A. Y. Bijlsma, C. G. Meskers, C. H. Ling, M. Narici, S. E. Kurrle et al., Defining sarcopenia: the impact of different diagnostic criteria on the prevalence of sarcopenia in a large middle aged cohort, Age (Dordr), vol.35, issue.3, pp.871-81, 2013.

P. Szulc, F. Munoz, F. Marchand, R. Chapurlat, and P. D. Delmas, Rapid loss of appendicular skeletal muscle mass is associated with higher all-cause mortality in older men: the prospective MINOS study, Am J Clin Nutr, issue.5, pp.1227-1263, 2010.

A. A. Sayer, S. M. Robinson, H. P. Patel, T. Shavlakadze, C. Cooper et al., New horizons in the pathogenesis, diagnosis and management of sarcopenia, Age Ageing, vol.42, issue.2, pp.145-150, 2013.

I. Janssen, S. B. Heymsfield, Z. M. Wang, and R. Ross, Skeletal muscle mass and distribution in 468 men and women aged 18-88 yr, J Appl Physiol, vol.89, issue.1, pp.81-89, 2000.

S. Carosio, M. G. Berardinelli, M. Aucello, and A. Musaro, Impact of ageing on muscle cell regeneration, Ageing Res Rev, vol.10, issue.1, pp.35-42, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00978647

M. V. Narici and N. Maffulli, Sarcopenia: characteristics, mechanisms and functional significance, Br Med Bull, vol.95, pp.139-59, 2010.

K. F. Petersen, D. Befroy, S. Dufour, J. Dziura, C. Ariyan et al., Mitochondrial dysfunction in the elderly: possible role in insulin resistance, Science, vol.300, issue.5622, pp.1140-1142, 2003.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke et al., Decline in skeletal muscle mitochondrial function with aging in humans, Proc Natl Acad Sci, vol.102, issue.15, pp.5618-5641, 2005.

C. K. Lee, R. G. Klopp, R. Weindruch, and T. A. Prolla, Gene expression profile of aging and its retardation by caloric restriction, Science, vol.285, issue.5432, pp.1390-1393, 1999.

M. A. Baraibar, M. Gueugneau, S. Duguez, G. Butler-browne, D. Bechet et al., Expression and modification proteomics during skeletal muscle ageing, Biogerontology, vol.14, issue.3, pp.339-52, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543981

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE, Proteomics, vol.19, issue.9, pp.2004-2024, 2005.

P. Doran, K. O'connell, J. Gannon, M. Kavanagh, and K. Ohlendieck, Opposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysis, Proteomics, vol.8, issue.2, pp.364-77, 2008.

J. Gannon, P. Doran, A. Kirwan, and K. Ohlendieck, Drastic increase of myosin light chain MLC-2 in senescent skeletal muscle indicates fast-to-slow fibre transition in sarcopenia of old age, Eur J Cell Biol, vol.88, issue.11, pp.685-700, 2009.

K. O'connell and K. Ohlendieck, Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle, Proteomics, vol.9, issue.24, pp.5509-5533, 2009.

C. Gelfi, A. Vigano, M. Ripamonti, A. Pontoglio, S. Begum et al., The human muscle proteome in aging, J Proteome Res, vol.5, issue.6, pp.1344-53, 2006.

H. Fischer, T. Gustafsson, C. J. Sundberg, J. Norrbom, M. Ekman et al., Fatty acid binding protein 4 in human skeletal muscle, Biochem Biophys Res Commun, vol.346, issue.1, pp.125-155, 2006.

J. J. Luiken, D. P. Koonen, W. A. Coumans, M. M. Pelsers, B. Binas et al., Long-chain fatty acid uptake by skeletal muscle is impaired in homozygous, but not heterozygous, heart-type-FABP null mice, Lipids, vol.38, issue.4, pp.491-497, 2003.

E. Erbay, V. R. Babaev, J. R. Mayers, L. Makowski, K. N. Charles et al., Reducing endoplasmic reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis, Nat Med, vol.15, issue.12, pp.1383-91, 2009.

S. G. Rhee, H. A. Woo, I. S. Kil, and S. H. Bae, Peroxiredoxin functions as a peroxidase and a regulator and sensor of local peroxides, J Biol Chem, vol.287, issue.7, pp.4403-4413, 2012.

M. M. Wilhelmus, P. G. Nijland, B. Drukarch, H. E. De-vries, and J. Van-horssen, Involvement and interplay of Parkin, PINK1, and DJ1 in neurodegenerative and neuroinflammatory disorders, Free Radic Biol Med, vol.53, issue.4, pp.983-92, 2012.

E. Andres-mateos, C. Perier, L. Zhang, B. Blanchard-fillion, T. M. Greco et al., DJ-1 gene deletion reveals that DJ-1 is an atypical peroxiredoxin-like peroxidase, Proc Natl Acad Sci, vol.104, issue.37, pp.14807-14819, 2007.

K. J. Thomas, M. K. Mccoy, J. Blackinton, A. Beilina, M. Van-der-brug et al., DJ-1 acts in parallel to the PINK1/parkin pathway to control mitochondrial function and autophagy, Hum Mol Genet, vol.20, issue.1, pp.40-50, 2011.

M. Altun, E. Edstrom, E. Spooner, A. Flores-moralez, E. Bergman et al., Iron load and redox stress in skeletal muscle of aged rats, Muscle Nerve, vol.36, issue.2, pp.223-256, 2007.

G. R. Budas, M. H. Disatnik, C. H. Chen, and D. Mochly-rosen, Activation of aldehyde dehydrogenase 2 (ALDH2) confers cardioprotection in protein kinase C epsilon (PKCvarepsilon) knockout mice, J Mol Cell Cardiol, vol.48, issue.4, pp.757-64, 2010.

O. A. Barski, S. M. Tipparaju, A. Bhatnagar, K. V. Ramana, and . Aldose-reductase, The aldo-keto reductase superfamily and its role in drug metabolism and detoxification, Biomol Concepts, vol.40, issue.4, pp.103-114, 2008.

S. Pandey, S. K. Srivastava, and K. V. Ramana, A potential therapeutic role for aldose reductase inhibitors in the treatment of endotoxin-related inflammatory diseases, Expert Opin Investig Drugs, vol.21, issue.3, pp.329-368, 2012.

A. Yoshida, A. Rzhetsky, L. C. Hsu, C. Chang, C. Forte-mcrobbie et al., Human glutamic-gamma-semialdehyde dehydrogenase. Kinetic mechanism, Eur J Biochem, vol.251, issue.3, pp.935-978, 1989.

H. H. Kampinga, J. Hageman, M. J. Vos, H. Kubota, R. M. Tanguay et al., Guidelines for the nomenclature of the human heat shock proteins, Cell Stress Chaperones, vol.14, issue.1, pp.105-116, 2009.

M. J. Vos, J. Hageman, S. Carra, and H. H. Kampinga, Structural and functional diversities between members of the human HSPB, HSPH, HSPA, and DNAJ chaperone families, Biochemistry, vol.47, issue.27, pp.7001-7012, 2008.

P. Doran, J. Gannon, K. O'connell, and K. Ohlendieck, Aging skeletal muscle shows a drastic increase in the small heat shock proteins alphaB-crystallin/HspB5 and cvHsp/HspB7, Eur J Cell Biol, vol.86, issue.10, pp.629-669, 2007.

E. V. Mymrikov, A. S. Seit-nebi, and N. B. Gusev, Large potentials of small heat shock proteins, Physiol Rev, vol.91, issue.4, pp.1123-59, 2011.

J. Acunzo, M. Katsogiannou, and P. Rocchi, Small heat shock proteins HSP27 (HspB1), alphaB-crystallin (HspB5) and HSP22 (HspB8) as regulators of cell death, Int J Biochem Cell Biol, vol.44, issue.10, pp.1622-1653, 2012.

Y. E. Kim, M. S. Hipp, A. Bracher, M. Hayer-hartl, and F. U. Hartl, Molecular chaperone functions in protein folding and proteostasis, Annu Rev Biochem, vol.82, pp.323-55, 2013.

S. M. Senf, S. L. Dodd, J. M. Mcclung, and A. R. Judge, Hsp70 overexpression inhibits NF-kappaB and Foxo3a transcriptional activities and prevents skeletal muscle atrophy, Faseb J, vol.22, issue.11, pp.3836-3881, 2008.

A. Mcardle, W. H. Dillmann, R. Mestril, J. A. Faulkner, and M. J. Jackson, Overexpression of HSP70 in mouse skeletal muscle protects against muscle damage and agerelated muscle dysfunction, Faseb J, vol.18, issue.2, pp.355-362, 2004.

E. H. Miyabara, J. L. Martin, T. M. Griffin, A. S. Moriscot, and R. Mestril, Overexpression of inducible 70-kDa heat shock protein in mouse attenuates skeletal muscle damage induced by cryolesioning, Am J Physiol Cell Physiol, vol.290, issue.4, pp.1128-1166, 2006.

F. E. Indig, J. J. Partridge, C. Von-kobbe, M. I. Aladjem, M. Latterich et al., Werner syndrome protein directly binds to the AAA ATPase p97/VCP in an ATP-dependent fashion, J Struct Biol, vol.146, issue.1-2, pp.251-260, 2004.

J. Kim, T. Lowe, and T. Hoppe, Protein quality control gets muscle into shape, Trends Cell Biol, vol.18, issue.6, pp.264-72, 2008.

Y. Ye, Diverse functions with a common regulator: ubiquitin takes command of an AAA ATPase, J Struct Biol, vol.156, issue.1, pp.29-40, 2006.

R. Krick, S. Bremer, E. Welter, P. Schlotterhose, Y. Muehe et al., Cdc48/p97 and Shp1/p47 regulate autophagosome biogenesis in concert with ubiquitin-like Atg8, J Cell Biol, vol.190, issue.6, pp.965-73, 2010.

A. Tanaka, M. M. Cleland, S. Xu, D. P. Narendra, D. F. Suen et al., Proteasome and p97 mediate mitophagy and degradation of mitofusins induced by Parkin, J Cell Biol, issue.7, pp.1367-80, 2010.

S. Bergink, W. Toussaint, M. S. Luijsterburg, C. Dinant, S. Alekseev et al., Recognition of DNA damage by XPC coincides with disruption of the XPC-RAD23 complex, J Cell Biol, vol.196, issue.6, pp.681-689, 2012.

S. L. Wade and D. T. Auble, The Rad23 ubiquitin receptor, the proteasome and functional specificity in transcriptional control, Transcription, vol.1, issue.1, pp.22-28, 2010.

F. Demarchi and C. Schneider, The calpain system as a modulator of stress/damage response, Cell Cycle, vol.6, issue.2, pp.136-144, 2007.

F. Cataldo, L. Y. Peche, E. Klaric, C. Brancolini, M. P. Myers et al., CAPNS1 regulates USP1 stability and maintenance of genome integrity, Mol Cell Biol, vol.33, issue.12, pp.2485-96, 2013.

R. L. Mellgren, W. Zhang, K. Miyake, and P. L. Mcneil, Calpain is required for the rapid, calcium-dependent repair of wounded plasma membrane, J Biol Chem, vol.282, issue.4, pp.2567-75, 2007.

B. P. Fairfax, F. O. Vannberg, J. Radhakrishnan, H. Hakonarson, B. J. Keating et al., An integrated expression phenotype mapping approach defines common variants in LEP, ALOX15 and CAPNS1 associated with induction of IL-6, Hum Mol Genet, vol.19, issue.4, pp.720-750, 2010.

Y. Li, J. Ma, H. Zhu, M. Singh, D. Hill et al., Targeted inhibition of calpain reduces myocardial hypertrophy and fibrosis in mouse models of type 1 diabetes, Diabetes, vol.60, issue.11, pp.2985-94, 2011.

A. Y. Bijlsma, C. G. Meskers, C. H. Ling, M. Narici, S. E. Kurrle et al., Proteomic study of calpain interacting proteins during skeletal muscle aging, Biochimie, vol.92, pp.1923-1933, 2010.

A. J. Cruz-jentoft, J. P. Baeyens, J. M. Bauer, Y. Boirie, T. Cederholm et al., Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People, vol.39, pp.412-423, 2010.

P. Szulc, R. Chapurlat, and P. D. Dumas, Accelerated bone loss, but not low periosteal expansion, is associated with higher all-cause mortality in older men, prospective MINOS study, J. Men's Health, vol.7, pp.199-210, 2010.

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Differential proteome analysis of aging in rat skeletal muscle, FASEB J, vol.19, pp.1143-1145, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680775

P. Doran, J. Gannon, K. O'connell, and K. Ohlendieck, Aging skeletal muscle shows a drastic decrease in the small heat shock proteins ?B-crystallin/HspB5 and cvHsp/HspB7, Eur. J. Cell Biol, vol.86, pp.629-640, 2007.

K. O'connell, J. Gannon, P. Doran, and K. Ohlendieck, Proteomic profiling reveals a severely perturbed protein expression pattern in aged skeletal muscle, Int. J. Mol. Med, vol.20, pp.145-153, 2007.

C. Gelfi, A. Vigano, M. Ripamonti, A. Pontoglio, S. Begum et al., The human muscle proteome in aging, J. Proteome Res, vol.5, pp.1344-1353, 2006.

L. Staunton, M. Zweyer, D. Swandulla, and K. Ohlendieck, Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle, Int. J. Mol. Med, vol.30, pp.723-733, 2012.

K. Short, M. L. Bigelow, J. Kahl, R. Singh, J. C. Coenen-schimke et al., Decline in skeletal muscle mitochondrial function with aging in humans, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.5618-5623, 2005.

J. Gannon, L. Staunton, K. O'connell, P. Doran, and K. Ohlendieck, Phosphoproteomic analysis of aged skeletal muscle, Int. J. Mol. Med, vol.22, pp.33-42, 2008.

K. O'connell, P. Doran, J. Gannon, and K. Ohlendieck, Lectinbased proteomic profiling of aged skeletal muscle: Decreased pyruvate kinase isozyme M1 exhibits drastically increased levels of N-glycosylation, Eur. J. Cell Biol, vol.87, pp.793-805, 2008.

J. Gannon and K. Ohlendieck, Subproteomic analysis of basic proteins in aged skeletal muscle following offgel pre-fractionation, Mol. Med. Reports, vol.5, pp.993-1000, 2012.

C. Brule, E. Dargelos, R. Diallo, A. Listrat, D. Bechet et al., Proteomic study of calpain interacting proteins during skeletal muscle aging, Biochimie, vol.90, pp.359-368, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02668382

M. A. Baraibar, M. Gueugneau, S. Duguez, G. Butler-browne, D. Bechet et al., Expression and modification proteomics during skeletal muscle ageing, Biogerontology, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543981

K. Hojlund, Z. Yi, H. Hwang, B. Bowen, N. Lefort et al., Characterization of the human skeletal muscle proteome by one-dimensional gel electrophoresis and HPLC-ESI-MS/MS, Mol. Cell.Proteomics, vol.7, pp.257-267, 2008.

A. F. Martin, M. Rabinowitz, R. Blough, G. Prior, Z. et al., Measurements of half-life of rat cardiac myosin heavy chain with leucyl-tRNA used as precursor pool, J. Biol. Chem, vol.252, pp.3422-3429, 1977.

G. Neti, S. M. Novak, V. F. Thompson, and D. E. Goll, Properties of easily releasable myofilaments: are they the first step in myofibrillar protein turnover?, Am. J. Cell Physiol, vol.296, pp.1383-1390, 2009.

T. Sayd, M. Morzel, C. Chambon, M. Franck, P. Figwer et al., Proteome analysis of the sarcoplasmic fraction of pig semimembranosus muscle: Implications on meat color development, J. Agricultural Food Chem, vol.24, pp.2732-2737, 2006.

U. K. Laemmli, Cleavage structural proteins during the assembly of the head of bacteriophage T4, Nature, vol.227, pp.680-685, 1970.

D. Szklarczyk, A. Franceschini, M. Kuhn, M. Simonovic, A. Roth et al., The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored, Nucleic Acid Res, vol.39, pp.561-568, 2011.

W. Zhu, J. W. Smith, and C. M. Huang, Mass spectrometry-based label-free quantitative proteomics, J. Med. Biotechnol, vol.840518, pp.1-6, 2010.

M. Altun, E. Edstrom, E. Spooner, A. Flores-moralez, E. Bergman et al., Iron load and redox stress in skeletal muscle of aged rats, Muscle Nerve, vol.36, pp.223-233, 2007.

P. Donoghue, L. Staunton, E. Mullen, G. Manning, and K. Ohlendieck, DIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult versus aged gastrocnemius tissue, J. Proteomics, vol.73, pp.1441-1453, 2010.

O. Pastoris, F. Boschi, M. Verri, P. Baiardi, G. Felzani et al., The effects of aging on enzyme activities and metabolite concentrations in skeletal muscle from sedentary male and female subjects, Exp. Gerontol, vol.35, pp.95-104, 2000.

A. Lombardi, E. Silvestri, F. Cioffi, R. Senese, A. Lanni et al., Defining the transcriptomic and proteomic profiles of rat ageing skeletal muscle by the use of a cDNA array, 2D-and Blue native-PAGE approach, J. Proteomics, vol.72, pp.708-721, 2009.

S. Papa, Mitochondrial oxidative phosphorylation changes in the life span. Molecular aspects and physiopathological implications, Biochim. Biophys. Acta, vol.87, p.105, 1996.

A. K. Samland and G. A. Sprenger, Transaldolase: From biochemistry to human disease, Int. J. Biochem. Cell Biol, vol.41, pp.1482-1494, 2009.

S. Violante, L. Ijlst, J. Ruiter, J. Koster, H. Van-lenthe et al., Substrate specificity of human carnitine acetyltransferase: Implications for fatty acid and branched-chain amino acid metabolism, Biochim. Biophys. Acta, vol.1832, pp.773-779, 2013.

D. M. Muoio, R. C. Noland, J. P. Kovalic, S. E. Seiler, M. N. Davies et al., Muscle specific deletion of carnitine acetyltransferase compromises glucose tolerance and metabolic flexibility, Cell Metabolism, vol.15, pp.764-777, 2012.

R. L. Smathers and D. R. Petersen, The human fatty acid-binding protein family: Evolutionary divergences and functions, Human Genomics, vol.5, pp.170-191, 2011.

H. Fisher, T. Gustafsson, C. J. Sundberg, J. Norrbom, M. Ekman et al., Fatty acid binding protein 4 in human skeletal muscle, Biochem. Biophys. Res. Commun, vol.346, pp.125-130, 2006.

A. A. Vandervoort, Aging of the human neuromuscular system, Muscle Nerve, vol.25, pp.17-25, 2002.

P. Doran, K. O'connell, J. Gannon, M. Kavanagh, and K. Ohlendieck, Opposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysis, Proteomics, vol.8, pp.364-377, 2008.

A. Belgrano, L. Rakicevic, L. Mittempergher, S. Campanaro, V. C. Martinelli et al., Multi-tasking role of the mechanosensing protein Ankrd2 in the signaling network of striated muscle, vol.6, p.25519, 2011.

A. Pallavicini, S. Kojic, C. Bean, M. Vainzof, M. Salamon et al., Characterization of human skeletal muscle Ankrd2, Biochem. Biophys. Res. Commun, vol.285, pp.378-386, 2001.

Y. Tsukamoto, T. Senda, T. Nakano, C. Nakada, T. Hida et al., Arpp, a new homolog of carp, is preferentially expressed in type 1 skeletal muscle fibers and is markedly induced by denervation, Lab. Invest, vol.82, pp.645-655, 2002.

E. Edstrom, M. Altun, E. Bergman, H. Johnson, S. Kullberg et al., Factors contributing to neuromuscular impairment and sarcopenia during aging, Physiol. Behav, vol.92, pp.129-135, 2007.

M. Krugern and W. A. Linke, The giant protein titin: A regulatory node that integrates myocyte signaling pathways, J. Biol. Chem, vol.286, pp.9905-9912, 2011.

