Skip to main content
Top
Published in: Endocrine 1/2013

01-02-2013 | Review

Atrogin-1, MuRF-1, and sarcopenia

Authors: Jonathan P. Gumucio, Christopher L. Mendias

Published in: Endocrine | Issue 1/2013

Login to get access

Abstract

Sarcopenia is one of the leading causes of disability in the elderly. Despite the growing prevalence of sarcopenia, the molecular mechanisms that control aging-related changes in muscle mass are not fully understood. The ubiquitin proteasome system is one of the major pathways that regulate muscle protein degradation, and this system plays a central role in controlling muscle size. Atrogin-1 and MuRF-1 are two E3 ubiquitin ligases that are important regulators of ubiquitin-mediated protein degradation in skeletal muscle. In this review, we will discuss: (i) aging-related changes to skeletal muscle structure and function; (ii) the regulation of protein synthesis and protein degradation by IGF-1, TGF-β, and myostatin, with emphasis on the control of atrogin-1 and MuRF-1 expression; and (iii) the potential for modulating atrogin-1 and MuRF-1 expression to treat or prevent sarcopenia.
Literature
1.
go back to reference I.H. Rosenberg, Summary comments. Am. J. Clin. Nutr. 50(5), 1231–1233 (1989) I.H. Rosenberg, Summary comments. Am. J. Clin. Nutr. 50(5), 1231–1233 (1989)
2.
go back to reference R.A. Fielding, B. Vellas, W.J. Evans, S. Bhasin, J.E. Morley, A.B. Newman, G. Abellan van Kan, S. Andrieu, J. Bauer, D. Breuille, T. Cederholm, J. Chandler, C. De Meynard, L. Donini, T. Harris, A. Kannt, F. Keime Guibert, G. Onder, D. Papanicolaou, Y. Rolland, D. Rooks, C. Sieber, E. Souhami, S. Verlaan, M. Zamboni, Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia. J. Am. Med. Dir. Assoc. 12(4), 249–256 (2011). doi:10.1016/j.jamda.2011.01.003 PubMedCrossRef R.A. Fielding, B. Vellas, W.J. Evans, S. Bhasin, J.E. Morley, A.B. Newman, G. Abellan van Kan, S. Andrieu, J. Bauer, D. Breuille, T. Cederholm, J. Chandler, C. De Meynard, L. Donini, T. Harris, A. Kannt, F. Keime Guibert, G. Onder, D. Papanicolaou, Y. Rolland, D. Rooks, C. Sieber, E. Souhami, S. Verlaan, M. Zamboni, Sarcopenia: an undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia. J. Am. Med. Dir. Assoc. 12(4), 249–256 (2011). doi:10.​1016/​j.​jamda.​2011.​01.​003 PubMedCrossRef
4.
go back to reference C.J. Evans, C.-F. Chiou, K.A. Fitzgerald, W.J. Evans, B.R. Ferrell, W. Dale, L.P. Fried, S.R. Gandra, B. Dennee-Sommers, D.L. Patrick, Development of a new patient-reported outcome measure in sarcopenia. J. Am. Med. Dir. Assoc. 12(3), 226–233 (2011). doi:10.1016/j.jamda.2010.09.010 PubMedCrossRef C.J. Evans, C.-F. Chiou, K.A. Fitzgerald, W.J. Evans, B.R. Ferrell, W. Dale, L.P. Fried, S.R. Gandra, B. Dennee-Sommers, D.L. Patrick, Development of a new patient-reported outcome measure in sarcopenia. J. Am. Med. Dir. Assoc. 12(3), 226–233 (2011). doi:10.​1016/​j.​jamda.​2010.​09.​010 PubMedCrossRef
5.
go back to reference I. Janssen, D.S. Shepard, P.T. Katzmarzyk, R. Roubenoff, The healthcare costs of sarcopenia in the United States. J. Am. Geriatr. Soc. 52(1), 80–85 (2004)PubMedCrossRef I. Janssen, D.S. Shepard, P.T. Katzmarzyk, R. Roubenoff, The healthcare costs of sarcopenia in the United States. J. Am. Geriatr. Soc. 52(1), 80–85 (2004)PubMedCrossRef
6.
8.
go back to reference I. Janssen, S.B. Heymsfield, Z.M. Wang, R. Ross, Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J. Appl. Physiol. 89(1), 81–88 (2000)PubMed I. Janssen, S.B. Heymsfield, Z.M. Wang, R. Ross, Skeletal muscle mass and distribution in 468 men and women aged 18–88 yr. J. Appl. Physiol. 89(1), 81–88 (2000)PubMed
9.
go back to reference J. Lexell, Human aging, muscle mass, and fiber type composition. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 50, 11–16 (1995) J. Lexell, Human aging, muscle mass, and fiber type composition. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 50, 11–16 (1995)
12.
go back to reference D.R. Claflin, L.M. Larkin, P.S. Cederna, J.F. Horowitz, N.B. Alexander, N.M. Cole, A.T. Galecki, S. Chen, L.V. Nyquist, B.M. Carlson, J.A. Faulkner, J.A. Ashton-Miller, Effects of high- and low-velocity resistance training on the contractile properties of skeletal muscle fibers from young and older humans. J. Appl. Physiol. 111(4), 1021–1030 (2011). doi:10.1152/japplphysiol.01119.2010 PubMedCrossRef D.R. Claflin, L.M. Larkin, P.S. Cederna, J.F. Horowitz, N.B. Alexander, N.M. Cole, A.T. Galecki, S. Chen, L.V. Nyquist, B.M. Carlson, J.A. Faulkner, J.A. Ashton-Miller, Effects of high- and low-velocity resistance training on the contractile properties of skeletal muscle fibers from young and older humans. J. Appl. Physiol. 111(4), 1021–1030 (2011). doi:10.​1152/​japplphysiol.​01119.​2010 PubMedCrossRef
13.
