76.PubMedCrossRef Corona BT, et al. Junctophilin damage contributes to early strength deficits and EC coupling failure after eccentric contractions. Am J Physiol Cell Physiol. 2010;298:C365–76.PubMedCrossRef
216.
go back to reference Ito K, et al. Deficiency of triad junction and contraction in mutant skeletal muscle lacking junctophilin type 1. J Cell Biol. 2001;154:1059–67.PubMedCrossRef Ito K, et al. Deficiency of triad junction and contraction in mutant skeletal muscle lacking junctophilin type 1. J Cell Biol. 2001;154:1059–67.PubMedCrossRef
217.
go back to reference Nishi M, et al. Abnormal features in skeletal muscle from mice lacking mitsugumin29. J Cell Biol. 1999;147:1473–80.PubMedCrossRef Nishi M, et al. Abnormal features in skeletal muscle from mice lacking mitsugumin29. J Cell Biol. 1999;147:1473–80.PubMedCrossRef
218.
go back to reference Woo JS, et al. S165F mutation of junctophilin 2 affects Ca2+ signalling in skeletal muscle. Biochem J. 2010;427:125–34.PubMedCrossRef Woo JS, et al. S165F mutation of junctophilin 2 affects Ca2+ signalling in skeletal muscle. Biochem J. 2010;427:125–34.PubMedCrossRef
219.
go back to reference Kurebayashi N, et al. Changes in Ca2+ handling in adult MG29-deficient skeletal muscle. Biochem Biophys Res Commun. 2003;310:1266–72.PubMedCrossRef Kurebayashi N, et al. Changes in Ca2+ handling in adult MG29-deficient skeletal muscle. Biochem Biophys Res Commun. 2003;310:1266–72.PubMedCrossRef
220.
go back to reference Weisleder N, et al. Muscle aging is associated with compromised Ca2+ spark signaling and segregated intracellular Ca2+ release. J Cell Biol. 2006;174:639–45.PubMedCrossRef Weisleder N, et al. Muscle aging is associated with compromised Ca2+ spark signaling and segregated intracellular Ca2+ release. J Cell Biol. 2006;174:639–45.PubMedCrossRef
221.
go back to reference Thornton AM, et al. Store-operated Ca2+ entry (SOCE) contributes to normal skeletal muscle contractility in young but not in aged skeletal muscle. Aging (Albany NY). 2011;3:621–34. Thornton AM, et al. Store-operated Ca2+ entry (SOCE) contributes to normal skeletal muscle contractility in young but not in aged skeletal muscle. Aging (Albany NY). 2011;3:621–34.
222.
go back to reference Musaro A, Dobrowolny G, Rosenthal N. The neuroprotective effects of a locally acting IGF-1 isoform. Exp Gerontol. 2007;42:76–80.PubMedCrossRef Musaro A, Dobrowolny G, Rosenthal N. The neuroprotective effects of a locally acting IGF-1 isoform. Exp Gerontol. 2007;42:76–80.PubMedCrossRef
223.
go back to reference Payne AM, et al. Motor neurone targeting of IGF-1 prevents specific force decline in ageing mouse muscle. J Physiol. 2006;570:283–94.PubMed Payne AM, et al. Motor neurone targeting of IGF-1 prevents specific force decline in ageing mouse muscle. J Physiol. 2006;570:283–94.PubMed
224.
go back to reference Payne AM, et al. Motor neuron targeting of IGF-1 attenuates age-related external Ca2+-dependent skeletal muscle contraction in senescent mice. Exp Gerontol. 2007;42:309–19.PubMedCrossRef Payne AM, et al. Motor neuron targeting of IGF-1 attenuates age-related external Ca2+-dependent skeletal muscle contraction in senescent mice. Exp Gerontol. 2007;42:309–19.PubMedCrossRef
226.
go back to reference Boesze-Battaglia K, Schimmel R. Cell membrane lipid composition and distribution: implications for cell function and lessons learned from photoreceptors and platelets. J Exp Biol. 1997;200:2927–36.PubMed Boesze-Battaglia K, Schimmel R. Cell membrane lipid composition and distribution: implications for cell function and lessons learned from photoreceptors and platelets. J Exp Biol. 1997;200:2927–36.PubMed
227.
go back to reference Blazyk J, Wu CJ, Wu SC. Correlation between lipid fluidity and tryptic susceptibility of Ca2+-ATPase in sarcoplasmic reticulum membranes. J Biol Chem. 1985;260:4845–9.PubMed Blazyk J, Wu CJ, Wu SC. Correlation between lipid fluidity and tryptic susceptibility of Ca2+-ATPase in sarcoplasmic reticulum membranes. J Biol Chem. 1985;260:4845–9.PubMed
228.
go back to reference Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985;26:1015–35.PubMed Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985;26:1015–35.PubMed
229.
go back to reference Grinna LS. Age-related alterations in membrane lipid and protein interactions: Arrhenius studies of microsomal glucose-6-phosphatase. Gerontology. 1977;23:342–9.PubMedCrossRef Grinna LS. Age-related alterations in membrane lipid and protein interactions: Arrhenius studies of microsomal glucose-6-phosphatase. Gerontology. 1977;23:342–9.PubMedCrossRef
230.
go back to reference Ferrington DA, et al. Decreased conformational stability of the sarcoplasmic reticulum Ca-ATPase in aged skeletal muscle. Biochim Biophys Acta. 1997;1330:233–47.PubMedCrossRef Ferrington DA, et al. Decreased conformational stability of the sarcoplasmic reticulum Ca-ATPase in aged skeletal muscle. Biochim Biophys Acta. 1997;1330:233–47.PubMedCrossRef
231.
go back to reference Krainev AG, et al. Adaptive changes in lipid composition of skeletal sarcoplasmic reticulum membranes associated with aging. Biochim Biophys Acta. 1995;1235:406–18.PubMedCrossRef Krainev AG, et al. Adaptive changes in lipid composition of skeletal sarcoplasmic reticulum membranes associated with aging. Biochim Biophys Acta. 1995;1235:406–18.PubMedCrossRef
232.
go back to reference von Haehling S, et al. Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. J Cachex Sarcopenia Muscle. 2010;1:7–8.CrossRef von Haehling S, et al. Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. J Cachex Sarcopenia Muscle. 2010;1:7–8.CrossRef
233.
go back to reference Clark BC, Taylor JL. Age-related changes in motor cortical properties and voluntary activation of skeletal muscle. Curr Aging Sci. 2011;4:193–200. Clark BC, Taylor JL. Age-related changes in motor cortical properties and voluntary activation of skeletal muscle. Curr Aging Sci. 2011;4:193–200.
234.
go back to reference Kido A, Tanaka N, Stein RB. Spinal excitation and inhibition decrease as humans age. Can J Physiol Pharmacol. 2004;82:238–48.PubMedCrossRef Kido A, Tanaka N, Stein RB. Spinal excitation and inhibition decrease as humans age. Can J Physiol Pharmacol. 2004;82:238–48.PubMedCrossRef
Metadata
Title
Evolving concepts on the age-related changes in “muscle quality”
Authors
David W. Russ
Kimberly Gregg-Cornell
Matthew J. Conaway
Brian C. Clark
Publication date
01-06-2012
Publisher
Springer-Verlag
Published in
Journal of Cachexia, Sarcopenia and Muscle / Issue 2/2012
Print ISSN: 2190-5991
Electronic ISSN: 2190-6009
DOI
https://doi.org/10.1007/s13539-011-0054-2

Other articles of this Issue 2/2012

Journal of Cachexia, Sarcopenia and Muscle 2/2012 Go to the issue