Skeletal muscle mitochondria and aging: a review

Courtney M Peterson, Darcy L Johannsen, Eric Ravussin, Courtney M Peterson, Darcy L Johannsen, Eric Ravussin

Abstract

Aging is characterized by a progressive loss of muscle mass and muscle strength. Declines in skeletal muscle mitochondria are thought to play a primary role in this process. Mitochondria are the major producers of reactive oxygen species, which damage DNA, proteins, and lipids if not rapidly quenched. Animal and human studies typically show that skeletal muscle mitochondria are altered with aging, including increased mutations in mitochondrial DNA, decreased activity of some mitochondrial enzymes, altered respiration with reduced maximal capacity at least in sedentary individuals, and reduced total mitochondrial content with increased morphological changes. However, there has been much controversy over measurements of mitochondrial energy production, which may largely be explained by differences in approach and by whether physical activity is controlled for. These changes may in turn alter mitochondrial dynamics, such as fusion and fission rates, and mitochondrially induced apoptosis, which may also lead to net muscle fiber loss and age-related sarcopenia. Fortunately, strategies such as exercise and caloric restriction that reduce oxidative damage also improve mitochondrial function. While these strategies may not completely prevent the primary effects of aging, they may help to attenuate the rate of decline.

Figures

Figure 1
Figure 1
This cartoon describes the changes in skeletal muscle with aging on the right side of the figure. Both the mass and function of skeletal muscle are decreased in elderly people. Furthermore, at the mitochondrial level, the number of mitochondria is decreased in parallel with changes in mitochondrial morphology. Mitochondrial DNA, oxidative capacity, biogenesis, and autophagy are all decreased in conjunction with an increased number of DNA mutations and increased levels of apoptosis. Finally, oxidative stress is increased in the muscles of elderly people in association with cellular lipid, protein, and DNA damage. The bottom left of the cartoon shows that exercise, caloric restriction, caloric restriction mimetics, and antioxidants can all delay the aging of skeletal muscle.
Figure 2
Figure 2
Mitochondrial processes are both static (a) and dynamic (b). (a) depicts the classical movement of electrons along complexes I–IV embedded in the inner mitochondrial membrane with the generation of a proton gradient (membrane potential). The proton gradient causes hydrogen ions to flow back into the mitochondrial matrix via complex V (ATP synthase), producing ATP in the process. (b) depicts the processes of mitochondrial fusion and fission. Mitochondria can undergo constriction and division (1), mediated by Drp1, which bonds and localizes to the constriction site via an interaction with the receptor-like protein Fis1 (2). During fusion, a tether of the Mfn1/2 to collateral mitochondrial Mfn1/2 conjoins the outer membranes. Opa1, an inner membrane GTPase protein, facilitates the fusion of the inner membrane, cristae formation, and unifying of compartments.
Figure 3
Figure 3
The electron transport system (ETS) of the inner mitochondrial membrane is the primary site of reactive oxygen species (ROS) production and therefore the main source of oxidative stress (damage to proteins, lipids, and DNA) in the mitochondria and in the cell. Free radical superoxide anions (O2 ∙−) are generated when electrons are donated from complexes I and III of the ETS to O2 instead of the appropriate ETS subunit. 2–4% of total oxygen consumption may go toward the production of ROS instead of energy as ATP. Scavenging enzymes represent an important mitochondrial defense mechanism against oxidative stress by neutralizing O2 ∙− within the mitochondrial matrix (superoxide dismutase; MnSOD = SOD2) and catalyzing the reduction of mitochondrial SOD2-generated H2O2 to nontoxic H2O in the mitochondria and the cell (glutathione peroxidase and catalase). Mitochondria in young muscle (a) are numerous and efficient. With age (b), muscle mitochondria become less numerous and seem to develop impaired function associated with reduced oxidative capacity. Through lifestyle changes such as exercise, and caloric restriction, and caloric restriction mimetics, we hypothesize that antioxidative enzymes are upregulated, and that most of the above impairments in aged muscle may be improved (c).

References

    1. Frontera WR, Hughes VA, Lutz KJ, Evans WJ. A cross-sectional study of muscle strength and mass in 45- to 78-yr-old men and women. Journal of Applied Physiology. 1991;71(2):644–650.
    1. Hughes VA, Frontera WR, Wood M, et al. Longitudinal muscle strength changes in older adults: influence of muscle mass, physical activity, and health. The Journals of Gerontology A. 2001;56(5):B209–B217.
    1. Goodpaster BH, Park SW, Harris TB, et al. The loss of skeletal muscle strength, mass, and quality in older adults: the Health, Aging and Body Composition Study. Journals of Gerontology A. 2006;61(10):1059–1064.
    1. Fiatarone MA, O'Neill EF, Ryan ND, Clements KM, Solares GR, Nelson ME. Exercise training and nutritional supplementation for physical frailty in very elderly people. The New England journal of medicine. 1994;330(25):1769–1775.
    1. Goodpaster BH, Chomentowski P, Ward BK, et al. Effects of physical activity on strength and skeletal muscle fat infiltration in older adults: a randomized controlled trial. Journal of Applied Physiology. 2008;105(5):1498–1503.
    1. Nelson ME, Fiatarone MA, Morganti CM, Trice I, Greenberg RA, Evans WJ. Effects of high-intensity strength training on multiple risk factors for osteoporotic fractures: a randomized controlled trial. Journal of the American Medical Association. 1994;272(24):1909–1914.
    1. Metter EJ, Lynch N, Conwit R, Lindle R, Tobin J, Hurley B. Muscle quality and age: cross-sectional and longitudinal comparisons. The Journals of Gerontology A. 1999;54(5):B207–B218.
    1. Cree MG, Newcomer BR, Katsanos CS, et al. Intramuscular and liver triglycerides are increased in the elderly. The Journal of Clinical Endocrinology and Metabolism. 2004;89(8):3864–3871.
    1. Hipkiss AR. Mitochondrial dysfunction, proteotoxicity, and aging: causes or effects, and the possible impact of NAD+-controlled protein glycation. Advances in Clinical Chemistry. 2010;50:123–150.
    1. Johannsen DL, Conley KE, Bajpeyi S, et al. Ectopic lipid accumulation and reduced glucose tolerance in elderly adults are accompanied by altered skeletal muscle mitochondrial activity. The Journal of Clinical Endocrinology and Metabolism. 2012;97(1):242–250.
    1. Saraste M. Oxidative phosphorylation at the fin de siecle. Science. 1999;283(5407):1488–1493.
    1. Holloszy JO. Skeletal muscle,“mitochondrial deficiency” does not mediate insulin resistance. The American Journal of Clinical Nutrition. 2009;89(1):463S–466S.
    1. Miller BF, Robinson MM, Bruss MD, Hellerstein M, Hamilton KL. A comprehensive assessment of mitochondrial protein synthesis and cellular proliferation with age and caloric restriction. Aging Cell. 2012;11(1):150–161.
    1. Shigenaga MK, Hagen TM, Ames BN. Oxidative damage and mitochondrial decay in aging. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(23):10771–10778.
    1. Terman A, Kurz T, Navratil M, Arriaga EA, Brunk UT. Mitochondrial turnover and aging of long-lived postmitotic cells: the mitochondrial-lysosomal axis theory of aging. Antioxidants and Redox Signaling. 2010;12(4):503–535.
    1. Twig G, Hyde B, Shirihai OS. Mitochondrial fusion, fission and autophagy as a quality control axis: the bioenergetic view. Biochimica et Biophysica Acta. 2008;1777(9):1092–1097.