B. Dahlmann, M. Rutschmann, and H. Reinauer, Effect of starvation or treatment with corticosterone on the amount of easily releasable myofilaments in rat skeletal muscles, Biochem. J, vol.234, pp.659-664, 1986.

A. G. Grieve, S. E. Moss, and M. J. Hayed, Annexin A2 at the interface of actin and membrane dynamics: A focus on its roles in endocytosis and cell polarization, Int. J. Cell Biol, 2012.

D. O. Furst, L. G. Goldfarb, R. A. Kley, M. Vorgerd, M. Olive et al., Filamin C-related myopathies: pathology and mech, 2013.

K. G. Alberti, R. H. Eckel, S. M. Grundy, P. Z. Zimmet, J. I. Cleeman et al., Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute, American Heart Association; World Heart, vol.120, pp.1640-1645, 2009.

J. L. Andersen, Muscle fibre type adaptation in the elderly human muscle, Scand J Med Sci Sports, vol.13, pp.40-47, 2003.

J. L. Andersen, G. Terzis, and K. A. , Increase in the degree of coexpression of myosin heavy chain isoforms in skeletal muscle fibers of the very old, Muscle & nerve, vol.22, pp.449-454, 1999.

J. C. Barthelemy, V. Pichot, V. Dauphinot, C. S. Laurent, B. Garcin et al., Autonomic nervous system activity and decline as prognostic indicators of cardiovascular and cerebrovascular events: the 'PROOF' Study. Study design and population sample, Associations with sleep-related breathing disorders: the 'SYNAPSE' Study, vol.29, pp.18-28, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-01248812

M. Borkman, L. H. Storlien, D. A. Pan, A. B. Jenkins, D. J. Chisholm et al., The relation between insulin sensitivity and the fatty-acid composition of skeletal-muscle phospholipids, N Engl J Med, vol.328, pp.238-244, 1993.

M. J. Campbell, A. J. Mccomas, and F. Petito, Physiological changes in ageing muscles, J Neurol Neurosurg Psychiatry, vol.36, pp.174-182, 1973.

A. R. Coggan, R. J. Spina, D. S. King, M. A. Rogers, M. Brown et al., Skeletal muscle adaptations to endurance training in 60-to 70-yr-old men and women, J Appl Physiol, vol.72, pp.1780-1786, 1992.

J. D. Crane, M. C. Devries, A. Safdar, M. J. Hamadeh, and T. Ma, The effect of aging on human skeletal muscle mitochondrial and intramyocellular lipid ultrastructure, J Gerontol A Biol Sci Med Sci, vol.65, pp.119-128, 2010.

M. G. Cree, B. R. Newcomer, C. S. Katsanos, M. Sheffield-moore, D. Chinkes et al., Intramuscular and liver triglycerides are increased in the elderly, The Journal of clinical endocrinology and metabolism, vol.89, pp.3864-3871, 2004.

G. D'antona, M. A. Pellegrino, R. Adami, R. Rossi, C. N. Carlizzi et al., The effect of ageing and immobilization on structure and function of human skeletal muscle fibres, The Journal of physiology, vol.552, pp.499-511, 2003.

H. C. Dreyer, C. E. Blanco, F. R. Sattler, and E. T. Schroeder, Satellite cell numbers in young and older men 24 hours after eccentric exercise, Muscle & nerve, vol.33, pp.242-253, 2006.

E. S. Ford, C. Li, and G. Zhao, Prevalence and correlates of metabolic syndrome based on a harmonious definition among adults in the US, J Diabetes, vol.2, pp.180-193, 2010.

W. R. Frontera, V. A. Hughes, R. A. Fielding, M. A. Fiatarone, W. J. Evans et al., Aging of skeletal muscle: a 12-yr longitudinal study, J Appl Physiol, vol.88, pp.1321-1326, 2000.

M. Garet, J. C. Barthelemy, F. Degache, F. Costes, A. Da-costa et al., A questionnaire-based assessment of daily physical activity in heart failure, European journal of heart failure, vol.6, pp.577-584, 2004.

T. P. Gavin, H. W. Stallings, K. A. Zwetsloot, L. M. Westerkamp, N. A. Ryan et al., Lower capillary density but no difference in VEGF expression in obese vs. lean young skeletal muscle in humans, J Appl Physiol, vol.98, pp.315-321, 2005.

B. H. Goodpaster, J. He, S. Watkins, and K. De, Skeletal muscle lipid content and insulin resistance: evidence for a paradox in endurance-trained athletes, The Journal of clinical endocrinology and metabolism, vol.86, pp.5755-5761, 2001.

S. M. Grundy, Metabolic syndrome: a multiplex cardiovascular risk factor, The Journal of clinical endocrinology and metabolism, vol.92, pp.399-404, 2007.

J. He, S. Watkins, and K. De, Skeletal muscle lipid content and oxidative enzyme activity in relation to muscle fiber type in type 2 diabetes and obesity, Diabetes, vol.50, pp.817-823, 2001.

J. W. Helge and D. F. , Effect of training on muscle triacylglycerol and structural lipids: a relation to insulin sensitivity?, Diabetes, vol.52, pp.1881-1887, 2003.

B. Isomaa, M. Henricsson, M. Lehto, C. Forsblom, S. Karanko et al., Chronic diabetic complications in patients with MODY3 diabetes, Diabetologia, vol.41, pp.467-473, 1998.

D. L. Johannsen, K. E. Conley, S. Bajpeyi, M. Punyanitya, D. Gallagher 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, vol.97, pp.242-250, 2012.

P. R. Jones and J. Pearson, Anthropometric determination of leg fat and muscle plus bone volumes in young male and female adults, The Journal of physiology, vol.204, pp.63-66, 1969.

J. S. Kim, K. W. Hinchcliff, M. Yamaguchi, L. A. Beard, C. D. Markert et al., Age-related changes in metabolic properties of equine skeletal muscle associated with muscle plasticity, Vet J, vol.169, pp.397-403, 2005.

S. Kirkeby and C. Garbarsch, Aging affects different human muscles in various ways. An image analysis of the histomorphometric characteristics of fiber types in human masseter and vastus lateralis muscles from young adults and the very old, Histology and histopathology, vol.15, pp.61-71, 2000.

D. J. Kosek, J. S. Kim, J. K. Petrella, J. M. Cross, and M. M. Bamman, Efficacy of 3 days/wk resistance training on myofiber hypertrophy and myogenic mechanisms in young vs. older adults, J Appl Physiol, vol.101, pp.531-544, 2006.

T. Lang, T. Streeper, P. Cawthon, K. Baldwin, D. R. Taaffe et al., Sarcopenia: etiology, clinical consequences, intervention, and assessment, Osteoporos Int, vol.21, pp.543-559, 2010.

L. Larsson, X. Li, and F. Wr, Effects of aging on shortening velocity and myosin isoform composition in single human skeletal muscle cells, The American journal of physiology, vol.272, pp.638-649, 1997.

W. S. Lee, W. H. Cheung, L. Qin, N. Tang, and K. S. Leung, Age-associated decrease of type IIA/B human skeletal muscle fibers, Clin Orthop Relat Res, vol.450, pp.231-237, 2006.

J. Lexell and D. Dy, The occurrence of fibre-type grouping in healthy human muscle: a quantitative study of cross-sections of whole vastus lateralis from men between 15 and 83 years, Acta Neuropathol, vol.81, pp.377-381, 1991.

J. Lexell, C. C. Taylor, and M. Sjostrom, 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, vol.84, pp.275-294, 1988.

P. Malenfant, D. R. Joanisse, R. Theriault, B. H. Goodpaster, D. E. Kelley et al., Fat content in individual muscle fibers of lean and obese subjects, Int J Obes Relat Metab Disord, vol.25, pp.1316-1321, 2001.

B. Meunier, B. Picard, T. Astruc, and R. Labas, Development of image analysis tool for the classification of muscle fibre type using immunohistochemical staining, Histochem Cell Biol, vol.134, pp.307-317, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02660249

C. M. Peterson, D. L. Johannsen, and R. E. , Skeletal muscle mitochondria and aging: a review, J Aging Res, 2012.

R. Rezzani, L. Rodella, G. Corsetti, and R. Bianchi, Does methylene blue protect the kidney tissues from damage induced by ciclosporin A treatment?, Nephron, vol.89, pp.329-336, 2001.

A. P. Russell, G. Gastaldi, E. Bobbioni-harsch, P. Arboit, C. Gobelet et al., Lipid peroxidation in skeletal muscle of obese as compared to endurance-trained humans: a case of good vs. bad lipids?, FEBS letters, vol.551, pp.104-106, 2003.

T. Sato, H. Akatsuka, K. Kito, Y. Tokoro, H. Tauchi et al., Age changes in size and number of muscle fibers in human minor pectoral muscle, Mech Ageing Dev, vol.28, pp.99-109, 1984.

S. Schiaffino, Fibre types in skeletal muscle: a personal account, Acta Physiol (Oxf), vol.199, pp.451-463, 2010.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke et al., Decline in skeletal muscle mitochondrial function with aging in humans, Proc Natl Acad Sci U S A, vol.102, pp.5618-5623, 2005.

J. A. Simoneau, J. H. Veerkamp, L. P. Turcotte, and K. De, Markers of capacity to utilize fatty acids in human skeletal muscle: relation to insulin resistance and obesity and effects of weight loss, Faseb J, vol.13, pp.2051-2060, 1999.

M. Trevisan, J. Liu, F. B. Bahsas, and M. A. , Syndrome X and mortality: a population-based study. Risk Factor and Life Expectancy Research Group, Am J Epidemiol, vol.148, pp.958-966, 1998.

L. B. Verdijk, R. Koopman, G. Schaart, K. Meijer, H. H. Savelberg et al., Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly, Am J Physiol Endocrinol Metab, vol.292, pp.151-157, 2007.

J. Verney, F. Kadi, M. A. Saafi, K. Piehl-aulin, and D. C. , Combined lower body endurance and upper body resistance training improves performance and health parameters in healthy active elderly, Eur J Appl Physiol, vol.97, pp.288-297, 2006.

M. J. Watt and A. J. Hoy, Lipid metabolism in skeletal muscle: generation of adaptive and maladaptive intracellular signals for cellular function, Am J Physiol Endocrinol Metab, vol.302, pp.1315-1328, 2012.

E. J. Yang, S. Lim, J. Y. Lim, K. W. Kim, H. C. Jang et al., Association between muscle strength and metabolic syndrome in older Korean men and women: the Korean Longitudinal Study on Health and Aging, Metabolism, vol.61, pp.317-324, 2012.

K. G. Alberti, R. H. Eckel, S. M. Grundy, P. Z. Zimmet, J. I. Cleeman et al., Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute, American Heart Association; World Heart, vol.120, pp.1640-1645, 2009.

J. L. Andersen, Muscle fibre type adaptation in the elderly human muscle. Scandinavian journal of medicine & science in sports, vol.13, pp.40-47, 2003.

K. Aukland and R. K. Reed, Interstitial-lymphatic mechanisms in the control of extracellular fluid volume, Physiological reviews, vol.73, pp.1-78, 1993.

J. A. Babraj, D. J. Cuthbertson, K. Smith, H. Langberg, B. Miller et al., Collagen synthesis in human musculoskeletal tissues and skin, American journal of physiology. Endocrinology and metabolism, vol.289, pp.864-869, 2005.

J. C. Barthelemy, V. Pichot, V. Dauphinot, C. S. Laurent, B. Garcin et al., Autonomic nervous system activity and decline as prognostic indicators of cardiovascular and cerebrovascular events: the 'PROOF' Study. Study design and population sample, Neuroepidemiology, vol.29, pp.18-28, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-01248812

S. E. Bearden, Effect of aging on the structure and function of skeletal muscle microvascular networks, Microcirculation, vol.13, pp.279-288, 2006.

C. Cardillo, U. Campia, C. M. Kilcoyne, M. B. Bryant, and J. A. Panza, Improved endotheliumdependent vasodilation after blockade of endothelin receptors in patients with essential hypertension, Circulation, vol.105, pp.452-456, 2002.

N. Charifi, F. Kadi, L. Feasson, F. Costes, A. Geyssant et al., Enhancement of microvessel tortuosity in the vastus lateralis muscle of old men in response to endurance training, The Journal of physiology, vol.554, pp.559-569, 2004.

M. Charles, N. Charifi, J. Verney, V. Pichot, L. Feasson et al., Effect of endurance training on muscle microvascular filtration capacity and vascular bed morphometry in the elderly, Acta Physiol (Oxf), vol.187, pp.399-406, 2006.

I. I. Chen, R. L. Prewitt, and R. F. Dowell, Microvascular rarefaction in spontaneously hypertensive rat cremaster muscle. The American journal of physiology, vol.241, pp.306-310, 1981.

A. R. Coggan, R. J. Spina, D. S. King, M. A. Rogers, M. Brown et al., Histochemical and enzymatic comparison of the gastrocnemius muscle of young and elderly men and women, Journal of gerontology, vol.47, pp.71-76, 1992.

A. R. Coggan, R. J. Spina, D. S. King, M. A. Rogers, M. Brown et al., Skeletal muscle adaptations to endurance training in 60-to 70-yr-old men and women, J Appl Physiol, vol.72, pp.1780-1786, 1985.

A. N. Croley, K. A. Zwetsloot, L. M. Westerkamp, N. A. Ryan, A. M. Pendergast et al., Lower capillarization, VEGF protein, and VEGF mRNA response to acute exercise in the vastus lateralis muscle of aged vs. young women, J Appl Physiol, vol.99, pp.1872-1879, 1985.

F. Feihl, L. Liaudet, B. I. Levy, and B. Waeber, Hypertension and microvascular remodelling, Cardiovascular research, vol.78, pp.274-285, 2008.

F. Feihl, L. Liaudet, B. Waeber, and B. I. Levy, Hypertension: a disease of the microcirculation? Hypertension, vol.48, pp.1012-1017, 2006.

E. S. Ford, C. Li, and G. Zhao, Prevalence and correlates of metabolic syndrome based on a harmonious definition among adults in the US, Journal of diabetes, vol.2, pp.180-193, 2010.

W. R. Frontera, V. A. Hughes, R. A. Fielding, M. A. Fiatarone, W. J. Evans et al., Aging of skeletal muscle: a 12-yr longitudinal study, J Appl Physiol, vol.88, pp.1321-1326, 1985.

Y. Gao, T. Y. Kostrominova, J. A. Faulkner, and A. S. Wineman, Age-related changes in the mechanical properties of the epimysium in skeletal muscles of rats, Journal of biomechanics, vol.41, pp.465-469, 2008.

M. Garet, J. C. Barthelemy, F. Degache, F. Costes, A. Da-costa et al., A questionnaire-based assessment of daily physical activity in heart failure, European journal of heart failure, vol.6, pp.577-584, 2004.

S. M. Grundy, Metabolic syndrome: a multiplex cardiovascular risk factor, J Clin Endocrinol Metab, vol.92, pp.399-404, 2007.

A. H. Hansen, J. J. Nielsen, B. Saltin, and Y. Hellsten, Exercise training normalizes skeletal muscle vascular endothelial growth factor levels in patients with essential hypertension, Journal of hypertension, vol.28, pp.1176-1185, 2010.

A. H. Hansen, M. Nyberg, J. Bangsbo, B. Saltin, and Y. Hellsten, Exercise training alters the balance between vasoactive compounds in skeletal muscle of individuals with essential hypertension, Hypertension, vol.58, pp.943-949, 2011.

B. A. Harris, The influence of endurance and resistance exercise on muscle capillarization in the elderly: a review, Acta physiologica Scandinavica, vol.185, pp.89-97, 2005.

J. M. Haus, J. A. Carrithers, S. W. Trappe, and T. A. Trappe, Collagen, cross-linking, and advanced glycation end products in aging human skeletal muscle, J Appl Physiol, vol.103, pp.2068-2076, 1985.

A. Hedman, R. Reneland, and H. O. Lithell, Alterations in skeletal muscle morphology in glucose-tolerant elderly hypertensive men: relationship to development of hypertension and heart rate, Journal of hypertension, vol.18, pp.559-565, 2000.

H. A. Henrich, W. Romen, W. Heimgartner, E. Hartung, and F. Baumer, Capillary rarefaction characteristic of the skeletal muscle of hypertensive patients, Klinische Wochenschrift, vol.66, pp.54-60, 1988.

R. T. Hepple, A new measurement of tissue capillarity: the capillary-to-fibre perimeter exchange index. Canadian journal of applied physiology = Revue canadienne de physiologie appliquee, vol.22, pp.11-22, 1997.

R. T. Hepple, S. L. Mackinnon, J. M. Goodman, S. G. Thomas, and M. J. Plyley, Resistance and aerobic training in older men: effects on VO2peak and the capillary supply to skeletal muscle, J Appl Physiol, vol.82, pp.1305-1310, 1985.

R. T. Hepple and O. Mathieu-costello, Estimating the size of the capillary-to-fiber interface in skeletal muscle: a comparison of methods, J Appl Physiol, vol.91, pp.2150-2156, 1985.

N. Hernandez, S. H. Torres, H. J. Finol, and O. Vera, Capillary changes in skeletal muscle of patients with essential hypertension. The Anatomical record, vol.256, pp.425-432, 1999.

J. Janacek, E. Cvetko, L. Kubinova, L. Travnik, and I. Erzen, A novel method for evaluation of capillarity in human skeletal muscles from confocal 3D images. Microvascular research, vol.81, pp.231-238, 2011.

P. R. Jones and J. Pearson, Anthropometric determination of leg fat and muscle plus bone volumes in young male and female adults, The Journal of physiology, vol.204, pp.63-66, 1969.

A. M. Jonk, A. J. Houben, R. T. De-jongh, E. H. Serne, N. C. Schaper et al., , 2007.

, Microvascular dysfunction in obesity: a potential mechanism in the pathogenesis of obesity-associated insulin resistance and hypertension, Physiology (Bethesda), vol.22, pp.252-260

B. Meunier, B. Picard, T. Astruc, and R. Labas, Development of image analysis tool for the classification of muscle fibre type using immunohistochemical staining, Histochem Cell Biol, vol.134, pp.307-317, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02660249

V. M. Monnier, G. T. Mustata, K. L. Biemel, O. Reihl, M. O. Lederer et al., Cross-linking of the extracellular matrix by the maillard reaction in aging and diabetes: an update on "a puzzle nearing resolution, Annals of the New York Academy of Sciences, vol.1043, pp.533-544, 2005.

B. J. Nicklas, I. Leng, O. Delbono, D. W. Kitzman, A. P. Marsh et al., Relationship of physical function to vastus lateralis capillary density and metabolic enzyme activity in elderly men and women, Aging clinical and experimental research, vol.20, pp.302-309, 2008.

J. A. Panza, C. E. Garcia, C. M. Kilcoyne, A. A. Quyyumi, and R. O. Cannon, Impaired endothelium-dependent vasodilation in patients with essential hypertension. Evidence that nitric oxide abnormality is not localized to a single signal transduction pathway, Circulation, vol.91, pp.1732-1738, 1995.

C. M. Peterson, D. L. Johannsen, and E. Ravussin, Skeletal muscle mitochondria and aging: a review, Journal of aging research, 2012.

D. C. Poole and O. Mathieu-costello, Relationship between fiber capillarization and mitochondrial volume density in control and trained rat soleus and plantaris muscles, Microcirculation, vol.3, pp.175-186, 1996.

R. L. Prewitt, I. I. Chen, and R. Dowell, Development of microvascular rarefaction in the spontaneously hypertensive rat. The American journal of physiology, vol.243, pp.243-251, 1982.

D. N. Proctor, W. E. Sinning, J. M. Walro, G. C. Sieck, and P. W. Lemon, Oxidative capacity of human muscle fiber types: effects of age and training status, J Appl Physiol, vol.78, pp.2033-2038, 1985.

P. P. Purslow, The structure and functional significance of variations in the connective tissue within muscle. Comparative biochemistry and physiology. Part A, Molecular & integrative physiology, vol.133, pp.947-966, 2002.

