A. J. Cruz-jentoft, G. Bahat, J. Bauer, Y. Boirie, O. Bruyère et al., Sarcopenia: revised European consensus on definition and diagnosis, Age Ageing, vol.48, pp.16-31, 2019.

P. Kittiskulnam, J. J. Carrero, G. M. Chertow, G. A. Kaysen, C. Delgado et al., Sarcopenia among patients receiving hemodialysis: weighing the evidence, J Cachexia Sarcopenia Muscle, vol.8, pp.57-68, 2017.

S. Von-haehling, J. E. Morley, and S. D. Anker, An overview of sarcopenia: facts and numbers on prevalence and clinical impact, J Cachexia Sarcopenia Muscle, vol.1, pp.129-162, 2010.

R. A. Pereira, A. C. Cordeiro, C. M. Avesani, J. J. Carrero, B. Lindholm et al., Sarcopenia in chronic kidney disease on conservative therapy: prevalence and association with mortality, Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.30, pp.1718-1743, 2015.

B. C. Clark and T. M. Manini, Sarcopenia =/= dynapenia, J Gerontol A Biol Sci Med Sci, vol.63, pp.829-863, 2008.

P. Stenvinkel and T. J. Ekström, Epigenetics and the uremic phenotype: a matter of balance, Contrib Nephrol, vol.161, pp.55-62, 2008.

J. J. Carrero and P. Stenvinkel, The vulnerable man: impact of testosterone deficiency on the uraemic phenotype, Nephrol Dial Transplant, vol.27, pp.4030-4071, 2012.

J. J. Carrero, K. L. Johansen, B. Lindholm, P. Stenvinkel, L. Cuppari et al., Screening for muscle wasting and dysfunction in patients with chronic kidney disease, Kidney Int, vol.90, pp.53-66, 2016.

D. Marcelli, L. A. Usvyat, P. Kotanko, I. Bayh, B. Canaud et al., Body composition and survival in dialysis patients: results from an international cohort study, Clin J Am Soc Nephrol CJASN, vol.10, pp.1192-200, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02005484

N. Terrier, I. Jaussent, A. Dupuy, M. M. Delcourt, C. Chalabi et al., Creatinine index and transthyretin as additive predictors of mortality in haemodialysis patients, Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.23, pp.345-53, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00170013

J. Souweine, A. Boudet, L. Chenine, H. Leray, A. Rodriguez et al., Standardized Method to Measure Muscle Force at the Bedside in Hemodialysis Patients, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.27, pp.194-200, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01774962

P. Kittiskulnam, G. M. Chertow, J. J. Carrero, C. Delgado, G. A. Kaysen et al., Sarcopenia and its individual criteria are associated, in part, with mortality among patients on hemodialysis, Kidney Int, vol.92, pp.238-285, 2017.

M. K. Han, A. Agusti, P. M. Calverley, B. R. Celli, G. Criner et al., Chronic obstructive pulmonary disease phenotypes: the future of COPD, Am J Respir Crit Care Med, vol.182, pp.598-604, 2010.

S. E. Wenzel, Asthma phenotypes: the evolution from clinical to molecular approaches, Nat Med, vol.18, pp.716-741, 2012.

D. N. Cooper, M. Krawczak, C. Polychronakos, C. Tyler-smith, and H. Kehrer-sawatzki, Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease, Hum Genet, vol.132, pp.1077-130, 2013.

L. J. Garred, D. L. Barichello, B. C. Canaud, and W. G. Mccready, Simple equations for protein catabolic rate determination from pre dialysis and post dialysis blood urea nitrogen, ASAIO J Am Soc Artif Intern Organs, vol.41, pp.889-95, 19921995.

J. T. Daugirdas, Second generation logarithmic estimates of single-pool variable volume Kt/V: an analysis of error, J Am Soc Nephrol JASN, vol.4, pp.1205-1218, 1993.

B. Canaud, A. Granger-vallée, N. Molinari, L. Chenine, H. Leray-moragues et al., Creatinine index as a surrogate of lean body mass derived from urea Kt/V, predialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS One, vol.9, p.93286, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02637906

N. Anderton, A. Giri, G. Wei, R. L. Marcus, X. Chen et al., Sedentary Behavior in Individuals With Diabetic Chronic Kidney Disease and Maintenance Hemodialysis, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.25, pp.364-70, 2015.

M. Tamaki, K. Miyashita, S. Wakino, M. Mitsuishi, K. Hayashi et al., Chronic kidney disease reduces muscle mitochondria and exercise endurance and its exacerbation by dietary protein through inactivation of pyruvate dehydrogenase, Kidney Int, vol.85, pp.1330-1339, 2014.

L. J. Smyth, S. Duffy, A. P. Maxwell, and A. J. Mcknight, Genetic and epigenetic factors influencing chronic kidney disease, Am J Physiol-Ren Physiol, vol.307, pp.757-76, 2014.

A. C. Webster, E. V. Nagler, R. L. Morton, and P. Masson, Chronic Kidney Disease, The Lancet, vol.389, pp.1238-52, 2017.

R. E. Ahonen, J. Mäkitie, and B. Kock, Striated muscle capillaries in uremic patients and in renal transplant recipients, Arch Intern Med, vol.141, pp.867-876, 1981.

A. J. Shah, V. Sahgal, A. P. Quintanilla, V. Subramani, H. Singh et al., Muscle in chronic uremia--a histochemical and morphometric study of human quadriceps muscle biopsies, Clin Neuropathol, vol.2, pp.83-92, 1983.

K. L. Johansen, T. Shubert, J. Doyle, B. Soher, G. K. Sakkas et al., Muscle atrophy in patients receiving hemodialysis: effects on muscle strength, muscle quality, and physical function, Kidney Int, vol.63, pp.291-298, 2003.

M. I. Lewis, M. Fournier, H. Wang, T. W. Storer, R. Casaburi et al., Metabolic and morphometric profile of muscle fibers in chronic hemodialysis patients, J Appl Physiol, vol.112, pp.72-80, 2012.

J. L. Gamboa, F. T. Billings, M. T. Bojanowski, L. A. Gilliam, C. Yu et al., Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease, Physiol Rep, vol.4, 2016.

Y. Y. Zhang, L. J. Gu, J. Huang, M. C. Cai, H. L. Yu et al., CKD autophagy activation and skeletal muscle atrophy-a preliminary study of mitophagy and inflammation, Eur J Clin Nutr, vol.73, pp.950-60, 2019.

J. Aniort, A. Stella, C. Philipponnet, A. Poyet, C. Polge et al., Muscle wasting in patients with end-stage renal disease or early-stage lung cancer: common mechanisms at work, J Cachexia Sarcopenia Muscle, vol.10, pp.323-360, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01882339

C. Clark, B. , L. Taylor, and J. , Age-Related Changes in Motor Cortical Properties and Voluntary Activation of Skeletal Muscle, Curr Aging Sci, vol.4, pp.192-201, 2011.

R. A. Mcgregor, D. Cameron-smith, and S. D. Poppitt, It is not just muscle mass: a review of muscle quality, composition and metabolism during ageing as determinants of muscle function and mobility in later life, Longev Heal, vol.3, 2014.

J. J. Carrero, K. L. Johansen, B. Lindholm, P. Stenvinkel, L. Cuppari et al., Screening for muscle wasting and dysfunction in patients with chronic kidney disease, Kidney Int, vol.90, pp.53-66, 2016.

N. Isoyama, A. R. Qureshi, C. M. Avesani, B. Lindholm, P. Bàràny et al., Comparative associations of muscle mass and muscle strength with mortality in dialysis patients, Clin J Am Soc Nephrol CJASN, vol.9, pp.1720-1728, 2014.

R. A. Pereira, A. C. Cordeiro, C. M. Avesani, J. J. Carrero, B. Lindholm et al., Sarcopenia in chronic kidney disease on conservative therapy: prevalence and association with mortality, Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.30, pp.1718-1725, 2015.

X. S. Cheng, J. N. Myers, G. M. Chertow, R. Rabkin, K. N. Chan et al., Prehabilitation for kidney transplant candidates: Is it time?, Clin Transplant, vol.31, p.13020, 2017.

M. A. Mcadams-demarco, Y. H. Van-pilsum-rasmussen, S. Schrack, J. Haugen, C. E. Chu et al., Prehabilitation prior to kidney transplantation: Results from a pilot study, Clin Transplant, vol.33, p.13450, 2019.

R. Chowdhury, N. M. Peel, M. Krosch, and R. E. Hubbard, Frailty and chronic kidney disease: A systematic review, Arch Gerontol Geriatr, vol.68, pp.135-142, 2017.

S. Heiwe and S. H. Jacobson, Exercise training in adults with CKD: a systematic review and meta-analysis, Am J Kidney Dis Off J Natl Kidney Found, vol.64, pp.383-393, 2014.

R. A. Mcgregor, D. Cameron-smith, and S. D. Poppitt, It is not just muscle mass: a review of muscle quality, composition and metabolism during ageing as determinants of muscle function and mobility in later life, Longev Heal, vol.3, 2014.

A. C. Webster, E. V. Nagler, R. L. Morton, and P. Masson, Chronic Kidney Disease, The Lancet, vol.389, pp.1238-1252, 2017.

L. J. Smyth, S. Duffy, A. P. Maxwell, and A. J. Mcknight, Genetic and epigenetic factors influencing chronic kidney disease, Am J Physiol-Ren Physiol, vol.307, pp.757-776, 2014.

M. Panaye, A. Kolko-labadens, C. Lasseur, J. Paillasseur, M. P. Guillodo et al., Phenotypes influencing low physical activity in maintenance dialysis, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.25, pp.31-39, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01850601

C. G. Musso and J. R. Jauregui, Macías Núñez JF. Frailty phenotype and chronic kidney disease: a review of the literature, Int Urol Nephrol, vol.47, pp.1801-1807, 2015.

P. Stenvinkel and T. J. Ekström, Epigenetics and the uremic phenotype: a matter of balance, Contrib Nephrol, vol.161, pp.55-62, 2008.

J. Souweine, N. Kuster, L. Chenine, A. Rodriguez, L. Patrier et al., Physical inactivity and protein energy wasting play independent roles in muscle weakness in maintenance haemodialysis patients, PloS One, vol.13, p.200061, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01853078

C. Polge, A. Heng, M. Jarzaguet, S. Ventadour, A. Claustre et al., Muscle actin is polyubiquitinylated in vitro and in vivo and targeted for breakdown by the E3 ligase MuRF1, FASEB J Off Publ Fed Am Soc Exp Biol, vol.25, pp.3790-3802, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02420104

K. G. Avin, N. X. Chen, J. M. Organ, C. Zarse, K. O'neill et al., Skeletal Muscle Regeneration and Oxidative Stress Are Altered in Chronic Kidney Disease, PLOS ONE, vol.11, p.159411, 2016.

X. Moreau-gaudry, G. Jean, L. Genet, D. Lataillade, E. Legrand et al., A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.24, pp.395-400, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01858992

D. Fouque, K. Kalantar-zadeh, J. Kopple, N. Cano, P. Chauveau et al., A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease, Kidney Int, vol.73, pp.391-398, 2008.

R. E. Ahonen, J. Mäkitie, and B. Kock, Striated muscle capillaries in uremic patients and in renal transplant recipients, Arch Intern Med, vol.141, pp.867-869, 1981.

A. J. Shah, V. Sahgal, A. P. Quintanilla, V. Subramani, H. Singh et al., Muscle in chronic uremia--a histochemical and morphometric study of human quadriceps muscle biopsies, Clin Neuropathol, vol.2, pp.83-89, 1983.

K. L. Johansen, T. Shubert, J. Doyle, B. Soher, G. K. Sakkas et al., Muscle atrophy in patients receiving hemodialysis: effects on muscle strength, muscle quality, and physical function, Kidney Int, vol.63, pp.291-297, 2003.

S. W. Lee, G. Dai, Z. Hu, X. Wang, J. Du et al., Regulation of muscle protein degradation: coordinated control of apoptotic and ubiquitin-proteasome systems by phosphatidylinositol 3 kinase, J Am Soc Nephrol JASN, vol.15, pp.1537-1545, 2004.