go back to reference J. Lexell, C.C. Taylor, M. Sjostrom, What is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15- to 83-year-old men. J. Neurol. Sci. 84(2–3), 275–294 (1988)PubMedCrossRef J. Lexell, C.C. Taylor, M. Sjostrom, What is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15- to 83-year-old men. J. Neurol. Sci. 84(2–3), 275–294 (1988)PubMedCrossRef
15.
go back to reference T.J. Hawke, D.J. Garry, Myogenic satellite cells: physiology to molecular biology. J. Appl. Physiol. 91(2), 534–551 (2001)PubMed T.J. Hawke, D.J. Garry, Myogenic satellite cells: physiology to molecular biology. J. Appl. Physiol. 91(2), 534–551 (2001)PubMed
16.
go back to reference I.M. Conboy, T.A. Rando, Aging, stem cells and tissue regeneration: lessons from muscle. Cell Cycle 4(3), 407–410 (2005)PubMedCrossRef I.M. Conboy, T.A. Rando, Aging, stem cells and tissue regeneration: lessons from muscle. Cell Cycle 4(3), 407–410 (2005)PubMedCrossRef
18.
go back to reference B.M. Carlson, J.A. Faulkner, Muscle transplantation between young and old rats: age of host determines recovery. Am. J. Physiol. 256(6 Pt 1), C1262–C1266 (1989)PubMed B.M. Carlson, J.A. Faulkner, Muscle transplantation between young and old rats: age of host determines recovery. Am. J. Physiol. 256(6 Pt 1), C1262–C1266 (1989)PubMed
19.
go back to reference I.M. Conboy, M.J. Conboy, A.J. Wagers, E.R. Girma, I.L. Weissman, T.A. Rando, Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433(7027), 760–764 (2005). doi:10.1038/nature03260 PubMedCrossRef I.M. Conboy, M.J. Conboy, A.J. Wagers, E.R. Girma, I.L. Weissman, T.A. Rando, Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433(7027), 760–764 (2005). doi:10.​1038/​nature03260 PubMedCrossRef
20.
go back to reference V. Mouly, A. Aamiri, A. Bigot, R.N. Cooper, S. Di Donna, D. Furling, T. Gidaro, V. Jacquemin, K. Mamchaoui, E. Negroni, S. Périé, V. Renault, S.D. Silva-Barbosa, G.S. Butler-Browne, The mitotic clock in skeletal muscle regeneration, disease and cell mediated gene therapy. Acta Physiol. Scand. 184(1), 3–15 (2005). doi:10.1111/j.1365-201X.2005.01417.x PubMedCrossRef V. Mouly, A. Aamiri, A. Bigot, R.N. Cooper, S. Di Donna, D. Furling, T. Gidaro, V. Jacquemin, K. Mamchaoui, E. Negroni, S. Périé, V. Renault, S.D. Silva-Barbosa, G.S. Butler-Browne, The mitotic clock in skeletal muscle regeneration, disease and cell mediated gene therapy. Acta Physiol. Scand. 184(1), 3–15 (2005). doi:10.​1111/​j.​1365-201X.​2005.​01417.​x PubMedCrossRef
21.
go back to reference R.M. Reznick, H. Zong, J. Li, K. Morino, I.K. Moore, H.J. Yu, Z.X. Liu, J. Dong, K.J. Mustard, S.A. Hawley, D. Befroy, M. Pypaert, D.G. Hardie, L.H. Young, G.I. Shulman, Aging-associated reductions in AMP-activated protein kinase activity and mitochondrial biogenesis. Cell Metab. 5(2), 151–156 (2007). doi:10.1016/j.cmet.2007.01.008 PubMedCrossRef R.M. Reznick, H. Zong, J. Li, K. Morino, I.K. Moore, H.J. Yu, Z.X. Liu, J. Dong, K.J. Mustard, S.A. Hawley, D. Befroy, M. Pypaert, D.G. Hardie, L.H. Young, G.I. Shulman, Aging-associated reductions in AMP-activated protein kinase activity and mitochondrial biogenesis. Cell Metab. 5(2), 151–156 (2007). doi:10.​1016/​j.​cmet.​2007.​01.​008 PubMedCrossRef
22.
go back to reference U.T. Brunk, A. Terman, The mitochondrial-lysosomal axis theory of aging: accumulation of damaged mitochondria as a result of imperfect autophagocytosis. Eur. J. Biochem. 269(8), 1996–2002 (2002)PubMedCrossRef U.T. Brunk, A. Terman, The mitochondrial-lysosomal axis theory of aging: accumulation of damaged mitochondria as a result of imperfect autophagocytosis. Eur. J. Biochem. 269(8), 1996–2002 (2002)PubMedCrossRef
24.
go back to reference E. Marzetti, J.C.Y. Hwang, H.A. Lees, S.E. Wohlgemuth, E.E. Dupont-Versteegden, C.S. Carter, R. Bernabei, C. Leeuwenburgh, Mitochondrial death effectors: relevance to sarcopenia and disuse muscle atrophy. Biochim. Biophys. Acta 1800(3), 235–244 (2010). doi:10.1016/j.bbagen.2009.05.007 PubMedCrossRef E. Marzetti, J.C.Y. Hwang, H.A. Lees, S.E. Wohlgemuth, E.E. Dupont-Versteegden, C.S. Carter, R. Bernabei, C. Leeuwenburgh, Mitochondrial death effectors: relevance to sarcopenia and disuse muscle atrophy. Biochim. Biophys. Acta 1800(3), 235–244 (2010). doi:10.​1016/​j.​bbagen.​2009.​05.​007 PubMedCrossRef
25.