K. S. Ramaswamy, M. L. Palmer, J. H. Van-der-meulen, A. Renoux, T. Y. Kostrominova et al., Lateral transmission of force is impaired in skeletal muscles of dystrophic mice and very old rats, The Journal of physiology, vol.589, pp.1195-1208, 2011.

R. K. Reed and K. Rubin, Transcapillary exchange: role and importance of the interstitial fluid pressure and the extracellular matrix, Cardiovascular research, vol.87, pp.211-217, 2010.

R. Rezzani, L. Rodella, G. Corsetti, and R. Bianchi, Does methylene blue protect the kidney tissues from damage induced by ciclosporin A treatment?, Nephron, vol.89, pp.329-336, 2001.

N. A. Ryan, K. A. Zwetsloot, L. M. Westerkamp, R. C. Hickner, W. E. Pofahl et al., Lower skeletal muscle capillarization and VEGF expression in aged vs. young men, J Appl Physiol, vol.100, pp.178-185, 1985.

E. H. Serne, R. T. De-jongh, E. C. Eringa, R. G. Ij, and C. D. Stehouwer, Microvascular dysfunction: a potential pathophysiological role in the metabolic syndrome, Hypertension, vol.50, pp.204-211, 2007.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke 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, vol.102, pp.5618-5623, 2005.

S. M. Sullivan and R. N. Pittman, Relationship between mitochondrial volume density and capillarity in hamster muscles. The American journal of physiology, vol.252, pp.149-155, 1987.

J. Verney, F. Kadi, M. A. Saafi, K. Piehl-aulin, and C. Denis, Combined lower body endurance and upper body resistance training improves performance and health parameters in healthy active elderly, Eur J Appl Physiol, vol.97, pp.288-297, 2006.

L. Vincent, L. Feasson, S. Oyono-enguelle, V. Banimbek, C. Denis et al., Remodeling of skeletal muscle microvasculature in sickle cell trait and alpha-thalassemia, American journal of physiology. Heart and circulatory physiology, vol.298, pp.375-384, 2010.

H. Wang, A. Listrat, B. Meunier, M. Gueugneau, C. Coudy-gandilhon et al., Apoptosis in capillary endothelial cells in ageing skeletal muscle, Aging cell, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543483

J. Lexell, C. C. Taylor, and M. Sjostrom, 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, vol.84, issue.2-3, pp.275-94, 1988.

J. Lexell, Human aging, muscle mass, and fiber type composition, J Gerontol A Biol Sci Med Sci, vol.50, pp.11-17, 1995.

G. Goldspink, Age-related loss of muscle mass and strength, J Aging Res, p.158279, 2012.

E. S. Ford, W. H. Giles, and W. H. Dietz, Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey, Jama, vol.287, issue.3, pp.356-365, 2002.

K. G. Alberti, R. H. Eckel, S. M. Grundy, P. Z. Zimmet, J. I. Cleeman et al., Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute, American Heart Association; World Heart, vol.120, issue.16, pp.1640-1645, 2009.

J. L. Andersen, Muscle fibre type adaptation in the elderly human muscle, Scand J Med Sci Sports, vol.13, issue.1, pp.40-47, 2003.

L. B. Verdijk, R. Koopman, G. Schaart, K. Meijer, H. H. Savelberg et al., Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly, Am J Physiol Endocrinol Metab, vol.292, issue.1, pp.151-158, 2007.

M. J. Campbell, A. J. Mccomas, and F. Petito, Physiological changes in ageing muscles

, J Neurol Neurosurg Psychiatry, vol.36, issue.2, pp.174-82, 1973.

C. J. Mcneil, T. J. Doherty, D. W. Stashuk, and C. L. Rice, Motor unit number estimates in the tibialis anterior muscle of young, old, and very old men, Muscle Nerve, vol.31, issue.4, pp.461-468, 2005.

I. M. Conboy, M. J. Conboy, A. J. Wagers, E. R. Girma, I. L. Weissman et al., Rejuvenation of aged progenitor cells by exposure to a young systemic environment, Nature, vol.433, issue.7027, pp.760-764, 2005.

M. Bencze, E. Negroni, D. Vallese, H. Yacoub-youssef, S. Chaouch et al., Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation, Mol Ther, vol.20, issue.11, pp.2168-79, 2012.

P. P. Purslow, The structure and functional significance of variations in the connective tissue within muscle, Comp Biochem Physiol A Mol Integr Physiol, vol.133, issue.4, pp.947-66, 2002.

Y. Gao, T. Y. Kostrominova, J. A. Faulkner, and A. S. Wineman, Age-related changes in the mechanical properties of the epimysium in skeletal muscles of rats, J Biomech, vol.41, issue.2, pp.465-474, 2008.

K. S. Ramaswamy, M. L. Palmer, J. H. Van-der-meulen, A. Renoux, T. Y. Kostrominova et al., Lateral transmission of force is impaired in skeletal muscles of dystrophic mice and very old rats, J Physiol, vol.589, pp.1195-208, 2011.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke et al., Decline in skeletal muscle mitochondrial function with aging in humans, Proc Natl Acad Sci, vol.102, issue.15, pp.5618-5641, 2005.

C. M. Peterson, D. L. Johannsen, and E. Ravussin, Skeletal muscle mitochondria and aging: a review, J Aging Res, 2012.

J. D. Crane, M. C. Devries, A. Safdar, M. J. Hamadeh, and M. A. Tarnopolsky, The effect of aging on human skeletal muscle mitochondrial and intramyocellular lipid ultrastructure, J Gerontol A Biol Sci Med Sci, vol.65, issue.2, pp.119-147, 2010.

M. J. Watt and A. J. Hoy, Lipid metabolism in skeletal muscle: generation of adaptive and maladaptive intracellular signals for cellular function, Am J Physiol Endocrinol Metab, vol.302, issue.11, pp.1315-1343, 2012.

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE, Proteomics, vol.19, issue.9, pp.2004-2024, 2005.

J. Gannon, P. Doran, A. Kirwan, and K. Ohlendieck, Drastic increase of myosin light chain MLC-2 in senescent skeletal muscle indicates fast-to-slow fibre transition in sarcopenia of old age, Eur J Cell Biol, vol.88, issue.11, pp.685-700, 2009.

K. O'connell and K. Ohlendieck, Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle, Proteomics, vol.9, issue.24, pp.5509-5533, 2009.

C. Gelfi, A. Vigano, M. Ripamonti, A. Pontoglio, S. Begum et al., The human muscle proteome in aging, J Proteome Res, vol.5, issue.6, pp.1344-53, 2006.

L. Staunton, M. Zweyer, D. Swandulla, and K. Ohlendieck, Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle, Int J Mol Med, vol.30, issue.4, pp.723-756, 2012.

J. C. Barthelemy, V. Pichot, V. Dauphinot, S. Celle, B. Laurent et al., Autonomic nervous system activity and decline as prognostic indicators of cardiovascular and cerebrovascular events: the 'PROOF' Study. Study design and population sample, Neuroepidemiology, vol.29, issue.1-2, pp.18-28, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-01248812

S. M. Grundy, Metabolic syndrome: a multiplex cardiovascular risk factor, J Clin Endocrinol Metab, vol.92, issue.2, pp.399-404, 2007.

T. Sayd, C. Chambon, E. Laville, B. Lebret, H. Gilbert et al., Early postmortem sarcoplasmic proteome of porcine muscle related to lipid oxidation in aged and cooked meat, Food Chem, vol.135, issue.4, pp.2238-2282
URL : https://hal.archives-ouvertes.fr/hal-01267820

R. Gilbert, J. A. Cohen, S. Pardo, A. Basu, and D. A. Fischman, Identification of the Aband localization domain of myosin binding proteins C and H (MyBP-C, MyBP-H) in skeletal muscle, J Cell Sci, vol.112, pp.69-79, 1999.

B. Wei and J. P. Jin, Troponin T isoforms and posttranscriptional modifications: Evolution, regulation and function, Arch Biochem Biophys, vol.505, issue.2, pp.144-54, 2011.

A. V. Gomes, J. D. Potter, and D. Szczesna-cordary, The role of troponins in muscle contraction, IUBMB Life, vol.54, issue.6, pp.323-356, 2002.

P. K. Luther, The vertebrate muscle Z-disc: sarcomere anchor for structure and signalling, J Muscle Res Cell Motil, vol.30, issue.5-6, pp.171-85, 2009.

I. Papa, C. Astier, O. Kwiatek, F. Raynaud, C. Bonnal et al., Alpha actinin-CapZ, an anchoring complex for thin filaments in Z-line, J Muscle Res Cell Motil, vol.20, issue.2, pp.187-97, 1999.

C. T. Pappas, N. Bhattacharya, J. A. Cooper, and C. C. Gregorio, Nebulin interacts with CapZ and regulates thin filament architecture within the Z-disc, Mol Biol Cell, vol.19, issue.5, pp.1837-1884, 2008.

P. B. Agrawal, M. Joshi, T. Savic, Z. Chen, and A. H. Beggs, Normal myofibrillar development followed by progressive sarcomeric disruption with actin accumulations in a mouse Cfl2 knockout demonstrates requirement of cofilin-2 for muscle maintenance, Hum Mol Genet, vol.21, issue.10, pp.2341-56, 2012.

A. Pallavicini, S. Kojic, C. Bean, M. Vainzof, M. Salamon et al., Characterization of human skeletal muscle Ankrd2, Biochem Biophys Res Commun, vol.285, issue.2, pp.378-86, 2001.

C. Hayashi, Y. Ono, N. Doi, F. Kitamura, M. Tagami et al., Multiple molecular interactions implicate the connectin/titin N2A region as a modulating scaffold for p94/calpain 3 activity in skeletal muscle, J Biol Chem, vol.283, issue.21, pp.14801-14815, 2008.

A. Belgrano, L. Rakicevic, L. Mittempergher, S. Campanaro, V. C. Martinelli et al., Arpp, a new homolog of carp, is preferentially expressed in type 1 skeletal muscle fibers and is markedly induced by denervation, Lab Invest, vol.6, issue.10, pp.645-55, 2002.

K. Ohlendieck, Proteomic Profiling of Fast-To-Slow Muscle Transitions during Aging, Front Physiol, issue.2, p.105, 2011.

N. Frey, D. Frank, S. Lippl, C. Kuhn, H. Kogler et al., Calsarcin-2 deficiency increases exercise capacity in mice through calcineurin/NFAT activation, J Clin Invest, vol.118, issue.11, pp.3598-608, 2008.

A. Bouter, C. Gounou, R. Berat, S. Tan, B. Gallois et al., Phosphoglycerate mutase deficiency with tubular aggregates in a patient from Panama, Muscle Nerve, vol.2, issue.1, pp.138-178, 2011.

C. Ryan and J. Radziuk, Distinguishable substrate pools for muscle glyconeogenesis in lactate-supplemented recovery from exercise, Am J Physiol, vol.269, issue.3, pp.538-50, 1995.

T. T. Gleeson, Post-exercise lactate metabolism: a comparative review of sites, pathways, and regulation, Annu Rev Physiol, vol.58, pp.565-81, 1996.

Y. Li, R. K. Dash, J. Kim, G. M. Saidel, and M. E. Cabrera, Role of NADH/NAD+ transport activity and glycogen store on skeletal muscle energy metabolism during exercise: in silico studies, Am J Physiol Cell Physiol, vol.296, issue.1, pp.25-46, 2009.

P. G. Schantz and J. Henriksson, Enzyme levels of the NADH shuttle systems: measurements in isolated muscle fibres from humans of differing physical activity, Acta Physiol Scand, vol.129, issue.4, pp.505-520, 1987.

H. Fischer, T. Gustafsson, C. J. Sundberg, J. Norrbom, M. Ekman et al., Fatty acid binding protein 4 in human skeletal muscle, Biochem Biophys Res Commun, vol.346, issue.1, pp.125-155, 2006.

J. F. Glatz, F. G. Schaap, B. Binas, A. Bonen, G. J. Van-der-vusse et al., Cytoplasmic fatty acid-binding protein facilitates fatty acid utilization by skeletal muscle, Acta Physiol Scand, vol.178, issue.4, pp.367-71, 2003.

J. J. Luiken, D. P. Koonen, W. A. Coumans, M. M. Pelsers, B. Binas et al., Long-chain fatty acid uptake by skeletal muscle is impaired in homozygous, but not heterozygous, heart-type-FABP null mice, Lipids, vol.38, issue.4, pp.491-497, 2003.

A. K. Saenger, T. V. Nguyen, J. Vockley, and M. T. Stankovich, Biochemical and electrochemical characterization of two variant human short-chain acyl-CoA dehydrogenases, Biochemistry, vol.44, issue.49, pp.16035-16077, 2005.

M. A. Baraibar, M. Gueugneau, S. Duguez, G. Butler-browne, D. Bechet et al., Expression and modification proteomics during skeletal muscle ageing, Biogerontology, vol.14, issue.3, pp.339-52, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543981

Y. Liu, W. Qi, A. Richardson, H. Van-remmen, Y. Ikeno et al., Oxidative damage associated with obesity is prevented by overexpression of CuZn-or Mnsuperoxide dismutase, Biochem Biophys Res Commun, vol.438, issue.1, pp.78-83, 2013.

S. G. Rhee, H. A. Woo, I. S. Kil, and S. H. Bae, Peroxiredoxin functions as a peroxidase and a regulator and sensor of local peroxides, J Biol Chem, vol.287, issue.7, pp.4403-4413, 2012.

A. Lombardi, E. Silvestri, F. Cioffi, R. Senese, A. Lanni et al., Defining the transcriptomic and proteomic profiles of rat ageing skeletal muscle by the use of a cDNA array, 2D-and Blue native-PAGE approach, J Proteomics, vol.72, issue.4, pp.708-729, 2009.

P. J. Thornalley, Protein and nucleotide damage by glyoxal and methylglyoxal in physiological systems--role in ageing and disease, Drug Metabol Drug Interact, vol.23, issue.1-2, pp.125-50, 2008.

F. Orosz, J. Olah, and J. Ovadi, Triosephosphate isomerase deficiency: new insights into an enigmatic disease, Biochim Biophys Acta, vol.1792, issue.12, pp.1168-74, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00537257

A. R. Hipkiss, Energy metabolism and ageing regulation: metabolically driven deamidation of triosephosphate isomerase may contribute to proteostatic dysfunction, Ageing Res Rev, vol.10, issue.4, pp.498-502, 2011.

H. H. Kampinga, J. Hageman, M. J. Vos, H. Kubota, R. M. Tanguay et al., Guidelines for the nomenclature of the human heat shock proteins, Cell Stress Chaperones, vol.14, issue.1, pp.105-116, 2009.

Y. E. Kim, M. S. Hipp, A. Bracher, M. Hayer-hartl, and F. U. Hartl, Molecular chaperone functions in protein folding and proteostasis, Annu Rev Biochem, vol.82, pp.323-55, 2013.

A. Mcardle, W. H. Dillmann, R. Mestril, J. A. Faulkner, and M. J. Jackson, Overexpression of HSP70 in mouse skeletal muscle protects against muscle damage and agerelated muscle dysfunction, Faseb J, vol.18, issue.2, pp.355-362, 2004.

S. M. Senf, S. L. Dodd, J. M. Mcclung, and A. R. Judge, Hsp70 overexpression inhibits NF-kappaB and Foxo3a transcriptional activities and prevents skeletal muscle atrophy, Faseb J, vol.22, issue.11, pp.3836-3881, 2008.

O. Iosefson, S. Sharon, P. Goloubinoff, and A. Azem, Reactivation of protein aggregates by mortalin and Tid1--the human mitochondrial Hsp70 chaperone system, Cell Stress Chaperones, vol.17, issue.1, pp.57-66, 2012.

K. O'connell, J. Gannon, P. Doran, and K. Ohlendieck, Proteomic profiling reveals a severely perturbed protein expression pattern in aged skeletal muscle, Int J Mol Med, vol.20, issue.2, pp.145-53, 2007.

P. Doran, J. Gannon, K. O'connell, and K. Ohlendieck, Aging skeletal muscle shows a drastic increase in the small heat shock proteins alphaB-crystallin/HspB5 and cvHsp/HspB7, Eur J Cell Biol, vol.86, issue.10, pp.629-669, 2007.

P. Doran, P. Donoghue, K. O'connell, J. Gannon, and K. Ohlendieck, Proteomics of skeletal muscle aging, Proteomics, vol.9, issue.4, pp.989-1003, 2009.

K. A. Won, R. J. Schumacher, G. W. Farr, A. L. Horwich, and S. I. Reed, Maturation of human cyclin E requires the function of eukaryotic chaperonin CCT, Mol Cell Biol, vol.18, issue.12, pp.7584-7593, 1998.

L. Theron, M. Gueugneau, C. Coudy, D. Viala, A. Bijlsma et al., Label-free quantitative protein profiling of vastus lateralis muscle during human aging, Mol Cell Proteomics, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00986095

K. H. Strucksberg, K. Tangavelou, R. Schroder, and C. S. Clemen, Proteasomal activity in skeletal muscle: a matter of assay design, muscle type, and age, Anal Biochem, issue.2, pp.225-234, 2010.

R. Medina, S. S. Wing, A. Haas, and A. L. Goldberg, Activation of the ubiquitin-ATPdependent proteolytic system in skeletal muscle during fasting and denervation atrophy, Biomed Biochim Acta, vol.50, issue.4-6, pp.347-56, 1991.

S. H. Lecker, A. L. Goldberg, and W. E. Mitch, Protein degradation by the ubiquitinproteasome pathway in normal and disease states, J Am Soc Nephrol, vol.17, issue.7, pp.1807-1826, 2006.

S. H. Lecker, R. T. Jagoe, A. Gilbert, M. Gomes, V. Baracos et al., Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression, Faseb J, vol.18, issue.1, pp.39-51, 2004.

S. S. Wing, Deubiquitinases in skeletal muscle atrophy, Int J Biochem Cell Biol, vol.45, issue.10, pp.2130-2135, 2013.

C. Cai, H. Masumiya, N. Weisleder, N. Matsuda, M. Nishi et al., MG53 nucleates assembly of cell membrane repair machinery, Nat Cell Biol, issue.11, pp.56-64, 2009.

H. Zhu, P. Lin, G. De, K. H. Choi, H. Takeshima et al., Polymerase transcriptase release factor (PTRF) anchors MG53 protein to cell injury site for initiation of membrane repair, J Biol Chem, vol.286, issue.15, pp.12820-12824, 2011.

M. M. Hill, M. Bastiani, R. Luetterforst, M. Kirkham, A. Kirkham et al., PTRF-Cavin, a conserved cytoplasmic protein required for caveola formation and function, Cell, vol.132, issue.1, pp.113-137, 2008.

S. E. Bearden, A. Rogowska-wrzesinska, P. Roepstorff, A. L. Bulteau, and B. Friguet, Effect of aging on the structure and function of skeletal muscle microvascular networks, References Ahmed EK, vol.13, issue.4, pp.252-272, 2006.

D. C. Andersson, M. J. Betzenhauser, S. Reiken, A. C. Meli, A. Umanskaya et al., Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging, Cell Metab, vol.14, pp.196-207, 2011.

R. Bandopadhyay and A. E. Kingsbury, The expression of DJ-1 (PARK7) in normal human CNS and idiopathic Parkinson's disease, Brain, vol.127, pp.420-430, 2004.

M. A. Baraibar and B. Friguet, Changes of the proteasomal system during the aging process, Prog Mol Biol Transl Sci, vol.109, pp.249-275, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01543991

M. A. Baraibar and B. Friguet, Oxidative proteome modifications target specific cellular pathways during oxidative stress, cellular senescence and aging, Exp Gerontol, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543979

M. A. Baraibar, J. Hyzewicz, A. Rogowska-wrzesinska, R. Ladouce, P. Roepstorff et al., Oxidative stress-induced proteome alterations target different cellular pathways in human myoblasts, Free Radic Biol Med, vol.51, issue.8, pp.1522-1532, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02148122

M. A. Baraibar, L. Liu, E. K. Ahmed, and B. Friguet, Protein oxidative damage at the crossroads of cellular senescence, aging, and age-related diseases, Oxid Med Cell Longev, p.919832, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01543992

M. A. Baraibar, A. G. Barbeito, B. B. Muhoberac, and R. Vidal, A mutant light-chain ferritin that causes neurodegeneration has enhanced propensity toward oxidative damage, Free Radic Biol Med, vol.52, issue.9, pp.1692-1697, 2012.