J. Aniort, A. Stella, C. Philipponnet, A. Poyet, C. Polge et al., Muscle wasting in patients with end-stage renal disease or early-stage lung cancer: common mechanisms at work, J Cachexia Sarcopenia Muscle, vol.10, pp.323-337, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01882339

M. I. Lewis, M. Fournier, H. Wang, T. W. Storer, R. Casaburi et al., Metabolic and morphometric profile of muscle fibers in chronic hemodialysis patients, J Appl Physiol Bethesda Md, vol.112, pp.72-78, 1985.

J. L. Gamboa, F. T. Billings, M. T. Bojanowski, L. A. Gilliam, C. Yu et al., Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease, Physiol Rep, vol.4, 2016.

Y. Y. Zhang, L. J. Gu, J. Huang, M. C. Cai, H. L. Yu et al., CKD autophagy activation and skeletal muscle atrophy-a preliminary study of mitophagy and inflammation, Eur J Clin Nutr, vol.73, pp.950-960, 2019.

R. J. Youle and D. P. Narendra, Mechanisms of mitophagy, Nat Rev Mol Cell Biol, vol.12, pp.9-14, 2011.

Z. Su, J. D. Klein, J. Du, H. A. Franch, L. Zhang et al., Chronic kidney disease induces autophagy leading to dysfunction of mitochondria in skeletal muscle, Am J Physiol-Ren Physiol, vol.312, pp.1128-1140, 2017.

B. Roshanravan, B. Kestenbaum, J. Gamboa, S. A. Jubrias, E. Ayers et al., Am J Kidney Dis Off J Natl Kidney Found, vol.68, pp.658-659, 2016.

D. N. Cooper, M. Krawczak, C. Polychronakos, C. Tyler-smith, and H. Kehrer-sawatzki, Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease, Hum Genet, vol.132, pp.1077-1130, 2013.

P. Andersen, R. P. Adams, G. Sjøgaard, A. Thorboe, and B. Saltin, Dynamic knee extension as model for study of isolated exercising muscle in humans, J Appl Physiol Bethesda Md, vol.59, pp.1647-1653, 19851985.

I. Serres, V. Gautier, A. Varray, and C. Préfaut, Impaired skeletal muscle endurance related to physical inactivity and altered lung function in COPD patients, Chest, vol.113, pp.900-905, 1998.
URL : https://hal.archives-ouvertes.fr/hal-01625242

A. Couillard, C. Koechlin, J. P. Cristol, A. Varray, and C. Prefaut, Evidence of local exerciseinduced systemic oxidative stress in chronic obstructive pulmonary disease patients, Eur Respir J, vol.20, pp.1123-1129, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01622943

N. Anderton, A. Giri, G. Wei, R. L. Marcus, X. Chen et al., Sedentary Behavior in Individuals With Diabetic Chronic Kidney Disease and Maintenance Hemodialysis, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.25, pp.364-370, 2015.

F. Gouzi, A. Abdellaoui, N. Molinari, E. Pinot, B. Ayoub et al., Fiber atrophy, oxidative stress, and oxidative fiber reduction are the attributes of different phenotypes in chronic obstructive pulmonary disease patients, J Appl Physiol Bethesda Md, vol.115, pp.1796-1805, 1985.
URL : https://hal.archives-ouvertes.fr/hal-01623970

M. Hayot, A. Michaud, C. Koechlin, M. Caron, P. Leblanc et al., Skeletal muscle microbiopsy: a validation study of a minimally invasive technique, Eur Respir J, vol.25, pp.431-440, 2005.

N. Terrier, I. Jaussent, A. Dupuy, M. M. Delcourt, C. Chalabi et al., Creatinine index and transthyretin as additive predictors of mortality in haemodialysis patients, Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.23, pp.345-353, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00170013

B. Canaud, Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS One
URL : https://hal.archives-ouvertes.fr/hal-02637906

E. Van-den-ham, J. P. Kooman, A. Schols, F. Nieman, J. D. Does et al., Similarities in skeletal muscle strength and exercise capacity between renal transplant and hemodialysis patients, Am J Transplant Off J Am Soc Transplant Am Soc Transpl Surg, vol.5, pp.1957-1965, 2005.

D. Marcelli, K. Brand, P. Ponce, A. Milkowski, C. Marelli et al., Longitudinal Changes in Body Composition in Patients After Initiation of Hemodialysis Therapy: Results From an International Cohort, J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.26, pp.72-80, 2016.

R. W. Bohannon, J. Smith, D. Hull, D. Palmeri, and R. Barnhard, Deficits in lower extremity muscle and gait performance among renal transplant candidates, Arch Phys Med Rehabil, vol.76, pp.547-551, 1995.

V. O. Leal, M. B. Stockler-pinto, N. E. Farage, L. N. Aranha, D. Fouque et al., Handgrip strength and its dialysis determinants in hemodialysis patients, Nutrition, vol.27, pp.1125-1129, 2011.

B. T. Workeneh and W. E. Mitch, Review of muscle wasting associated with chronic kidney disease, Am J Clin Nutr, vol.91, pp.1128-1132, 2010.

D. P. Lipkin, A. J. Scriven, T. Crake, and P. A. Poole-wilson, Six minute walking test for assessing exercise capacity in chronic heart failure, BMJ, vol.292, pp.653-655, 1986.

H. Watz, B. Waschki, T. Meyer, and H. Magnussen, Physical activity in patients with COPD, Eur Respir J, vol.33, pp.262-272, 2008.

P. Esposito, F. Furini, T. Rampino, M. Gregorini, L. Petrucci et al., Assessment of physical performance and quality of life in kidney-transplanted patients: a crosssectional study, Clin Kidney J, p.102, 2016.

M. Tamaki, K. Miyashita, S. Wakino, M. Mitsuishi, K. Hayashi et al., Chronic kidney disease reduces muscle mitochondria and exercise endurance and its exacerbation by dietary protein through inactivation of pyruvate dehydrogenase, Kidney Int, vol.85, pp.1330-1339, 2014.

D. Sousa, E. Veksler, V. Bigard, X. Mateo, P. Ventura-clapier et al., Heart failure affects mitochondrial but not myofibrillar intrinsic properties of skeletal muscle, Circulation, vol.102, pp.1847-1853, 2000.

M. Picard, R. Godin, M. Sinnreich, J. Baril, J. Bourbeau et al., The mitochondrial phenotype of peripheral muscle in chronic obstructive pulmonary disease: disuse or dysfunction?, Am J Respir Crit Care Med, vol.178, pp.1040-1047, 2008.

R. E. Ahonen, Striated muscle ultrastructure in uremic patients and in renal transplant recipients, Acta Neuropathol (Berl), vol.50, pp.163-166, 1980.

P. Stenvinkel, J. J. Carrero, F. Von-walden, T. A. Ikizler, and G. A. Nader, Muscle wasting in endstage renal disease promulgates premature death: established, emerging and potential novel treatment strategies, Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, vol.31, pp.1070-1077, 2016.

G. Garibotto, R. Russo, A. Sofia, M. R. Sala, C. Robaudo et al., Skeletal muscle protein synthesis and degradation in patients with chronic renal failure, Kidney Int, vol.45, pp.1432-1439, 1994.

D. Adey, R. Kumar, J. T. Mccarthy, and K. S. Nair, Reduced synthesis of muscle proteins in chronic renal failure, Am J Physiol Endocrinol Metab, vol.278, pp.219-225, 2000.

X. H. Wang and W. E. Mitch, Mechanisms of muscle wasting in chronic kidney disease, Nat Rev Nephrol, vol.10, pp.504-516, 2014.

F. Raynaud, E. Fernandez, G. Coulis, A. L. Vignon, X. Bleimling et al., Calpain 1-titin interactions concentrate calpain 1 in the Z-band edges and in the N2-line region within the skeletal myofibril, FEBS J, vol.272, pp.2578-2590, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02680943

X. H. Wang, L. Zhang, W. E. Mitch, J. M. Ledoux, J. Hu et al., Caspase-3 cleaves specific 19 S proteasome subunits in skeletal muscle stimulating proteasome activity, J Biol Chem, vol.285, pp.21249-21257, 2010.

P. Esposito, L. Porta, E. Calatroni, M. Grignano, M. A. Milanesi et al., Modulation of Myostatin/Hepatocyte Growth Factor Balance by Different Hemodialysis Modalities, BioMed Res Int, p.7635459, 2017.

D. Wang, Y. Yang, R. Huang, Z. Zhang, and X. Lin, Myostatin Activates the Ubiquitin-Proteasome and Autophagy-Lysosome Systems Contributing to Muscle Wasting in Chronic Kidney Disease, Oxid Med Cell Longev, vol.2015, pp.1-18, 2015.

C. Castaneda, P. L. Gordon, K. L. Uhlin, A. S. Levey, J. J. Kehayias et al., Resistance training to counteract the catabolism of a low-protein diet in patients with chronic renal insufficiency. A randomized, controlled trial, Ann Intern Med, vol.135, pp.965-976, 2001.

D. M. Mancini, E. Coyle, A. Coggan, J. Beltz, N. Ferraro et al., Contribution of intrinsic skeletal muscle changes to 31P NMR skeletal muscle metabolic abnormalities in patients with chronic heart failure, Circulation, vol.80, pp.1338-1346, 1989.

P. G. Yazdi, H. Moradi, J. Yang, P. H. Wang, and N. D. Vaziri, Skeletal muscle mitochondrial depletion and dysfunction in chronic kidney disease, Int J Clin Exp Med, vol.6, pp.532-539, 2013.

C. Xu, A. Kasimumali, X. Guo, R. Lu, K. Xie et al., Reduction of mitochondria and up regulation of pyruvate dehydrogenase kinase 4 of skeletal muscle in patients with chronic kidney disease, Nephrology, 2019.

M. S. Hickey, J. O. Carey, J. L. Azevedo, J. A. Houmard, W. J. Pories et al., Skeletal muscle fiber composition is related to adiposity and in vitro glucose transport rate in humans, Am J Physiol-Endocrinol Metab, 1995.

D. E. Kelley, J. He, E. V. Menshikova, and V. B. Ritov, Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes, Diabetes, vol.51, pp.2944-2950, 2002.

M. Hesselink, V. Schrauwen-hinderling, and P. Schrauwen, Skeletal muscle mitochondria as a target to prevent or treat type 2 diabetes mellitus, Nat Rev Endocrinol, vol.12, pp.633-645, 2016.

A. Chacon-cabrera, H. Lund-palau, J. Gea, and E. Barreiro, Time-Course of Muscle Mass Loss, Damage, and Proteolysis in Gastrocnemius following Unloading and Reloading: Implications in Chronic Diseases, PloS One, vol.11, p.164951, 2016.

A. Bigard, E. Boehm, V. Veksler, P. Mateo, K. Anflous et al., Muscle Unloading Induces Slow to Fast Transitions in Myofibrillar but not Mitochondrial Properties. Relevance to Skeletal Muscle Abnormalities in Heart Failure, J Mol Cell Cardiol, vol.30, pp.2391-2401, 1998.

T. Lin, V. Wu, and C. Wang, Autophagy in Chronic Kidney Diseases. Cells, vol.8, p.61, 2019.

J. J. Carrero, P. Stenvinkel, L. Cuppari, T. A. Ikizler, K. Kalantar-zadeh et al., Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM), J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found, vol.23, pp.77-90, 2013.

J. Satriano and K. Sharma, Autophagy and metabolic changes in obesity-related chronic kidney disease, Nephrol Dial Transplant, vol.28, pp.29-36, 2013.

Y. Takabatake, T. Kimura, A. Takahashi, and Y. Isaka, Autophagy and the kidney: health and disease, Nephrol Dial Transplant, vol.29, pp.1639-1647, 2014.

R. Singh and A. M. Cuervo, Autophagy in the Cellular Energetic Balance, Cell Metab, vol.13, pp.495-504, 2011.

X. Deng, Y. Xie, and A. Zhang, Advance of autophagy in chronic kidney diseases, Ren Fail, vol.39, pp.306-313, 2017.

T. Vellai, Autophagy genes and ageing, Cell Death Differ, vol.16, pp.94-102, 2009.

J. M. Campistol, Uremic myopathy, Kidney Int, vol.62, pp.1901-1913, 2002.

I. H. Fahal, Uraemic sarcopenia: aetiology and implications, Nephrol Dial Transplant, vol.29, pp.1655-1665, 2014.

M. Piasecki, A. Ireland, J. Piasecki, D. W. Stashuk, A. Swiecicka et al., Failure to expand the motor unit size to compensate for declining motor unit numbers distinguishes sarcopenic from non-sarcopenic older men, J Physiol, vol.596, pp.1627-1637, 2018.