    1. Twig G, Elorza A, Molina AJ, et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. The EMBO Journal. 2008;27(2):433–446.
    1. Beregi E, Regius O, Huttl T, Gobl Z. Age-related changes in the skeletal muscle cells. Zeitschrift fur Gerontologie. 1988;21(2):83–86.
    1. Poggi P, Marchetti C, Scelsi R. Automatic morphometric analysis of skeletal muscle fibers in the aging man. Anatomical Record. 1987;217(1):30–34.
    1. Conley KE, Jubrias SA, Esselman PC. Oxidative capacity and ageing in human muscle. The Journal of Physiology. 2000;5261, part 1:203–210.
    1. Crane JD, Devries MC, Safdar A, Hamadeh MJ, Tarnopolsky MA. The effect of aging on human skeletal muscle mitochondrial and intramyocellular lipid ultrastructure. The Journals of Gerontology A. 2010;65(2):119–128.
    1. Nakamura S, Takamura T, Matsuzawa-Nagata N, et al. Palmitate induces insulin resistance in H4IIEC3 hepatocytes through reactive oxygen species produced by mitochondria. The Journal of Biological Chemistry. 2009;284(22):14809–14818.
    1. Harman D. Aging: a theory based on free radical and radiation chemistry. Journal of Gerontology. 1956;11(3):298–300.
    1. Harman D. Free radical theory of aging: an update: increasing the functional life span. Annals of the New York Academy of Sciences. 2006;1067:10–21.
    1. Chabi B, Ljubicic V, Menzies KJ, Huang JH, Saleem A, Hood DA. Mitochondrial function and apoptotic susceptibility in aging skeletal muscle. Aging Cell. 2008;7(1):2–12.
    1. Hütter E, Skovbro M, Lener B, et al. Oxidative stress and mitochondrial impairment can be separated from lipofuscin accumulation in aged human skeletal muscle. Aging Cell. 2007;6(2):245–256.
    1. Mansouri A, Muller FL, Liu Y, et al. Alterations in mitochondrial function, hydrogen peroxide release and oxidative damage in mouse hind-limb skeletal muscle during aging. Mechanisms of Ageing and Development. 2006;127(3):298–306.
    1. Yarian CS, Rebrin I, Sohal RS. Aconitase and ATP synthase are targets of malondialdehyde modification and undergo an age-related decrease in activity in mouse heart mitochondria. Biochemical and Biophysical Research Communications. 2005;330(1):151–156.
    1. Pesce V, Cormio A, Fracasso F, et al. Age-related mitochondrial genotypic and phenotypic alterations in human skeletal muscle. Free Radical Biology and Medicine. 2001;30(11):1223–1233.
    1. Beal MF. Oxidatively modified proteins in aging and disease. Free Radical Biology & Medicine. 2002;32(9):797–803.
    1. Staunton L, O'Connell K, Ohlendieck K. Proteomic profiling of mitochondrial enzymes during skeletal muscle aging. Journal of Aging Research. 2011;2011:9 pages.908035
    1. Kushnareva Y, Murphy AN, Andreyev A. Complex I-mediated reactive oxygen species generation: modulation by cytochrome c and NAD(P)+ oxidation-reduction state. The Biochemical Journal. 2002;368, part 2:545–553.
    1. Barreiro E, Coronell C, Laviña B, Ramírez-Sarmiento A, Orozco-Levi M, Gea J. Aging, sex differences, and oxidative stress in human respiratory and limb muscles. Free Radical Biology and Medicine. 2006;41(5):797–809.
    1. Ji LL, Dillon D, Wu E. Alteration of antioxidant enzymes with aging in rat skeletal muscle and liver. The American Journal of Physiology. 1990;258(4, part 2):918–923.
    1. Luhtala TA, Roecker EB, Pugh T, Feuers RJ, Weindruch R. Dietary restriction attenuates age-related increases in rat skeletal muscle antioxidant enzyme activities. Journals of Gerontology. 1994;49(5):B231–B238.
    1. Armeni T, Principato G, Quiles JL, Pieri C, Bompadre S, Battino M. Mitochondrial dysfunctions during aging: vitamin E deficiency or caloric restriction—two different ways of modulating stress. Journal of Bioenergetics and Biomembranes. 2003;35(2):181–191.
    1. Ren J, Li Q, Wu S, Li SY, Babcock SA. Cardiac overexpression of antioxidant catalase attenuates aging-induced cardiomyocyte relaxation dysfunction. Mechanisms of Ageing and Development. 2007;128(3):276–285.
    1. Xia E, Rao G, Van Remmen H, Heydari AR, Richardson A. Activities of antioxidant enzymes in various tissues of male Fischer 344 rats are altered by food restriction. Journal of Nutrition. 1995;125(2):195–201.
    1. Safdar A, Hamadeh MJ, Kaczor JJ, Raha S, Debeer J, Tarnopolsky MA. Aberrant mitochondrial homeostasis in the skeletal muscle of sedentary older adults. PLoS ONE. 2010;5(5):p. e10778.
    1. Lee HY, Choi CS, Birkenfeld AL, Alves TC, Jornayvaz FR, Jurczak MJ. Targeted expression of catalase to mitochondria prevents age-associated reductions in mitochondrial function and insulin resistance. Cell Metabolism. 2010;12(6):668–674.
    1. Iossa S, Mollica MP, Lionetti L, Crescenzo R, Tasso R, Liverini G. A possible link between skeletal muscle mitochondrial efficiency and age-induced insulin resistance. Diabetes. 2004;53(11):2861–2866.
    1. Marcinek DJ, Schenkman KA, Ciesielski WA, Lee D, Conley KE. Reduced mitochondrial coupling in vivo alters cellular energetics in aged mouse skeletal muscle. The Journal of Physiology. 2005;569, part 2:467–473.
    1. Amara CE, Shankland EG, Jubrias SA, Marcinek DJ, Kushmerick MJ, Conley KE. Mild mitochondrial uncoupling impacts cellular aging in human muscles in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(3):1057–1062.
    1. Lopez MF, Kristal BS, Chernokalskaya E, Lazarev A, Shestopalov AI, Bogdanova A, et al. High-throughput profiling of the mitochondrial proteome using affinity fractionation and automation. Electrophoresis. 2000;21(16):3427–3440.
    1. Calvo S, Jain M, Xie X, Sheth SA, Chang B, Goldberger OA, et al. Systematic identification of human mitochondrial disease genes through integrative genomics. Nature Genetics. 2006;38(5):576–582.
    1. Anderson S, Bankier AT, Barrell BG, de Bruijn MH, Coulson AR, Drouin J, et al. Sequence and organization of the human mitochondrial genome. Nature. 1981;290(5806):457–465.
    1. Menshikova EV, Ritov VB, Fairfull L, Ferrell RE, Kelley DE, Goodpaster BH. Effects of exercise on mitochondrial content and function in aging human skeletal muscle. Journals of Gerontology A. 2006;61(6):534–540.
    1. Short KR, Bigelow ML, Kahl J, Singh R, Coenen-Schimke J, Raghavakaimal S, et al. Decline in skeletal muscle mitochondrial function with aging in humans. . Proceedings of the National Academy of Sciences of the United States of America. 2005;102(15):5618–5623.
    1. Welle S, Bhatt K, Shah B, Needler N, Delehanty JM, Thornton CA. Reduced amount of mitochondrial DNA in aged human muscle. Journal of Applied Physiology. 2003;94(4):1479–1484.