M. A. Baraibar, R. Ladouce, and B. Friguet, A method for detecting and/or quantifying carbonylated proteins, vol.27, 2012.

E. Barreiro and S. N. Hussain, Protein carbonylation in skeletal muscles: impact on function, Antioxid Redox Signal, vol.12, issue.3, pp.417-429, 2010.

, Biogerontology, vol.14, pp.339-352, 2013.

E. Barreiro, V. Coronell, and B. Laviña, Aging, sex differences, and oxidative stress in human respiratory and limb muscles, Free Radic Biol Med, vol.41, issue.5, pp.797-809, 2006.

R. N. Baumgartner, K. M. Koehler, D. Gallagher, L. Romero, S. B. Heymsfield et al., Epidemiology of sarcopenia among the elderly in New Mexico, Am J Epidemiol, vol.147, issue.8, pp.755-763, 1998.

B. S. Berlett and E. R. Stadtman, Protein oxidation in aging, disease, and oxidative stress, J Biol Chem, vol.272, issue.33, pp.20313-20316, 1997.

B. Biteau, J. Labarre, and M. B. Toledano, ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin, Nature, vol.425, issue.6961, pp.980-984, 2003.

N. Breusing and T. Grune, Regulation of proteasome-mediated protein degradation during oxidative stress and aging, Biol Chem, vol.389, pp.203-209, 2008.

D. Capitanio, M. Vasso, C. Fania, M. Moriggi, A. Viganò et al., Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE, Proteomics, vol.9, 2004.

C. Y. Chan, J. C. Mcdermott, and K. W. Siu, Secretome analysis of skeletal myogenesis using SILAC and shotgun proteomics, 2011.

, Int J Proteomics, p.329467, 2011.

K. B. Choksi and J. Papaconstantinou, Age-related alterations in oxidatively damaged proteins of mouse heart mitochondrial electron transport chain complexes, Free Radic Biol Med, vol.44, issue.10, pp.1795-1805, 2008.

K. A. Clark, A. S. Mcelhinny, M. C. Beckerle, and C. C. Gregorio, Striated muscle cytoarchitecture: an intricate web of form and function, Annu Rev Cell Dev Biol, vol.18, pp.637-706, 2002.

P. Donoghue, L. Staunton, E. Mullen, G. Manning, and K. Ohlendieck, DIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult versus aged gastrocnemius tissue, J Proteomics, vol.73, issue.8, pp.1441-1453, 2010.

P. Doran, J. Gannon, K. O'connell, and K. Ohlendieck, Ageing skeletal muscle shows a drastic increase in the small heat shock proteins B-crystallin/HspB5 and cvHsp/HspB7, Eur J Cell Biol, vol.86, pp.629-640, 2007.

P. Doran, K. O'connell, J. Gannon, M. Kavanagh, and K. Ohlendieck, Opposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysis, Proteomics, vol.8, pp.364-377, 2008.

W. Droge, Free radicals in the physiological control of cell function, Physiol Rev, vol.82, pp.47-95, 2002.

S. Duguez, W. Duddy, H. Johnston, J. Laine, L. Bihan et al., Dystrophin deficiency leads to disturbance of LAMP1-vesicle-associated protein secretion, Cell Mol Life Sci, 2013.

T. D. Engerson, T. G. Mckelvey, D. B. Rhyne, E. B. Boggio, S. J. Snyder et al., Conversion of xanthine dehydrogenase to oxidase in ischemic rat tissues, J Clin Invest, vol.79, pp.1564-1570, 1987.

D. Engler, Hypothesis: Musculin is a hormone secreted by skeletal muscle, the body's largest endocrine organ. Evidence for actions on the endocrine pancreas to restrain the beta-cell mass and to inhibit insulin secretion and on the hypothalamus to co-ordinate the neuroendocrine and appetite responses to exercise, Acta Biomed, vol.78, issue.1, pp.156-206, 2007.

W. J. Evans, What is sarcopenia?, J Gerontol A Biol Sci Med Sci 50 Spec, pp.5-8, 1995.

M. Fedorova, N. Kuleva, and R. Hoffmann, Identification, quantification, and functional aspects of skeletal muscle protein-carbonylation in vivo during acute oxidative stress, J Proteome Res, vol.9, issue.5, pp.2516-2526, 2010.

L. F. Ferreira and M. B. Reid, Muscle-derived ROS and thiol regulation in muscle fatigue, J Appl Physiol, vol.104, pp.853-860, 2008.

B. Friguet, Aging of proteins and the proteasome, Prog Mol Subcell Biol, vol.29, pp.17-33, 2002.

B. Friguet, Oxidized protein degradation and repair in ageing and oxidative stress, FEBS Lett, vol.580, issue.12, pp.2910-2916, 2006.

B. Friguet, A. L. Bulteau, N. Chondrogianni, and M. Conconi, Petropoulos I (2000) Protein degradation by the proteasome and its implications in aging, Ann N Y Acad Sci, vol.908, pp.143-154

N. A. Fugere, D. A. Ferrington, and L. V. Thompson, Protein nitration with aging in the rat semimembranosus and soleus muscles, J Gerontol Ser A Biol Sci Med Sci, vol.61, issue.8, pp.806-812, 2006.

J. Gannon, L. Staunton, O. Connell, K. Doran, P. Ohlendieck et al., Phosphoproteomic analysis of aged skeletal muscle, Int J Mol Med, vol.22, pp.33-42, 2008.

J. Gannon, P. Doran, A. Kirwan, and K. Ohlendieck, Drastic increase of myosin light chain MLC-2 in senescent skeletal muscle indicates fast-to-slow fibre transition in sarcopenia of old age, Eur J Cell Biol, vol.88, pp.685-700, 2009.

C. Gelfi, A. Vigano, M. Ripamonti, A. Pontoglio, S. Begum et al., The human muscle proteome in aging, J Proteome Res, vol.5, pp.1344-1353, 2006.

P. Gianni, K. J. Jan, M. J. Douglas, P. M. Stuart, and M. A. Tarnopolsky, Oxidative stress and the mitochondrial theory of aging in human skeletal muscle, Exp Gerontol, vol.39, issue.9, pp.1391-1400, 2004.

J. M. Gilmore and M. P. Washburn, Advances in shotgun proteomics and the analysis of membrane proteomes, J Proteomics, vol.73, issue.11, pp.2078-2091, 2010.

C. Giulivi, N. J. Traaseth, and K. J. Davies, Tyrosine oxidation products: analysis and biological relevance, Amino Acids, vol.25, issue.3-4, pp.227-232, 2003.

L. G. Goldfarb and M. C. Dalakas, Tragedy in a heartbeat: malfunctioning desmin causes skeletal and cardiac muscle disease, J Clin Invest, vol.119, pp.1806-1813, 2009.

A. Gorg, O. Drews, C. Luck, F. Weiland, and W. Weiss, Electrophoresis, vol.30, issue.1, pp.122-132, 2009.

A. Görg, W. Weiss, and M. J. Dunn, Current two-dimensional electrophoresis technology for proteomics, Proteomics, vol.4, pp.3665-3685, 2004.

J. Henningsen, K. T. Rigbolt, B. Blagoev, and B. K. Pedersen, Kratchmarova I (2010) Dynamics of the skeletal muscle secretome during myoblast differentiation, Mol Cell Proteomics, vol.9, issue.11, pp.2482-2496

K. Hojlund, Z. Yi, H. Hwang, B. Bowen, N. Lefort et al., Characterization of the human skeletal muscle proteome by onedimensional gel electrophoresis and HPLC-ESI-MS/MS, Mol Cell Proteomics, vol.7, pp.257-267, 2008.

S. Horiuchi and N. Araki, Advanced glycation end products of the Maillard reaction and their relation to aging, Gerontology, vol.40, issue.2, pp.10-15, 1994.

M. J. Jackson, S. Papa, and J. Bolanos, Antioxidants, reactive oxygen and nitrogen species, gene induction and mitochondrial function, Mol Aspects Med, vol.23, pp.209-285, 2002.

J. Kanski, S. J. Hong, and C. Schöneich, Proteomic analysis of protein nitration in ageing skeletal muscle and identification of nitrotyrosine-containing sequences in vivo by nanoelectrospray ionization tandem mass spectrometry, J Biol Chem, vol.280, pp.24261-24266, 2005.

H. Kondo, M. Miura, and Y. Itokawa, Oxidative stress in skeletal muscle atrophied by immobilization, Acta Physiol Scand, vol.142, pp.527-528, 1991.

H. Kondo, K. Nishino, and Y. Itokawa, Hydroxyl radical generation in skeletal muscle atrophied by immobilization, FEBS Lett, vol.349, pp.169-172, 1994.

L. Bihan, M. C. Bigot, A. Jensen, S. S. Dennis, J. L. Rogowska-wrzesinska et al., Indepth analysis of the secretome identifies three major independent secretory pathways in differentiating human myoblasts, J Proteomics, vol.77, pp.344-356, 2012.

N. Lefort, Z. Yi, B. Bowen, B. Glancy, D. Filippis et al., Proteome profile of functional mitochondria from human skeletal muscle using one-dimensional gel electrophoresis and HPLC ESI-MS/MS, J Proteomics, vol.72, pp.1046-1060, 2009.

R. L. Levine and E. R. Stadtman, Oxidative modification of proteins during aging, Exp Gerontol, vol.36, issue.9, pp.1495-1502, 2001.

R. L. Levine, N. Wehr, J. A. Williams, E. R. Stadtman, and E. Shacter, Determination of carbonyl groups in oxidized proteins, Methods Mol Biol, vol.99, pp.15-24, 2000.

A. Lombardi, E. Silvestri, F. Cioffi, R. Senese, A. Lanni et al., Defining the transcriptomic and proteomic profiles of rat ageing skeletal muscle by the use of a cDNA array, 2D-and Blue native-PAGE approach, J Proteomics, vol.72, pp.708-721, 2009.

A. G. Madian and F. E. Regnier, Proteomic identification of carbonylated proteins and their oxidation sites, J Proteome Res, vol.9, issue.8, pp.3766-3780, 2010.

B. Marzani, G. Felzani, R. G. Bellomo, J. Vecchiet, and F. Marzatico, Human muscle ageing: ROS-mediated alterations in rectus abdominis and vastus lateralis muscles, Exp Gerontol, vol.40, pp.959-965, 2005.

B. Meunier, E. Dumas, I. Piec, D. Béchet, M. Hébraud et al., Assessment of hierarchical clustering methodologies for proteomic data mining, J Proteome Res, vol.6, pp.358-366, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02657777

R. Moreau, S. H. Heath, C. E. Doneanu, J. G. Lindsay, and T. M. Hagen, Age-related increase in 4-hydroxynonenal adduction to rat heart alpha-ketoglutarate dehydrogenase does not cause loss of its catalytic activity, Antioxid Redox Signal, vol.5, issue.5, pp.517-527, 2003.

K. O'connell and K. Ohlendieck, Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle, Proteomics, vol.9, pp.5509-5524, 2009.

O. Pansarasa, B. Castagna, J. Colombi, J. Vecchiet, C. Felzani et al., Age and sex differences in human skeletal muscle: role of reactive oxygen species, Free Radic Res, vol.33, issue.3, pp.287-293, 2000.

I. Petropoulos and B. Friguet, Maintenance of proteins and aging: the role of oxidized protein repair, Free Radic Res, vol.40, issue.12, pp.1269-1276, 2006.

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Differential proteome analysis of aging in rat skeletal muscle, FASEB J, vol.19, issue.9, pp.1143-1145, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680775

S. K. Powers and M. J. Jackson, Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production, Physiol Rev, vol.88, pp.1243-1276, 2008.

T. Rabilloud, Two-dimensional gel electrophoresis in proteomics: old, old fashioned, but it still climbs up the mountains, Proteomics, vol.2, pp.3-10, 2002.

T. Rabilloud and C. Lelong, Two-dimensional gel electrophoresis in proteomics: a tutorial, J Proteomics, vol.74, issue.10, pp.1829-1841, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00623443

T. Rabilloud, M. Heller, F. Gasnier, S. Luche, C. Rey et al., Proteomics analysis of cellular response to oxidative stress: evidence for in vivo overoxidation of peroxiredoxins at their active site, J Biol Chem, vol.277, pp.19396-19401, 2002.

M. B. Reid, Nitric oxide, reactive oxygen species, and skeletal muscle contraction, Med Sci Sports Exerc, vol.33, pp.371-376, 2001.

A. Roca-rivada, O. Al-massadi, C. Castelao, L. L. Senin, J. Alonso et al., Muscle tissue as an endocrine organ: comparative secretome profiling of slow-oxidative and fast-glycolytic rat muscle explants and its variation with exercise, J Proteomics, vol.75, issue.17, pp.5414-5425, 2012.

A. Rogowska-wrzesinska, L. Bihan, M. C. Thaysen-andersen, M. Roepstorff, and P. , 2D gels still have a niche in proteomics, J Proteomics, 2013.

S. Schiaffino, Fibre types in skeletal muscle: a personal account, Acta Physiol, vol.199, issue.4, pp.451-463, 2010.

D. Shenton, J. B. Smirnova, J. N. Selley, K. Carroll, S. J. Hubbard et al., Global translational responses to oxidative stress impact upon multiple levels of protein synthesis, J Biol Chem, vol.281, pp.29011-29021, 2006.

C. Shindoh, A. Dimarco, D. Nethery, and G. Supinski, Effect of PEG-superoxide dismutase on the diaphragmatic response to endotoxin, Am Rev Respir Dis, vol.145, pp.1350-1354, 1992.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke et al., Decline in skeletal muscle mitochondrial function with ageing in humans, Proc Natl Acad Sci, vol.102, pp.5618-5623, 2005.

T. Snijders, L. B. Verdijk, and L. J. Van-loon, The impact of sarcopenia and exercise training on skeletal muscle satellite cells, Ageing Res Rev, vol.8, issue.4, pp.328-338, 2009.

L. M. Snow, N. A. Fugere, and L. V. Thompson, Advanced glycation end-product accumulation and associated protein modification in type II skeletal muscle with ageing, J Gerontol Ser A Biol Sci Med Sci, vol.62, issue.11, pp.1204-1210, 2007.

E. R. Stadtman and R. L. Levine, Free radical-mediated oxidation of free amino acids and amino acid residues in proteins, Amino Acids, vol.25, issue.3-4, pp.207-218, 2003.

L. Staunton, M. Zweyer, D. Swandulla, and K. Ohlendieck, Mass spectrometry-based proteomic analysis of middle, 2012.

, Biogerontology, p.14, 2013.

, Int J Mol Med, vol.30, pp.723-733

L. V. Thompson, D. Durand, N. A. Fugere, and D. A. Ferrington, Myosin and actin expression and oxidation in aging muscle, J Appl Physiol, vol.101, issue.6, pp.1581-1587, 2006.

M. Ü-nlü and J. S. Minden, Difference gel electrophoresis. In: Walker JM (ed) The protein protocols handbook, pp.185-196, 2002.

M. A. Whidden, J. M. Mcclung, D. J. Falk, M. B. Hudson, A. J. Smuder et al., Xanthine oxidase contributes to mechanical ventilation-induced diaphragmatic oxidative stress and contractile dysfunction, J Appl Physiol, vol.106, pp.385-394, 2009.

L. J. Yan and M. J. Forster, Chemical probes for analysis of carbonylated proteins: a review, J Chromatogr B Anal Technol Biomed Life Sci, vol.879, pp.1308-1315, 2011.

J. H. Yoon, J. Kim, P. Song, T. G. Lee, P. G. Suh et al., Secretomics for skeletal muscle cells: a discovery of novel regulators?, Adv Biol Regul, vol.52, issue.2, pp.340-350, 2012.

D. L. References-allen, J. K. Linderman, R. R. Roy, A. J. Bigbee, R. E. Grindeland et al., Apoptosis: a mechanism contributing to remodeling of skeletal muscle in response to hindlimb unweighting, Am. J. Physiol, vol.273, pp.579-587, 1997.

M. Alsharidah, N. R. Lazarus, T. E. George, C. C. Agley, C. P. Velloso et al., Primary human muscle precursor cells obtained from young and old donors produce similar proliferative, differentiation and senescent profiles in culture, Aging Cell, vol.12, pp.333-344, 2013.

J. L. Andersen, Muscle fibre type adaptation in the elderly human muscle. Scand, J. Med. Sci. Sports, vol.13, pp.40-47, 2003.

K. Asai, R. K. Kudej, Y. T. Shen, G. P. Yang, G. Takagi et al., , p.Peripheral, 2000.

, Apoptosis in capillary endothelial cells in old muscle, H. Wang et al. vascular endothelial dysfunction and apoptosis in old monkeys, Arterioscler. Thromb. Vasc. Biol, vol.20, pp.1493-1499

M. A. Baraibar, M. Gueugneau, S. Duguez, G. Butler-browne, . Bé-chet-d et al., Proteomics of muscle protein modifications during aging, Biogerontology, vol.14, pp.339-352, 2013.

J. C. Bruusgaard and K. Gundersen, In vivo time-lapse microscopy reveals no loss of murine myonuclei during weeks of muscle atrophy, J. Clin. Invest, vol.118, pp.1450-1457, 2008.

J. C. Bruusgaard, K. Liestøl, and K. Gundersen, Distribution of myonuclei and microtubules in live muscle fibers of young, middle-aged, and old mice, J. Appl. Physiol, vol.100, pp.2024-2030, 2006.

C. Christov, F. Chré-tien, R. Abou-khalil, G. Bassez, G. Vallet et al., Muscle satellite cells and endothelial cells: close neighbors and privileged partners, Mol. Biol. Cell, vol.18, pp.1397-1409, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00128985

C. A. Collins, P. S. Zammit, A. P. Ruiz, J. E. Morgan, and T. A. Partridge, A population of myogenic stem cells that survives skeletal muscle aging, Stem Cells, vol.25, pp.885-894, 2007.

L. Combaret, D. Dardevet, D. Bé-chet, D. Taillandier, L. Mosoni et al., Skeletal muscle proteolysis in aging, Curr. Opin. Clin. Nutr. Metab. Care, vol.12, pp.37-41, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02661511

G. Gobé, J. Browning, T. Howard, N. Hogg, C. Winterford et al., Apoptosis occurs in endothelial cells during hypertension-induced microvascular rarefaction, J. Struct. Biol, vol.118, pp.63-72, 1997.

M. D. Herrera, C. Mingorance, R. Rodr-iguez-rodr-iguez, and M. Alvarez-de-sotomayor, Endothelial dysfunction and aging: an update, Ageing Res. Rev, vol.9, pp.142-152, 2010.

C. Ibebunjo, J. M. Chick, T. Kendall, J. K. Eash, C. Li et al., Genomic and proteomic profiling reveals reduced mitochondrial function and disruption of the neuromuscular junction driving rat sarcopenia, Mol. Cell. Biol, vol.33, pp.194-212, 2013.

R. V. Iozzo, J. J. Zoeller, N. , and A. , Basement membrane proteoglycans: modulators par excellence of cancer growth and angiogenesis, Mol. Cells, vol.27, pp.503-513, 2009.

S. S. Jejurikar, E. A. Henkelman, P. S. Cederna, C. L. Marcelo, M. G. Urbanchek et al., Aging increases the susceptibility of skeletal muscle derived satellite cells to apoptosis, Exp. Gerontol, vol.41, pp.828-836, 2006.

T. W. Kragstrup, M. Kjaer, and A. L. Mackey, Structural, biochemical, cellular, and functional changes in skeletal muscle extracellular matrix with aging. Scand, J. Med. Sci. Sports, vol.21, pp.749-757, 2011.

S. W. Lamberts, A. W. Van-den-beld, and A. J. Van-der-lely, The endocrinology of aging, Science, vol.278, pp.419-424, 1997.