, Rapport sur la situation mondiale des maladies non transmissibles, 2014.

, WHO | Noncommunicable diseases country profiles 2018, 2019.

, Prévention des maladies chroniques un investissement vital. Organisation mondiale de la Santé, 2006.

B. Silva and G. , Maladies chroniques : vers un changement du paradigme des soins, Santé Publique, vol.1, pp.9-11, 2015.

, Global Recommendations on Physical Activity for Health [Internet]. World Health Organization, 2010.

, Global health risks: mortality and burden of disease attributable to selected major risks. World Health Organization, 2009.

, Stratégies de prévention de l'obésité de l'enfant dans la population

P. M. Cawthon, L. M. Marshall, and Y. Michael, Frailty in older men: prevalence, progression, and relationship with mortality, Journal of the American Geriatrics Society, vol.55, pp.1216-1223, 2007.

S. K. Inouye, S. Studenski, M. E. Tinetti, and G. A. Kuchel, Geriatric syndromes: clinical, research, and policy implications of a core geriatric concept, Journal of the American Geriatrics Society, vol.55, pp.780-791, 2007.

E. Topinková, Aging, disability and frailty, Annals of Nutrition & Metabolism, vol.52, pp.6-11, 2008.

I. H. Rosenberg, Summary comments, The American Journal of Clinical Nutrition, vol.50, pp.1231-1233, 1989.

E. Volpi, B. Mittendorfer, B. B. Rasmussen, and R. R. Wolfe, The response of muscle protein anabolism to combined hyperaminoacidemia and glucose-induced hyperinsulinemia is impaired in the elderly, The Journal of Clinical Endocrinology and Metabolism, vol.85, pp.4481-4490, 2000.

E. Volpi, H. Kobayashi, M. Sheffield-moore, B. Mittendorfer, and R. R. Wolfe, Essential amino acids are primarily responsible for the amino acid stimulation of muscle protein anabolism in healthy elderly adults, The American Journal of Clinical Nutrition, vol.78, pp.250-258, 2003.

F. Lauretani, C. R. Russo, and S. Bandinelli, Age-associated changes in skeletal muscles and their effect on mobility: an operational diagnosis of sarcopenia, Journal of Applied Physiology, vol.95, pp.1851-1860, 1985.

A. J. Cruz-jentoft, G. Bahat, and J. Bauer, Sarcopenia: revised European consensus on definition and diagnosis, Age and Ageing, vol.48, pp.16-31, 2019.

A. J. Cruz-jentoft, F. Landi, and S. M. Schneider, Prevalence of and interventions for sarcopenia in ageing adults: a systematic review, Report of the International Sarcopenia Initiative (EWGSOP and IWGS), vol.43, pp.748-759, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02639708

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.

P. Kortebein, A. Ferrando, J. Lombeida, R. Wolfe, and W. J. Evans, JAMA, vol.297, pp.1772-1774, 2007.

J. E. Morley, D. R. Thomas, and M. Wilson, Cachexia: pathophysiology and clinical relevance, The American Journal of Clinical Nutrition, vol.83, pp.735-743, 2006.

P. Lang, J. Michel, and D. Zekry, Frailty syndrome: a transitional state in a dynamic process, Gerontology, vol.55, pp.539-549, 2009.

L. P. Fried, C. M. Tangen, and J. Walston, Frailty in older adults: evidence for a phenotype, Biological Sciences and Medical Sciences, vol.56, pp.146-156, 2001.

J. E. Morley, B. Vellas, and G. A. Van-kan, Frailty consensus: a call to action, Journal of the American Medical Directors Association, vol.14, pp.392-397, 2013.

A. Clegg, J. Young, S. Iliffe, M. O. Rikkert, and K. Rockwood, Frailty in elderly people, Lancet, vol.381, pp.752-762, 2013.

R. Guthold, G. A. Stevens, L. M. Riley, and F. C. Bull, Worldwide trends in insufficient physical activity from 2001 to 2016: a pooled analysis of 358 population-based surveys with 1·9 million participants, The Lancet. Global Health, vol.6, pp.1077-1086, 2018.

S. A. Carlson, J. E. Fulton, M. Pratt, Z. Yang, and E. K. Adams, Inadequate Physical Activity and Health Care Expenditures in the United States, Progress in Cardiovascular Diseases, vol.57, pp.315-323, 2015.

, World Health Organization. Global action plan for the prevention and control of noncommunicable diseases, 2013.

G. Kojima, A. Liljas, and S. Iliffe, Frailty syndrome: implications and challenges for health care policy, Risk management and healthcare policy, vol.12, pp.23-30, 2019.

D. Ding, K. D. Lawson, and T. L. Kolbe-alexander, The economic burden of physical inactivity: a global analysis of major non-communicable diseases, Lancet, vol.388, pp.1311-1324, 2016.

, World Health Organization. More active people for a healthier world: global action plan on physical, 2018.

C. Sayer and A. A. Sarcopenia, Lancet, vol.393, pp.2636-2646, 2019.

E. Dent, J. E. Morley, and C. Aj, International Clinical Practice Guidelines for Sarcopenia (ICFSR): Screening, Diagnosis and Management. The journal of nutrition, health & aging, vol.22, pp.1148-1161, 2018.

M. Locquet, C. Beaudart, J. Reginster, J. Petermans, and O. Bruyère, Comparison of the performance of five screening methods for sarcopenia, Clinical epidemiology, vol.10, pp.71-82, 2017.

S. Yu, K. Khow, A. D. Jadczak, and R. Visvanathan, Clinical Screening Tools for Sarcopenia and Its Management, Current Gerontology and Geriatrics Research, vol.2016, pp.1-10, 2016.

Y. Lv, J. Yuan, and C. Mao, Association of Body Mass Index With Disability in Activities of Daily Living Among Chinese Adults 80 Years of Age or Older, JAMA Network Open, vol.1, pp.181915-181915, 2018.

I. Janssen, S. B. Heymsfield, R. N. Baumgartner, and R. Ross, Estimation of skeletal muscle mass by bioelectrical impedance analysis, Journal of Applied Physiology, vol.89, pp.465-471, 1985.

G. B. Forbes and G. J. Bruining, Urinary creatinine excretion and lean body mass, The American Journal of Clinical Nutrition, vol.29, pp.1359-1366, 1976.

S. B. Heymsfield, C. Arteaga, C. Mcmanus, J. Smith, and S. Moffitt, Measurement of muscle mass in humans: validity of the 24-hour urinary creatinine method, The American Journal of Clinical Nutrition, vol.37, pp.478-494, 1983.

S. B. Heymsfield, M. C. Gonzalez, J. Lu, G. Jia, and J. Zheng, Skeletal muscle mass and quality: evolution of modern measurement concepts in the context of sarcopenia, Proceedings of the Nutrition Society, vol.74, pp.355-366, 2015.

N. Terrier, I. Jaussent, and A. Dupuy, Creatinine index and transthyretin as additive predictors of mortality in haemodialysis patients. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association, European Renal Association, vol.23, pp.345-353, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00170013

B. Canaud, A. Granger-vallée, and N. Molinari, Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS one, vol.9, p.93286, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02637906

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

K. Norman, N. Stobäus, M. C. Gonzalez, J. Schulzke, and M. Pirlich, Hand grip strength: outcome predictor and marker of nutritional status, Clinical Nutrition, vol.30, pp.135-142, 2011.

F. Gouzi, C. Préfaut, and A. Abdellaoui, Evidence of an early physical activity reduction in chronic obstructive pulmonary disease patients, Archives of physical medicine and rehabilitation, vol.92, pp.1611-1617, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01623976

F. Gouzi, A. Abdellaoui, and N. Molinari, Fiber atrophy, oxidative stress, and oxidative fiber reduction are the attributes of different phenotypes in chronic obstructive pulmonary disease patients, Journal of Applied Physiology, vol.115, pp.1796-1805, 1985.
URL : https://hal.archives-ouvertes.fr/hal-01623970

J. M. Guralnik, E. M. Simonsick, and L. Ferrucci, A short physical performance battery assessing lower extremity function: association with self-reported disability and prediction of mortality and nursing home admission, Journal of Gerontology, vol.49, pp.85-94, 1994.

L. Kang, P. Han, and J. Wang, Timed Up and Go Test can predict recurrent falls: a longitudinal study of the community-dwelling elderly in China, Clinical interventions in aging, vol.12, pp.2009-2016, 2017.

E. Barry, R. Galvin, C. Keogh, F. Horgan, and T. Fahey, Is the Timed Up and Go test a useful predictor of risk of falls in community dwelling older adults: a systematic review and meta-analysis, BMC geriatrics, vol.14, p.14, 2014.

A. Corsonello, F. Lattanzio, and C. Pedone, Prognostic significance of the short physical performance battery in older patients discharged from acute care hospitals, Rejuvenation Research, vol.15, pp.41-48, 2012.

J. M. Pritchard, C. C. Kennedy, and S. Karampatos, Measuring frailty in clinical practice: a comparison of physical frailty assessment methods in a geriatric out-patient clinic, BMC Geriatrics, vol.17, p.264, 2017.

E. Nordin, N. Lindelöf, E. Rosendahl, J. Jensen, and L. Lundin-olsson, Prognostic validity of the Timed Up-and-Go test, a modified Get-Up-and-Go test, staff's global judgement and fall history in evaluating fall risk in residential care facilities, Age and Ageing, vol.37, pp.442-448, 2008.

M. Pfeifer, M. Sinaki, and P. Geusens, Musculoskeletal Rehabilitation in Osteoporosis: A Review. Journal of Bone and Mineral Research, vol.19, pp.1208-1214, 2004.

A. Trombetti, M. Hars, F. R. Herrmann, R. W. Kressig, S. Ferrari et al., Effect of music-based multitask training on gait, balance, and fall risk in elderly people: a randomized controlled trial, Archives of Internal Medicine, vol.171, pp.525-533, 2011.

T. M. Gill, D. I. Baker, M. Gottschalk, P. N. Peduzzi, H. Allore et al., A program to prevent functional decline in physically frail, elderly persons who live at home, The New England Journal of Medicine, vol.347, pp.1068-1074, 2002.

K. Nagai, T. Miyamato, and A. Okamae, Physical activity combined with resistance training reduces symptoms of frailty in older adults: A randomized controlled trial, Archives of Gerontology and Geriatrics, vol.76, pp.41-47, 2018.

M. A. Fiatarone, E. F. O'neill, and N. D. Ryan, Exercise training and nutritional supplementation for physical frailty in very elderly people, The New England Journal of Medicine, vol.330, pp.1769-1775, 1994.

D. Paddon-jones and B. B. Rasmussen, Dietary protein recommendations and the prevention of sarcopenia. Current Opinion in, Clinical Nutrition and Metabolic Care, vol.12, pp.86-90, 2009.

R. Saran, B. Robinson, and K. C. Abbott, US Renal Data System 2017 Annual Data Report: Epidemiology of Kidney Disease in the United States, American journal of kidney diseases : the official journal of the National Kidney Foundation, vol.71, pp.7-7, 2018.

M. A. Kaballo, M. Canney, O. Kelly, P. Williams, Y. O'seaghdha et al., A comparative analysis of survival of patients on dialysis and after kidney transplantation, Clinical Kidney Journal, vol.11, pp.389-393, 2018.

E. J. Kouidi, Central and peripheral adaptations to physical training in patients with endstage renal disease. Sports medicine (Auckland, N.Z.), vol.31, pp.651-665, 2001.

G. Serratrice, M. Toga, H. Roux, A. Murisasco, and G. De-bisschop,

, La Presse Medicale, vol.75, pp.1835-1838, 1967.

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

I. H. Fahal, Uraemic sarcopenia: aetiology and implications, Nephrology Dialysis Transplantation, vol.29, pp.1655-1665, 2014.

N. Isoyama, A. R. Qureshi, and C. M. Avesani, Comparative associations of muscle mass and muscle strength with mortality in dialysis patients, Clinical journal of the American Society of Nephrology: CJASN, vol.9, pp.1720-1728, 2014.

D. Marcelli, L. A. Usvyat, and P. Kotanko, Body composition and survival in dialysis patients: results from an international cohort study, Clinical journal of the American Society of Nephrology: CJASN, vol.10, pp.1192-1200, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02005484

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.