    1. Lanza IR, Short DK, Short KR, Raghavakaimal S, Basu R, Joyner MJ, et al. Endurance exercise as a countermeasure for aging. Diabetes. 2008;57(11):2933–2942.
    1. Barrientos A, Casademont J, Cardellach F, et al. Qualitative and quantitative changes in skeletal muscle mtDNA and expression of mitochondrial-encoded genes in the human aging process. Biochemical and Molecular Medicine. 1997;62(2):165–171.
    1. Barazzoni R, Short KR, Nair KS. Effects of aging on mitochondrial DNA copy number and cytochrome c oxidase gene expression in rat skeletal muscle, liver, and heart. Journal of Biological Chemistry. 2000;275(5):3343–3347.
    1. McKenzie D, Bua E, McKiernan S, Cao Z, Wanagat J, Aiken JM. Mitochondrial DNA deletion mutations: a causal role in sarcopenia. European Journal of Biochemistry. 2002;269(8):2010–2015.
    1. Khrapko K, Vijg J. Mitochondrial DNA mutations and aging: devils in the details? Trends in Genetics. 2009;25(2):91–98.
    1. Bua E, Johnson J, Herbst A, et al. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. American Journal of Human Genetics. 2006;79(3):469–480.
    1. Chabi B, Mousson de Camaret B, Chevrollier A, Boisgard S, Stepien G. Random mtDNA deletions and functional consequence in aged human skeletal muscle. Biochemical and Biophysical Research Communications. 2005;332(2):542–549.
    1. Melov S, Shoffner JM, Kaufman A, Wallace DC. Marked increase in the number and variety of mitochondrial DNA rearrangements in aging human skeletal muscle. Nucleic Acids Research. 1995;23(20):4122–4126.
    1. Cooper JM, Mann VM, Schapira AHV. Analyses of mitochondrial respiratory chain function and mitochondrial DNA deletion in human skeletal muscle: effect of ageing. Journal of the Neurological Sciences. 1992;113(1):91–98.
    1. Pallotti F, Chen X, Bonilla E, Schon EA. Evidence that specific mtDNA point mutations may not accumulate in skeletal muscle during normal human aging. American Journal of Human Genetics. 1996;59(3):591–602.
    1. Wang Y, Michikawa Y, Mallidis C, Bai Y, Woodhouse L, Yarasheski KE, et al. Muscle-specific mutations accumulate with aging in critical human mtDNA control sites for replication. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(7):4022–4027.
    1. Michikawa Y, Mazzucchelli F, Bresolin N, Scarlato G, Attardi G. Aging-dependent large accumulation of point mutations in the human mtDNA control region for replication. Science. 1999;286(5440):774–779.
    1. Wanagat J, Cao Z, Pathare P, Aiken JM. Mitochondrial DNA deletion mutations colocalize with segmental electron transport system abnormalities, muscle fiber atrophy, fiber splitting, and oxidative damage in sarcopenia. The FASEB Journal. 2001;15(2):322–332.
    1. Torii K, Sugiyama S, Tanaka M, Takagi K, Hanaki Y, Iida K, et al. Aging-associated deletions of human diaphragmatic mitochondrial DNA. American Journal of Respiratory Cell and Molecular Biology. 1992;6(5):543–549.
    1. Fattoretti P, Vecchiet J, Felzani G, et al. Succinic dehydrogenase activity in human muscle mitochondria during aging: a quantitative cytochemical investigation. Mechanisms of Ageing and Development. 2001;122(15):1841–1848.
    1. Bohr VA, Stevnsner T, de Souza-Pinto NC. Mitochondrial DNA repair of oxidative damage in mammalian cells. Gene. 2002;286(1):127–134.
    1. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(2):514–519.
    1. Larsson NG. Somatic mitochondrial DNA mutations in mammalian aging. Annual Review of Biochemistry. 2010;79:683–706.
    1. Meissner C. Mutations of mitochondrial DNA—cause or consequence of the ageing process? Z Gerontol Geriatr. 2007;40(5):325–333.
    1. Hiona A, Leeuwenburgh C. The role of mitochondrial DNA mutations in aging and sarcopenia: implications for the mitochondrial vicious cycle theory of aging. Experimental Gerontology. 2008;43(1):24–33.
    1. Conley KE, Marcinek DJ, Villarin J. Mitochondrial dysfunction and age. Current Opinion in Clinical Nutrition and Metabolic Care. 2007;10(6):688–692.
    1. Brierley EJ, Johnson MA, Lightowlers RN, James OF, Turnbull DM. Role of mitochondrial DNA mutations in human aging: implications for the central nervous system and muscle. Annals of Neurology. 1998;43(2):217–223.
    1. Pesce V, Cormio A, Fracasso F, et al. Age-related mitochondrial genotypic and phenotypic alterations in human skeletal muscle. Free Radical Biology and Medicine. 2001;30(11):1223–1233.
    1. Fayet G, Jansson M, Sternberg D, et al. Ageing muscle: clonal expansions of mitochondrial DNA point mutations and deletions cause focal impairment of mitochondrial function. Neuromuscular Disorders. 2002;12(5):484–493.
    1. Lee HC, Pang CY, Hsu HS, Wei YH. Ageing-associated tandem duplications in the D-loop of mitochondrial DNA of human muscle. FEBS Letters. 1994;354(1):79–83.
    1. Rooyackers OE, Adey DB, Ades PA, Nair KS. Effect of age on in vivo rates of mitochondrial protein synthesis in human skeletal muscle. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(26):15364–15369.
    1. Trifunovic A, Wredenberg A, Falkenberg M, et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature. 2004;429(6990):417–423.
    1. Kujoth GC, Hiona A, Pugh TD, et al. Medicine: mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science. 2005;309(5733):481–484.
    1. Hiona A, Sanz A, Kujoth GC, et al. Mitochondrial DNA mutations induce mitochondrial dysfunction, apoptosis and sarcopenia in skeletal muscle of mitochondrial DNA mutator mice. PLoS ONE. 2010;5(7)e11468
    1. Boffoli D, Scacco SC, Vergari R, Solarino G, Santacroce G, Papa S. Decline with age of the respiratory chain activity in human skeletal muscle. Biochimica et Biophysica Acta. 1994;1226(1):73–82.
    1. Welle S, Bhatt K, Thornton CA. High-abundance mRNAs in human muscle: comparison between young and old. Journal of Applied Physiology. 2000;89(1):297–304.
    1. Welle S, Brooks AI, Delehanty JM, et al. Skeletal muscle gene expression profiles in 20–29 year old and 65–71 year old women. Experimental Gerontology. 2004;39(3):369–377.
    1. Zahn JM, Sonu R, Vogel H, Crane E, Mazan-Mamczarz K, Rabkin R. Transcriptional profiling of aging in human muscle reveals a common aging signature. PLoS Genetics. 2006;2(7):p. e115.
    1. Welle S, Brooks AI, Delehanty JM, Needler N, Thornton CA. Gene expression profile of aging in human muscle. Physiol Genomics. 2003;14(2):149–159.
    1. Melov S, Tarnopolsky MA, Beckman K, Felkey K, Hubbard A. Resistance exercise reverses aging in human skeletal muscle. PLoS ONE. 2007;2(5):p. e465.
    1. Kayo T, Allison DB, Weindruch R, Prolla TA. Influences of aging and caloric restriction on the transcriptional profile of skeletal muscle from rhesus monkeys. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(9):5093–5098.