P. Laplante, M. A. Raymond, G. Gagnon, N. Vigneault, A. Sasseville et al., Novel fibrogenic pathways are activated in response to endothelial apoptosis: implications in the pathophysiology of systemic sclerosis, J. Immunol, vol.174, pp.5740-5749, 2005.

B. D. Larsen, S. Rampalli, L. E. Burns, S. Brunette, F. J. Dilworth et al., Caspase 3/caspase-activated DNase promote cell differentiation by inducing DNA strand breaks, Proc. Natl Acad. Sci. USA, vol.107, pp.4230-4235, 2010.

J. Lexell, Evidence for nervous system degeneration with advancing age, J. Nutr, vol.127, pp.1011-1013, 1997.

L. T. Malmgren, C. E. Jones, and L. M. Bookman, Muscle fiber and satellite cell apoptosis in the aging human thyroarytenoid muscle: a stereological study with confocal laser scanning microscopy, Otolaryngol. Head Neck Surg, vol.125, pp.34-39, 2001.

Y. Margaron, L. Bostan, J. Y. Exposito, M. Malbouyres, A. M. Trunfio-sfarghiu et al., Tenascin-X increases the stiffness of collagen gels without affecting fibrillogenesis, Biophys. Chem, vol.147, pp.87-91, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00466917

E. Marzetti, G. Privitera, V. Simili, S. E. Wohlgemuth, L. Aulisa et al., Multiple pathways to the same end: mechanisms of myonuclear apoptosis in sarcopenia of aging, ScientificWorldJournal, vol.10, pp.340-349, 2010.

M. A. Murray and N. Robbins, Cell proliferation in denervated muscle: time course, distribution and relation to disuse, Neuroscience, vol.7, pp.1817-1822, 1982.

B. Pé-ault, M. Rudnicki, Y. Torrente, G. Cossu, J. P. Tremblay et al., Stem and progenitor cells in skeletal muscle development, maintenance, and therapy, Mol. Ther, vol.15, pp.867-877, 2007.

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Differential proteome analysis of aging in rat skeletal muscle, FASEB J, vol.19, pp.1143-1145, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680775

M. Podhorska-okolow, M. Sandri, S. Zampieri, B. Brun, K. Rossini et al., Apoptosis of myofibres and satellite cells: exercise-induced damage in skeletal muscle of the mouse, Neuropathol. Appl. Neurobiol, vol.24, pp.518-531, 1998.

J. R. Privratsky, D. K. Newman, and P. J. Newman, PECAM-1: conflicts of interest in inflammation, Life Sci, vol.87, pp.69-82, 2010.

M. D. Rees, J. M. Whitelock, E. Malle, C. Y. Chuang, R. V. Iozzo et al., Myeloperoxidase-derived oxidants selectively disrupt the protein core of the heparan sulfate proteoglycan perlecan, Matrix Biol, vol.29, pp.63-73, 2010.

V. Renault, L. E. Thorne, P. O. Eriksson, G. Butler-browne, and V. Mouly, Regenerative potential of human skeletal muscle during aging, Aging Cell, vol.1, pp.132-139, 2002.

M. M. De-resende, S. L. Amaral, D. H. Munzenmaier, and A. S. Greene, Role of endothelial cell apoptosis in regulation of skeletal muscle angiogenesis during high and low salt intake, Physiol. Genomics, vol.13, pp.325-335, 2006.

M. Sandri, C. Minetti, M. Pedemonte, and U. Carraro, Apoptotic myonuclei in human Duchenne muscular dystrophy, Lab. Invest, vol.78, pp.1005-1016, 1998.

J. Scharner and P. S. Zammit, The muscle satellite cell at 50: the formative years, Skelet. Muscle, vol.1, pp.28-40, 2011.

H. Schmalbruch and U. Hellhammer, The number of nuclei in adult rat muscles with special reference to satellite, Anat. Rec, vol.189, pp.169-175, 1977.

T. Shavlakadze, J. Mcgeachie, and M. D. Grounds, Delayed but excellent myogenic stem cell response of regenerating geriatric skeletal muscles in mice, Biogerontology, vol.11, pp.363-376, 2010.

G. Shefer, D. P. Van-de-mark, J. B. Richardson, and Z. Yablonka-reuveni, Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle, Dev. Biol, vol.294, pp.50-66, 2006.

H. Tullberg-reinert and G. Jundt, In situ measurement of collagen synthesis by human bone cells with a Sirius Red-based colorimetric microassay: effects of transforming growth factor b2 and ascorbic acid 2-phosphate, Histochem. Cell Biol, vol.112, pp.271-276, 1999.

G. Valdez, J. C. Tapia, H. Kang, G. D. Clemenson, F. H. Gage et al., Attenuation of age-related changes in mouse neuromuscular synapses by caloric restriction and exercise, Proc. Natl Acad. Sci. USA, vol.107, pp.14863-14868, 2010.

G. Vescovo, R. Zennaro, M. Sandri, U. Carraro, C. Leprotti et al., Apoptosis of skeletal muscle myofibers and interstitial cells in experimental heart failure, J. Mol. Cell. Cardiol, vol.30, pp.2449-2459, 1998.

A. Virdis, L. Ghiadoni, C. Giannarelli, and S. Taddei, Endothelial dysfunction and vascular disease in later life, Maturitas, vol.67, pp.20-24, 2010.

N. C. Voermans, C. G. B?-onnemann, P. A. Huijing, B. C. Hamel, T. H. Van-kuppevelt et al., Clinical and molecular overlap between myopathies and inherited connective tissue diseases, Neuromuscul. Disord, vol.18, pp.843-856, 2008.

J. M. Whitelock, J. Melrose, and R. V. Iozzo, Diverse cell signaling events modulated by perlecan, Biochemistry, vol.47, pp.11174-11183, 2008.

J. F. Woessner, The determination of hydroxyproline in tissue and protein samples containing small proportions of amino acid, Arch. Biochem. Biophys, vol.93, pp.440-448, 1961.

Z. Yablonka-reuveni and J. E. Anderson, Satellite cells from dystrophic (Mdx) mice display accelerated differentiation in primary cultures and in isolated myofibers, Dev. Dyn, vol.235, pp.203-212, 2006.

N. O. Yarovaya, L. Kramarova, J. Borg, S. A. Kovalenko, A. Caragounis et al., Age-related atrophy of rat soleus muscle is accompanied by changes in fibre type composition, bioenergy decline and mtDNA rearrangements, Biogerontology, vol.3, pp.25-27, 2002.

H. Wang, Apoptosis in capillary endothelial cells in old muscle

, The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd. Références bibliographiques Références bibliographiques, 2013.

A. Agrawal, S. Agrawal, and S. Gupta, Dendritic cells in human aging, Experimental gerontology, vol.42, pp.421-426, 2007.

L. Al-qusairi and J. Laporte, T-tubule biogenesis and triad formation in skeletal muscle and implication in human diseases, Skeletal muscle, vol.1, p.26, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00614419

K. G. Alberti, R. H. Eckel, S. M. Grundy, P. Z. Zimmet, J. I. Cleeman et al., Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute, American Heart Association; World Heart, vol.120, pp.1640-1645, 2009.

K. G. Alberti, P. Zimmet, and J. Shaw, The metabolic syndrome--a new worldwide definition, Lancet, vol.366, pp.1059-1062, 2005.

K. G. Alberti and P. Z. Zimmet, Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation, Diabetic medicine : a journal of the British Diabetic Association, vol.15, pp.539-553, 1998.

R. E. Allen and L. K. Boxhorn, Regulation of skeletal muscle satellite cell proliferation and differentiation by transforming growth factor-beta, insulin-like growth factor I, and fibroblast growth factor, Journal of cellular physiology, vol.138, pp.311-315, 1989.

M. Alsharidah, N. R. Lazarus, T. E. George, C. C. Agley, C. P. Velloso et al., Primary human muscle precursor cells obtained from young and old donors produce similar proliferative, differentiation and senescent profiles in culture, Aging cell, vol.12, pp.333-344, 2013.

J. L. Andersen, Muscle fibre type adaptation in the elderly human muscle, Scandinavian journal of medicine & science in sports, vol.13, pp.40-47, 2003.

J. L. Andersen, G. Terzis, and A. Kryger, Increase in the degree of coexpression of myosin heavy chain isoforms in skeletal muscle fibers of the very old, Muscle & nerve, vol.22, pp.449-454, 1999.

Y. Aydar, P. Balogh, J. G. Tew, and A. K. Szakal, Age-related depression of FDC accessory functions and CD21 ligand-mediated repair of co-stimulation, European journal of immunology, vol.32, pp.2817-2826, 2002.

F. Azizi, P. Salehi, A. Etemadi, and S. Zahedi-asl, Prevalence of metabolic syndrome in an urban population: Tehran Lipid and Glucose Study, Diabetes research and clinical practice, vol.61, pp.29-37, 2003.

A. J. Bailey, Molecular mechanisms of ageing in connective tissues, Mechanisms of ageing and development, vol.122, pp.735-755, 2001.

B. Balkau and M. A. Charles, Comment on the provisional report from the WHO consultation. European Group for the Study of Insulin Resistance (EGIR), British Diabetic Association, vol.16, pp.442-443, 1999.

Z. Z. Bao, M. Lakonishok, S. Kaufman, and A. F. Horwitz, Alpha 7 beta 1 integrin is a component of the myotendinous junction on skeletal muscle, Journal of cell science, vol.106, issue.2, pp.579-589, 1993.

M. A. Baraibar, M. Gueugneau, S. Duguez, G. Butler-browne, D. Bechet et al., Expression and modification proteomics during skeletal muscle ageing, Biogerontology, vol.14, pp.339-352, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01543981

R. N. Baumgartner, K. M. Koehler, D. Gallagher, L. Romero, S. B. Heymsfield et al., Epidemiology of sarcopenia among the elderly in New Mexico, American journal of epidemiology, vol.147, pp.755-763, 1998.

J. A. Baur, K. J. Pearson, N. L. Price, H. A. Jamieson, C. Lerin et al., Resveratrol improves health and survival of mice on a high-calorie diet, Nature, vol.444, pp.337-342, 2006.

S. M. Baylor and S. Hollingworth, Intracellular calcium movements during excitationcontraction coupling in mammalian slow-twitch and fast-twitch muscle fibers, The Journal of general physiology, vol.139, pp.261-272, 2012.

E. J. Behringer and S. S. Segal, Spreading the signal for vasodilatation: implications for skeletal muscle blood flow control and the effects of ageing, The Journal of physiology, vol.590, pp.6277-6284, 2012.

M. Bencze, E. Negroni, D. Vallese, H. Yacoub-youssef, S. Chaouch et al., Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation, Molecular therapy : the journal of the American Society of Gene Therapy, vol.20, pp.2168-2179, 2012.

A. Y. Bijlsma, C. G. Meskers, C. H. Ling, M. Narici, S. E. Kurrle et al., Defining sarcopenia: the impact of different diagnostic criteria on the prevalence of sarcopenia in a large middle aged cohort, Age (Dordr), vol.35, pp.871-881, 2013.

A. Y. Bijlsma, C. G. Meskers, N. Van-den-eshof, R. G. Westendorp, S. Sipila et al., Diagnostic criteria for sarcopenia and physical performance, 2013.

A. Y. Bijlsma, C. G. Meskers, D. Van-heemst, R. G. Westendorp, A. J. De-craen et al., Diagnostic criteria for sarcopenia relate differently to insulin resistance, Age, 2013.

A. Y. Bijlsma, C. G. Meskers, R. G. Westendorp, and A. B. Maier, Chronology of age-related disease definitions: osteoporosis and sarcopenia, Ageing research reviews, vol.11, pp.320-324, 2012.

J. W. Bijlsma, F. Berenbaum, and F. P. Lafeber, Osteoarthritis: an update with relevance for clinical practice, Lancet, vol.377, pp.2115-2126, 2011.

A. Blanchard, V. Ohanian, and D. Critchley, The structure and function of alpha-actinin, Journal of muscle research and cell motility, vol.10, pp.280-289, 1989.

N. Blanpin and O. Chardon, Projections de la population à l'horizon 2060, 2010.

R. J. Bloch and H. Gonzalez-serratos, Lateral force transmission across costameres in skeletal muscle, Exercise and sport sciences reviews, vol.31, pp.73-78, 2003.

W. Bloom and D. W. Fawcett, A Textbook of Histology: By W.Bloom and D.W.Fawcett, 9 edn, 1962.

D. Boffoli, S. C. Scacco, R. Vergari, G. Solarino, G. Santacroce et al., Decline with age of the respiratory chain activity in human skeletal muscle, Biochimica et biophysica acta, vol.1226, pp.73-82, 1994.

T. K. Borg and J. B. Caulfield, Morphology of connective tissue in skeletal muscle, Tissue & cell, vol.12, pp.197-207, 1980.

F. Borrego, M. C. Alonso, M. D. Galiani, J. Carracedo, R. Ramirez et al., NK phenotypic markers and IL2 response in NK cells from elderly people, Experimental gerontology, vol.34, pp.253-265, 1999.

S. E. Borst, C. F. Conover, and G. J. Bagby, Association of resistin with visceral fat and muscle insulin resistance, Cytokine, vol.32, pp.39-44, 2005.

C. Bos, B. Stoll, H. Fouillet, C. Gaudichon, X. Guan et al., Postprandial intestinal and whole body nitrogen kinetics and distribution in piglets fed a single meal, American journal of physiology Endocrinology and metabolism, vol.288, pp.436-446, 2005.

M. J. Bossingham, N. S. Carnell, and W. W. Campbell, Water balance, hydration status, and fatfree mass hydration in younger and older adults, The American journal of clinical nutrition, vol.81, pp.1342-1350, 2005.

A. S. Brack and T. A. Rando, Intrinsic changes and extrinsic influences of myogenic stem cell function during aging, Stem cell reviews, vol.3, pp.226-237, 2007.

M. K. Brawer, Testosterone replacement in men with andropause: an overview, Reviews in urology, vol.6, pp.9-15, 2004.

K. A. Britton, J. M. Massaro, J. M. Murabito, B. E. Kreger, U. Hoffmann et al., Body fat distribution, incident cardiovascular disease, cancer, and all-cause mortality, Journal of the American College of Cardiology, vol.62, pp.921-925, 2013.

F. Broglio, E. Arvat, A. Benso, C. Gottero, F. Prodam et al., Ghrelin: much more than a natural growth hormone secretagogue, The Israel Medical Association journal : IMAJ, vol.4, pp.607-613, 2002.

M. H. Brooke and K. K. Kaiser, Three "myosin adenosine triphosphatase" systems: the nature of their pH lability and sulfhydryl dependence. The journal of histochemistry and cytochemistry : official journal of the, Histochemistry Society, vol.18, pp.670-672, 1970.

H. Bruunsgaard, K. Andersen-ranberg, J. Hjelmborg, B. K. Pedersen, and B. Jeune, Elevated levels of tumor necrosis factor alpha and mortality in centenarians, The American journal of medicine, vol.115, pp.278-283, 2003.

J. C. Bruusgaard, K. Liestol, and K. Gundersen, Distribution of myonuclei and microtubules in live muscle fibers of young, middle-aged, and old mice, J Appl Physiol, vol.100, pp.2024-2030, 1985.

E. Bua, J. Johnson, A. Herbst, B. Delong, D. Mckenzie et al., , 2006.

, Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers, American journal of human genetics, vol.79, pp.469-480

T. W. Buford, S. D. Anton, A. R. Judge, E. Marzetti, S. E. Wohlgemuth et al., Models of accelerated sarcopenia: critical pieces for solving the puzzle of age-related muscle atrophy, Ageing research reviews, vol.9, pp.369-383, 2010.

D. A. Calderwood, S. J. Shattil, and M. H. Ginsberg, Integrins and actin filaments: reciprocal regulation of cell adhesion and signaling, The Journal of biological chemistry, vol.275, pp.22607-22610, 2000.

R. Calvani, A. M. Joseph, P. J. Adhihetty, A. Miccheli, M. Bossola et al., Mitochondrial pathways in sarcopenia of aging and disuse muscle atrophy, Biological chemistry, vol.394, pp.393-414, 2013.

A. J. Cameron, J. E. Shaw, and P. Z. Zimmet, The metabolic syndrome: prevalence in worldwide populations, Endocrinology and metabolism clinics of North America, vol.33, pp.351-375, 2004.

M. J. Campbell, A. J. Mccomas, and F. Petito, Physiological changes in ageing muscles, Journal of neurology, vol.36, pp.174-182, 1973.

W. W. Campbell and H. J. Leidy, Dietary protein and resistance training effects on muscle and body composition in older persons, Journal of the American College of Nutrition, vol.26, pp.696-703, 2007.

D. Capitanio, M. Vasso, C. Fania, M. Moriggi, A. Vigano et al., Comparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGE, Proteomics, vol.9, pp.2004-2020, 2009.

C. Cardillo, U. Campia, C. M. Kilcoyne, M. B. Bryant, and J. A. Panza, Improved endotheliumdependent vasodilation after blockade of endothelin receptors in patients with essential hypertension, Circulation, vol.105, pp.452-456, 2002.

B. M. Carlson, E. I. Dedkov, A. B. Borisov, and J. A. Faulkner, Skeletal muscle regeneration in very old rats. The journals of gerontology Series A, Biological sciences and medical sciences, vol.56, pp.224-233, 2001.

W. T. Cefalu, Z. Q. Wang, S. Werbel, A. Bell-farrow, J. R. Crouse et al., Contribution of visceral fat mass to the insulin resistance of aging, Metabolism: clinical and experimental, vol.44, pp.954-959, 1995.

B. Chabi, M. De-camaret, B. Chevrollier, A. Boisgard, S. Stepien et al., Random mtDNA deletions and functional consequence in aged human skeletal muscle, Biochemical and biophysical research communications, vol.332, pp.542-549, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02682861

B. H. Chang and L. Chan, Regulation of Triglyceride Metabolism. III. Emerging role of lipid droplet protein ADFP in health and disease, American journal of physiology Gastrointestinal and liver physiology, vol.292, pp.1465-1468, 2007.

S. B. Charge and M. A. Rudnicki, Cellular and molecular regulation of muscle regeneration, Physiological reviews, vol.84, pp.209-238, 2004.

N. Charifi, F. Kadi, L. Feasson, F. Costes, A. Geyssant et al., Enhancement of microvessel tortuosity in the vastus lateralis muscle of old men in response to endurance training, The Journal of physiology, vol.554, pp.559-569, 2004.

B. Charvet, F. Ruggiero, L. Guellec, and D. , The development of the myotendinous junction. A review. Muscles, ligaments and tendons journal, vol.2, pp.53-63, 2012.

L. Chow, A. From, and E. Seaquist, Skeletal muscle insulin resistance: the interplay of local lipid excess and mitochondrial dysfunction, Metabolism: clinical and experimental, vol.59, pp.70-85, 2010.

T. A. Churchward-venne, L. Breen, and S. M. Phillips, Alterations in human muscle protein metabolism with aging: Protein and exercise as countermeasures to offset sarcopenia, Biofactors Clamann HP, vol.73, pp.830-843, 1993.

B. C. Clark and T. M. Manini, Sarcopenia =/= dynapenia. The journals of gerontology Series A, Biological sciences and medical sciences, vol.63, pp.829-834, 2008.

R. Close, Properties of motor units in fast and slow skeletal muscles of the rat, The Journal of physiology, vol.193, pp.45-55, 1967.

A. R. Coggan, R. J. Spina, D. S. King, M. A. Rogers, M. Brown et al., Histochemical and enzymatic comparison of the gastrocnemius muscle of young and elderly men and women, Journal of gerontology, vol.47, pp.71-76, 1992.

P. Cohen, D. R. Clemmons, and R. G. Rosenfeld, Does the GH-IGF axis play a role in cancer pathogenesis? Growth hormone & IGF research : official journal of the Growth Hormone Research Society and the, International IGF Research Society, vol.10, pp.297-305, 2000.

C. A. Collins, P. S. Zammit, A. P. Ruiz, J. E. Morgan, and T. A. Partridge, A population of myogenic stem cells that survives skeletal muscle aging, Stem Cells, vol.25, pp.885-894, 2007.