M. R. Deschenes, Effects of aging on muscle fibre type and size, Sports Medicine, vol.34, pp.809-824, 2004.

N. Clyne, M. Esbjörnsson, E. Jansson, T. Jogestrand, L. E. Lins et al., Effects of renal failure on skeletal muscle, Nephron, vol.63, pp.395-399, 1993.

I. H. Fahal, G. M. Bell, J. M. Bone, and R. H. Edwards, Physiological abnormalities of skeletal muscle in dialysis patients. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association, European Renal Association, vol.12, pp.119-127, 1997.

A. V. Crowe, A. Mcardle, and F. Mcardle, Markers of oxidative stress in the skeletal muscle of patients on haemodialysis, Nephrology Dialysis Transplantation, vol.22, pp.1177-1183, 2007.

M. I. Lewis, M. Fournier, and H. Wang, Metabolic and morphometric profile of muscle fibers in chronic hemodialysis patients, Journal of Applied Physiology, vol.112, pp.72-78, 2012.

J. M. Campistol, Uremic myopathy, Kidney International, vol.62, pp.1901-1913, 2002.

J. L. Gamboa, F. T. Billings, and M. T. Bojanowski, Mitochondrial dysfunction and oxidative stress in patients with chronic kidney disease, Physiological Reports, vol.4, 2016.

B. Roshanravan, B. Kestenbaum, and J. Gamboa, CKD and Muscle Mitochondrial Energetics. American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.68, pp.658-659, 2016.

K. G. Avin, N. X. Chen, and J. M. Organ, Skeletal Muscle Regeneration and Oxidative Stress Are Altered in Chronic Kidney Disease, PLOS ONE, vol.11, p.159411, 2016.

C. Xu, A. Kasimumali, and X. Guo, Reduction of mitochondria and up regulation of pyruvate dehydrogenase kinase 4 of skeletal muscle in patients with chronic kidney disease, Nephrology [Internet], 2019.

Y. Y. Zhang, L. J. Gu, and J. Huang, CKD autophagy activation and skeletal muscle atrophy-a preliminary study of mitophagy and inflammation, European Journal of Clinical Nutrition, vol.73, pp.950-960, 2019.

J. Aniort, A. Stella, and C. Philipponnet, Muscle wasting in patients with end-stage renal disease or early-stage lung cancer: common mechanisms at work, Journal of Cachexia, Sarcopenia and Muscle, vol.10, pp.323-337, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01882339

L. Zhang, X. H. Wang, H. Wang, J. Du, and W. E. Mitch, Satellite cell dysfunction and impaired IGF-1 signaling cause CKD-induced muscle atrophy, Journal of the American Society of Nephrology: JASN, vol.21, pp.419-427, 2010.

J. E. Friedman, G. L. Dohm, and C. W. Elton, Muscle insulin resistance in uremic humans: glucose transport, glucose transporters, and insulin receptors, American Journal of Physiology-Endocrinology and Metabolism, 1991.

G. K. Sakkas, Atrophy of non-locomotor muscle in patients with end-stage renal failure, Nephrology Dialysis Transplantation, vol.18, pp.2074-2081, 2003.

M. Panaye, A. Kolko-labadens, and C. Lasseur, Phenotypes influencing low physical activity in maintenance dialysis, Journal of Renal Nutrition: The Official Journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.25, pp.31-39, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01850601

D. Sousa, E. Veksler, V. Bigard, X. Mateo, P. Ventura-clapier et al., Heart failure affects mitochondrial but not myofibrillar intrinsic properties of skeletal muscle, Circulation, vol.102, pp.1847-1853, 2000.

P. Pomiès, J. Rodriguez, and M. Blaquière, Reduced myotube diameter, atrophic signalling and elevated oxidative stress in cultured satellite cells from COPD patients, Journal of Cellular and Molecular Medicine, vol.19, pp.175-186, 2015.

A. Chacon-cabrera, H. Lund-palau, J. Gea, and E. Barreiro, Time-Course of Muscle Mass Loss, Damage, and Proteolysis in Gastrocnemius following Unloading and Reloading: Implications in Chronic Diseases, PloS One, vol.11, p.164951, 2016.

P. Kittiskulnam, G. M. Chertow, J. J. Carrero, C. Delgado, G. A. Kaysen et al., Sarcopenia and its individual criteria are associated, in part, with mortality among patients on hemodialysis, Kidney International, vol.92, pp.238-247, 2017.

S. E. Wenzel, Asthma phenotypes: the evolution from clinical to molecular approaches, Nature Medicine, vol.18, pp.716-725, 2012.

J. P. Kooman, P. Kotanko, A. Schols, P. G. Shiels, and P. Stenvinkel, Chronic kidney disease and premature ageing, Nature Reviews. Nephrology, vol.10, pp.732-742, 2014.

X. H. Wang and W. E. Mitch, Mechanisms of muscle wasting in chronic kidney disease, Nature Reviews. Nephrology, vol.10, pp.504-516, 2014.

P. Stenvinkel and T. J. Ekström, Epigenetics and the uremic phenotype: a matter of balance, Contributions to Nephrology, vol.161, pp.55-62, 2008.

J. J. Carrero and P. Stenvinkel, The vulnerable man: impact of testosterone deficiency on the uraemic phenotype, Nephrology Dialysis Transplantation, vol.27, pp.4030-4041, 2012.

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

P. Kittiskulnam, J. J. Carrero, G. M. Chertow, G. A. Kaysen, C. Delgado et al., Sarcopenia among patients receiving hemodialysis: weighing the evidence, Journal of Cachexia, Sarcopenia and Muscle, vol.8, pp.57-68, 2017.

J. Souweine, N. Kuster, and L. Chenine, Physical inactivity and protein energy wasting play independent roles in muscle weakness in maintenance haemodialysis patients, PloS One, vol.13, p.200061, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01853078

B. C. Clark, T. M. Manini, and . Sarcopenia-=/=-dynapenia, Biological Sciences and Medical Sciences, vol.63, pp.829-834, 2008.

J. J. Carrero, K. L. Johansen, B. Lindholm, P. Stenvinkel, L. Cuppari et al., Screening for muscle wasting and dysfunction in patients with chronic kidney disease, Kidney International, 2016.

V. O. Leal, M. B. Stockler-pinto, and N. E. Farage, Handgrip strength and its dialysis determinants in hemodialysis patients, Nutrition, vol.27, pp.1125-1129, 2011.

C. Beaudart, M. Zaaria, F. Pasleau, J. Reginster, and O. Bruyère, Health Outcomes of Sarcopenia: A Systematic Review and Meta-Analysis, PloS one, vol.12, pp.169548-0169548, 2017.

K. Norman and L. Otten, Financial impact of sarcopenia or low muscle mass -A short review, Clinical Nutrition, vol.38, pp.1489-1495, 2019.

S. Von-haehling, J. E. Morley, and S. D. Anker, An overview of sarcopenia: facts and numbers on prevalence and clinical impact, Journal of Cachexia, Sarcopenia and Muscle, vol.1, pp.129-133, 2010.

T. R. Komatsu, F. S. Borim, A. L. Neri, and L. P. Corona, Association of dynapenia, obesity and chronic diseases with all-cause mortality of community-dwelling older adults: A path analysis, Geriatrics & Gerontology International, vol.19, pp.108-112, 2019.

K. L. Johansen, T. Shubert, J. Doyle, B. Soher, G. K. Sakkas et al., Muscle atrophy in patients receiving hemodialysis: effects on muscle strength, muscle quality, and physical function, Kidney International, vol.63, pp.291-297, 2003.

J. Kim, S. R. Choi, and M. J. Choi, Prevalence of and factors associated with sarcopenia in elderly patients with end-stage renal disease, Clinical Nutrition, vol.33, pp.64-68, 2014.

R. N. Foley, C. Wang, A. Ishani, A. J. Collins, and A. M. Murray, Kidney function and sarcopenia in the United States general population: NHANES III, American Journal of Nephrology, vol.27, pp.279-286, 2007.

M. Doma?ski and K. Ciechanowski, Sarcopenia: a major challenge in elderly patients with end-stage renal disease, Journal of aging research, vol.2012, p.754739, 2012.

W. R. Frontera and J. Ochala, Skeletal Muscle: A Brief Review of Structure and Function, Calcified Tissue International, vol.96, pp.183-195, 2015.

B. M. Carlson, The regeneration of skeletal muscle -a review, American Journal of Anatomy, vol.137, pp.119-149, 1973.

, Cellular and Molecular Regulation of Muscle Regeneration | Physiological Reviews, 2019.

D. J. Wilkinson, M. Piasecki, and P. J. Atherton, The age-related loss of skeletal muscle mass and function: Measurement and physiology of muscle fibre atrophy and muscle fibre loss in humans, Ageing Research Reviews, vol.47, pp.123-132, 2018.

R. R. Wolfe, The underappreciated role of muscle in health and disease1?3

A. Bigard, E. Boehm, V. Veksler, P. Mateo, K. Anflous et al., Muscle Unloading Induces Slow to Fast Transitions in Myofibrillar but not Mitochondrial Properties. Relevance to Skeletal Muscle Abnormalities in Heart Failure, Journal of Molecular and Cellular Cardiology, vol.30, pp.2391-2401, 1998.

P. Chauveau, K. Moreau, C. Lasseur, D. Fouque, C. Combe et al., Sarcopénie et myopathie urémique : similitudes et différences, Néphrologie & Thérapeutique, vol.12, pp.71-75, 2016.

D. Gallagher, E. Ruts, and M. Visser, Weight stability masks sarcopenia in elderly men and women, American Journal of Physiology. Endocrinology and Metabolism, vol.279, pp.366-375, 2000.

D. Marcelli, K. Brand, and P. Ponce, Longitudinal Changes in Body Composition in Patients After Initiation of Hemodialysis Therapy: Results From an International Cohort, Journal of Renal Nutrition: The Official Journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.26, pp.72-80, 2016.

B. H. Goodpaster, S. W. Park, and T. B. Harris, The loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition study, Biological Sciences and Medical Sciences, vol.61, pp.1059-1064, 2006.

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

D. Sit, A. K. Kadiroglu, H. Kayabasi, and M. E. Yilmaz, The prevalence of insulin resistance in nondiabetic nonobese patients with chronic kidney disease, Advances in Therapy, vol.23, pp.988-998, 2006.

C. S. Carter, J. N. Justice, and L. Thompson, Lipotoxicity, aging, and muscle contractility: does fiber type matter?, GeroScience, vol.41, pp.297-308, 2019.

S. W. Lee, G. Dai, Z. Hu, X. Wang, J. Du et al., Regulation of muscle protein degradation: coordinated control of apoptotic and ubiquitin-proteasome systems by phosphatidylinositol 3 kinase, Journal of the American Society of Nephrology: JASN, vol.15, pp.1537-1545, 2004.

V. R. Rajan and W. E. Mitch, Muscle wasting in chronic kidney disease: the role of the ubiquitin proteasome system and its clinical impact, Pediatric Nephrology, vol.23, pp.527-535, 2008.

P. Balagopal, O. E. Rooyackers, D. B. Adey, P. A. Ades, and K. S. Nair, Effects of aging on in vivo synthesis of skeletal muscle myosin heavy-chain and sarcoplasmic protein in humans, The American Journal of Physiology, vol.273, pp.790-800, 1997.

A. Tournadre, G. Vial, F. Capel, M. Soubrier, Y. Boirie et al., Revue du Rhumatisme, vol.86, pp.39-45, 2019.

J. J. Carrero, M. Chmielewski, and J. Axelsson, Muscle atrophy, inflammation and clinical outcome in incident and prevalent dialysis patients, Clinical nutrition, vol.27, pp.557-564, 2008.

M. Piasecki, A. Ireland, and J. Piasecki, Failure to expand the motor unit size to compensate for declining motor unit numbers distinguishes sarcopenic from nonsarcopenic older men, The Journal of Physiology, vol.596, pp.1627-1637, 2018.

T. J. Doherty, Invited review: Aging and sarcopenia, Journal of applied physiology, vol.95, pp.1717-1727, 2003.

G. Garibotto, R. Russo, and A. Sofia, Skeletal muscle protein synthesis and degradation in patients with chronic renal failure, Kidney International, vol.45, pp.1432-1439, 1994.

. Ckd-evaluation, -. Management, and . Kdigo, , 2019.

D. Adey, R. Kumar, J. T. Mccarthy, and K. S. Nair, Reduced synthesis of muscle proteins in chronic renal failure, American journal of physiology. Endocrinology and metabolism, vol.278, pp.219-225, 2000.