go back to reference A. Musaro, K. McCullagh, A. Paul, L. Houghton, G. Dobrowolny, M. Molinaro, E.R. Barton, H.L. Sweeney, N. Rosenthal, Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat. Genet. 27(2), 195–200 (2001). doi:10.1038/84839 PubMedCrossRef A. Musaro, K. McCullagh, A. Paul, L. Houghton, G. Dobrowolny, M. Molinaro, E.R. Barton, H.L. Sweeney, N. Rosenthal, Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat. Genet. 27(2), 195–200 (2001). doi:10.​1038/​84839 PubMedCrossRef
26.
go back to reference M.D. Mavalli, D.J. DiGirolamo, Y. Fan, R.C. Riddle, K.S. Campbell, T. van Groen, S.J. Frank, M.A. Sperling, K.A. Esser, M.M. Bamman, T.L. Clemens, Distinct growth hormone receptor signaling modes regulate skeletal muscle development and insulin sensitivity in mice. J. Clin. Investig. 120(11), 4007–4020 (2010). doi:10.1172/JCI42447 PubMedCrossRef M.D. Mavalli, D.J. DiGirolamo, Y. Fan, R.C. Riddle, K.S. Campbell, T. van Groen, S.J. Frank, M.A. Sperling, K.A. Esser, M.M. Bamman, T.L. Clemens, Distinct growth hormone receptor signaling modes regulate skeletal muscle development and insulin sensitivity in mice. J. Clin. Investig. 120(11), 4007–4020 (2010). doi:10.​1172/​JCI42447 PubMedCrossRef
27.
go back to reference Mozzetti Marone, D. Ritis, Scambia Pierelli, Semiquantitative RT-PCR analysis to assess the expression levels of multiple transcripts from the same sample. Biol Proced Online 3, 19–25 (2001). doi:10.1251/bpo20 PubMedCrossRef Mozzetti Marone, D. Ritis, Scambia Pierelli, Semiquantitative RT-PCR analysis to assess the expression levels of multiple transcripts from the same sample. Biol Proced Online 3, 19–25 (2001). doi:10.​1251/​bpo20 PubMedCrossRef
30.
go back to reference M. Andjelkovic, D.R. Alessi, R. Meier, A. Fernandez, N.J. Lamb, M. Frech, P. Cron, P. Cohen, J.M. Lucocq, B.A. Hemmings, Role of translocation in the activation and function of protein kinase B. J. Biol. Chem. 272(50), 31515–31524 (1997)PubMedCrossRef M. Andjelkovic, D.R. Alessi, R. Meier, A. Fernandez, N.J. Lamb, M. Frech, P. Cron, P. Cohen, J.M. Lucocq, B.A. Hemmings, Role of translocation in the activation and function of protein kinase B. J. Biol. Chem. 272(50), 31515–31524 (1997)PubMedCrossRef
33.
go back to reference R.W. Matheny, B.C. Nindl, M.L. Adamo, Minireview: mechano-growth factor: a putative product of IGF-I gene expression involved in tissue repair and regeneration. Endocrinology 151(3), 865–875 (2010). doi:10.1210/en.2009-1217 PubMedCrossRef R.W. Matheny, B.C. Nindl, M.L. Adamo, Minireview: mechano-growth factor: a putative product of IGF-I gene expression involved in tissue repair and regeneration. Endocrinology 151(3), 865–875 (2010). doi:10.​1210/​en.​2009-1217 PubMedCrossRef
34.
go back to reference M. Vinciguerra, A. Musarò, N. Rosenthal, Regulation of muscle atrophy in aging and disease. Adv. Exp. Med. Biol. 694, 211–233 (2010)PubMedCrossRef M. Vinciguerra, A. Musarò, N. Rosenthal, Regulation of muscle atrophy in aging and disease. Adv. Exp. Med. Biol. 694, 211–233 (2010)PubMedCrossRef
35.
go back to reference S.C. Bodine, E. Latres, S. Baumhueter, V.K. Lai, L. Nunez, B.A. Clarke, W.T. Poueymirou, F.J. Panaro, E. Na, K. Dharmarajan, Z.Q. Pan, D.M. Valenzuela, T.M. DeChiara, T.N. Stitt, G.D. Yancopoulos, D.J. Glass, Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294(5547), 1704–1708 (2001). doi:10.1126/science.1065874 PubMedCrossRef S.C. Bodine, E. Latres, S. Baumhueter, V.K. Lai, L. Nunez, B.A. Clarke, W.T. Poueymirou, F.J. Panaro, E. Na, K. Dharmarajan, Z.Q. Pan, D.M. Valenzuela, T.M. DeChiara, T.N. Stitt, G.D. Yancopoulos, D.J. Glass, Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294(5547), 1704–1708 (2001). doi:10.​1126/​science.​1065874 PubMedCrossRef
36.
go back to reference C. Rommel, S.C. Bodine, B.A. Clarke, R. Rossman, L. Nunez, T.N. Stitt, G.D. Yancopoulos, D.J. Glass, Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat. Cell Biol. 3(11), 1009–1013 (2001). doi:10.1038/ncb1101-1009 PubMedCrossRef C. Rommel, S.C. Bodine, B.A. Clarke, R. Rossman, L. Nunez, T.N. Stitt, G.D. Yancopoulos, D.J. Glass, Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat. Cell Biol. 3(11), 1009–1013 (2001). doi:10.​1038/​ncb1101-1009 PubMedCrossRef
38.
go back to reference Y.-H. Song, M. Godard, Y. Li, S.R. Richmond, N. Rosenthal, P. Delafontaine, Insulin-like growth factor I-mediated skeletal muscle hypertrophy is characterized by increased mTOR-p70S6K signaling without increased Akt phosphorylation. J. Investig. Med. 53(3), 135–142 (2005)PubMedCrossRef Y.-H. Song, M. Godard, Y. Li, S.R. Richmond, N. Rosenthal, P. Delafontaine, Insulin-like growth factor I-mediated skeletal muscle hypertrophy is characterized by increased mTOR-p70S6K signaling without increased Akt phosphorylation. J. Investig. Med. 53(3), 135–142 (2005)PubMedCrossRef
40.