    1. Picard M, Ritchie D, Wright KJ, et al. Mitochondrial functional impairment with aging is exaggerated in isolated mitochondria compared to permeabilized myofibers. Aging Cell. 2010;9(6):1032–1046.
    1. de Magalhaes JP, Curado J, Church GM. Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics. 2009;25(7):875–881.
    1. Lyons CN, Mathieu-Costello O, Moyes CD. Regulation of skeletal muscle mitochondrial content during aging. Journals of Gerontology A. 2006;61(1):3–13.
    1. Giresi PG, Stevenson EJ, Theilhaber J, et al. Identification of a molecular signature of sarcopenia. Physiological Genomics. 2005;21:253–263.
    1. Mootha VK, Bunkenborg J, Olsen JV, et al. Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell. 2003;115(5):629–640.
    1. Combaret L, Dardevet D, Béchet D, Taillandier D, Mosoni L, Attaix D. Skeletal muscle proteolysis in aging. Current Opinion in Clinical Nutrition and Metabolic Care. 2009;12(1):37–41.
    1. Irving BA, Robinson MM, Nair KS. Age effect on myocellular remodeling: response to exercise and nutrition in humans. Ageing Research Reviews. 2012;11(3):374–389.
    1. Gianni P, Jan KJ, Douglas MJ, Stuart PM, Tarnopolsky MA. Oxidative stress and the mitochondrial theory of aging in human skeletal muscle. Experimental Gerontology. 2004;39(9):1391–400.
    1. O’Connell K, Ohlendieck K. Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle. Proteomics. 2009;9(24):5509–5524.
    1. Piec I, Listrat A, Alliot J, Chambon C, Taylor RG, Bechet D. Differential proteome analysis of aging in rat skeletal muscle. The FASEB Journal. 2005;19(9):1143–1145.
    1. Ghosh S, Lertwattanarak R, Lefort N, Molina-Carrion M, Joya-Galeana J, Bowen BP. Reduction in reactive oxygen species production by mitochondria from elderly subjects with normal and impaired glucose tolerance. Diabetes. 2011;60(8):2051–2060.
    1. Short KR, Vittone JL, Bigelow ML, et al. Impact of aerobic exercise training on age-related changes in insulin sensitivity and muscle oxidative capacity. Diabetes. 2003;52(8):1888–1896.
    1. Alves RMP, Vitorino R, Figueiredo P, Duarte JA, Ferreira R, Amado F. Lifelong physical activity modulation of the skeletal muscle mitochondrial proteome in mice. Journals of Gerontology A. 2010;65(8):832–842.
    1. Aspnes LE, Lee CM, Weindruch R, Chung SS, Roecker EB, Aiken JM. Caloric restriction reduces fiber loss and mitochondrial abnormalities in aged rat muscle. The FASEB Journal. 1997;11(7):573–581.
    1. McKiernan SH, Colman RJ, Aiken E, et al. Cellular adaptation contributes to calorie restriction-induced preservation of skeletal muscle in aged rhesus monkeys. Experimental Gerontology. 2012;47(3):229–236.
    1. Brierley EJ, Johnson MA, James OF, Turnbull DM. Effects of physical activity and age on mitochondrial function. Monthly Journal of the Association of Physicians. 1996;89(4):251–258.
    1. Gelfi C, Vigano A, Ripamonti M, et al. The human muscle proteome in aging. Journal of Proteome Research. 2006;5(6):1344–1353.
    1. Donoghue P, Staunton L, Mullen E, Manning G, Ohlendieck K. DIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult versus aged gastrocnemius tissue. Journal of Proteomics. 2010;73(8):1441–1453.
    1. Tonkonogi M, Fernström M, Walsh B, et al. Reduced oxidative power but unchanged antioxidative capacity in skeletal muscle from aged humans. Pflugers Archiv European Journal of Physiology. 2003;446(2):261–269.
    1. Coggan AR, Spina RJ, King DS, et al. Histochemical and enzymatic comparison of the gastrocnemius muscle of young and elderly men and women. Journals of Gerontology. 1992;47(3):B71–B76.
    1. Lenaz G, Bovina C, Castelluccio C, et al. Mitochondrial complex I defects in aging. Molecular and Cellular Biochemistry. 1997;174(1-2):329–333.
    1. Kumaran S, Subathra M, Balu M, Panneerselvam C. Age-associated decreased activities of mitochondrial electron transport chain complexes in heart and skeletal muscle: role of L-carnitine. Chemico-Biological Interactions. 2004;148(1-2):11–18.
    1. Kerner J, Turkaly PJ, Minkler PE, Hoppel CL. Aging skeletal muscle mitochondria in the rat: decreased uncoupling protein-3 content. American Journal of Physiology. 2001;281(5):E1054–E1062.
    1. Mansouri A, Muller FL, Liu Y, et al. Alterations in mitochondrial function, hydrogen peroxide release and oxidative damage in mouse hind-limb skeletal muscle during aging. Mechanisms of Ageing and Development. 2006;127(3):298–306.
    1. Hepple RT, Baker DJ, McConkey M, Murynka T, Norris R. Caloric restriction protects mitochondrial function with aging in skeletal and cardiac muscles. Rejuvenation Research. 2006;9(2):219–222.
    1. Hepple RT, Baker DJ, Kaczor JJ, Krause DJ. Long-term caloric restriction abrogates the age-related decline in skeletal muscle aerobic function. The FASEB Journal. 2005;19(10):1320–1322.
    1. Baker DJ, Betik AC, Krause DJ, Hepple RT. No decline in skeletal muscle oxidative capacity with aging in long-term calorically restricted rats: effects are independent of mitochondrial DNA integrity. Journals of Gerontology A. 2006;61(7):675–684.
    1. Capel F, Rimbert V, Lioger D, et al. Due to reverse electron transfer, mitochondrial H2O2 release increases with age in human vastus lateralis muscle although oxidative capacity is preserved. Mechanisms of Ageing and Development. 2005;126(4):505–511.
    1. Pastoris O, Boschi F, Verri M, et al. The effects of aging on enzyme activities and metabolite concentrations in skeletal muscle from sedentary male and female subjects. Experimental Gerontology. 2000;35(1):95–104.
    1. Rasmussen UF, Krustrup P, Kjær M, Rasmussen HN. Experimental evidence against the mitochondrial theory of aging A study of isolated human skeletal muscle mitochondria. Experimental Gerontology. 2003;38(8):877–886.
    1. Larsson L, Sjodin B, Karlsson J. Histochemical and biochemical changes in human skeletal muscle with age in sedentary males, age 22–65 yrs. Acta Physiologica Scandinavica. 1978;103(1):31–39.
    1. Rasmussen UF, Krustrup P, Kjaer M, Rasmussen HN. Human skeletal muscle mitochondrial metabolism in youth and senescence: no signs of functional changes in ATP formation and mitochondrial oxidative capacity. Pflugers Archiv European Journal of Physiology. 2003;446(2):270–278.
    1. McCully KK, Fielding RA, Evans WJ, Leigh JS, Posner JD. Relationships between in vivo and in vitro measurements of metabolism in young and old human calf muscles. Journal of Applied Physiology. 1993;75(2):813–819.
    1. Chilibeck PD, McCreary CR, Marsh GD, et al. Evaluation of muscle oxidative potential by 31P-MRS during incremental exercise in old and young humans. European Journal of Applied Physiology and Occupational Physiology. 1998;78(5):460–465.
    1. McCully KK, Forciea MA, Hack LM, et al. Muscle metabolism in older subjects using 31P magnetic resonance spectroscopy. Canadian Journal of Physiology and Pharmacology. 1991;69(5):576–580.