L. B. Verdijk, R. Koopman, G. Schaart, K. Meijer, 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.

E. Carmeli, R. Coleman, and A. Z. Reznick, The biochemistry of aging muscle, Experimental Gerontology, vol.37, pp.477-489, 2002.

A. M. Corsi, L. Ferrucci, A. Gozzini, A. Tanini, and M. L. Brandi, Myostatin polymorphisms and age-related sarcopenia in the Italian population, Journal of the American Geriatrics Society, vol.50, p.1463, 2002.

A. G. Williams, M. P. Rayson, and M. Jubb, The ACE gene and muscle performance, Nature, vol.403, pp.614-614, 2000.

C. Annweiler, A. Schott, G. Berrut, B. Fantino, and O. Beauchet, Vitamin D-related changes in physical performance: A systematic review. The journal of nutrition, health & aging, vol.13, pp.893-898, 2009.

L. A. Herndon, P. J. Schmeissner, and J. M. Dudaronek, Stochastic and genetic factors influence tissue-specific decline in ageing C. elegans, Nature, vol.419, pp.808-814, 2002.

P. Lim, Y. Ma, and Y. Cheng, Mitochondrial DNA mutations and oxidative damage in skeletal muscle of patients with chronic uremia, Journal of biomedical science, vol.9, pp.549-560, 2002.

P. Kortebein, A. Ferrando, J. Lombeida, R. Wolfe, and W. J. Evans, Effect of 10 days of bed rest on skeletal muscle in healthy older adults, JAMA, vol.297, pp.1772-1774, 2007.

E. Edström, M. Altun, and E. Bergman, Factors contributing to neuromuscular impairment and sarcopenia during aging, Physiology & Behavior, vol.92, pp.129-135, 2007.

R. E. Ahonen, Striated muscle ultrastructure in uremic patients and in renal transplant recipients, Acta Neuropathologica, vol.50, pp.163-166, 1980.

D. Mckenzie, E. Bua, S. Mckiernan, Z. Cao, and J. M. Aiken, Mitochondrial DNA deletion mutations, European Journal of Biochemistry, vol.269, pp.2010-2015, 2002.

D. L. Waters, P. G. Mullins, C. R. Qualls, D. Raj, C. Gasparovic et al., Mitochondrial function in physically active elders with sarcopenia, Mechanisms of ageing and development, vol.130, pp.315-319, 2009.

E. Marzetti, R. Calvani, and M. Cesari, 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.

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, The FASEB Journal, vol.15, pp.322-332, 2001.

S. Fulle, F. Protasi, D. Tano, and G. , The contribution of reactive oxygen species to sarcopenia and muscle ageing, Experimental Gerontology, vol.39, pp.17-24, 2004.

R. Meex, V. B. Schrauwen-hinderling, and E. Moonen-kornips, Restoration of Muscle Mitochondrial Function and Metabolic Flexibility in Type 2 Diabetes by Exercise Training Is Paralleled by Increased Myocellular Fat Storage and Improved Insulin Sensitivity, Diabetes, vol.59, pp.572-579, 2010.

R. M. Dodds, K. Davies, and A. Granic, Mitochondrial respiratory chain function and content are preserved in the skeletal muscle of active very old men and women, Experimental Gerontology, vol.113, pp.80-85, 2018.

J. E. Morley, The diagnosis of late life hypogonadism, The Aging Male, vol.10, pp.217-220, 2007.

S. Bhasin, W. E. Taylor, and R. Singh, The mechanisms of androgen effects on body composition: mesenchymal pluripotent cell as the target of androgen action, Biological Sciences and Medical Sciences, vol.58, pp.1103-1110, 2003.

P. J. Snyder, S. Bhasin, and G. R. Cunningham, Lessons From the Testosterone Trials. Endocrine Reviews, vol.39, pp.369-386, 2018.

E. T. Schroeder, A. F. Vallejo, and L. Zheng, Six-Week Improvements in Muscle Mass and Strength During Androgen Therapy in Older Men, The Journals of Gerontology: Series A, vol.60, pp.1586-1592, 2005.

D. E. Jacobsen, M. M. Samson, S. Kezic, and H. Verhaar, Postmenopausal HRT and tibolone in relation to muscle strength and body composition, Maturitas, vol.58, pp.7-18, 2007.

P. Garcia-alfaro, S. Garcia, I. Rodríguez, F. Tresserra, and F. R. Pérez-lópez, Factors related to muscle strength in postmenopausal women aged younger than 65 years with normal vitamin D status, Climacteric: The Journal of the International Menopause Society, vol.22, pp.390-394, 2019.

M. L. Maltais, J. Desroches, and I. J. Dionne, Changes in muscle mass and strength after menopause, p.13

R. Boland, Role of vitamin D in skeletal muscle function, Endocrine Reviews, vol.7, pp.434-448, 1986.

M. Garcia, M. Seelaender, A. Sotiropoulos, D. Coletti, and A. H. Lancha, Vitamin D, muscle recovery, sarcopenia, cachexia, and muscle atrophy, Nutrition, vol.60, pp.66-69, 2019.

, Addressing the Musculoskeletal Components of Fracture Risk with Calcium and Vitamin D: A Review of the Evidence | SpringerLink, 2019.

M. Visser, D. Deeg, and P. Lips, Longitudinal Aging Study Amsterdam. Low vitamin D and high parathyroid hormone levels as determinants of loss of muscle strength and muscle mass (sarcopenia): the Longitudinal Aging Study Amsterdam, The Journal of Clinical Endocrinology and Metabolism, vol.88, pp.5766-5772, 2003.

W. Chang, C. Wu, and L. Hsu, Serum vitamin D, intact parathyroid hormone, and Fetuin A concentrations were associated with geriatric sarcopenia and cardiac hypertrophy, Scientific Reports, vol.7, p.40996, 2017.

T. Sikjaer, L. Rolighed, A. Hess, A. Fuglsang-frederiksen, L. Mosekilde et al., Effects of PTH(1-84) therapy on muscle function and quality of life in hypoparathyroidism: results from a randomized controlled trial. Osteoporosis international: a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the, USA, vol.25, pp.1717-1726, 2014.

M. Pfeifer, B. Begerow, and H. W. Minne, Vitamin D and Muscle Function, p.8, 2002.

P. Stenvinkel, J. J. Carrero, F. Von-walden, T. A. Ikizler, and G. A. Nader, Muscle wasting in endstage renal disease promulgates premature death: established, emerging and potential novel treatment strategies. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association, European Renal Association, vol.31, pp.1070-1077, 2016.

K. Makki, P. Froguel, and I. Wolowczuk, Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines, ISRN inflammation, p.139239, 2013.

M. E. Rodríguez-ortiz, A. Canalejo, and C. Herencia, Magnesium modulates parathyroid hormone secretion and upregulates parathyroid receptor expression at moderately low calcium concentration. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association, European Renal Association, vol.29, pp.282-289, 2014.

E. M. Onal, B. Afsar, A. Covic, N. D. Vaziri, and M. Kanbay, Gut microbiota and inflammation in chronic kidney disease and their roles in the development of cardiovascular disease, Hypertension Research, vol.42, pp.123-140, 2019.

K. Kalantar-zadeh, T. A. Ikizler, G. Block, M. M. Avram, and J. D. Kopple, Malnutritioninflammation complex syndrome in dialysis patients: causes and consequences, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.42, pp.864-881, 2003.

B. P. Oberg, E. Mcmenamin, and F. L. Lucas, Increased prevalence of oxidant stress and inflammation in patients with moderate to severe chronic kidney disease, Kidney International, vol.65, pp.1009-1016, 2004.

G. Pertosa, G. Grandaliano, L. Gesualdo, and F. P. Schena, Clinical relevance of cytokine production in hemodialysis, Kidney International. Supplement, vol.76, pp.104-111, 2000.

D. Rios, M. B. Pinheiro, and W. V. De-oliveira-junior, Cytokine Signature in End-Stage Renal Disease Patients on Hemodialysis. Disease Markers, 2017.

R. Schindler, R. Senf, and U. Frei, Influencing the inflammatory response of haemodialysis patients by cytokine elimination using large-pore membranes, Nephrology Dialysis Transplantation, vol.17, pp.17-19, 2002.

L. F. Fried, J. S. Lee, and M. Shlipak, Chronic kidney disease and functional limitation in older people: health, aging and body composition study, Journal of the American Geriatrics Society, vol.54, pp.750-756, 2006.

N. Mangner, A. Linke, and A. Oberbach, Exercise Training Prevents TNF-? Induced Loss of Force in the Diaphragm of Mice, PLOS ONE, vol.8, p.52274, 2013.

L. A. Schaap, S. Pluijm, D. Deeg, and M. Visser, Inflammatory markers and loss of muscle mass (sarcopenia) and strength, The American Journal of Medicine, vol.119, pp.526-535, 2006.

R. Roubenoff and V. A. Hughes, Sarcopenia: current concepts, Biological Sciences and Medical Sciences, vol.55, pp.716-724, 2000.

L. Koppe, D. Fouque, and K. Kalantar-zadeh, Kidney cachexia or protein-energy wasting in chronic kidney disease: facts and numbers, Sarcopenia and Muscle, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02195232

X. Moreau-gaudry, G. Jean, and L. Genet, A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients, Journal of Renal Nutrition: The Official Journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.24, pp.395-400, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01858992

J. J. Carrero, Identification of Patients With Eating Disorders: Clinical and Biochemical Signs of Appetite Loss in Dialysis Patients, Journal of Renal Nutrition, vol.19, pp.10-15, 2009.

S. Bataille, J. Bon, and A. Kolko, Prise en charge pratique de la dénutrition des patients hémodialysés en France : enquête nationale, Néphrologie & Thérapeutique, vol.15, pp.136-142, 2019.

A. Ikizler, H. Greene, L. Wingard, A. Parker, and M. Hakim, Spontaneous Dietary Protein Intake During Progression of Chronic Renal Failure1'2, Journal of the American Society of Nephrology, vol.5, p.6

W. W. Campbell, T. A. Trappe, R. R. Wolfe, and W. J. Evans, The recommended dietary allowance for protein may not be adequate for older people to maintain skeletal muscle, Biological Sciences and Medical Sciences, vol.56, pp.373-380, 2001.

M. Bonnefoy, C. Cornu, and N. S. , The effects of exercise and protein-energy supplements on body composition and muscle function in frail elderly individuals: a long-term controlled randomised study, The British Journal of Nutrition, vol.89, pp.731-739, 2003.
URL : https://hal.archives-ouvertes.fr/hal-02357899

R. R. Wolfe, The role of dietary protein in optimizing muscle mass, function and health outcomes in older individuals, The British Journal of Nutrition, vol.108, issue.2, pp.88-93, 2012.

S. J. Peterson and M. Mozer, Differentiating Sarcopenia and Cachexia Among Patients With Cancer, Nutrition in Clinical Practice, vol.32, pp.30-39, 2017.

D. Fouque, K. Kalantar-zadeh, and J. Kopple, A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease, Kidney International, vol.73, pp.391-398, 2008.

X. Moreau-gaudry, G. Jean, and L. Genet, A simple protein-energy wasting score predicts survival in maintenance hemodialysis patients, Journal of Renal Nutrition: The Official Journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.24, pp.395-400, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01858992

J. J. Carrero, P. Stenvinkel, and L. Cuppari, Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM), Renal Nutrition of the National Kidney Foundation, vol.23, pp.77-90, 2013.

J. A. Kraut and N. E. Madias, Metabolic Acidosis of CKD: An Update, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.67, pp.307-317, 2016.

J. A. Kraut and N. E. Madias, Adverse Effects of the Metabolic Acidosis of Chronic Kidney Disease, Advances in Chronic Kidney Disease, vol.24, pp.289-297, 2017.

R. Mehrotra, J. D. Kopple, and M. Wolfson, Metabolic acidosis in maintenance dialysis patients: Clinical considerations, Kidney International, vol.64, pp.13-25, 2003.

Y. Zha and Q. Qian, Protein Nutrition and Malnutrition in CKD and ESRD, Nutrients, vol.9, 2017.

E. Hultman, D. Canale, S. Sjöholm, and H. , Effect of induced metabolic acidosis on intracellular pH, buffer capacity and contraction force of human skeletal muscle, Clinical Science, vol.69, pp.505-510, 1985.