C. M. Colombo, R. M. Macedo, M. M. Fernandes-silva, A. M. Caporal, A. E. Stinghen et al., Short-term effects of moderate intensity physical activity in patients with metabolic syndrome, Einstein (Sao Paulo), vol.11, pp.324-330, 2013.

L. Combaret, D. Dardevet, D. Bechet, D. Taillandier, L. Mosoni et al., Skeletal muscle proteolysis in aging, Current opinion in clinical nutrition and metabolic care, vol.12, pp.37-41, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02661511

I. M. Conboy, M. J. Conboy, G. M. Smythe, and T. A. Rando, Notch-mediated restoration of regenerative potential to aged muscle, Science, vol.302, pp.1575-1577, 2003.

I. M. Conboy, M. J. Conboy, A. J. Wagers, E. R. Girma, I. L. Weissman et al., Rejuvenation of aged progenitor cells by exposure to a young systemic environment, Nature, vol.433, pp.760-764, 2005.

K. E. Conley, S. A. Jubrias, and P. C. Esselman, Oxidative capacity and ageing in human muscle, The Journal of physiology 526 Pt, vol.1, pp.203-210, 2000.

K. E. Conley, D. J. Marcinek, and J. Villarin, Mitochondrial dysfunction and age, Current opinion in clinical nutrition and metabolic care, vol.10, pp.688-692, 2007.

M. Conte, F. Vasuri, G. Trisolino, E. Bellavista, A. Santoro et al., Increased Plin2 expression in human skeletal muscle is associated with sarcopenia and muscle weakness, PloS one, vol.8, p.73709, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01030818

C. Cooper, R. Fielding, M. Visser, L. J. Van-loon, Y. Rolland et al., Tools in the assessment of sarcopenia, Calcified tissue international, vol.93, pp.201-210, 2013.

R. Cooper, D. Kuh, and R. Hardy, Objectively measured physical capability levels and mortality: systematic review and meta-analysis, BMJ, vol.341, p.4467, 2010.

R. N. Cooper, S. Tajbakhsh, V. Mouly, G. Cossu, M. Buckingham et al., In vivo satellite cell activation via Myf5 and MyoD in regenerating mouse skeletal muscle, Journal of cell science, vol.112, pp.2895-2901, 1999.

E. Corpas, S. M. Harman, M. A. Pineyro, R. Roberson, and M. R. Blackman, Growth hormone (GH)-releasing hormone-(1-29) twice daily reverses the decreased GH and insulin-like growth factor-I levels in old men, The Journal of clinical endocrinology and metabolism, vol.75, pp.530-535, 1992.

J. D. Crane, M. C. Devries, A. Safdar, M. J. Hamadeh, and M. A. Tarnopolsky, The effect of aging on human skeletal muscle mitochondrial and intramyocellular lipid ultrastructure. The journals of gerontology Series A, Biological sciences and medical sciences, vol.65, pp.119-128, 2010.

M. G. Cree, B. R. Newcomer, C. S. Katsanos, M. Sheffield-moore, D. Chinkes et al., Intramuscular and liver triglycerides are increased in the elderly, The Journal of clinical endocrinology and metabolism, vol.89, pp.3864-3871, 2004.

A. N. Croley, K. A. Zwetsloot, L. M. Westerkamp, N. A. Ryan, A. M. Pendergast et al., Lower capillarization, VEGF protein, and VEGF mRNA response to acute exercise in the vastus lateralis muscle of aged vs. young women, J Appl Physiol, vol.99, pp.1872-1879, 1985.

G. D'antona, M. A. Pellegrino, R. Adami, R. Rossi, C. N. Carlizzi et al., The effect of ageing and immobilization on structure and function of human skeletal muscle fibres, The Journal of physiology, vol.552, pp.499-511, 2003.

G. R. Dagenais, R. G. Tancredi, and K. L. Zierler, Free fatty acid oxidation by forearm muscle at rest, and evidence for an intramuscular lipid pool in the human forearm, The Journal of clinical investigation, vol.58, pp.421-431, 1976.

D. P. Delev, . Kostadinova, . Ii, I. D. Kostadinov, and D. K. Ubenova, Physiological and clinical characteristics of andropause, Folia medica, vol.51, pp.15-22, 2009.

T. C. Delgado, Glutamate and GABA in Appetite Regulation, Frontiers in endocrinology, vol.4, p.103, 2013.

M. J. Delmonico, T. B. Harris, J. S. Lee, M. Visser, M. Nevitt et al., Alternative definitions of sarcopenia, lower extremity performance, and functional impairment with aging in older men and women, Journal of the American Geriatrics Society, vol.55, pp.769-774, 2007.

O. Demontiero, C. Vidal, and G. Duque, Aging and bone loss: new insights for the clinician, Therapeutic advances in musculoskeletal disease, vol.4, pp.61-76, 2012.

C. Denardi, S. Ausoni, P. Moretti, L. Gorza, M. Velleca et al., Type 2X-myosin heavy chain is coded by a muscle fiber type-specific and developmentally regulated gene, The Journal of cell biology, vol.123, pp.823-835, 1993.

Y. H. Deng, H. Wang, and H. S. Zhang, Determination of amino acid neurotransmitters in human cerebrospinal fluid and saliva by capillary electrophoresis with laser-induced fluorescence detection, Journal of separation science, vol.31, pp.3088-3097, 2008.

F. A. Dinenno, N. M. Dietz, and M. J. Joyner, Aging and forearm postjunctional alphaadrenergic vasoconstriction in healthy men, Circulation, vol.106, pp.1349-1354, 2002.

F. A. Dinenno and M. J. Joyner, Alpha-adrenergic control of skeletal muscle circulation at rest and during exercise in aging humans, Microcirculation, vol.13, pp.329-341, 2006.

F. A. Dinenno, D. R. Seals, C. A. Desouza, and H. Tanaka, Age-related decreases in basal limb blood flow in humans: time course, determinants and habitual exercise effects, The Journal of physiology, vol.531, pp.573-579, 2001.

T. J. Doherty, Invited review: Aging and sarcopenia, J Appl Physiol, vol.95, pp.1717-1727, 1985.

T. J. Doherty and W. F. Brown, The estimated numbers and relative sizes of thenar motor units as selected by multiple point stimulation in young and older adults, Muscle & nerve, vol.16, pp.355-366, 1993.

A. J. Donato, L. B. Gano, I. Eskurza, A. E. Silver, P. E. Gates et al., Vascular endothelial dysfunction with aging: endothelin-1 and endothelial nitric oxide synthase, American journal of physiology Heart and circulatory physiology, vol.297, pp.425-432, 2009.

P. Doran, K. O'connell, J. Gannon, M. Kavanagh, and K. Ohlendieck, Opposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysis, Proteomics, vol.8, pp.364-377, 2008.

B. Dormont, M. Grignon, and H. Huber, Health expenditure growth: reassessing the threat of ageing, Health economics, vol.15, pp.947-963, 2006.
URL : https://hal.archives-ouvertes.fr/halshs-00181605

B. Dormont and H. Hubert, Vieillissement de la population et croissance des dépenses de santé, Collection Recherches, 2012.

H. C. Dreyer, C. E. Blanco, F. R. Sattler, E. T. Schroeder, and R. A. Wiswell, Satellite cell numbers in young and older men 24 hours after eccentric exercise, Muscle & nerve, vol.33, pp.242-253, 2006.

J. Duchateau and R. M. Enoka, Human motor unit recordings: origins and insight into the integrated motor system, Brain research, vol.1409, pp.42-61, 2011.

J. Dupâquier, Le vieillissement de la population dans le monde, 2006.

D. Dykiert, G. Der, J. M. Starr, and I. J. Deary, Age differences in intra-individual variability in simple and choice reaction time: systematic review and meta-analysis, PloS one, vol.7, p.45759, 2012.

J. O. Ebbert and M. D. Jensen, Fat depots, free fatty acids, and dyslipidemia, Nutrients, vol.5, pp.498-508, 2013.

T. J. Ebert, B. J. Morgan, J. A. Barney, T. Denahan, and J. J. Smith, Effects of aging on baroreflex regulation of sympathetic activity in humans, The American journal of physiology, vol.263, pp.798-803, 1992.

R. H. Eckel, S. M. Grundy, and P. Z. Zimmet, The metabolic syndrome, Lancet, vol.365, pp.1415-1428, 2005.

, Economic UNDo, Division SAP (2010) World Population Ageing 2009: United Nations, Department of Economic and Social Affairs

E. Edstrom and B. Ulfhake, Sarcopenia is not due to lack of regenerative drive in senescent skeletal muscle, Aging cell, vol.4, pp.65-77, 2005.

D. Einhorn, G. M. Reaven, R. H. Cobin, E. Ford, O. P. Ganda et al., American College of Endocrinology position statement on the insulin resistance syndrome. Endocrine practice : official journal of the American College of Endocrinology and the, American Association of Clinical Endocrinologists, vol.9, pp.237-252, 2003.

S. Ennion, &. Sant, J. Pereira, A. J. Sargeant, A. Young et al., Characterization of human skeletal muscle fibres according to the myosin heavy chains they express, Journal of muscle research and cell motility, vol.16, pp.35-43, 1995.

J. Epelbaum, Neuroendocrinology and aging, Journal of neuroendocrinology, vol.20, pp.808-811, 2008.

W. B. Ershler and E. T. Keller, Age-associated increased interleukin-6 gene expression, latelife diseases, and frailty, Annual review of medicine, vol.51, pp.245-270, 2000.

J. A. Faulkner, L. M. Larkin, D. R. Claflin, and S. V. Brooks, Age-related changes in the structure and function of skeletal muscles, Clinical and experimental pharmacology & physiology, vol.34, pp.1091-1096, 2007.

F. Feihl, L. Liaudet, B. Waeber, and B. I. Levy, Hypertension: a disease of the microcirculation?, Hypertension, vol.48, pp.1012-1017, 2006.

E. Ferrannini, S. M. Haffner, B. D. Mitchell, and M. P. Stern, Hyperinsulinaemia: the key feature of a cardiovascular and metabolic syndrome, Diabetologia, vol.34, pp.416-422, 1991.

E. S. Ford, Risks for all-cause mortality, cardiovascular disease, and diabetes associated with the metabolic syndrome: a summary of the evidence, Diabetes care, vol.28, pp.1769-1778, 2005.

E. S. Ford, W. H. Giles, and W. H. Dietz, Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey, JAMA : the journal of the American Medical Association, vol.287, pp.356-359, 2002.

E. S. Ford, C. Li, and G. Zhao, Prevalence and correlates of metabolic syndrome based on a harmonious definition among adults in the US, Journal of diabetes, vol.2, pp.180-193, 2010.

A. Fournier, S. Mesrine, M. C. Boutron-ruault, and F. Clavel-chapelon, Estrogen-progestagen menopausal hormone therapy and breast cancer: does delay from menopause onset to treatment initiation influence risks, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, vol.27, pp.5138-5143, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-01319999

C. Franceschi, M. Bonafe, S. Valensin, F. Olivieri, D. Luca et al., Inflamm-aging. An evolutionary perspective on immunosenescence, Annals of the New York Academy of Sciences, vol.908, pp.244-254, 2000.

W. R. Frontera, V. A. Hughes, R. A. Fielding, M. A. Fiatarone, W. J. Evans et al., Aging of skeletal muscle: a 12-yr longitudinal study, J Appl Physiol, vol.88, pp.1321-1326, 1985.

I. Gabriely, X. H. Ma, X. M. Yang, G. Atzmon, M. W. Rajala et al., Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process, Diabetes, vol.51, pp.2951-2958, 2002.

A. S. Gami, B. J. Witt, D. E. Howard, P. J. Erwin, L. A. Gami et al., Metabolic syndrome and risk of incident cardiovascular events and death: a systematic review and meta-analysis of longitudinal studies, Journal of the American College of Cardiology, vol.49, pp.403-414, 2007.

J. Gannon, P. Doran, A. Kirwan, and K. Ohlendieck, Drastic increase of myosin light chain MLC-2 in senescent skeletal muscle indicates fast-to-slow fibre transition in sarcopenia of old age, Eur J Cell Biol, vol.88, pp.685-700, 2009.

Y. Gao, T. Y. Kostrominova, J. A. Faulkner, and A. S. Wineman, Age-related changes in the mechanical properties of the epimysium in skeletal muscles of rats, Journal of biomechanics, vol.41, pp.465-469, 2008.

T. P. Gavin, R. S. Ruster, J. A. Carrithers, K. A. Zwetsloot, R. M. Kraus et al., No difference in the skeletal muscle angiogenic response to aerobic exercise training between young and aged men, The Journal of physiology, vol.585, pp.231-239, 2007.

C. Gelfi, A. Vigano, M. Ripamonti, A. Pontoglio, S. Begum et al., The human muscle proteome in aging, Journal of proteome research, vol.5, pp.1344-1353, 2006.

E. Ghigo, E. Arvat, L. Gianotti, J. Ramunni, L. Divito et al., Human aging and the GH-IGF-I axis, Journal of pediatric endocrinology & metabolism : JPEM, vol.9, pp.271-278, 1996.

M. C. Gibson and E. Schultz, Age-related differences in absolute numbers of skeletal muscle satellite cells, Muscle & nerve, vol.6, pp.574-580, 1983.

R. Giordano, L. Bonelli, E. Marinazzo, E. Ghigo, and E. Arvat, Growth hormone treatment in human ageing: benefits and risks, Hormones (Athens), vol.7, pp.133-139, 2008.

G. Goldspink, K. Fernandes, P. E. Williams, and D. J. Wells, Age-related changes in collagen gene expression in the muscles of mdx dystrophic and normal mice, Neuromuscular disorders : NMD, vol.4, pp.183-191, 1994.

A. V. Gomes, J. D. Potter, and D. Szczesna-cordary, The role of troponins in muscle contraction, IUBMB life, vol.54, pp.323-333, 2002.

B. H. Goodpaster, J. He, S. Watkins, and D. E. Kelley, Skeletal muscle lipid content and insulin resistance: evidence for a paradox in endurance-trained athletes, The Journal of clinical endocrinology and metabolism, vol.86, pp.5755-5761, 2001.

B. H. Goodpaster, S. W. Park, T. B. Harris, S. B. Kritchevsky, M. Nevitt et al., The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study. The journals of gerontology Series A, Biological sciences and medical sciences, vol.61, pp.1059-1064, 2006.

G. H. Goossens, The role of adipose tissue dysfunction in the pathogenesis of obesityrelated insulin resistance, Physiology & behavior, vol.94, pp.206-218, 2008.

A. M. Gordon, E. Homsher, and M. Regnier, Regulation of contraction in striated muscle, Physiological reviews, vol.80, pp.853-924, 2000.

D. Grady, S. M. Rubin, D. B. Petitti, C. S. Fox, D. Black et al., Hormone therapy to prevent disease and prolong life in postmenopausal women, Annals of internal medicine, vol.117, pp.1016-1037, 1992.

H. L. Granzier and S. Labeit, Titin and its associated proteins: the third myofilament system of the sarcomere, Advances in protein chemistry, vol.71, pp.89-119, 2005.

H. J. Groot, J. D. Trinity, G. Layec, M. J. Rossman, S. J. Ives et al., Perfusion pressure and movement-induced hyperemia: evidence of limited vascular function and vasodilatory reserve with age, American journal of physiology Heart and circulatory physiology, vol.304, pp.610-619, 2013.

S. M. Grundy, Atherogenic dyslipidemia associated with metabolic syndrome and insulin resistance, Clinical cornerstone, vol.8, issue.1, pp.21-27, 2006.

S. M. Grundy, Metabolic syndrome: a multiplex cardiovascular risk factor, The Journal of clinical endocrinology and metabolism, vol.92, pp.399-404, 2007.

S. M. Grundy, Metabolic syndrome pandemic, Arteriosclerosis, thrombosis, and vascular biology, vol.28, pp.629-636, 2008.

S. M. Grundy, J. I. Cleeman, S. R. Daniels, K. A. Donato, R. H. Eckel et al., Diagnosis and management of the metabolic syndrome. An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Executive summary, Cardiology in review, vol.13, pp.322-327, 2005.

C. Guillet and Y. Boirie, Insulin resistance: a contributing factor to age-related muscle mass loss?, Diabetes & metabolism, vol.31, issue.2, pp.5-20, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02679999

Y. Guo, K. R. Cordes, R. V. Farese, J. Walther, and T. C. , Lipid droplets at a glance, Journal of cell science, vol.122, pp.749-752, 2009.

K. Hakkinen, W. J. Kraemer, R. U. Newton, and M. Alen, Changes in electromyographic activity, muscle fibre and force production characteristics during heavy resistance/power strength training in middle-aged and older men and women, Acta physiologica Scandinavica, vol.171, pp.51-62, 2001.

A. H. Hansen, J. J. Nielsen, B. Saltin, and Y. Hellsten, Exercise training normalizes skeletal muscle vascular endothelial growth factor levels in patients with essential hypertension, Journal of hypertension, vol.28, pp.1176-1185, 2010.

A. H. Hansen, M. Nyberg, J. Bangsbo, B. Saltin, and Y. Hellsten, Exercise training alters the balance between vasoactive compounds in skeletal muscle of individuals with essential hypertension, Hypertension, vol.58, pp.943-949, 2011.

D. Harman, Free radical theory of aging: an update: increasing the functional life span, Annals of the New York Academy of Sciences, vol.1067, pp.10-21, 2006.

D. E. Harrison, R. Strong, Z. D. Sharp, J. F. Nelson, C. M. Astle et al., Rapamycin fed late in life extends lifespan in genetically heterogeneous mice, Nature, vol.460, pp.392-395, 2009.

A. Hedman, R. Reneland, and H. O. Lithell, Alterations in skeletal muscle morphology in glucose-tolerant elderly hypertensive men: relationship to development of hypertension and heart rate, Journal of hypertension, vol.18, pp.559-565, 2000.

H. A. Henrich, W. Romen, W. Heimgartner, E. Hartung, and F. Baumer, Capillary rarefaction characteristic of the skeletal muscle of hypertensive patients, Klinische Wochenschrift, vol.66, pp.54-60, 1988.

N. Hernandez, S. H. Torres, H. J. Finol, and O. Vera, Capillary changes in skeletal muscle of patients with essential hypertension, The Anatomical record, vol.256, pp.425-432, 1999.

A. G. Hindle, M. Horning, J. A. Mellish, and J. M. Lawler, Diving into old age: muscular senescence in a large-bodied, long-lived mammal, the Weddell seal (Leptonychotes weddellii), The Journal of experimental biology, vol.212, pp.790-796, 2009.

A. Hiona and C. Leeuwenburgh, The role of mitochondrial DNA mutations in aging and sarcopenia: implications for the mitochondrial vicious cycle theory of aging, Experimental gerontology, vol.43, pp.24-33, 2008.

A. Hiona, A. Sanz, G. C. Kujoth, R. Pamplona, A. Y. Seo et al., Mitochondrial DNA mutations induce mitochondrial dysfunction, apoptosis and sarcopenia in skeletal muscle of mitochondrial DNA mutator mice, PloS one, vol.5, p.11468, 2010.

E. F. Hodson-tole and J. M. Wakeling, Motor unit recruitment for dynamic tasks: current understanding and future directions, Journal of comparative physiology B, Biochemical, systemic, and environmental physiology, vol.179, pp.57-66, 2009.

K. Hojlund, Z. Yi, H. Hwang, B. Bowen, N. Lefort et al., Characterization of the human skeletal muscle proteome by onedimensional gel electrophoresis and HPLC-ESI-MS/MS, Molecular & cellular proteomics : MCP, vol.7, pp.257-267, 2008.

H. Hotta and S. Uchida, Aging of the autonomic nervous system and possible improvements in autonomic activity using somatic afferent stimulation, Geriatrics & gerontology international, vol.10, pp.127-136, 2010.