    1. Houmard JA, Weidner ML, Gavigan KE, Tyndall GL, Hickey MS, Alshami A. Fiber type and citrate synthase activity in the human gastrocnemius and vastus lateralis with aging. Journal of Applied Physiology. 1998;85(4):1337–1341.
    1. Trounce I, Byrne E, Marzuki S. Decline in skeletal muscle mitochondrial respiratory chain function: possible factor in ageing. The Lancet. 1989;1(8639):637–639.
    1. Schunk K, Pitton M, Düber C, Kersjes W, Schadmand-Fischer S, Thelen M. Dynamic phosphorus-31 magnetic resonance spectroscopy of the quadriceps muscle: effects of age and sex on spectroscopic results. Investigative Radiology. 1999;34(2):116–125.
    1. Taylor DJ, Kemp GJ, Thompson CH, Radda GK. Ageing: effects on oxidative function of skeletal muscle in vivo. Molecular and Cellular Biochemistry. 1997;174(1-2):1321–1322.
    1. Marcinek DJ, Schenkman KA, Ciesielski WA, Lee D, Conley KE. Reduced mitochondrial coupling in vivo alters cellular energetics in aged mouse skeletal muscle. The Journal of Physiology. 2005;569, part 2:467–473.
    1. Barrientos A, Casademont J, Rötig A, et al. Absence of relationship between the level of electron transport chain activities and aging in human skeletal muscle. Biochemical and Biophysical Research Communications. 1996;229(2):536–539.
    1. Brierly EJ, Johnson MA, Bowman A, et al. Mitochondrial function in muscle from elderly athletes. Annals of Neurology. 1997;41(1):114–116.
    1. Larsen RG, Callahan DM, Foulis SA, Kent-Braun JA. Age-related changes in oxidative capacity differ between locomotory muscles and are associated with physical activity behavior. Applied Physiology, Nutrition and Metabolism. 2012;37(1):88–99.
    1. Waters DL, Brooks WM, Qualls CR, Baumgartner RN. Skeletal muscle mitochondrial function and lean body mass in healthy exercising elderly. Mechanisms of Ageing and Development. 2003;124(3):301–309.
    1. Petersen KF, Befroy D, Dufour S, et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science. 2003;300(5622):1140–1142.
    1. Proctor DN, Sinning WE, Walro JM, Sieck GC, Lemon PWR. Oxidative capacity of human muscle fiber types: effects of age and training status. Journal of Applied Physiology. 1995;78(6):2033–2038.
    1. Rimbert V, Boirie Y, Bedu M, Hocquette JF, Ritz P, Morio B. Muscle fat oxidative capacity is not impaired by age but by physical inactivity: association with insulin sensitivity. The FASEB Journal. 2004;18(6):737–739.
    1. Figueiredo PA, Powers SK, Ferreira RM, Amado F, Appell HJ, Duarte JA. Impact of lifelong sedentary behavior on mitochondrial function of mice skeletal muscle. The Journals of Gerontology A. 2009;64(9):927–939.
    1. Kent-Braun JA, Ng AV. Skeletal muscle oxidative capacity in young and older women and men. Journal of Applied Physiology. 2000;89(3):1072–1078.
    1. Lanza IR, Befroy DE, Kent-Braun JA. Age-related changes in ATP-producing pathways in human skeletal muscle in vivo. Journal of Applied Physiology. 2005;99(5):1736–1744.
    1. Lanza IR, Larsen RG, Kent-Braun JA. Effects of old age on human skeletal muscle energetics during fatiguing contractions with and without blood flow. The Journal of Physiology. 2007;583, part 3:1093–1105.
    1. Lopez-Lluch G, Irusta PM, Navas P, de Cabo R. Mitochondrial biogenesis and healthy aging. Experimental Gerontology. 2008;43(9):813–819.
    1. Hood DA, Irrcher I, Ljubicic V, Joseph AM. Coordination of metabolic plasticity in skeletal muscle. The Journal of Experimental Biology. 2006;209, part 2:2265–2275.
    1. Jager S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(29):12017–12022.
    1. Puigserver P, Rhee J, Lin J, et al. Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator-1. Molecular Cell. 2001;8(5):971–982.
    1. Knutti D, Kressler D, Kralli A. Regulation of the transcriptional coactivator PGC-1 via MAPK-sensitive interaction with a repressor. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(17):9713–9718.
    1. Akimoto T, Pohnert SC, Li P, et al. Exercise stimulates Pgc-1alpha transcription in skeletal muscle through activation of the p38 MAPK pathway. The Journal of Biological Chemistry. 2005;280(20):19587–19593.
    1. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature. 2005;434(7029):113–118.
    1. Gerhart-Hines Z, Rodgers JT, Bare O, et al. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. The EMBO Journal. 2007;26(7):1913–1923.
    1. Wu Z, Huang X, Feng Y, et al. Transducer of regulated CREB-binding proteins (TORCs) induce PGC-1α transcription and mitochondrial biogenesis in muscle cells. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(39):14379–14384.
    1. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998;92(6):829–839.
    1. Puigserver P, Adelmant G, Wu Z, et al. Activation of PPARγ coactivator-1 through transcription factor docking. Science. 1999;286(5443):1368–1371.
    1. Virbasius JV, Virbasius CA, Scarpulla RC. Identity of GABP with NRF-2, a multisubunit activator of cytochrome oxidase expression, reveals a cellular role for and ETS domain activator of viral promoters. Genes and Development. 1993;7(3):380–392.
    1. Virbasius CMA, Virbasius JV, Scarpulla RC. NRF-1, an activator involved in nuclear-mitochondrial interactions, utilizes a new DNA-binding domain conserved in a family of developmental regulators. Genes and Development. 1993;7(12 A):2431–2445.
    1. Scarpulla RC. Nuclear control of respiratory chain expression by nuclear respiratory factors and PGC-1-related coactivator. Annals of the New York Academy of Sciences. 2008;1147:321–334.
    1. Virbasius JV, Scarpulla RC. Activation of the human mitochondrial transcription factor A gene by nuclear respiratory factors: a potential regulatory link between nuclear and mitochondrial gene expression in organelle biogenesis. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(4):1309–1313.
    1. Hood DA, Joseph AM. Mitochondrial assembly: protein import. The Proceedings of the Nutrition Society. 2004;63(2):293–300.
    1. Diaz F, Moraes CT. Mitochondrial biogenesis and turnover. Cell Calcium. 2008;44(1):24–35.
    1. Hood DA, Adhihetty PJ, Colavecchia M, et al. Mitochondrial biogenesis and the role of the protein import pathway. Medicine and Science in Sports and Exercise. 2003;35(1):86–94.
    1. Neupert W, Herrmann JM. Translocation of proteins into mitochondria. Annual Review of Biochemistry. 2007;76:723–749.
    1. Gleyzer N, Vercauteren K, Scarpulla RC. Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators. Molecular and Cellular Biology. 2005;25(4):1354–1366.
    1. Reznick RM, Zong H, Li J, et al. Aging-associated reductions in AMP-activated protein kinase activity and mitochondrial biogenesis. Cell Metabolism. 2007;5(2):151–156.
    1. Wenz T, Rossi SG, Rotundo RL, Spiegelman BM, Moraes CT. Increased muscle PGC-1α expression protects from sarcopenia and metabolic disease during aging. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(48):20405–20410.
    1. Bevilacqua L, Ramsey JJ, Hagopian K, Weindruch R, Harper ME. Effects of short- and medium-term calorie restriction on muscle mitochondrial proton leak and reactive oxygen species production. American Journal of Physiology. 2004;286(5):E852–E861.