41.
go back to reference B. Raught, F. Peiretti, A.C. Gingras, M. Livingstone, D. Shahbazian, G.L. Mayeur, R.D. Polakiewicz, N. Sonenberg, J.W. Hershey, Phosphorylation of eucaryotic translation initiation factor 4B Ser422 is modulated by S6 kinases. EMBO J. 23(8), 1761–1769 (2004). doi:10.1038/sj.emboj.7600193 PubMedCrossRef B. Raught, F. Peiretti, A.C. Gingras, M. Livingstone, D. Shahbazian, G.L. Mayeur, R.D. Polakiewicz, N. Sonenberg, J.W. Hershey, Phosphorylation of eucaryotic translation initiation factor 4B Ser422 is modulated by S6 kinases. EMBO J. 23(8), 1761–1769 (2004). doi:10.​1038/​sj.​emboj.​7600193 PubMedCrossRef
42.
43.
go back to reference K. Hara, K. Yonezawa, M.T. Kozlowski, T. Sugimoto, K. Andrabi, Q.P. Weng, M. Kasuga, I. Nishimoto, J. Avruch, Regulation of eIF-4E BP1 phosphorylation by mTOR. J. Biol. Chem. 272(42), 26457–26463 (1997)PubMedCrossRef K. Hara, K. Yonezawa, M.T. Kozlowski, T. Sugimoto, K. Andrabi, Q.P. Weng, M. Kasuga, I. Nishimoto, J. Avruch, Regulation of eIF-4E BP1 phosphorylation by mTOR. J. Biol. Chem. 272(42), 26457–26463 (1997)PubMedCrossRef
44.
go back to reference Y. Izumiya, T. Hopkins, C. Morris, K. Sato, L. Zeng, J. Viereck, J.A. Hamilton, N. Ouchi, N.K. LeBrasseur, K. Walsh, Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metab. 7(2), 159–172 (2008). doi:10.1016/j.cmet.2007.11.003 PubMedCrossRef Y. Izumiya, T. Hopkins, C. Morris, K. Sato, L. Zeng, J. Viereck, J.A. Hamilton, N. Ouchi, N.K. LeBrasseur, K. Walsh, Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metab. 7(2), 159–172 (2008). doi:10.​1016/​j.​cmet.​2007.​11.​003 PubMedCrossRef
45.
go back to reference G. Pallafacchina, E. Calabria, A.L. Serrano, J.M. Kalhovde, S. Schiaffino, A protein kinase B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type specification. Proc. Natl. Acad. Sci. USA 99(14), 9213–9218 (2002). doi:10.1073/pnas.142166599 PubMedCrossRef G. Pallafacchina, E. Calabria, A.L. Serrano, J.M. Kalhovde, S. Schiaffino, A protein kinase B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type specification. Proc. Natl. Acad. Sci. USA 99(14), 9213–9218 (2002). doi:10.​1073/​pnas.​142166599 PubMedCrossRef
47.
go back to reference E. Marzetti, G. Privitera, V. Simili, S.E. Wohlgemuth, L. Aulisa, M. Pahor, C. Leeuwenburgh, Multiple pathways to the same end: mechanisms of myonuclear apoptosis in sarcopenia of aging. Scientific World J. 10, 340–349 (2010). doi:10.1100/tsw.2010.27 CrossRef E. Marzetti, G. Privitera, V. Simili, S.E. Wohlgemuth, L. Aulisa, M. Pahor, C. Leeuwenburgh, Multiple pathways to the same end: mechanisms of myonuclear apoptosis in sarcopenia of aging. Scientific World J. 10, 340–349 (2010). doi:10.​1100/​tsw.​2010.​27 CrossRef
48.
go back to reference T.W. Buford, S.D. Anton, A.R. Judge, E. Marzetti, S.E. Wohlgemuth, C.S. Carter, C. Leeuwenburgh, M. Pahor, T.M. Manini, Models of accelerated sarcopenia: critical pieces for solving the puzzle of age-related muscle atrophy. Ageing Res. Rev. 9(4), 369–383 (2010). doi:10.1016/j.arr.2010.04.004 PubMedCrossRef T.W. Buford, S.D. Anton, A.R. Judge, E. Marzetti, S.E. Wohlgemuth, C.S. Carter, C. Leeuwenburgh, M. Pahor, T.M. Manini, Models of accelerated sarcopenia: critical pieces for solving the puzzle of age-related muscle atrophy. Ageing Res. Rev. 9(4), 369–383 (2010). doi:10.​1016/​j.​arr.​2010.​04.​004 PubMedCrossRef
52.
go back to reference T.A. Zimmers, M.V. Davies, L.G. Koniaris, P. Haynes, A.F. Esquela, K.N. Tomkinson, A.C. McPherron, N.M. Wolfman, S.-J. Lee, Induction of cachexia in mice by systemically administered myostatin. Science 296(5572), 1486–1488 (2002). doi:10.1126/science.1069525 PubMedCrossRef T.A. Zimmers, M.V. Davies, L.G. Koniaris, P. Haynes, A.F. Esquela, K.N. Tomkinson, A.C. McPherron, N.M. Wolfman, S.-J. Lee, Induction of cachexia in mice by systemically administered myostatin. Science 296(5572), 1486–1488 (2002). doi:10.​1126/​science.​1069525 PubMedCrossRef
53.
go back to reference C.L. Mendias, J.P. Gumucio, M.E. Davis, C.W. Bromley, C.S. Davis, S.V. Brooks, Transforming growth factor-beta induces skeletal muscle atrophy and fibrosis through the induction of atrogin-1 and scleraxis. Muscle Nerve 45(1), 55–59 (2012). doi:10.1002/mus.22232 PubMedCrossRef C.L. Mendias, J.P. Gumucio, M.E. Davis, C.W. Bromley, C.S. Davis, S.V. Brooks, Transforming growth factor-beta induces skeletal muscle atrophy and fibrosis through the induction of atrogin-1 and scleraxis. Muscle Nerve 45(1), 55–59 (2012). doi:10.​1002/​mus.​22232 PubMedCrossRef
54.