J. A. Houmard, M. L. Weidner, K. E. Gavigan, G. L. Tyndall, M. S. Hickey et al., Fiber type and citrate synthase activity in the human gastrocnemius and vastus lateralis with aging, J Appl Physiol, vol.85, pp.1337-1341, 1985.

D. F. Hultsch, S. W. Macdonald, and R. A. Dixon, Variability in reaction time performance of younger and older adults, The journals of gerontology Series B, Psychological sciences and social sciences, vol.57, pp.101-115, 2002.

E. Hutter, M. Skovbro, B. Lener, C. Prats, R. Rabol et al., Oxidative stress and mitochondrial impairment can be separated from lipofuscin accumulation in aged human skeletal muscle, Aging cell, vol.6, pp.245-256, 2007.

A. F. Huxley, Muscle structure and theories of contraction, Progress in biophysics and biophysical chemistry, vol.7, pp.255-318, 1957.

H. Ishikawa, Formation of elaborate networks of T-system tubules in cultured skeletal muscle with special reference to the T-system formation, The Journal of cell biology, vol.38, pp.51-66, 1968.

M. P. Jacob, Extracellular matrix and vascular ageing, Medecine sciences : M/S, vol.22, pp.273-278, 2006.

C. M. Jankowski, W. S. Gozansky, R. E. Van-pelt, P. Wolfe, R. S. Schwartz et al., Oral dehydroepiandrosterone replacement in older adults: effects on central adiposity, glucose metabolism and blood lipids, Clinical endocrinology, vol.75, pp.456-463, 2011.

I. Janssen, S. B. Heymsfield, and R. Ross, Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability, Journal of the American Geriatrics Society, vol.50, pp.889-896, 2002.

I. Janssen, D. S. Shepard, P. T. Katzmarzyk, and R. Roubenoff, The healthcare costs of sarcopenia in the United States, Journal of the American Geriatrics Society, vol.52, pp.80-85, 2004.

D. L. Johannsen and E. Ravussin, Can increased muscle ROS scavenging keep older animals young and metabolically fit, Cell metabolism, vol.12, pp.557-558, 2010.

R. Jurca, M. J. Lamonte, C. E. Barlow, J. B. Kampert, T. S. Church et al., Association of muscular strength with incidence of metabolic syndrome in men, Medicine and science in sports and exercise, vol.37, pp.1849-1855, 2005.

F. Kadi, N. Charifi, C. Denis, and J. Lexell, Satellite cells and myonuclei in young and elderly women and men, Muscle & nerve, vol.29, pp.120-127, 2004.

I. Karch, M. Olszowska, T. Pajak, L. Drapisz, S. Luszczak et al., The effect of physical activity on serum levels of selected biomarkers of atherosclerosis, Kardiologia polska, vol.71, pp.55-60, 2013.

J. S. Kim, D. J. Kosek, J. K. Petrella, J. M. Cross, and M. M. Bamman, Resting and load-induced levels of myogenic gene transcripts differ between older adults with demonstrable sarcopenia and young men and women, J Appl Physiol, vol.99, pp.2149-2158, 1985.

K. G. Kinsella, H. Wan, and C. Usbot, An Aging World, vol.9, p.95, 2008.

S. Kirkeby and C. Garbarsch, Aging affects different human muscles in various ways. An image analysis of the histomorphometric characteristics of fiber types in human masseter and vastus lateralis muscles from young adults and the very old, Histology and histopathology, vol.15, pp.61-71, 2000.

M. Kjaer, Role of extracellular matrix in adaptation of tendon and skeletal muscle to mechanical loading, Physiological reviews, vol.84, pp.649-698, 2004.

H. Klitgaard, M. Mantoni, S. Schiaffino, S. Ausoni, L. Gorza et al., ) Function, morphology and protein expression of ageing skeletal muscle: a cross-sectional study of elderly men with different training backgrounds, Acta physiologica Scandinavica, vol.140, pp.41-54, 1990.

D. J. Kosek, J. S. Kim, J. K. Petrella, J. M. Cross, and M. M. Bamman, Efficacy of 3 days/wk resistance training on myofiber hypertrophy and myogenic mechanisms in young vs. older adults, J Appl Physiol, vol.101, pp.531-544, 1985.

T. W. Kragstrup, M. Kjaer, and A. L. Mackey, Structural, biochemical, cellular, and functional changes in skeletal muscle extracellular matrix with aging, Scandinavian journal of medicine & science in sports, vol.21, pp.749-757, 2011.

M. Kruger, J. Wright, and K. Wang, Nebulin as a length regulator of thin filaments of vertebrate skeletal muscles: correlation of thin filament length, nebulin size, and epitope profile, The Journal of cell biology, vol.115, pp.97-107, 1991.

Y. Kushnareva, A. N. Murphy, and A. Andreyev, Complex I-mediated reactive oxygen species generation: modulation by cytochrome c and NAD(P)+ oxidation-reduction state, The Biochemical journal, vol.368, pp.545-553, 2002.

F. Labrie, A. Belanger, J. Simard, L. Van, and C. Labrie, DHEA and peripheral androgen and estrogen formation: intracinology, Annals of the New York Academy of Sciences, vol.774, pp.16-28, 1995.

E. G. Lakatta, Changes in cardiovascular function with aging, European heart journal, p.11, 1990.

C. Suppl, , pp.22-29

E. G. Lakatta, Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part III: cellular and molecular clues to heart and arterial aging, Circulation, vol.107, pp.490-497, 2003.

J. T. Lanner, D. K. Georgiou, A. D. Joshi, and S. L. Hamilton, Ryanodine receptors: structure, expression, molecular details, and function in calcium release, Cold Spring Harbor perspectives in biology, vol.2, p.3996, 2010.

P. M. Lansdorp, W. Dragowska, T. E. Thomas, M. T. Little, and H. Mayani, Age-related decline in proliferative potential of purified stem cell candidates, Blood cells, vol.20, pp.380-371, 1994.

I. R. Lanza, D. K. Short, K. R. Short, S. Raghavakaimal, R. Basu et al., Endurance exercise as a countermeasure for aging, Diabetes, vol.57, pp.2933-2942, 2008.

R. G. Larsen, D. M. Callahan, S. A. Foulis, and J. A. Kent-braun, Age-related changes in oxidative capacity differ between locomotory muscles and are associated with physical activity behavior. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme, vol.37, pp.88-99, 2012.

L. Larsson, L. Edstrom, B. Lindegren, L. Gorza, and S. Schiaffino, MHC composition and enzyme-histochemical and physiological properties of a novel fast-twitch motor unit type, The American journal of physiology, vol.261, pp.93-101, 1991.

L. Larsson, X. Li, and W. R. Frontera, Effects of aging on shortening velocity and myosin isoform composition in single human skeletal muscle cells, The American journal of physiology, vol.272, pp.638-649, 1997.

J. E. Layne and M. E. Nelson, The effects of progressive resistance training on bone density: a review, Medicine and science in sports and exercise, vol.31, pp.25-30, 1999.

L. Garrec, M. A. Bouvet, M. Koubi, and M. , Les comptes nationaux de la santé en 2011, Etudes et Résultats. DRESS, 2012.

L. Lay, S. Dugail, and I. , Connecting lipid droplet biology and the metabolic syndrome, Progress in lipid research, vol.48, pp.191-195, 2009.

H. Y. Lee, C. S. Choi, A. L. Birkenfeld, T. C. Alves, F. R. Jornayvaz et al., Targeted expression of catalase to mitochondria prevents age-associated reductions in mitochondrial function and insulin resistance, Cell metabolism, vol.12, pp.668-674, 2010.

S. Lee, S. Y. Jeong, W. C. Lim, S. Kim, Y. Y. Park et al., Mitochondrial fission and fusion mediators, hFis1 and OPA1, modulate cellular senescence, The Journal of biological chemistry, vol.282, pp.22977-22983, 2007.

S. H. Lee and K. J. Min, Caloric restriction and its mimetics, BMB reports, vol.46, pp.181-187, 2013.

J. Lexell and D. Y. Downham, The occurrence of fibre-type grouping in healthy human muscle: a quantitative study of cross-sections of whole vastus lateralis from men between 15 and 83 years, Acta neuropathologica, vol.81, pp.377-381, 1991.

J. Lexell, K. Henriksson-larsen, B. Winblad, and M. Sjostrom, Distribution of different fiber types in human skeletal muscles: effects of aging studied in whole muscle cross sections, Muscle & nerve, vol.6, pp.588-595, 1983.

J. Lexell and C. C. Taylor, Variability in muscle fibre areas in whole human quadriceps muscle: effects of increasing age, Journal of anatomy, vol.174, pp.239-249, 1991.

J. Lexell, C. C. Taylor, and M. Sjostrom, 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, Journal of the neurological sciences, vol.84, pp.275-294, 1988.

C. Leyvraz, C. Verdumo, and V. Giusti, , 2008.

, Revue medicale suisse, vol.4, pp.844-847

A. Listrat, B. Picard, and Y. Geay, Age-related changes and location of type I, III, IV, V and VI collagens during development of four foetal skeletal muscles of double-muscled and normal bovine animals, Tissue & cell, vol.31, pp.17-27, 1999.
URL : https://hal.archives-ouvertes.fr/hal-02688751

J. Lloberas and A. Celada, Effect of aging on macrophage function, Experimental gerontology, vol.37, pp.1325-1331, 2002.

A. Lombardi, E. Silvestri, F. Cioffi, R. Senese, A. Lanni et al., Defining the transcriptomic and proteomic profiles of rat ageing skeletal muscle by the use of a cDNA array, 2D-and Blue native-PAGE approach, Journal of proteomics, vol.72, pp.708-721, 2009.

B. B. Lowell and G. I. Shulman, Mitochondrial dysfunction and type 2 diabetes, Science, vol.307, pp.384-387, 2005.

X. Lu, J. Gruia-gray, N. G. Copeland, D. J. Gilbert, N. A. Jenkins et al., The murine perilipin gene: the lipid droplet-associated perilipins derive from tissue-specific, mRNA splice variants and define a gene family of ancient origin, Mammalian genome : official journal of the International Mammalian Genome Society, vol.12, pp.741-749, 2001.

R. W. Lymn and E. W. Taylor, Mechanism of adenosine triphosphate hydrolysis by actomyosin, Biochemistry, vol.10, pp.4617-4624, 1971.

S. Martin and R. G. Parton, Lipid droplets: a unified view of a dynamic organelle, Nature reviews Molecular cell biology, vol.7, pp.373-378, 2006.

E. Marzetti, R. Calvani, M. Cesari, T. W. Buford, M. Lorenzi et al., Mitochondrial dysfunction and sarcopenia of aging: from signaling pathways to clinical trials, The international journal of biochemistry & cell biology, vol.45, pp.2288-2301, 2013.

A. Mauro, Satellite cell of skeletal muscle fibers, The Journal of biophysical and biochemical cytology, vol.9, pp.493-495, 1961.

R. Mayne and R. D. Sanderson, The extracellular matrix of skeletal muscle, Collagen and related research, vol.5, pp.449-468, 1985.

W. D. Mcardle, F. L. Katch, and V. L. Katch, Exercice physiology: Energy, nutrition and human performance, 2007.

K. M. Mccormick, K. M. Baldwin, and F. Schachat, Coordinate changes in C protein and myosin expression during skeletal muscle hypertrophy, The American journal of physiology, vol.267, pp.443-449, 1994.

A. S. Mcelhinny, S. T. Kazmierski, S. Labeit, and C. C. Gregorio, Nebulin: the nebulous, multifunctional giant of striated muscle, Trends in cardiovascular medicine, vol.13, pp.195-201, 2003.

I. B. Mcinnes and G. Schett, Cytokines in the pathogenesis of rheumatoid arthritis, Nature reviews Immunology, vol.7, pp.429-442, 2007.

B. R. Mckay, C. E. O'reilly, S. M. Phillips, M. A. Tarnopolsky, and G. Parise, Co-expression of IGF-1 family members with myogenic regulatory factors following acute damaging musclelengthening contractions in humans, The Journal of physiology, vol.586, pp.5549-5560, 2008.

S. H. Mckiernan, E. Bua, J. Mcgorray, and J. Aiken, Early-onset calorie restriction conserves fiber number in aging rat skeletal muscle, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, vol.18, pp.580-581, 2004.

C. J. Mcneil, T. J. Doherty, D. W. Stashuk, and C. L. Rice, Motor unit number estimates in the tibialis anterior muscle of young, old, and very old men, Muscle & nerve, vol.31, pp.461-467, 2005.

S. Mekrami and T. N. Brignol, Le muscle squelettique. Repères Savoir & Comprendre. Association Française contre les Myopathies, 2003.

J. F. Merlen, The physiology of microcirculation, Phlebologie, vol.29, pp.237-243, 1976.

B. J. Merry, Molecular mechanisms linking calorie restriction and longevity, The international journal of biochemistry & cell biology, vol.34, pp.1340-1354, 2002.

W. K. Mitchell, J. Williams, P. Atherton, M. Larvin, J. Lund et al., Sarcopenia, dynapenia, and the impact of advancing age on human skeletal muscle size and strength; a quantitative review, Frontiers in physiology, vol.3, p.260, 2012.

V. M. Monnier, G. T. Mustata, K. L. Biemel, O. Reihl, M. O. Lederer et al., , 2005.

, Cross-linking of the extracellular matrix by the maillard reaction in aging and diabetes: an update on "a puzzle nearing resolution, Annals of the New York Academy of Sciences, vol.1043, pp.533-544

M. K. Moon, B. J. Cho, Y. J. Lee, S. H. Choi, S. Lim et al., The effects of chronic exercise on the inflammatory cytokines interleukin-6 and tumor necrosis factoralpha are different with age. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme, vol.37, pp.631-636, 2012.

S. S. Moon, Low skeletal muscle mass is associated with insulin resistance, diabetes, and metabolic syndrome in the Korean population: The Korea National Health and Nutrition Examination Survey (KNHANES), Endocrine journal, pp.2009-2010, 2013.

I. Moppett, Basic principles of control of regional blood flow in vascular beds, Surgery, vol.30, pp.365-369, 2012.

F. Mora, G. Segovia, D. Arco, A. De-blas, M. Garrido et al., Stress, neurotransmitters, corticosterone and body-brain integration, Brain research, vol.1476, pp.71-85, 2012.

J. A. Morais, S. Chevalier, and R. Gougeon, Protein turnover and requirements in the healthy and frail elderly. The journal of nutrition, health & aging, vol.10, pp.272-283, 2006.

J. H. Morrison and P. R. Hof, Life and death of neurons in the aging brain, Science, vol.278, pp.412-419, 1997.

J. M. Muller-delp, A. N. Gurovich, D. D. Christou, and C. Leeuwenburgh, Redox balance in the aging microcirculation: new friends, new foes, and new clinical directions, vol.19, pp.19-28, 2012.

J. M. Muller-delp, S. A. Spier, M. W. Ramsey, and M. D. Delp, Aging impairs endotheliumdependent vasodilation in rat skeletal muscle arterioles, American journal of physiology Heart and circulatory physiology, vol.283, pp.1662-1672, 2002.

A. Musaro, C. De-angelis, M. G. Germani, A. Ciccarelli, C. Molinaro et al., Enhanced expression of myogenic regulatory genes in aging skeletal muscle, Experimental cell research, vol.221, pp.241-248, 1995.

T. Nakamura and R. L. Prewitt, Effect of NG-monomethyl L-arginine on endotheliumdependent relaxation in arterioles of one-kidney, one clip hypertensive rats, Hypertension, vol.17, pp.875-880, 1991.

M. V. Narici and N. Maffulli, Sarcopenia: characteristics, mechanisms and functional significance, British medical bulletin, vol.95, pp.139-159, 2010.

. Ncpe/atpiii, Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report, Circulation, vol.106, pp.3143-3421, 2002.

D. M. Needham, Red and white muscles, Physiological reviews, vol.6, pp.1-27, 1926.

S. C. Newcomer, U. A. Leuenberger, C. S. Hogeman, and D. N. Proctor, Heterogeneous vasodilator responses of human limbs: influence of age and habitual endurance training, American journal of physiology Heart and circulatory physiology, vol.289, pp.308-315, 2005.

A. B. Newman, V. Kupelian, M. Visser, E. Simonsick, B. Goodpaster et al., Sarcopenia: alternative definitions and associations with lower extremity function, Journal of the American Geriatrics Society, vol.51, pp.1602-1609, 2003.

J. G. Nickerson, H. Alkhateeb, C. R. Benton, J. Lally, J. Nickerson et al., Greater transport efficiencies of the membrane fatty acid transporters FAT/CD36 and FATP4 compared with FABPpm and FATP1 and differential effects on fatty acid esterification and oxidation in rat skeletal muscle, The Journal of biological chemistry, vol.284, pp.16522-16530, 2009.

R. Nilwik, T. Snijders, M. Leenders, B. B. Groen, J. Van-kranenburg et al., The decline in skeletal muscle mass with aging is mainly attributed to a reduction in type II muscle fiber size, Experimental gerontology, vol.48, pp.492-498, 2013.

S. K. Nishiyama, D. W. Wray, and R. S. Richardson, Aging affects vascular structure and function in a limb-specific manner, J Appl Physiol, vol.105, pp.1661-1670, 1985.

P. Novak and T. Soukup, Calsequestrin distribution, structure and function, its role in normal and pathological situations and the effect of thyroid hormones, Academia Scientiarum Bohemoslovaca, vol.60, pp.439-452, 2011.

K. O'connell and K. Ohlendieck, Proteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscle, Proteomics, vol.9, pp.5509-5524, 2009.

L. O'mahony, J. Holland, J. Jackson, C. Feighery, T. P. Hennessy et al., Quantitative intracellular cytokine measurement: age-related changes in proinflammatory cytokine production, Clinical and experimental immunology, vol.113, pp.213-219, 1998.

T. Okagaki, F. E. Weber, D. A. Fischman, K. T. Vaughan, T. Mikawa et al., The major myosin-binding domain of skeletal muscle MyBP-C (C protein) resides in the COOHterminal, immunoglobulin C2 motif, The Journal of cell biology, vol.123, pp.619-626, 1993.

G. Olivetti, G. Giordano, D. Corradi, M. Melissari, C. Lagrasta et al., Gender differences and aging: effects on the human heart, Journal of the American College of Cardiology, vol.26, pp.1068-1079, 1995.

B. Onken and M. Driscoll, Metformin induces a dietary restriction-like state and the oxidative stress response to extend C. elegans Healthspan via AMPK, LKB1, and SKN-1, PloS one, vol.5, p.8758, 2010.

E. F. Pace-schott and R. M. Spencer, Age-related changes in the cognitive function of sleep, Progress in brain research, vol.191, pp.75-89, 2011.

D. A. Pan, S. Lillioja, A. D. Kriketos, M. R. Milner, L. A. Baur et al., Skeletal muscle triglyceride levels are inversely related to insulin action, Diabetes, vol.46, pp.983-988, 1997.

J. A. Panza, C. E. Garcia, C. M. Kilcoyne, A. A. Quyyumi, and R. O. Cannon, Impaired endothelium-dependent vasodilation in patients with essential hypertension. Evidence that nitric oxide abnormality is not localized to a single signal transduction pathway, Circulation, vol.3, pp.1732-1738, 1995.

I. Papa, C. Astier, O. Kwiatek, F. Raynaud, C. Bonnal et al., Alpha actinin-CapZ, an anchoring complex for thin filaments in Z-line, Journal of muscle research and cell motility, vol.20, pp.187-197, 1999.

C. T. Pappas, N. Bhattacharya, J. A. Cooper, and C. C. Gregorio, Nebulin interacts with CapZ and regulates thin filament architecture within the Z-disc, Molecular biology of the cell, vol.19, pp.1837-1847, 2008.

J. V. Pardo, J. D. Siliciano, and S. W. Craig, A vinculin-containing cortical lattice in skeletal muscle: transverse lattice elements ("costameres") mark sites of attachment between myofibrils and sarcolemma, Proceedings of the National Academy of Sciences of the United States of America, vol.80, pp.1008-1012, 1983.