    1. Lezza AMS, Pesce V, Cormio A, et al. Increased expression of mitochondrial transcription factor A and nuclear respiratory factor-1 in skeletal muscle from aged human subjects. FEBS Letters. 2001;501(1–3):74–78.
    1. Frazier AE, Kiu C, Stojanovski D, Hoogenraad NJ, Ryan MT. Mitochondrial morphology and distribution in mammalian cells. Biological Chemistry. 2006;387(12):1551–1558.
    1. Benard G, Karbowski M. Mitochondrial fusion and division: regulation and role in cell viability. Seminars in Cell & Developmental Biology. 2009;20(3):365–374.
    1. Stojanovski D, Koutsopoulos OS, Okamoto K, Ryan MT. Levels of human Fis1 at the mitochondrial outer membrane regulate mitochondrial morphology. Journal of Cell Science. 2004;117, part 7:1201–1210.
    1. Chan DC. Mitochondrial fusion and fission in mammals. Annual Review of Cell and Developmental Biology. 2006;22:79–99.
    1. Romanello V, Guadagnin E, Gomes L, et al. Mitochondrial fission and remodelling contributes to muscle atrophy. The EMBO Journal. 2010;29(10):1774–1785.
    1. Scheckhuber CQ, Wanger RA, Mignat CA, Osiewacz HD. Unopposed mitochondrial fission leads to severe lifespan shortening. Cell Cycle. 2011;10(18):3105–3110.
    1. Lee S, Jeong SY, Lim WC, et al. Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate cellular senescence. Journal of Biological Chemistry. 2007;282(31):22977–22983.
    1. Smirnova E, Shurland DL, Ryazantsev SN, van der Bliek AM. A human dynamin-related protein controls the distribution of mitochondria. The Journal of Cell Biology. 1998;143(2):351–358.
    1. Ono T, Isobe K, Nakada K, Hayashi JI. Human cells are protected from mitochondrial dysfunction by complementation of DNA products in fused mitochondria. Nature Genetics. 2001;28(3):272–275.
    1. Sato A, Nakada K, Hayashi JI. Mitochondrial dynamics and aging: mitochondrial interaction preventing individuals from expression of respiratory deficiency caused by mutant mtDNA. Biochimica et Biophysica Acta. 2006;1763(5-6):473–481.
    1. Kowald A, Kirkwood TB. Evolution of the mitochondrial fusion-fission cycle and its role in aging. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(25):10237–10242.
    1. Kirkwood SP, Munn EA, Brooks GA. Mitochondrial reticulum in limb skeletal muscle. The American Journal of Physiology. 1986;3, part 1:C395–C402.
    1. Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. The Journal of Biological Chemistry. 2005;280(28):26185–26192.
    1. Bach D, Pich S, Soriano FX, et al. Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism: a novel regulatory mechanism altered in obesity. Journal of Biological Chemistry. 2003;278(19):17190–17197.
    1. Cipolat S, Rudka T, Hartmann D, et al. Mitochondrial rhomboid PARL regulates cytochrome c release during apoptosis via OPA1-dependent cristae remodeling. Cell. 2006;126(1):163–175.
    1. Duvezin-Caubet S, Jagasia R, Wagener J, et al. Proteolytic processing of OPA1 links mitochondrial dysfunction to alterations in mitochondrial morphology. Journal of Biological Chemistry. 2006;281(49):37972–37979.
    1. Chen H, Vermulst M, Wang YE, et al. Mitochondrial fusion is required for mtdna stability in skeletal muscle and tolerance of mtDNA mutations. Cell. 2010;141(2):280–289.
    1. Lodi R, Tonon C, Valentino ML, et al. Deficit of in vivo mitochondrial ATP production in OPA1-related dominant optic atrophy. Annals of Neurology. 2004;56(5):719–723.
    1. Kim I, Rodriguez-Enriquez S, Lemasters JJ. Selective degradation of mitochondria by mitophagy. Archives of Biochemistry and Biophysics. 2007;462(2):245–253.
    1. Wang K, Klionsky DJ. Mitochondria removal by autophagy. Autophagy. 2011;7(3):297–300.
    1. Okamoto K, Kondo-Okamoto N, Ohsumi Y. Mitochondria-anchored receptor Atg32 mediates degradation of mitochondria via selective autophagy. Developmental Cell. 2009;17(1):87–97.
    1. Kanki T. Nix, a receptor protein for mitophagy in mammals. Autophagy. 2010;6(3):433–435.
    1. Gu Y, Wang C, Cohen A. Effect of IGF-1 on the balance between autophagy of dysfunctional mitochondria and apoptosis. FEBS Letters. 2004;577(3):357–360.
    1. Zhang Y, Qi H, Taylor R, Xu W, Liu LF, Jin S. The role of autophagy in mitochondria maintenance: characterization of mitochondrial functions in autophagy-deficient S. cerevisiae strains. Autophagy. 2007;3(4):337–346.
    1. Cavallini G, Donati A, Taddei M, Bergamini E. Evidence for selective mitochondrial autophagy and failure in aging. Autophagy. 2007;3(1):26–27.
    1. Cuervo AM, Bergamini E, Brunk UT, Dröge W, Ffrench M, Terman A. Autophagy and aging: the importance of maintaining “clean” cells. Autophagy. 2005;1(3):131–140.
    1. Wohlgemuth SE, Seo AY, Marzetti E, Lees HA, Leeuwenburgh C. Skeletal muscle autophagy and apoptosis during aging: effects of calorie restriction and life-long exercise. Experimental Gerontology. 2010;45(2):138–148.
    1. Masiero E, Sandri M. Autophagy inhibition induces atrophy and myopathy in adult skeletal muscles. Autophagy. 2010;6(2):307–309.
    1. Wu JJ, Quijano C, Chen E, et al. Mitochondrial dysfunction and oxidative stress mediate the physiological impairment induced by the disruption of autophagy. Aging. 2009;1(4):425–437.
    1. Vellai T, Takacs-Vellai K, Sass M, Klionsky DJ. The regulation of aging: does autophagy underlie longevity? Trends in Cell Biology. 2009;19(10):487–494.
    1. Taylor EB, Rutter J. Mitochondrial quality control by the ubiquitin-proteasome system. Biochemical Society Transactions. 2011;39(5):1509–1513.
    1. Low P. The role of ubiquitin-proteasome system in ageing. General and Comparative Endocrinology. 2011;172(1):39–43.
    1. Altun M, Besche HC, Overkleeft HS, et al. Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. Journal of Biological Chemistry. 2010;285(51):39597–39608.
    1. Whitman SA, Wacker MJ, Richmond SR, Godard MP. Contributions of the ubiquitin-proteasome pathway and apoptosis to human skeletal muscle wasting with age. Pflugers Archiv European Journal of Physiology. 2005;450(6):437–446.
    1. Dalbo VJ, Roberts MD, Hassell SE, Brown RD, Kerksick CM. Effects of age on serum hormone concentrations and intramuscular proteolytic signaling before and after a single bout of resistance training. The Journal of Strength & Conditioning Research. 2011;25(1):1–9.
    1. Lee CK, Klopp RG, Weindruch R, Prolla TA. Gene expression profile of aging and its retardation by caloric restriction. Science. 1999;285(5432):1390–1393.
    1. Strucksberg KH, Tangavelou K, Schröder R, Clemen CS. Proteasomal activity in skeletal muscle: a matter of assay design, muscle type, and age. Analytical Biochemistry. 2010;399(2):225–229.