go back to reference S.J. Lee, Y.S. Lee, T.A. Zimmers, A. Soleimani, M.M. Matzuk, K. Tsuchida, R.D. Cohn, E.R. Barton, Regulation of muscle mass by follistatin and activins. Mol. Endocrinol. 24(10), 1998–2008 (2010). doi:10.1210/me.2010-0127 PubMedCrossRef S.J. Lee, Y.S. Lee, T.A. Zimmers, A. Soleimani, M.M. Matzuk, K. Tsuchida, R.D. Cohn, E.R. Barton, Regulation of muscle mass by follistatin and activins. Mol. Endocrinol. 24(10), 1998–2008 (2010). doi:10.​1210/​me.​2010-0127 PubMedCrossRef
58.
go back to reference Y. Shi, J. Massagué, Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113(6), 685–700 (2003)PubMedCrossRef Y. Shi, J. Massagué, Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113(6), 685–700 (2003)PubMedCrossRef
63.
go back to reference J. Seoane, H.-V. Le, L. Shen, S.A. Anderson, J. Massagué, Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117(2), 211–223 (2004)PubMedCrossRef J. Seoane, H.-V. Le, L. Shen, S.A. Anderson, J. Massagué, Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117(2), 211–223 (2004)PubMedCrossRef
65.
go back to reference A. Amirouche, A.-C. Durieux, S. Banzet, N. Koulmann, R. Bonnefoy, C. Mouret, X. Bigard, A. Peinnequin, D. Freyssenet, Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. Endocrinology 150(1), 286–294 (2009). doi:10.1210/en.2008-0959 PubMedCrossRef A. Amirouche, A.-C. Durieux, S. Banzet, N. Koulmann, R. Bonnefoy, C. Mouret, X. Bigard, A. Peinnequin, D. Freyssenet, Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. Endocrinology 150(1), 286–294 (2009). doi:10.​1210/​en.​2008-0959 PubMedCrossRef
66.
go back to reference C. McFarlane, E. Plummer, M. Thomas, A. Hennebry, M. Ashby, N. Ling, H. Smith, M. Sharma, R. Kambadur, Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. J. Cell. Physiol. 209(2), 501–514 (2006). doi:10.1002/jcp.20757 PubMedCrossRef C. McFarlane, E. Plummer, M. Thomas, A. Hennebry, M. Ashby, N. Ling, H. Smith, M. Sharma, R. Kambadur, Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. J. Cell. Physiol. 209(2), 501–514 (2006). doi:10.​1002/​jcp.​20757 PubMedCrossRef
67.
go back to reference A.U. Trendelenburg, A. Meyer, D. Rohner, J. Boyle, S. Hatakeyama, D.J. Glass, Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am. J. Physiol. Cell Physiol. 296(6), C1258–C1270 (2009). doi:10.1152/ajpcell.00105.2009 PubMedCrossRef A.U. Trendelenburg, A. Meyer, D. Rohner, J. Boyle, S. Hatakeyama, D.J. Glass, Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am. J. Physiol. Cell Physiol. 296(6), C1258–C1270 (2009). doi:10.​1152/​ajpcell.​00105.​2009 PubMedCrossRef
68.
go back to reference W. Yang, Y. Zhang, Y. Li, Z. Wu, 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. J. Biol. Chem. 282(6), 3799–3808 (2007). doi:10.1074/jbc.M610185200 PubMedCrossRef W. Yang, Y. Zhang, Y. Li, Z. Wu, 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. J. Biol. Chem. 282(6), 3799–3808 (2007). doi:10.​1074/​jbc.​M610185200 PubMedCrossRef
69.
go back to reference M.D. Gomes, S.H. Lecker, R.T. Jagoe, A. Navon, A.L. Goldberg, Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc. Natl. Acad. Sci. USA 98(25), 14440–14445 (2001). doi:10.1073/pnas.251541198 PubMedCrossRef M.D. Gomes, S.H. Lecker, R.T. Jagoe, A. Navon, A.L. Goldberg, Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc. Natl. Acad. Sci. USA 98(25), 14440–14445 (2001). doi:10.​1073/​pnas.​251541198 PubMedCrossRef
70.
go back to reference S. Lokireddy, C. McFarlane, X. Ge, H. Zhang, S.K. Sze, M. Sharma, R. Kambadur, Myostatin induces degradation of sarcomeric proteins through a Smad3 signaling mechanism during skeletal muscle wasting. Mol. Endocrinol. 25(11), 1936–1949 (2011). doi:10.1210/me.2011-1124 PubMedCrossRef S. Lokireddy, C. McFarlane, X. Ge, H. Zhang, S.K. Sze, M. Sharma, R. Kambadur, Myostatin induces degradation of sarcomeric proteins through a Smad3 signaling mechanism during skeletal muscle wasting. Mol. Endocrinol. 25(11), 1936–1949 (2011). doi:10.​1210/​me.​2011-1124 PubMedCrossRef
72.
go back to reference C.L. Mendias, E. Kayupov, J.R. Bradley, S.V. Brooks, D.R. Claflin, Decreased specific force and power production of muscle fibers from myostatin-deficient mice are associated with a suppression of protein degradation. J. Appl. Physiol. 111(1), 185–191 (2011). doi:10.1152/japplphysiol.00126.2011 PubMedCrossRef C.L. Mendias, E. Kayupov, J.R. Bradley, S.V. Brooks, D.R. Claflin, Decreased specific force and power production of muscle fibers from myostatin-deficient mice are associated with a suppression of protein degradation. J. Appl. Physiol. 111(1), 185–191 (2011). doi:10.​1152/​japplphysiol.​00126.​2011 PubMedCrossRef
73.