J. L. Bailey, B. K. England, R. C. Long, J. Weissman, and W. E. Mitch, Experimental acidemia and muscle cell pH in chronic acidosis and renal failure, American Journal of Physiology-Cell Physiology, vol.269, pp.706-712, 1995.

B. Canaud, J. Cristol, M. Morena, H. Leray-moragues, J. Bosc et al., Imbalance of oxidants and antioxidants in haemodialysis patients, Blood purification, vol.17, pp.99-106, 1999.

M. Morena, J. Cristol, L. Senécal, H. Leray-moragues, D. Krieter et al., Oxidative stress in hemodialysis patients: is NADPH oxidase complex the culprit?, Kidney international. Supplement, pp.109-114, 2002.

M. A. Aminzadeh, S. B. Nicholas, K. C. Norris, and N. D. Vaziri, Role of impaired Nrf2 activation in the pathogenesis of oxidative stress and inflammation in chronic tubulo-interstitial nephropathy. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association, European Renal Association, vol.28, pp.2038-2045, 2013.

D. Raj, E. A. Dominic, and A. Pai, Skeletal muscle, cytokines, and oxidative stress in end-stage renal disease, Kidney international, vol.68, pp.2338-2344, 2005.

M. Tamaki, K. Miyashita, S. Wakino, M. Mitsuishi, K. Hayashi et al., Chronic kidney disease reduces muscle mitochondria and exercise endurance and its exacerbation by dietary protein through inactivation of pyruvate dehydrogenase, Kidney International, vol.85, pp.1330-1339, 2014.

C. H. Thompson, G. J. Kemp, D. J. Taylor, J. G. Ledingham, G. K. Radda et al., No effect of blood transfusion on muscle metabolism, The Quarterly Journal of Medicine, vol.85, pp.897-899, 1992.

J. L. Teruel, R. Marcen, J. Navarro-antolin, A. Aguilera, G. Fernandez-juarez et al., Androgen versus erythropoietin for the treatment of anemia in hemodialyzed patients: a prospective study, Journal of the American Society of Nephrology: JASN, vol.7, pp.140-144, 1996.

X. H. Wang, J. Du, J. D. Klein, J. L. Bailey, and W. E. Mitch, Exercise ameliorates chronic kidney disease-induced defects in muscle protein metabolism and progenitor cell function, Kidney International, vol.76, pp.751-759, 2009.

W. Yang, Y. Zhang, Y. Li, Z. Wu, and D. Zhu, Myostatin induces cyclin D1 degradation to cause cell cycle arrest through a phosphatidylinositol 3-kinase/AKT/GSK-3 beta pathway and is antagonized by insulin-like growth factor 1, The Journal of Biological Chemistry, vol.282, pp.3799-3808, 2007.

P. Esposito, L. Porta, E. Calatroni, and M. , Modulation of Myostatin/Hepatocyte Growth Factor Balance by Different Hemodialysis Modalities, BioMed Research International, p.7635459, 2017.

D. Wang, Y. Yang, R. Huang, Z. Zhang, and X. Lin, Myostatin Activates the Ubiquitin-Proteasome and Autophagy-Lysosome Systems Contributing to Muscle Wasting in Chronic Kidney Disease, Oxidative Medicine and Cellular Longevity, vol.2015, pp.1-18, 2015.

L. Koppe, N. J. Pillon, and R. E. Vella, p-Cresyl sulfate promotes insulin resistance associated with CKD, Journal of the American Society of Nephrology: JASN, vol.24, pp.88-99, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00850482

F. C. Barreto, D. V. Barreto, S. Liabeuf, T. B. Drüeke, and Z. A. Massy, Effects of uremic toxins on vascular and bone remodeling, Seminars in Dialysis, vol.22, pp.433-437, 2009.

T. Sutra, M. Morena, A. Bargnoux, B. Caporiccio, B. Canaud et al., Superoxide production: a procalcifying cell signalling event in osteoblastic differentiation of vascular smooth muscle cells exposed to calcification media, Free Radical Research, vol.42, pp.789-797, 2008.

G. Caso and P. J. Garlick, Control of muscle protein kinetics by acid-base balance. Current Opinion in, Clinical Nutrition and Metabolic Care, vol.8, pp.73-76, 2005.

C. Verove, N. Maisonneuve, A. A. El, A. Boldron, and R. Azar, Effect of the correction of metabolic acidosis on nutritional status in elderly patients with chronic renal failure, Journal of renal nutrition : the official journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.12, pp.224-228, 2002.

H. Crossland, D. Constantin-teodosiu, S. M. Gardiner, D. Constantin, and P. L. Greenhaff, A potential role for Akt/FOXO signalling in both protein loss and the impairment of muscle carbohydrate oxidation during sepsis in rodent skeletal muscle, The Journal of Physiology, vol.586, pp.5589-5600, 2008.

E. Sato, T. Mori, and E. Mishima, Metabolic alterations by indoxyl sulfate in skeletal muscle induce uremic sarcopenia in chronic kidney disease, Scientific Reports, vol.6, p.36618, 2016.

J. E. Morley, A. M. Abbatecola, and J. M. Argiles, Sarcopenia With Limited Mobility: An International Consensus, Journal of the American Medical Directors Association, vol.12, pp.403-409, 2011.

M. D. Miller, M. Crotty, and L. C. Giles, Corrected arm muscle area: an independent predictor of long-term mortality in community-dwelling older adults, Journal of the American Geriatrics Society, vol.50, pp.1272-1277, 2002.

F. Gouzi, C. Préfaut, and A. Abdellaoui, Evidence of an early physical activity reduction in chronic obstructive pulmonary disease patients, Archives of physical medicine and rehabilitation, vol.92, pp.1611-1617, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01623976

J. B. Wikholm and R. W. Bohannon, Hand-held Dynamometer Measurements: Tester Strength Makes a Difference, The Journal of Orthopaedic and Sports Physical Therapy, vol.13, pp.191-198, 1991.

R. W. Bohannon, J. Kindig, G. Sabo, A. E. Duni, and P. Cram, Isometric knee extension force measured using a handheld dynamometer with and without belt-stabilization, Physiotherapy Theory and Practice, vol.28, pp.562-568, 2012.

J. Souweine, A. Boudet, and L. Chenine, Standardized Method to Measure Muscle Force at the Bedside in Hemodialysis Patients, Journal of Renal Nutrition: The Official Journal of the Council on Renal Nutrition of the National Kidney Foundation, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01774962

E. Dent, J. E. Morley, and C. Aj, Physical Frailty: ICFSR International Clinical Practice Guidelines for Identification and Management. The journal of nutrition, health & aging, vol.23, pp.771-787, 2019.

C. E. Haugen, N. M. Chu, and H. Ying, Frailty and Access to Kidney Transplantation, Clinical Journal of the American Society of Nephrology, vol.14, pp.576-582, 2019.

M. A. Mcadams-demarco, A. Law, and E. King, Frailty and Mortality in Kidney Transplant Recipients, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, vol.15, pp.149-154, 2015.

E. Van-den-ham, J. P. Kooman, and A. Schols, Similarities in skeletal muscle strength and exercise capacity between renal transplant and hemodialysis patients, American Journal of Transplantation: Official Journal of the American Society of Transplantation and the American Society of Transplant Surgeons, vol.5, pp.1957-1965, 2005.

S. P. Stam, M. F. Eisenga, and A. W. Gomes-neto, Muscle mass determined from urinary creatinine excretion rate, and muscle performance in renal transplant recipients, 2019.

I. Serres, V. Gautier, A. Varray, and C. Préfaut, Impaired skeletal muscle endurance related to physical inactivity and altered lung function in COPD patients, Chest, vol.113, pp.900-905, 1998.
URL : https://hal.archives-ouvertes.fr/hal-01625242

L. Androga, D. Sharma, A. Amodu, and M. K. Abramowitz, Sarcopenia, Obesity, and Mortality in US Adults With and Without Chronic Kidney Disease, Kidney International Reports, vol.2, pp.201-211, 2017.

A. P. Cerri, G. Bellelli, and A. Mazzone, Sarcopenia and malnutrition in acutely ill hospitalized elderly: Prevalence and outcomes, Clinical Nutrition, vol.34, pp.745-751, 2015.

P. Chauveau, K. Moreau, C. Lasseur, C. Combe, and M. Aparicio, Approches thérapeutiques communes de la sarcopénie du sujet âgé et de la myopathie urémique, Néphrologie & Thérapeutique, vol.13, pp.511-517, 2017.

M. I. Carter and P. S. Hinton, Physical Activity and Bone Health. Missouri Medicine, vol.111, pp.59-64, 2014.

F. Branca and S. Vatueña, Calcium, physical activity and bone health -building bones for a stronger future, Public Health Nutrition, vol.4, pp.117-123, 2001.

P. A. Quigley, T. Bulat, and S. Hart-hughes, Strategies to reduce risk of fall-related injuries in rehabilitation nursing. Rehabilitation Nursing: The Official Journal of the Association of Rehabilitation, Nurses, vol.32, pp.120-125, 2007.

, Recommandations mondiales sur l'activité physique pour la santé, 2010.

E. Aizen and E. Zlotver, Prediction of falls in rehabilitation and acute care geriatric setting, Journal of Clinical Gerontology and Geriatrics, vol.4, pp.57-61, 2013.

N. Anderton, A. Giri, and G. Wei, Sedentary Behavior in Individuals With Diabetic Chronic Kidney Disease and Maintenance Hemodialysis, Journal of Renal Nutrition, vol.25, pp.364-370, 2015.

M. A. Mcadams-demarco, Y. H. Van-pilsum-rasmussen, and S. , Prehabilitation prior to kidney transplantation: Results from a pilot study, Clinical Transplantation, vol.33, p.13450, 2019.

H. Akima, K. Kubo, and M. Imai, Inactivity and muscle: effect of resistance training during bed rest on muscle size in the lower limb, Acta Physiologica Scandinavica, vol.172, pp.269-278, 2001.

M. Roig, K. O'brien, and G. Kirk, The effects of eccentric versus concentric resistance training on muscle strength and mass in healthy adults: a systematic review with metaanalysis, British Journal of Sports Medicine, vol.43, pp.556-568, 2009.

K. Michael, Relationship of Skeletal Muscle Atrophy to Functional Status: A Systematic Research Review, Biological Research For Nursing, vol.2, pp.117-131, 2000.

D. L. Kirkman, P. Mullins, N. A. Junglee, M. Kumwenda, M. M. Jibani et al., Anabolic exercise in haemodialysis patients: a randomised controlled pilot study, Journal of Cachexia, Sarcopenia and Muscle, vol.5, pp.199-207, 2014.

V. Rigalleau, V. Blanchetier, and C. Combe, A low-protein diet improves insulin sensitivity of endogenous glucose production in predialytic uremic patients, The American Journal of Clinical Nutrition, vol.65, pp.1512-1516, 1997.

L. Koppe, E. Nyam, and K. Vivot, Urea impairs ? cell glycolysis and insulin secretion in chronic kidney disease, The Journal of Clinical Investigation, vol.126, pp.3598-3612, 2016.

D. Fouque, R. Horne, M. Cozzolino, and K. Kalantar-zadeh, Balancing nutrition and serum phosphorus in maintenance dialysis, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.64, pp.143-150, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01859016

A. R. Chang and C. Anderson, Dietary Phosphorus Intake and the Kidney, Annual review of nutrition, vol.37, pp.321-346, 2017.

J. D. Kopple, National kidney foundation K/DOQI clinical practice guidelines for nutrition in chronic renal failure, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.37, pp.66-70, 2001.

D. E. St-jules, K. Woolf, M. Pompeii, and M. A. Sevick, Exploring problems in following the hemodialysis diet, and their relation to energy and nutrient intakes: The Balance Wise Study, Journal of renal nutrition : the official journal of the Council on Renal Nutrition of the National Kidney Foundation, vol.26, pp.118-124, 2016.

K. Kalantar-zadeh, N. J. Cano, and K. Budde, Diets and enteral supplements for improving outcomes in chronic kidney disease, Nature Reviews. Nephrology, vol.7, pp.369-384, 2011.

C. Pasian, R. Azar, and D. Fouque,

, Nephrologie & Therapeutique, vol.12, pp.496-502, 2016.

N. Cano, D. Fouque, and H. Roth, Intradialytic parenteral nutrition does not improve survival in malnourished hemodialysis patients: a 2-year multicenter, prospective, randomized study, Journal of the American Society of Nephrology : JASN, vol.18, pp.2583-2591, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00388543

D. Fouque, M. Vennegoor, and P. Ter-wee, EBPG guideline on nutrition. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association -European Renal Association, vol.22, pp.45-87, 2007.