Y. Park, R. D. Prisby, B. J. Behnke, J. M. Dominguez, L. A. Lesniewski et al., Effects of aging, TNF-alpha, and exercise training on angiotensin IIinduced vasoconstriction of rat skeletal muscle arterioles, J Appl Physiol, vol.113, pp.1091-1100, 1985.

A. C. Pearson, C. V. Gudipati, and A. J. Labovitz, Effects of aging on left ventricular structure and function, American heart journal, vol.121, pp.871-875, 1991.

G. Perseghin, P. Scifo, D. Cobelli, F. Pagliato, E. Battezzati et al., Intramyocellular triglyceride content is a determinant of in vivo insulin resistance in humans: a 1H-13C nuclear magnetic resonance spectroscopy assessment in offspring of type 2 diabetic parents, Diabetes, vol.48, pp.1600-1606, 1999.

K. F. Petersen, D. Befroy, S. Dufour, J. Dziura, C. Ariyan et al., Mitochondrial dysfunction in the elderly: possible role in insulin resistance, Science, vol.300, pp.1140-1142, 2003.

K. F. Petersen, S. Dufour, D. B. Savage, S. Bilz, G. Solomon et al., The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.12587-12594, 2007.

C. M. Peterson, D. L. Johannsen, and E. Ravussin, Skeletal muscle mitochondria and aging: a review, Journal of aging research, vol.2012, p.194821, 2012.

B. J. Petrof, J. B. Shrager, H. H. Stedman, A. M. Kelly, and H. L. Sweeney, Dystrophin protects the sarcolemma from stresses developed during muscle contraction, Proceedings of the National Academy of Sciences of the United States of America, vol.90, pp.3710-3714, 1993.

D. Pette, H. Peuker, and R. S. Staron, The impact of biochemical methods for single muscle fibre analysis, Acta physiologica Scandinavica, vol.166, pp.261-277, 1999.

G. Pfister, D. Weiskopf, L. Lazuardi, R. D. Kovaiou, D. P. Cioca et al., Naive T cells in the elderly: are they still there?, Annals of the New York Academy of Sciences, vol.1067, pp.152-157, 2006.

M. Picard, D. Ritchie, K. J. Wright, C. Romestaing, M. M. Thomas et al., Mitochondrial functional impairment with aging is exaggerated in isolated mitochondria compared to permeabilized myofibers, Aging cell, vol.9, pp.1032-1046, 2010.

I. Piec, A. Listrat, J. Alliot, C. Chambon, R. G. Taylor et al., Differential proteome analysis of aging in rat skeletal muscle, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, vol.19, pp.1143-1145, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680775

P. Poggi, C. Marchetti, and R. Scelsi, Automatic morphometric analysis of skeletal muscle fibers in the aging man, The Anatomical record, vol.217, pp.30-34, 1987.

D. C. Poole and O. Mathieu-costello, Relationship between fiber capillarization and mitochondrial volume density in control and trained rat soleus and plantaris muscles, Microcirculation, vol.3, pp.175-186, 1996.

G. A. Porter, G. M. Dmytrenko, J. C. Winkelmann, and R. J. Bloch, Dystrophin colocalizes with beta-spectrin in distinct subsarcolemmal domains in mammalian skeletal muscle, The Journal of cell biology, vol.117, pp.997-1005, 1992.

H. J. Priebe, The aged cardiovascular risk patient, British journal of anaesthesia, vol.85, pp.763-778, 2000.

D. N. Proctor, W. E. Sinning, J. M. Walro, G. C. Sieck, and P. W. Lemon, Oxidative capacity of human muscle fiber types: effects of age and training status, J Appl Physiol, vol.78, pp.2033-2038, 1985.

M. K. Pugsley and R. Tabrizchi, The vascular system. An overview of structure and function, Journal of pharmacological and toxicological methods, vol.44, pp.333-340, 2000.

P. P. Purslow, The structure and functional significance of variations in the connective tissue within muscle. Comparative biochemistry and physiology Part A, Molecular & integrative physiology, vol.133, pp.947-966, 2002.

P. P. Purslow and V. C. Duance, The structure and function of intramuscular connective tissue. Connective tissue matrix, vol.2, pp.127-166, 1990.

K. S. Ramaswamy, M. L. Palmer, J. H. Van-der-meulen, A. Renoux, T. Y. Kostrominova et al., Lateral transmission of force is impaired in skeletal muscles of dystrophic mice and very old rats, The Journal of physiology, vol.589, pp.1195-1208, 2011.

I. Rayment, H. M. Holden, M. Whittaker, C. B. Yohn, M. Lorenz et al., Structure of the actin-myosin complex and its implications for muscle contraction, Science, vol.261, pp.58-65, 1993.

G. M. Reaven, Banting lecture 1988. Role of insulin resistance in human disease, Diabetes, vol.37, pp.1595-1607, 1988.

G. M. Reaven, Pathophysiology of insulin resistance in human disease, Physiological reviews, vol.75, pp.473-486, 1995.

V. Renault, L. E. Thornell, P. O. Eriksson, G. Butler-browne, and V. Mouly, Regenerative potential of human skeletal muscle during aging, Aging cell, vol.1, pp.132-139, 2002.

I. Robert-bobée, Projections de population pour la France métropolitaine à l'horizon 2050, 2006.

C. J. Rodrigues, R. Junior, A. J. Bohm, and G. M. , Effects of aging on muscle fibers and collagen content of the diaphragm: a comparison with the rectus abdominis muscle, Gerontology, vol.42, pp.218-228, 1996.

M. R. Roos, C. L. Rice, and A. A. Vandervoort, Age-related changes in motor unit function, Muscle & nerve, vol.20, pp.679-690, 1997.

I. H. Rosenberg, Sarcopenia: origins and clinical relevance, The Journal of nutrition, vol.127, pp.990-991, 1997.

D. Rossi, V. Barone, E. Giacomello, V. Cusimano, and V. Sorrentino, The sarcoplasmic reticulum: an organized patchwork of specialized domains, Traffic, vol.9, pp.1044-1049, 2008.

S. M. Roth, R. F. Ferrell, and B. F. Hurley, Strength training for the prevention and treatment of sarcopenia. The journal of nutrition, health & aging, vol.4, pp.143-155, 2000.

D. Rudman, A. G. Feller, H. S. Nagraj, G. A. Gergans, P. Y. Lalitha et al., Effects of human growth hormone in men over 60 years old, The New England journal of medicine, vol.323, pp.1-6, 1990.

N. A. Ryan, K. A. Zwetsloot, L. M. Westerkamp, R. C. Hickner, W. E. Pofahl et al., Lower skeletal muscle capillarization and VEGF expression in aged vs. young men, J Appl Physiol, vol.100, pp.178-185, 1985.

A. Safdar, M. J. Hamadeh, J. J. Kaczor, S. Raha, J. Debeer et al., Aberrant mitochondrial homeostasis in the skeletal muscle of sedentary older adults, PloS one, vol.5, p.10778, 2010.

S. Sajko, L. Kubinova, E. Cvetko, M. Kreft, A. Wernig et al., Frequency of Mcadherin-stained satellite cells declines in human muscles during aging. The journal of histochemistry and cytochemistry : official journal of the, Histochemistry Society, vol.52, pp.179-185, 2004.

J. R. Sanes, The basement membrane/basal lamina of skeletal muscle, The Journal of biological chemistry, vol.278, pp.12601-12604, 2003.

S. Schiaffino, Fibre types in skeletal muscle: a personal account, Acta Physiol (Oxf), vol.199, pp.451-463, 2010.

S. Schiaffino, V. Hanzlikova, and S. Pierobon, Relations between structure and function in rat skeletal muscle fibers, The Journal of cell biology, vol.47, pp.107-119, 1970.

S. Schiaffino and C. Reggiani, Fiber types in mammalian skeletal muscles, Physiological reviews, vol.91, pp.1447-1531, 2011.

J. J. Sciote and T. J. Morris, Skeletal muscle function and fibre types: the relationship between occlusal function and the phenotype of jaw-closing muscles in human, Journal of orthodontics, vol.27, pp.15-30, 2000.

D. L. Scott, F. Wolfe, and T. W. Huizinga, Lancet, vol.376, pp.1094-1108, 2010.

J. E. Scott, Proteoglycan:collagen interactions and subfibrillar structure in collagen fibrils. Implications in the development and ageing of connective tissues, Journal of anatomy, vol.169, pp.23-35, 1990.

P. Seale, L. A. Sabourin, A. Girgis-gabardo, A. Mansouri, P. Gruss et al., Pax7 is required for the specification of myogenic satellite cells, Cell, vol.102, pp.777-786, 2000.

D. R. Seals, K. L. Jablonski, and A. J. Donato, Aging and vascular endothelial function in humans, Clin Sci (Lond), vol.120, pp.357-375, 2011.

S. M. Senf, S. L. Dodd, J. M. Mcclung, and A. R. Judge, Hsp70 overexpression inhibits NF-kappaB and Foxo3a transcriptional activities and prevents skeletal muscle atrophy, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, vol.22, pp.3836-3845, 2008.

A. L. Serrano, M. Perez, A. Lucia, J. L. Chicharro, E. Quiroz-rothe et al., , 2001.

, Immunolabelling, histochemistry and in situ hybridisation in human skeletal muscle fibres to detect myosin heavy chain expression at the protein and mRNA level, Journal of anatomy, vol.199, pp.329-337

T. Shavlakadze, J. Mcgeachie, and M. D. Grounds, Delayed but excellent myogenic stem cell response of regenerating geriatric skeletal muscles in mice, Biogerontology, vol.11, pp.363-376, 2010.

C. S. Shaw, D. A. Jones, and A. J. Wagenmakers, Network distribution of mitochondria and lipid droplets in human muscle fibres, Histochemistry and cell biology, vol.129, pp.65-72, 2008.

G. Shefer, D. P. Van-de-mark, J. B. Richardson, and Z. Yablonka-reuveni, Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle, Developmental biology, vol.294, pp.50-66, 2006.

T. Shioi and Y. Inuzuka, Aging as a substrate of heart failure, Journal of cardiology, vol.60, pp.423-428, 2012.

K. R. Short, M. L. Bigelow, J. Kahl, R. Singh, J. Coenen-schimke 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, vol.102, pp.5618-5623, 2005.

G. I. Shulman, Cellular mechanisms of insulin resistance, The Journal of clinical investigation, vol.106, pp.171-176, 2000.

J. Y. Shyy and S. Chien, Role of integrins in cellular responses to mechanical stress and adhesion, Current opinion in cell biology, vol.9, pp.707-713, 1997.

D. R. Sinacore and E. A. Gulve, The role of skeletal muscle in glucose transport, glucose homeostasis, and insulin resistance: implications for physical therapy, Physical therapy, vol.73, pp.878-891, 1993.

W. Sipos, P. Pietschmann, M. Rauner, K. Kerschan-schindl, and J. Patsch, Pathophysiology of osteoporosis, Wien Med Wochenschr, vol.159, pp.230-234, 2009.

B. Sjoblom, A. Salmazo, and K. Djinovic-carugo, Alpha-actinin structure and regulation, Cellular and molecular life sciences : CMLS, vol.65, pp.2688-2701, 2008.

V. Smerdu, I. Karsch-mizrachi, M. Campione, L. Leinwand, and S. Schiaffino, Type IIx myosin heavy chain transcripts are expressed in type IIb fibers of human skeletal muscle, The American journal of physiology, vol.267, pp.1723-1728, 1994.

W. Song, H. B. Kwak, and J. M. Lawler, Exercise training attenuates age-induced changes in apoptotic signaling in rat skeletal muscle, Antioxidants & redox signaling, vol.8, pp.517-528, 2006.

L. L. Spriet, R. A. Howlett, and G. J. Heigenhauser, An enzymatic approach to lactate production in human skeletal muscle during exercise, Medicine and science in sports and exercise, vol.32, pp.756-763, 2000.

E. Stalberg, O. Borges, M. Ericsson, B. Essen-gustavsson, P. R. Fawcett et al., The quadriceps femoris muscle in 20-70-year-old subjects: relationship between knee extension torque, electrophysiological parameters, and muscle fiber characteristics, Muscle & nerve, vol.12, pp.382-389, 1989.

R. S. Staron and D. Pette, Nonuniform myosin expression along single fibers of chronically stimulated and contralateral rabbit tibialis anterior muscles, Pflugers Archiv : European journal of physiology, vol.409, pp.67-73, 1987.

L. Staunton, M. Zweyer, D. Swandulla, and K. Ohlendieck, Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle, Int J Mol Med, vol.30, pp.723-733, 2012.

M. P. Stern, K. Williams, C. Gonzalez-villalpando, K. J. Hunt, and S. M. Haffner, Does the metabolic syndrome improve identification of individuals at risk of type 2 diabetes and/or cardiovascular disease?, Diabetes care, vol.27, pp.2676-2681, 2004.

K. H. Strucksberg, K. Tangavelou, R. Schroder, and C. S. Clemen, Proteasomal activity in skeletal muscle: a matter of assay design, muscle type, and age, Analytical biochemistry, vol.399, pp.225-229, 2010.

L. H. Stryer, C. Freeman, A. Valerio, D. 'antona, G. Nisoli et al., Branched-chain amino acids, mitochondrial biogenesis, and healthspan: an evolutionary perspective, Biochemistry, vol.3, pp.464-478, 1995.

L. J. Van-loon and B. H. Goodpaster, Increased intramuscular lipid storage in the insulinresistant and endurance-trained state, Pflugers Archiv : European journal of physiology, vol.451, pp.606-616, 2006.

A. A. Vandervoort, Aging of the human neuromuscular system, Muscle & nerve, vol.25, pp.17-25, 2002.

L. B. Verdijk, B. G. Gleeson, R. A. Jonkers, K. Meijer, H. H. Savelberg et al., Skeletal muscle hypertrophy following resistance training is accompanied by a fiber type-specific increase in satellite cell content in elderly men, Biological sciences and medical sciences, vol.64, pp.332-339, 2009.

L. B. Verdijk, R. Koopman, G. Schaart, K. Meijer, H. H. Savelberg et al., Satellite cell content is specifically reduced in type II skeletal muscle fibers in the elderly, American journal of physiology Endocrinology and metabolism, vol.292, pp.151-157, 2007.

M. Vernay, B. Salanave, C. De-peretti, C. Druet, A. Malon et al., Metabolic syndrome and socioeconomic status in France: The French Nutrition and Health Survey, International journal of public health, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02644473

J. Verney, F. Kadi, N. Charifi, L. Feasson, M. A. Saafi et al., Effects of combined lower body endurance and upper body resistance training on the satellite cell pool in elderly subjects, Muscle & nerve, vol.38, pp.1147-1154, 2008.

D. C. Vieira, R. A. Tibana, V. Tajra, N. Dda, C. De-farias et al., Decreased functional capacity and muscle strength in elderly women with metabolic syndrome, Clinical interventions in aging, vol.8, pp.1377-1386, 2013.

D. T. Villareal and J. O. Holloszy, DHEA enhances effects of weight training on muscle mass and strength in elderly women and men, American journal of physiology Endocrinology and metabolism, vol.291, pp.1003-1008, 2006.

J. Vina, M. C. Gomez-cabrera, C. Borras, T. Froio, F. Sanchis-gomar et al., Mitochondrial biogenesis in exercise and in ageing, Advanced drug delivery reviews, vol.61, pp.1369-1374, 2009.

A. J. Wagenmakers, Muscle amino acid metabolism at rest and during exercise: role in human physiology and metabolism. Exercise and sport sciences reviews, vol.26, pp.287-314, 1998.

P. D. Wagner, I. M. Olfert, K. Tang, and E. C. Breen, Muscle-targeted deletion of VEGF and exercise capacity in mice, Respiratory physiology & neurobiology, vol.151, pp.159-166, 2006.

S. Wakil, Lipid Metabolism, 2012.

J. Wanagat, Z. Cao, P. Pathare, and J. M. Aiken, Mitochondrial DNA deletion mutations colocalize with segmental electron transport system abnormalities, muscle fiber atrophy, fiber splitting, and oxidative damage in sarcopenia, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, vol.15, pp.322-332, 2001.

J. C. Wang and M. Bennett, Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence, Circulation research, vol.111, pp.245-259, 2012.

K. Wang and D. J. Klionsky, Mitochondria removal by autophagy, Autophagy, vol.7, pp.297-300, 2011.

M. J. Watt and A. J. Hoy, Lipid metabolism in skeletal muscle: generation of adaptive and maladaptive intracellular signals for cellular function, American journal of physiology Endocrinology and metabolism, vol.302, pp.1315-1328, 2012.

D. Weiskopf, B. Weinberger, and B. Grubeck-loebenstein, The aging of the immune system. Transplant international : official journal of the European Society for, Organ Transplantation, vol.22, pp.1041-1050, 2009.

M. E. Weksler and P. Szabo, The effect of age on the B-cell repertoire, Journal of clinical immunology, vol.20, pp.240-249, 2000.

S. Welle, K. Bhatt, B. Shah, N. Needler, J. M. Delehanty et al., Reduced amount of mitochondrial DNA in aged human muscle, J Appl Physiol, vol.94, pp.1479-1484, 1985.

G. D. Wells, H. Selvadurai, and I. Tein, Bioenergetic provision of energy for muscular activity, Paediatric respiratory reviews, vol.10, pp.83-90, 2009.

C. Wenisch, S. Patruta, F. Daxbock, R. Krause, and W. Horl, Effect of age on human neutrophil function, Journal of leukocyte biology, vol.67, pp.40-45, 2000.

T. Wenz, S. G. Rossi, R. L. Rotundo, B. M. Spiegelman, and C. T. Moraes, Increased muscle PGC-1alpha expression protects from sarcopenia and metabolic disease during aging, Proceedings of the National Academy of Sciences of the United States of America, vol.106, pp.20405-20410, 2009.

H. A. Wieland, M. Michaelis, B. J. Kirschbaum, and K. A. Rudolphi, Osteoarthritis -an untreatable disease?, Nature reviews Drug discovery, vol.4, pp.331-344, 2005.

E. A. Wilkes, A. L. Selby, P. J. Atherton, R. Patel, D. Rankin et al., Blunting of insulin inhibition of proteolysis in legs of older subjects may contribute to age-related sarcopenia, The American journal of clinical nutrition, vol.90, pp.1343-1350, 2009.

P. W. Wilson, D. 'agostino, R. B. Parise, H. Sullivan, L. Meigs et al., Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus, Circulation, vol.112, pp.3066-3072, 2005.

P. M. Wise, Neuroendocrine modulation of the "menopause": insights into the aging brain, The American journal of physiology, vol.277, pp.965-970, 1999.

S. E. Wohlgemuth, A. Y. Seo, E. Marzetti, H. A. Lees, and C. Leeuwenburgh, Skeletal muscle autophagy and apoptosis during aging: effects of calorie restriction and life-long exercise, Experimental gerontology, vol.45, pp.138-148, 2010.

E. J. Yang, S. Lim, J. Y. Lim, K. W. Kim, H. C. Jang et al., Association between muscle strength and metabolic syndrome in older Korean men and women: the Korean Longitudinal Study on Health and Aging, Metabolism: clinical and experimental, vol.61, pp.317-324, 2012.

Z. Yang, E. C. Norton, and S. C. Stearns, Longevity and health care expenditures: the real reasons older people spend more. The journals of gerontology Series B, Psychological sciences and social sciences, vol.58, pp.2-10, 2003.

C. Zechner, L. Lai, J. F. Zechner, T. Geng, Z. Yan et al., Total skeletal muscle PGC-1 deficiency uncouples mitochondrial derangements from fiber type determination and insulin sensitivity, Cell metabolism, vol.12, pp.633-642, 2010.

R. Zechner, P. C. Kienesberger, G. Haemmerle, R. Zimmermann, and A. Lass, Adipose triglyceride lipase and the lipolytic catabolism of cellular fat stores, Journal of lipid research, vol.50, pp.3-21, 2009.

S. D. Zimmerman, R. J. Mccormick, R. K. Vadlamudi, and D. P. Thomas, Age and training alter collagen characteristics in fast-and slow-twitch rat limb muscle, J Appl Physiol, vol.75, pp.1670-1674, 1985.