go back to reference J.M. Sacheck, J.-P.K. Hyatt, A. Raffaello, R.T. Jagoe, R.R. Roy, V.R. Edgerton, S.H. Lecker, A.L. Goldberg, Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases. FASEB J. 21(1), 140–155 (2007). doi:10.1096/fj.06-6604com PubMedCrossRef J.M. Sacheck, J.-P.K. Hyatt, A. Raffaello, R.T. Jagoe, R.R. Roy, V.R. Edgerton, S.H. Lecker, A.L. Goldberg, Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases. FASEB J. 21(1), 140–155 (2007). doi:10.​1096/​fj.​06-6604com PubMedCrossRef
74.
go back to reference Y.-P. Li, Y. Chen, J. John, J. Moylan, B. Jin, D.L. Mann, M.B. Reid, TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle. FASEB J. 19(3), 362–370 (2005). doi:10.1096/fj.04-2364com PubMedCrossRef Y.-P. Li, Y. Chen, J. John, J. Moylan, B. Jin, D.L. Mann, M.B. Reid, TNF-alpha acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle. FASEB J. 19(3), 362–370 (2005). doi:10.​1096/​fj.​04-2364com PubMedCrossRef
76.
78.
go back to reference T.N. Stitt, D. Drujan, B.A. Clarke, F. Panaro, Y. Timofeyva, W.O. Kline, M. Gonzalez, G.D. Yancopoulos, D.J. Glass, The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol. Cell 14(3), 395–403 (2004)PubMedCrossRef T.N. Stitt, D. Drujan, B.A. Clarke, F. Panaro, Y. Timofeyva, W.O. Kline, M. Gonzalez, G.D. Yancopoulos, D.J. Glass, The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol. Cell 14(3), 395–403 (2004)PubMedCrossRef
79.
go back to reference A.R. Conery, Y. Cao, E.A. Thompson, C.M. Townsend, T.C. Ko, K. Luo, Akt interacts directly with Smad3 to regulate the sensitivity to TGF-beta induced apoptosis. Nat. Cell Biol. 6(4), 366–372 (2004)PubMedCrossRef A.R. Conery, Y. Cao, E.A. Thompson, C.M. Townsend, T.C. Ko, K. Luo, Akt interacts directly with Smad3 to regulate the sensitivity to TGF-beta induced apoptosis. Nat. Cell Biol. 6(4), 366–372 (2004)PubMedCrossRef
81.
go back to reference S. Clavel, A.-S. Coldefy, E. Kurkdjian, J. Salles, I. Margaritis, B. Derijard, Atrophy-related ubiquitin ligases, atrogin-1 and MuRF1 are up-regulated in aged rat Tibialis Anterior muscle. Mech. Ageing Dev. 127(10), 794–801 (2006). doi:10.1016/j.mad.2006.07.005 PubMedCrossRef S. Clavel, A.-S. Coldefy, E. Kurkdjian, J. Salles, I. Margaritis, B. Derijard, Atrophy-related ubiquitin ligases, atrogin-1 and MuRF1 are up-regulated in aged rat Tibialis Anterior muscle. Mech. Ageing Dev. 127(10), 794–801 (2006). doi:10.​1016/​j.​mad.​2006.​07.​005 PubMedCrossRef
82.
go back to reference E. Edström, M. Altun, M. Hägglund, B. Ulfhake, Atrogin-1/MAFbx and MuRF1 are downregulated in aging-related loss of skeletal muscle. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 61(7), 663–674 (2006)CrossRef E. Edström, M. Altun, M. Hägglund, B. Ulfhake, Atrogin-1/MAFbx and MuRF1 are downregulated in aging-related loss of skeletal muscle. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 61(7), 663–674 (2006)CrossRef
85.
go back to reference S.R. Kimball, J.P. O'Malley, J.C. Anthony, S.J. Crozier, L.S. Jefferson, Assessment of biomarkers of protein anabolism in skeletal muscle during the life span of the rat: sarcopenia despite elevated protein synthesis. Am. J. Physiol. Endocrinol. Metab. 287(4), E772–E780 (2004). doi:10.1152/ajpendo.00535.2003 PubMedCrossRef S.R. Kimball, J.P. O'Malley, J.C. Anthony, S.J. Crozier, L.S. Jefferson, Assessment of biomarkers of protein anabolism in skeletal muscle during the life span of the rat: sarcopenia despite elevated protein synthesis. Am. J. Physiol. Endocrinol. Metab. 287(4), E772–E780 (2004). doi:10.​1152/​ajpendo.​00535.​2003 PubMedCrossRef
86.
go back to reference A. Chalé-Rush, E.P. Morris, T.L. Kendall, N.E. Brooks, R.A. Fielding, Effects of chronic overload on muscle hypertrophy and mTOR signaling in young adult and aged rats. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 64(12), 1232–1239 (2009). doi:10.1093/gerona/glp146 CrossRef A. Chalé-Rush, E.P. Morris, T.L. Kendall, N.E. Brooks, R.A. Fielding, Effects of chronic overload on muscle hypertrophy and mTOR signaling in young adult and aged rats. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 64(12), 1232–1239 (2009). doi:10.​1093/​gerona/​glp146 CrossRef
87.
go back to reference K.G. O’Connor, J.D. Tobin, S.M. Harman, C.C. Plato, T.A. Roy, S.S. Sherman, M.R. Blackman, Serum levels of insulin-like growth factor-I are related to age and not to body composition in healthy women and men. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 53(3), M176–M182 (1998)CrossRef K.G. O’Connor, J.D. Tobin, S.M. Harman, C.C. Plato, T.A. Roy, S.S. Sherman, M.R. Blackman, Serum levels of insulin-like growth factor-I are related to age and not to body composition in healthy women and men. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 53(3), M176–M182 (1998)CrossRef
89.