C. Delgado and L. Frassetto, Hip Fractures in Hemodialysis Patients, American Journal of Kidney Diseases, vol.62, pp.653-655, 2013.

N. Zahed, S. Chehrazi, and K. Falaknasi, The evaluation of relationship between vitamin D and muscle power by micro manual muscle tester in end-stage renal disease patients. Saudi Journal of Kidney Diseases and Transplantation: An Official Publication of the Saudi Center for Organ Transplantation, vol.25, pp.998-1003, 2014.

H. Taskapan, O. Baysal, D. Karahan, B. Durmus, Z. Altay et al., Vitamin D and muscle strength, functional ability and balance in peritoneal dialysis patients with vitamin D deficiency, Clinical Nephrology, vol.76, pp.110-116, 2011.

J. D. Kopple, A. K. Cheung, and J. S. Christiansen, OPPORTUNITY™: a large-scale randomized clinical trial of growth hormone in hemodialysis patients. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association, European Renal Association, vol.26, pp.4095-4103, 2011.

G. Johannsson, B. A. Bengtsson, and J. Ahlmén, Double-blind, placebo-controlled study of growth hormone treatment in elderly patients undergoing chronic hemodialysis: anabolic effect and functional improvement, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.33, pp.709-717, 1999.

S. Heiwe and S. H. Jacobson, Exercise training in adults with CKD: a systematic review and meta-analysis, American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation, vol.64, pp.383-393, 2014.

K. M. Baldwin, T. P. White, and S. B. Arnaud, Musculoskeletal adaptations to weightlessness and development of effective countermeasures, Medicine and Science in Sports and Exercise, vol.28, pp.1247-1253, 1996.

C. Polge, A. Heng, and M. Jarzaguet, Muscle actin is polyubiquitinylated in vitro and in vivo and targeted for breakdown by the E3 ligase MuRF1, FASEB journal: official publication of the Federation of American Societies for Experimental Biology, vol.25, pp.3790-3802, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02420104

D. Glick, S. Barth, and K. F. Macleod, Autophagy: cellular and molecular mechanisms, The Journal of pathology, vol.221, pp.3-12, 2010.

B. Levine, N. Mizushima, and H. W. Virgin, Autophagy in immunity and inflammation, Nature, vol.469, pp.323-335, 2011.

Z. Su, J. D. Klein, and J. Du, Chronic kidney disease induces autophagy leading to dysfunction of mitochondria in skeletal muscle, American Journal of Physiology-Renal Physiology, vol.312, pp.1128-1140, 2017.

J. Kim, S. R. Choi, and M. J. Choi, Prevalence of and factors associated with sarcopenia in elderly patients with end-stage renal disease, Clinical nutrition, 2013.

R. J. Maughan, J. S. Watson, and J. Weir, Strength and cross-sectional area of human skeletal muscle, The Journal of Physiology, vol.338, pp.37-49, 1983.

E. Van-den-ham, J. P. Kooman, and A. Schols, Similarities in skeletal muscle strength and exercise capacity between renal transplant and hemodialysis patients, American Journal of Transplantation: Official Journal of the American Society of Transplantation and the American Society of Transplant Surgeons, vol.5, pp.1957-1965, 2005.

B. Canaud, A. Granger-vallée, and N. Molinari, Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS one, vol.9, p.93286, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02637906

J. M. Seymour, M. A. Spruit, and N. S. Hopkinson, The prevalence of quadriceps weakness in COPD and the relationship with disease severity, The European Respiratory Journal, vol.36, pp.81-88, 2010.

R. A. Pereira, A. C. Cordeiro, and C. M. Avesani, Sarcopenia in chronic kidney disease on conservative therapy: prevalence and association with mortality. Nephrology, Dialysis, Transplantation: Official Publication of the European Dialysis and Transplant Association, European Renal Association, vol.30, pp.1718-1725, 2015.

G. Baptista, A. Dupuy, and A. Jaussent, Low-grade chronic inflammation and superoxide anion production by NADPH oxidase are the main determinants of physical frailty in older adults, Free radical research, vol.46, pp.1108-1114, 2012.

P. Stenvinkel, O. Heimbürger, B. Lindholm, G. A. Kaysen, and J. Bergström, Are there two types of malnutrition in chronic renal failure? Evidence for relationships between malnutrition, inflammation and atherosclerosis (MIA syndrome). Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association, European Renal Association, vol.15, pp.953-960, 2000.

B. Canaud, A. Granger-vallée, and N. Molinari, Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS One, vol.9, p.93286, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02637906

B. C. Clark and T. M. Manini, What is dynapenia?, Nutrition, vol.28, pp.495-503, 2012.

M. K. Han, A. Agusti, and P. M. Calverley, Chronic obstructive pulmonary disease phenotypes: the future of COPD, American Journal of Respiratory and Critical Care Medicine, vol.182, pp.598-604, 2010.

L. Koppe, D. Fouque, and K. Kalantar-zadeh, Kidney cachexia or protein-energy wasting in chronic kidney disease: facts and numbers, Journal of Cachexia, Sarcopenia and Muscle, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02195232

J. J. Carrero, K. L. Johansen, B. Lindholm, P. Stenvinkel, L. Cuppari et al., Screening for muscle wasting and dysfunction in patients with chronic kidney disease, Kidney International, vol.90, pp.53-66, 2016.

M. I. Lewis, M. Fournier, and H. Wang, Metabolic and morphometric profile of muscle fibers in chronic hemodialysis patients, Journal of applied physiology, vol.112, pp.72-78, 1985.

J. L. Bailey, B. Zheng, Z. Hu, S. R. Price, and W. E. Mitch, Chronic kidney disease causes defects in signaling through the insulin receptor substrate/phosphatidylinositol 3-kinase/Akt pathway: implications for muscle atrophy, Journal of the American Society of Nephrology: JASN, vol.17, pp.1388-1394, 2006.

J. L. Bailey, X. Wang, B. K. England, S. R. Price, X. Ding et al., The acidosis of chronic renal failure activates muscle proteolysis in rats by augmenting transcription of genes encoding proteins of the ATP-dependent ubiquitin-proteasome pathway, The Journal of clinical investigation, vol.97, pp.1447-1453, 1996.

R. E. Ahonen, Light microscopic study of striated muscle in uremia, Acta Neuropathologica, vol.49, pp.51-55, 1980.

J. M. Garonzik-wang, P. Govindan, and J. W. Grinnan, Frailty and Delayed Graft Function in Kidney Transplant Recipients, Archives of Surgery, vol.147, pp.190-193, 2012.

M. A. Makary, D. L. Segev, and P. J. Pronovost, Frailty as a Predictor of Surgical Outcomes in Older Patients, Journal of the American College of Surgeons, vol.210, pp.901-908, 2010.

M. A. Mcadams-demarco, A. Law, and M. L. Salter, Frailty and Early Hospital Readmission after Kidney Transplantation, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons, vol.13, pp.2091-2095, 2013.

B. Granger and . Ma-santé, des moyens limités. Le Débat, vol.203, pp.98-108, 2019.

, The main objective of this study is to characterize frailty in chronic hemodialysis (CHD) patients

, Primary objective is to evaluate at inclusion the relationships between frailty (assessed by the short physical performance battery -SPPB -test score) and muscle dysfunction assessed by the quadriceps isometric maximal voluntary force (MVF)

, Secondary objectives are to identify the relationships between i) SPPB score and muscle mass at inclusion ii) SPPB score and PEW at inclusion iii) SPPB score and inflammation at inclusion iv) SPPB score and myokines at inclusion v) myokine at inclusion and SPPB score after 2 years of follow-up vi) myokine levels (and SPPB score) at inclusion and adverse event (mortality) decreased physiological reserves and poor response to stressors, main features of the frailty condition

, Thus, identifying frail patients is highly warranted in CHD patients. The Fried frailty criteria includes weakness, slowness, low level of physical activity, low energy or self-reported exhaustion and unintentional weight loss, vol.7

, It consists of three timed tasks: standing balance, walking speed, and chair stand tests. Every task may account a score between 0 and 4; the total SPPB score ranges from 0

, Muscle function assessed by muscle strength at the patient bedside is associated with physical inactivity. Weakness influences the independence in activities of daily living in these patients. Thus, factors like physical activity reduction, leading to deconditioning, can also be involved in frailty. Sarcopenia, defined as skeletal muscle weakness associated with reduced muscle mass, and dynapenia, defined as muscle weakness with normal mass, are emerging risk factor in CHD patients. Currently, no prognostic biomarkers predict muscle damage in these patients. In fact, during muscular dysfunction signaling pathways are altered. Beyong proteolysis activity and autophagy,myokines, cytokines that are produced and released by muscle cells with autocrine, paracrine and/or endocrine effects are modified. This signalling pathway impairment is currently explored in the laboratory (subject of my PhD thesis, Frailty could be associated with muscular dysfunction. Low muscle mass estimated by bioelectrical impedance analysis

, Beyond classical factors of frailty in elderly, some elements are specific to CHD patients. In fact, malnutrition and other factors including inflammation, low physical activity, metabolic acidosis, insulin resistance, may also be involved in muscle weakness of CHD patients, vol.11

, All these factors, which are related to protein energy wasting (PEW), characterized by a loss of systematic proteins, an hypercatabolic status and malnutrition play a role in both muscle mass and strength reduction, vol.13

, Recently, loss of muscle strength which is not related 6. REFERENCES

D. A. Goodkin, « Association of comorbid conditions and mortality in hemodialysis patients in Europe, Japan, and the United States: the Dialysis Outcomes and Practice Patterns Study (DOPPS), J. Am. Soc. Nephrol. JASN, vol.14, pp.3270-3277, 2003.

K. Kalantar-zadeh, T. A. Ikizler, G. Block, M. M. Avram, and J. D. Kopple, « Malnutrition-inflammation complex syndrome in dialysis patients: causes and consequences, Am. J. Kidney Dis. Off. J. Natl. Kidney Found, vol.42, issue.5, pp.864-881, 2003.

R. Chowdhury, N. M. Peel, M. Krosch, and R. E. Hubbard, « Frailty and chronic kidney disease: A systematic review, Arch. Gerontol. Geriatr, vol.68, pp.135-142, 2017.

J. C. Kim, K. Kalantar-zadeh, and J. D. Kopple, « Frailty and protein-energy wasting in elderly patients with end stage kidney disease, J. Am. Soc. Nephrol. JASN, vol.24, pp.337-351, 2013.

P. Lang, J. Michel, and D. Zekry, « Frailty syndrome: a transitional state in a dynamic process, Gerontology, vol.55, pp.539-549, 2009.

E. F. Binder, « Effects of exercise training on frailty in community-dwelling older adults: results of a randomized, J. Am. Geriatr. Soc, vol.50, pp.1921-1928, 2002.

L. P. Fried, « Frailty in older adults: evidence for a phenotype, J. Gerontol. A. Biol. Sci. Med. Sci, vol.56, pp.146-156, 2001.

Y. N. Hall, « Effects of six versus three times per week hemodialysis on physical performance, health, and functioning: Frequent Hemodialysis Network (FHN) randomized trials », Clin. J. Am. Soc. Nephrol. CJASN, vol.7, issue.5, pp.782-794, 2012.

P. Painter, L. Carlson, S. Carey, S. M. Paul, and J. Myll, « Physical functioning and health-related quality-of-life changes with exercise training in hemodialysis patients, Am. J. Kidney Dis. Off. J. Natl. Kidney Found, vol.35, pp.482-492, 2000.

J. M. Guralnik, « A short physical performance battery assessing lower extremity function: association with self-reported disability and prediction of mortality and nursing home admission, J. Gerontol, vol.49, issue.2, pp.85-94, 1994.

J. J. Carrero, « Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM), J. Ren. Nutr. Off. J. Counc. Ren. Nutr. Natl. Kidney Found, vol.23, issue.2, pp.77-90, 2013.

J. Souweine, « Physical inactivity and protein energy wasting play independent roles in muscle weakness in maintenance haemodialysis patients, PloS One, vol.13, p.200061, 2018.

N. Isoyama, « Comparative associations of muscle mass and muscle strength with mortality in dialysis patients, Clin. J. Am. Soc. Nephrol. CJASN, vol.9, pp.1720-1728, 2014.