    1. Cai D, Lee KK, Li M, Tang MK, Chan KM. Ubiquitin expression is up-regulated in human and rat skeletal muscles during aging. Archives of Biochemistry and Biophysics. 2004;425(1):42–50.
    1. Green DR, Reed JC. Mitochondria and apoptosis. Science. 1998;281(5381):1309–1312.
    1. Jeong SY, Seol DW. The role of mitochondria in apoptosis. BMB Reports. 2008;41(1):11–22.
    1. Marzetti E, Hwang JC, Lees HA, et al. Mitochondrial death effectors: relevance to sarcopenia and disuse muscle atrophy. Biochimica et Biophysica. 2010;1800(3):235–244.
    1. Suen DF, Norris KL, Youle RJ. Mitochondrial dynamics and apoptosis. Genes & Development. 2008;22(12):1577–1590.
    1. Dirks AJ, Leeuwenburgh C. The role of apoptosis in age-related skeletal muscle atrophy. Sports Medicine. 2005;35(6):473–483.
    1. Marzetti E, Leeuwenburgh C. Skeletal muscle apoptosis, sarcopenia and frailty at old age. Experimental Gerontology. 2006;41(12):1234–1238.
    1. Malmgren LT, Jones CE, Bookman LM. Muscle fiber and satellite cell apoptosis in the aging human thyroarytenoid muscle: a stereological study with confocal laser scanning microscopy. Otolaryngology. 2001;125(1):34–39.
    1. Koseki T, Inohara N, Chen S, Nunez G. ARC, an inhibitor of apoptosis expressed in skeletal muscle and heart that interacts selectively with caspases. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(9):5156–5160.
    1. Park SY, Kim HY, Lee JH, Yoon KH, Chang MS, Park SK. The age-dependent induction of apoptosis-inducing factor (AIF) in the human semitendinosus skeletal muscle. Cellular and Molecular Biology Letters. 2010;15(1):1–12.
    1. Tamilselvan J, Jayaraman G, Sivarajan K, Panneerselvam C. Age-dependent upregulation of p53 and cytochrome c release and susceptibility to apoptosis in skeletal muscle fiber of aged rats: role of carnitine and lipoic acid. Free Radical Biology and Medicine. 2007;43(12):1656–1669.
    1. Seo AY, Xu J, Servais S, et al. Mitochondrial iron accumulation with age and functional consequences. Aging Cell. 2008;7(5):706–716.
    1. Dirks A, Leeuwenburgh C. Apoptosis in skeletal muscle with aging. American Journal of Physiology Regulatory. 2002;282(2):519–527.
    1. Marzetti E, Wohlgemuth SE, Lees HA, Chung HY, Giovannini S, Leeuwenburgh C. Age-related activation of mitochondrial caspase-independent apoptotic signaling in rat gastrocnemius muscle. Mechanisms of Ageing and Development. 2008;129(9):542–549.
    1. Leeuwenburgh C, Gurley CM, Strotman BA, Dupont-Versteegden EE. Age-related differences in apoptosis with disuse atrophy in soleus muscle. American Journal of Physiology. 2005;288(5):R1288–R1296.
    1. Siu PM, Pistilli EE, Alway SE. Apoptotic responses to hindlimb suspension in gastrocnemius muscles from young adult and aged rats. American Journal of Physiology. 2005;289(4):R1015–R1026.
    1. Rice KM, Blough ER. Sarcopenia-related apoptosis is regulated differently in fast- and slow-twitch muscles of the aging F344/N × BN rat model. Mechanisms of Ageing and Development. 2006;127(8):670–679.
    1. McMillan EM, Quadrilatero J. Differential apoptosis-related protein expression, mitochondrial properties, proteolytic enzyme activity, and DNA fragmentation between skeletal muscles. American Journal of Physiology. 2011;300(3):R531–R543.
    1. Adhihetty PJ, Ljubicic V, Menzies KJ, Hood DA. Differential susceptibility of subsarcolemmal and intermyofibrillar mitochondria to apoptotic stimuli. American Journal of Physiology. 2005;289(4):C994–C1001.
    1. Holloszy JO. Biochemical adaptations in muscle. Effects of exercise on mitochondrial oxygen uptake and respiratory enzyme activity in skeletal muscle. Journal of Biological Chemistry. 1967;242(9):2278–2282.
    1. Holloszy JO. Adaptations of muscular tissue to training. Progress in Cardiovascular Diseases. 1976;18(6):445–458.
    1. Johannsen DL, DeLany JP, Frisard MI, et al. Physical activity in aging: comparison among young, aged, and nonagenarian individuals. Journal of Applied Physiology. 2008;105(2):495–501.
    1. Coggan AR, Spina RJ, King DS, et al. Histochemical and enzymatic comparison of the gastrocnemius muscle of young and elderly men and women. Journals of Gerontology. 1992;47(3):B71–B76.
    1. Cooper JM, Mann VM, Schapira AH. Analyses of mitochondrial respiratory chain function and mitochondrial DNA deletion in human skeletal muscle: effect of ageing. Journal of the Neurological Sciences. 1992;113(1):91–98.
    1. Short KR, Bigelow ML, Kahl J, et al. Decline in skeletal muscle mitochondrial function with aging in humans. Proceedings of the National Academy of Sciences. 2005;102(15):5618–5623.
    1. Jubrias SA, Esselman PC, Price LB, Cress ME, Conley KE. Large energetic adaptations of elderly muscle to resistance and endurance training. Journal of Applied Physiology. 2001;90(5):1663–1670.
    1. Menshikova EV, Ritov VB, Fairfull L, Ferrell RE, Kelley DE, Goodpaster BH. Effects of exercise on mitochondrial content and function in aging human skeletal muscle. The Journals of Gerontology A. 2006;61(6):534–540.
    1. Short KR. Mitochondrial ATP measurements. American Journal of Physiology Regulatory. 2004;287(1):R243–R245.
    1. Song W, Kwak HB, Lawler JM. Exercise training attenuates age-induced changes in apoptotic signaling in rat skeletal muscle. Antioxidants and Redox Signaling. 2006;8(3-4):517–528.
    1. Short KR, Vittone JL, Bigelow ML, Proctor DN, Nair KS. Age and aerobic exercise training effects on whole body and muscle protein metabolism. American Journal of Physiology. 2004;286(1):92–101.
    1. Leeuwenburgh C, Fiebig R, Chandwaney R, Ji Li Li Aging and exercise training in skeletal muscle: responses of glutathione and antioxidant enzyme systems. American Journal of Physiology. 1994;267(2, part 2):R439–R445.
    1. Parise G, Brose AN, Tarnopolsky MA. Resistance exercise training decreases oxidative damage to DNA and increases cytochrome oxidase activity in older adults. Experimental Gerontology. 2005;40(3):173–180.
    1. Parise G, Phillips SM, Kaczor JJ, Tarnopolsky MA. Antioxidant enzyme activity is up-regulated after unilateral resistance exercise training in older adults. Free Radical Biology and Medicine. 2005;39(2):289–295.
    1. Colman RJ, Anderson RM, Johnson SC, et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science. 2009;325(5937):201–204.
    1. Speakman JR, Mitchell SE. Caloric restriction. Molecular Aspects of Medicine. 2011;32(3):159–221.
    1. Merry BJ. Oxidative stress and mitochondrial function with aging—the effects of calorie restriction. Aging Cell. 2004;3(1):7–12.