go back to reference B. Léger, W. Derave, K. De Bock, P. Hespel, A.P. Russell, Human sarcopenia reveals an increase in SOCS-3 and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Res. 11(1), 163B–175B (2008). doi:10.1089/rej.2007.0588 CrossRef B. Léger, W. Derave, K. De Bock, P. Hespel, A.P. Russell, Human sarcopenia reveals an increase in SOCS-3 and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Res. 11(1), 163B–175B (2008). doi:10.​1089/​rej.​2007.​0588 CrossRef
90.
go back to reference C. Guillet, M. Prod’homme, M. Balage, P. Gachon, C. Giraudet, L. Morin, J. Grizard, Y. Boirie, Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans. FASEB J. 18(13), 1586–1587 (2004). doi:10.1096/fj.03-1341fje PubMed C. Guillet, M. Prod’homme, M. Balage, P. Gachon, C. Giraudet, L. Morin, J. Grizard, Y. Boirie, Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans. FASEB J. 18(13), 1586–1587 (2004). doi:10.​1096/​fj.​03-1341fje PubMed
91.
go back to reference M.J. Drummond, H.C. Dreyer, B. Pennings, C.S. Fry, S. Dhanani, E.L. Dillon, M. Sheffield-Moore, E. Volpi, B.B. Rasmussen, Skeletal muscle protein anabolic response to resistance exercise and essential amino acids is delayed with aging. J. Appl. Physiol. 104(5), 1452–1461 (2008). doi:10.1152/japplphysiol.00021.2008 PubMedCrossRef M.J. Drummond, H.C. Dreyer, B. Pennings, C.S. Fry, S. Dhanani, E.L. Dillon, M. Sheffield-Moore, E. Volpi, B.B. Rasmussen, Skeletal muscle protein anabolic response to resistance exercise and essential amino acids is delayed with aging. J. Appl. Physiol. 104(5), 1452–1461 (2008). doi:10.​1152/​japplphysiol.​00021.​2008 PubMedCrossRef
92.
go back to reference M.E. Carlson, M.J. Conboy, M. Hsu, L. Barchas, J. Jeong, A. Agrawal, A.J. Mikels, S. Agrawal, D.V. Schaffer, I.M. Conboy, Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 8(6), 676–689 (2009). doi:10.1111/j.1474-9726.2009.00517.x PubMedCrossRef M.E. Carlson, M.J. Conboy, M. Hsu, L. Barchas, J. Jeong, A. Agrawal, A.J. Mikels, S. Agrawal, D.V. Schaffer, I.M. Conboy, Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 8(6), 676–689 (2009). doi:10.​1111/​j.​1474-9726.​2009.​00517.​x PubMedCrossRef
94.
go back to reference M. Altun, H.C. Besche, H.S. Overkleeft, R. Piccirillo, M.J. Edelmann, B.M. Kessler, A.L. Goldberg, B. Ulfhake, Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. J. Biol. Chem. 285(51), 39597–39608 (2010). doi:10.1074/jbc.M110.129718 PubMedCrossRef M. Altun, H.C. Besche, H.S. Overkleeft, R. Piccirillo, M.J. Edelmann, B.M. Kessler, A.L. Goldberg, B. Ulfhake, Muscle wasting in aged, sarcopenic rats is associated with enhanced activity of the ubiquitin proteasome pathway. J. Biol. Chem. 285(51), 39597–39608 (2010). doi:10.​1074/​jbc.​M110.​129718 PubMedCrossRef
95.
97.
go back to reference A. Ratkevicius, A. Joyson, I. Selmer, T. Dhanani, C. Grierson, A.M. Tommasi, A. Devries, P. Rauchhaus, D. Crowther, S. Alesci, P. Yaworsky, F. Gilbert, T.W. Redpath, J. Brady, K.C.H. Fearon, D.M. Reid, C.A. Greig, H. Wackerhage, Serum concentrations of myostatin and myostatin-interacting proteins do not differ between young and sarcopenic elderly men. J. Gerontol. Ser. A Biol. Sci. Med. Sci. (2011). doi:10.1093/gerona/glr025 A. Ratkevicius, A. Joyson, I. Selmer, T. Dhanani, C. Grierson, A.M. Tommasi, A. Devries, P. Rauchhaus, D. Crowther, S. Alesci, P. Yaworsky, F. Gilbert, T.W. Redpath, J. Brady, K.C.H. Fearon, D.M. Reid, C.A. Greig, H. Wackerhage, Serum concentrations of myostatin and myostatin-interacting proteins do not differ between young and sarcopenic elderly men. J. Gerontol. Ser. A Biol. Sci. Med. Sci. (2011). doi:10.​1093/​gerona/​glr025
98.
go back to reference U. Raue, D. Slivka, B. Jemiolo, C. Hollon, S. Trappe, Proteolytic gene expression differs at rest and after resistance exercise between young and old women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 62(12), 1407–1412 (2007)CrossRef U. Raue, D. Slivka, B. Jemiolo, C. Hollon, S. Trappe, Proteolytic gene expression differs at rest and after resistance exercise between young and old women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 62(12), 1407–1412 (2007)CrossRef
99.
go back to reference S.A. Whitman, M.J. Wacker, S.R. Richmond, M.P. Godard, Contributions of the ubiquitin-proteasome pathway and apoptosis to human skeletal muscle wasting with age. Pflügers Archiv. 450(6), 437–446 (2005). doi:10.1007/s00424-005-1473-8 PubMedCrossRef S.A. Whitman, M.J. Wacker, S.R. Richmond, M.P. Godard, Contributions of the ubiquitin-proteasome pathway and apoptosis to human skeletal muscle wasting with age. Pflügers Archiv. 450(6), 437–446 (2005). doi:10.​1007/​s00424-005-1473-8 PubMedCrossRef
103.