D. Marcelli, « Body composition and survival in dialysis patients: results from an international cohort study », Clin. J. Am. Soc. Nephrol. CJASN, vol.10, pp.1192-1200, 2015.

B. Canaud, « Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS One, vol.9, p.93286, 2014.

A. J. Cruz-jentoft, Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People, vol.39, pp.412-423, 2010.

J. Souweine, « Standardized Method to Measure Muscle Force at the Bedside in Hemodialysis Patients, J. Ren. Nutr. Off. J. Counc. Ren. Nutr. Natl. Kidney Found, vol.27, pp.194-200, 2017.

N. Terrier, « Creatinine index and transthyretin as additive predictors of mortality in haemodialysis patients, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. -Eur. Ren. Assoc, vol.23, issue.1, pp.345-353, 2008.

D. Fouque, « A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease, Kidney Int, vol.73, pp.391-398, 2008.

Y. Obi, H. Qader, C. P. Kovesdy, and K. Kalantar-zadeh, Latest consensus and update on protein-energy wasting in chronic kidney disease, vol.18, pp.254-262, 2015.

M. J. Lee, « Irisin, a novel myokine is an independent predictor for sarcopenia and carotid atherosclerosis in dialysis patients, Atherosclerosis, vol.242, issue.2, pp.476-482, 2015.

D. Di-raimondo, A. Tuttolomondo, G. Musiari, C. Schimmenti, A. Angelo et al., « Are the Myokines the Mediators of Physical Activity-Induced Health Benefits? », Curr. Pharm. Des, vol.22, pp.3622-3647, 2016.

P. Esposito, Retombées / perspectives : L'impact de ce travail sera multiple : -Individuel : Améliorer la prise en charge des patients fragiles à risque de chute dans un centre spécialisé. -Global : Permettre de connaitre la prévalence des patients à risque de chute en hémodialyse -Médico-économique, Modulation of Myostatin/Hepatocyte Growth Factor Balance by Different Hemodialysis Modalities, vol.2017, p.7635459, 2017.

, Mots clefs : hémodialyse chronique, dysfonction musculaire, fragilité, sarcopénie EXPOSE DU PROJET DE RECHERCHE

, L'ensemble de ces processus participe à l'apparition d'un risque de chute qui reste mal connu chez les patients en hémodialyse chronique. Pourtant, les chutes répétées sont associées à une forte morbimortalité accélérant le processus de perte d'indépendance et d'autonomie et à un taux d'institutionnalisation élevé. Chez les patients en hémodialyse, la fragilité pourrait être liée à la dénutrition protéino-énergétique (Protein Energy Wasting) et à la faiblesse musculaire (diminution de la fonction musculaire). De plus les troubles minéraux et osseux des patients insuffisants rénaux chroniques augmentent le risque de complication des chutes répétées, La fragilité est un concept décrit initialement en gériatrie et mis en évidence récemment chez les patients en hémodialyse qui associe perte de poids, fatigue, lenteur de marche, faiblesse musculaire et faible activité physique (critères de Fried)

, propose un programme qui a pour but d'améliorer l'autonomie et ainsi réduire l'incidence des chutes des personnes âgées. Le short physical performance battery (SPPB), outil validé chez les patients hémodialysés chroniques

, Une intervention personnalisée des patients hémodialysés chroniques dans le centre Equilibre et Prévention de la Chute du CHU de Montpellier pourrait permettre une diminution du risque de chute et ainsi réduire la morbi-mortalité chez ces patients. Les patients à risque de chute pourront bénéficier d'une prise en charge globale et spécifique après une évaluation gérontologique standardisée menée dans le Centre Equilibre et Prévention de la Chute du CHU de Montpellier. Ni l'évaluation du risque de chute chez le dialysé, Le résultat de ce test est corrélé au risque d'institutionnalisation, d'hospitalisation, et de décès chez les personnes âgées

, Or les muscles squelettiques possèdent une activité autocrine et paracrine de signalisation, via la sécrétion de myokines. Ces dernières contrôlent l'homéostasie et l'intégrité musculaire mais aussi celles de différents organes (coeur, os, pancréas, système nerveux, ?)

, jugement principal : L'amélioration de l'autonomie fonctionnelle mesurée par le résultat du SPPB, 6 mois après l'évaluation initiale et la prise en charge personnalisée par le centre Equilibre et Prévention de la Chute

, Critères de jugements secondaires : Corrélation entre le taux de myokines et le SPPB à l'inclusion et après réhabilitation musculaire. Evaluation de l'aide au support nutritionnel « ReinBow ». Il sera évalué sur le nombre de patients consultants cet outil innovant, le nombre de consultations par semaine et la relation éventuelle entre consultation de l'application mobile et l'amélioration des indices nutritionnels

, Population : Patients majeurs hémodialysés chroniques à risque de chute

, Durée du projet : 12 mois

, Calendrier des évaluations : ? Période d'inclusion : 6 mois

M. Dam, F. Neelemaat, T. Struijk-wielinga, P. J. Weijs, and B. C. Van-jaarsveld, « Physical performance and protein-energy wasting in patients treated with nocturnal haemodialysis compared to conventional haemodialysis: protocol of the DiapriFIT study, BMC Nephrol, vol.18, issue.1, p.144, 2017.

K. Kalantar-zadeh, T. A. Ikizler, G. Block, M. M. Avram, and J. D. Kopple, « Malnutritioninflammation complex syndrome in dialysis patients: causes and consequences, Am. J. Kidney Dis. Off. J. Natl. Kidney Found, vol.42, issue.5, pp.864-881, 2003.

J. C. Kim, K. Kalantar-zadeh, and J. D. Kopple, « Frailty and protein-energy wasting in elderly patients with end stage kidney disease, J. Am. Soc. Nephrol. JASN, vol.24, pp.337-351, 2013.

R. Chowdhury, N. M. Peel, M. Krosch, and R. E. Hubbard, « Frailty and chronic kidney disease: A systematic review, Arch. Gerontol. Geriatr, vol.68, pp.135-142, 2017.

J. J. Carrero, « Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism

». and J. Ren, Nutr. Off. J. Counc. Ren. Nutr. Natl. Kidney Found, vol.23, issue.2, pp.77-90, 2013.

M. Panaye, « Phenotypes influencing low physical activity in maintenance dialysis, J. Ren. Nutr. Off. J. Counc. Ren. Nutr. Natl. Kidney Found, vol.25, issue.1, pp.31-39

S. Bataille, M. Serveaux, E. Carreno, N. Pedinielli, P. Darmon et al., « The diagnosis of sarcopenia is mainly driven by muscle mass in hemodialysis patients, Clin. Nutr. Edinb. Scotl, vol.36, pp.1654-1660, 2017.

N. Isoyama, « Comparative associations of muscle mass and muscle strength with mortality in dialysis patients, Clin. J. Am. Soc. Nephrol. CJASN, vol.9, pp.1720-1728, 2014.

P. Molina, « Vitamin D, a modulator of musculoskeletal health in chronic kidney disease, J. Cachexia Sarcopenia Muscle, vol.8, pp.686-701, 2017.

N. Boudville, C. Inderjeeth, G. J. Elder, and P. Glendenning, « Association between 25-hydroxyvitamin D, somatic muscle weakness and falls risk in end-stage renal failure, Clin. Endocrinol. (Oxf.), vol.73, pp.299-304, 2010.

H. Blain, « Effectiveness of a programme delivered in a falls clinic in preventing serious injuries in high-risk older adults: A pre-and post-intervention study, Maturitas, vol.122, pp.80-86, 2019.

Y. N. Hall, « Effects of six versus three times per week hemodialysis on physical performance, health, and functioning: Frequent Hemodialysis Network (FHN) randomized trials », Clin. J. Am. Soc. Nephrol. CJASN, vol.7, issue.5, pp.782-794, 2012.

P. Painter, L. Carlson, S. Carey, S. M. Paul, and J. Myll, « Physical functioning and healthrelated quality-of-life changes with exercise training in hemodialysis patients, Am. J. Kidney Dis. Off. J. Natl. Kidney Found, vol.35, pp.482-492, 2000.

K. Nagai, « Physical activity combined with resistance training reduces symptoms of frailty in older adults: A randomized controlled trial, Arch. Gerontol. Geriatr, vol.76, pp.41-47, 2018.

D. Di-raimondo, A. Tuttolomondo, G. Musiari, C. Schimmenti, A. Angelo et al., « Are the Myokines the Mediators of Physical Activity-Induced Health Benefits? », Curr. Pharm. Des, vol.22, pp.3622-3647, 2016.

M. J. Lee, « Irisin, a novel myokine is an independent predictor for sarcopenia and carotid atherosclerosis in dialysis patients, Atherosclerosis, vol.242, issue.2, pp.476-482, 2015.

B. K. Pedersen and M. A. Febbraio, « Muscles, exercise and obesity: skeletal muscle as a secretory organ, Nat. Rev. Endocrinol, vol.8, pp.457-465, 2012.

H. Peng, « Myokine mediated muscle-kidney crosstalk suppresses metabolic reprogramming and fibrosis in damaged kidneys, Nat. Commun, vol.8, issue.1, p.2017

B. K. Pedersen, « Physical activity and muscle-brain crosstalk », Nat. Rev. Endocrinol, vol.2019

B. Grygiel-górniak and M. Puszczewicz, « A review on irisin, a new protagonist that mediates muscle-adipose-bone-neuron connectivity », Eur. Rev. Med. Pharmacol. Sci, vol.21, pp.4687-4693, 2017.

P. Esposito, « Modulation of Myostatin/Hepatocyte Growth Factor Balance by Different Hemodialysis Modalities, BioMed Res. Int, vol.2017, p.7635459, 2017.

J. Souweine, « Standardized Method to Measure Muscle Force at the Bedside in Hemodialysis Patients, J. Ren. Nutr. Off. J. Counc. Ren. Nutr. Natl. Kidney Found, vol.27, pp.194-200, 2017.

J. Souweine, « Physical inactivity and protein energy wasting play independent roles in muscle weakness in maintenance haemodialysis patients, PloS One, vol.13, p.200061, 2018.

J. M. Guralnik, « A short physical performance battery assessing lower extremity function: association with self-reported disability and prediction of mortality and nursing home admission, J. Gerontol, vol.49, issue.2, pp.85-94, 1994.

A. Sawant and T. Overend, The Impact of Hemodialysis on Spatio-Temporal Characteristics of Gait and Role of Exercise: A Systematic Review, vol.5, 2017.

D. Marcelli, « Body composition and survival in dialysis patients: results from an international cohort study », Clin. J. Am. Soc. Nephrol. CJASN, vol.10, pp.1192-1200, 2015.

B. Canaud, « Creatinine index as a surrogate of lean body mass derived from urea Kt/V, pre-dialysis serum levels and anthropometric characteristics of haemodialysis patients, PloS One, vol.9, p.93286, 2014.

A. J. Cruz-jentoft, Sarcopenia: revised European consensus on definition and diagnosis, vol.48, pp.16-31, 2019.

B. C. Clark and T. M. Manini, What is dynapenia? », Nutr. Burbank Los Angel. Cty. Calif, vol.28, pp.495-503, 2012.

L. E. Voorrips, A. C. Ravelli, P. C. Dongelmans, P. Deurenberg, and W. , Van Staveren, « A physical activity questionnaire for the elderly, Med. Sci. Sports Exerc, vol.23, pp.974-979, 1991.

D. Fouque, « A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease, Kidney Int, vol.73, pp.391-398, 2008.

L. Posters, Poster: Muscle mass is predictive of mortality in obese renal transplant patients 2016: European muscle congress, Poster: Muscle force measurement in haemodialysis 2016: ERA-EDTA, Poster: Determinants of muscle strength in haemodialysis 2016 : SFNDT, Communication orale : Dysfonctions musculaire en hémodialyse chronique : quels en sont les déterminants ? PUBLICATIONS SCIENTIFIQUES, 2017.

J. S. Souweine, N. Kuster, L. Chenine, A. Rodriguez, L. Patrier et al., Physical inactivity and protein energy wasting play independent roles in muscleweakness in maintenance haemodialysis patients, PLoS One, vol.13, issue.8, p.200061, 2018.

J. S. Souweine, A. Boudet, L. Chenine, H. Leray, A. Rodriguez et al., Standardized Method to Measure Muscle Force at the Bedside in hemodialysis patients, J Ren Nutr, vol.27, pp.194-200, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01774962