    1. Zainal TA, Oberley TD, Allison DB, Szweda LI, Weindruch R. Caloric restriction of rhesus monkeys lowers oxidative damage in skeletal muscle. The FASEB Journal. 2000;14(12):1825–1836.
    1. Drew B, Dirks PA, Selman C, et al. Effects of aging and caloric restriction on mitochondrial energy production in gastrocnemius muscle and heart. American Journal of Physiology. 2003;284(2):R474–R480.
    1. Lass A, Sohal BH, Weindruch R, Forster MJ, Sohal RS. Caloric restriction prevents age-associated accrual of oxidative damage to mouse skeletal muscle mitochondria. Free Radical Biology and Medicine. 1998;25(9):1089–1097.
    1. Bevilacqua L, Ramsey JJ, Hagopian K, Weindruch R, Harper ME. Long-term caloric restriction increases UCP3 content but decreases proton leak and reactive oxygen species production in rat skeletal muscle mitochondria. American Journal of Physiology. 2005;289(3):E429–E438.
    1. Hamilton ML, Van Remmen H, Drake JA, et al. Does oxidative damage to DNA increase with age? Proceedings of the National Academy of Sciences of the United States of America. 2001;98(18):10469–10474.
    1. Usuki F, Yasutake A, Umehara F, Higuchi I. Beneficial effects of mild lifelong dietary restriction on skeletal muscle: prevention of age-related mitochondrial damage, morphological changes, and vulnerability to a chemical toxin. Acta Neuropathologica. 2004;108(1):1–9.
    1. Lee CM, Aspnes LE, Chung SS, Weindruch R, Aiken JM. Influences of caloric restriction on age-associated skeletal muscle fiber characteristics and mitochondrial changes in rats and mice. Annals of the New York Academy of Sciences. 1998;854:182–191.
    1. Civitarese AE, Carling S, Heilbronn LK, et al. Calorie restriction increases muscle mitochondrial biogenesis in healthy humans. PLoS Medicine. 2007;4(3):p. e76.
    1. Sreekumar R, Unnikrishnan J, Fu A, et al. Effects of caloric restriction on mitochondrial function and gene transcripts in rat muscle. American Journal of Physiology. 2002;283(1):E38–E43.
    1. Lopez-Lluch G, Hunt N, Jones B, et al. Calorie restriction induces mitochondrial biogenesis and bioenergetic efficiency. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(6):1768–1773.
    1. Lal SB, Ramsey JJ, Monemdjou S, Weindruch R, Harper ME. Effects of caloric restriction on skeletal muscle mitochondrial proton leak in aging rats. Journals of Gerontology A. 2001;56(3):B116–B122.
    1. Hancock CR, Han DH, Higashida K, Kim SH, Holloszy JO. Does calorie restriction induce mitochondrial biogenesis? A reevaluation. The FASEB Journal. 2011;25(2):785–791.
    1. Desai VG, Weindruch R, Hart RW, Feuers RJ. Influences of age and dietary restriction on gastrocnemius electron transport system activities in mice. Archives of Biochemistry and Biophysics. 1996;333(1):145–151.
    1. Bua E, McKiernan SH, Aiken JM. Calorie restriction limits the generation but not the progression of mitochondrial abnormalities in aging skeletal muscle. The FASEB Journal Biology. 2004;18(3):582–584.
    1. McKiernan SH, Colman RJ, Lopez M, et al. Caloric restriction delays aging-induced cellular phenotypes in rhesus monkey skeletal muscle. Experimental Gerontology. 2011;46(1):23–29.
    1. Gonzalez AA, Kumar R, Mulligan JD, Davis AJ, Weindruch R, Saupe KW. Metabolic adaptations to fasting and chronic caloric restriction in heart, muscle, and liver do not include changes in AMPK activity. American Journal of Physiology. 2004;287(5):E1032–E1037.
    1. Shinmura K, Tamaki K, Bolli R. Short-term caloric restriction improves ischemic tolerance independent of opening of ATP-sensitive K+ channels in both young and aged hearts. Journal of Molecular and Cellular Cardiology. 2005;39(2):285–296.
    1. Wang ZQ, Floyd ZE, Qin J, et al. Modulation of skeletal muscle insulin signaling with chronic caloric restriction in cynomolgus monkeys. Diabetes. 2009;58(7):1488–1498.
    1. Dirks AJ, Leeuwenburgh C. Aging and lifelong calorie restriction result in adaptations of skeletal muscle apoptosis repressor, apoptosis-inducing factor, X-linked inhibitor of apoptosis, caspase-3, and caspase-12. Free Radical Biology and Medicine. 2004;36(1):27–39.
    1. Marzetti E, Lawler JM, Hiona A, Manini T, Seo AY, Leeuwenburgh C. Modulation of age-induced apoptotic signaling and cellular remodeling by exercise and calorie restriction in skeletal muscle. Free Radical Biology and Medicine. 2008;44(2):160–168.
    1. Phillips T, Leeuwenburgh C. Muscle fiber specific apoptosis and TNF-alpha signaling in sarcopenia are attenuated by life-long calorie restriction. The FASEB Journal. 2005;19(6):668–670.
    1. Lee HY, Choi CS, Birkenfeld AL, et al. Targeted expression of catalase to mitochondria prevents age-associated reductions in mitochondrial function and insulin resistance. Cell Metabolism. 2010;12(6):668–674.
    1. Marzetti E, Lees HA, Wohlgemuth SE, Leeuwenburgh C. Sarcopenia of aging: underlying cellular mechanisms and protection by calorie restriction. BioFactors. 2009;35(1):28–35.
    1. Payne AM, Dodd SL, Leeuwenburgh C. Life-long calorie restriction in Fischer 344 rats attenuates age-related loss in skeletal muscle-specific force and reduces extracellular space. Journal of Applied Physiology. 2003;95(6):2554–2562.
    1. McKiernan SH, Bua E, McGorray J, Aiken J. Early-onset calorie restriction conserves fiber number in aging rat skeletal muscle. The FASEB Journal. 2004;18(3):580–581.
    1. Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α . Cell. 2006;127(6):1109–1122.
    1. Um JH, Park SJ, Kang H, et al. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes. 2010;59(3):554–563.
    1. Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–342.
    1. Timmers S, Konings E, Bilet L, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metabolism. 2011;14(5):612–622.
    1. Pacholec M, Bleasdale JE, Chrunyk B, et al. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. The Journal of Biological Chemistry. 2010;285(11):8340–8351.
    1. Barger JL, Kayo T, Vann JM, et al. A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS ONE. 2008;3(6):p. e2264.
    1. Ungvari Z, Sonntag WE, de Cabo R, Baur JA, Csiszar A. Mitochondrial protection by resveratrol. Exercise and Sport Sciences Reviews. 2011;39(3):128–132.
    1. Murase T, Haramizu S, Ota N, Hase T. Suppression of the aging-associated decline in physical performance by a combination of resveratrol intake and habitual exercise in senescence-accelerated mice. Biogerontology. 2009;10(4):423–434.
    1. Jackson JR, Ryan MJ, Hao Y, Alway SE. Mediation of endogenous antioxidant enzymes and apoptotic signaling by resveratrol following muscle disuse in the gastrocnemius muscles of young and old rats. American Journal of Physiology. 2010;299(6):R1572–R1581.
    1. Jackson JR, Ryan MJ, Alway SE. Long-term supplementation with resveratrol alleviates oxidative stress but does not attenuate sarcopenia in aged mice. The Journals of Gerontology A. 2011;66(7):751–764.

Source: PubMed

3
구독하다