go back to reference C.S. Fry, M.J. Drummond, E.L. Glynn, J.M. Dickinson, D.M. Gundermann, K.L. Timmerman, D.K. Walker, S. Dhanani, E. Volpi, B.B. Rasmussen, Aging impairs contraction-induced human skeletal muscle mTORC1 signaling and protein synthesis. Skelet. Muscle 1(1), 11 (2011). doi:10.1186/2044-5040-1-11 PubMedCrossRef C.S. Fry, M.J. Drummond, E.L. Glynn, J.M. Dickinson, D.M. Gundermann, K.L. Timmerman, D.K. Walker, S. Dhanani, E. Volpi, B.B. Rasmussen, Aging impairs contraction-induced human skeletal muscle mTORC1 signaling and protein synthesis. Skelet. Muscle 1(1), 11 (2011). doi:10.​1186/​2044-5040-1-11 PubMedCrossRef
105.
go back to reference S. Trappe, M. Godard, P. Gallagher, C. Carroll, G. Rowden, D. Porter, Resistance training improves single muscle fiber contractile function in older women. Am. J. Physiol. Cell Physiol. 281(2), C398–C406 (2001)PubMed S. Trappe, M. Godard, P. Gallagher, C. Carroll, G. Rowden, D. Porter, Resistance training improves single muscle fiber contractile function in older women. Am. J. Physiol. Cell Physiol. 281(2), C398–C406 (2001)PubMed
106.
go back to reference S. Trappe, D. Williamson, M. Godard, D. Porter, G. Rowden, D. Costill, Effect of resistance training on single muscle fiber contractile function in older men. J. Appl. Physiol. 89(1), 143–152 (2000)PubMed S. Trappe, D. Williamson, M. Godard, D. Porter, G. Rowden, D. Costill, Effect of resistance training on single muscle fiber contractile function in older men. J. Appl. Physiol. 89(1), 143–152 (2000)PubMed
107.
go back to reference D.L. Williamson, U. Raue, D.R. Slivka, S. Trappe, Resistance exercise, skeletal muscle FOXO3A, and 85-year-old women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 65(4), 335–343 (2010). doi:10.1093/gerona/glq005 CrossRef D.L. Williamson, U. Raue, D.R. Slivka, S. Trappe, Resistance exercise, skeletal muscle FOXO3A, and 85-year-old women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 65(4), 335–343 (2010). doi:10.​1093/​gerona/​glq005 CrossRef
108.
go back to reference A.R. Konopka, M.D. Douglass, L.A. Kaminsky, B. Jemiolo, T.A. Trappe, S. Trappe, M.P. Harber, Molecular adaptations to aerobic exercise training in skeletal muscle of older women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 65(11), 1201–1207 (2010). doi:10.1093/gerona/glq109 CrossRef A.R. Konopka, M.D. Douglass, L.A. Kaminsky, B. Jemiolo, T.A. Trappe, S. Trappe, M.P. Harber, Molecular adaptations to aerobic exercise training in skeletal muscle of older women. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 65(11), 1201–1207 (2010). doi:10.​1093/​gerona/​glq109 CrossRef
109.
go back to reference A.R. Konopka, T.A. Trappe, B. Jemiolo, S.W. Trappe, M.P. Harber, Myosin heavy chain plasticity in aging skeletal muscle with aerobic exercise training. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 66(8), 835–841 (2011). doi:10.1093/gerona/glr088 CrossRef A.R. Konopka, T.A. Trappe, B. Jemiolo, S.W. Trappe, M.P. Harber, Myosin heavy chain plasticity in aging skeletal muscle with aerobic exercise training. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 66(8), 835–841 (2011). doi:10.​1093/​gerona/​glr088 CrossRef
111.
go back to reference M.A. Williams, W.L. Haskell, P.A. Ades, E.A. Amsterdam, V. Bittner, B.A. Franklin, M. Gulanick, S.T. Laing, K.J. Stewart, Resistance exercise in individuals with and without cardiovascular disease: 2007 update: a scientific statement from the American Heart Association Council on Clinical Cardiology and Council on Nutrition, Physical Activity, and Metabolism. Circulation 116(5), 572–584 (2007). doi:10.1161/CIRCULATIONAHA.107.185214 PubMedCrossRef M.A. Williams, W.L. Haskell, P.A. Ades, E.A. Amsterdam, V. Bittner, B.A. Franklin, M. Gulanick, S.T. Laing, K.J. Stewart, Resistance exercise in individuals with and without cardiovascular disease: 2007 update: a scientific statement from the American Heart Association Council on Clinical Cardiology and Council on Nutrition, Physical Activity, and Metabolism. Circulation 116(5), 572–584 (2007). doi:10.​1161/​CIRCULATIONAHA.​107.​185214 PubMedCrossRef
112.
go back to reference G.E. Butterfield, J. Thompson, M.J. Rennie, R. Marcus, R.L. Hintz, A.R. Hoffman, Effect of rhGH and rhIGF-I treatment on protein utilization in elderly women. Am. J. Physiol. 272(1 Pt 1), E94–E99 (1997)PubMed G.E. Butterfield, J. Thompson, M.J. Rennie, R. Marcus, R.L. Hintz, A.R. Hoffman, Effect of rhGH and rhIGF-I treatment on protein utilization in elderly women. Am. J. Physiol. 272(1 Pt 1), E94–E99 (1997)PubMed
113.
go back to reference N. Lebrasseur, T. Schelhorn, B. Bernardo, P. Cosgrove, P. Loria, T. Brown, Myostatin inhibition enhances the effects of exercise on performance and metabolic outcomes in aged mice. J. Gerontol. Ser. A Biol. Sci. Med. Sci. (2009). doi:10.1093/gerona/glp068 N. Lebrasseur, T. Schelhorn, B. Bernardo, P. Cosgrove, P. Loria, T. Brown, Myostatin inhibition enhances the effects of exercise on performance and metabolic outcomes in aged mice. J. Gerontol. Ser. A Biol. Sci. Med. Sci. (2009). doi:10.​1093/​gerona/​glp068
114.
go back to reference K.T. Murphy, R. Koopman, T. Naim, B. Léger, J. Trieu, C. Ibebunjo, G.S. Lynch, Antibody-directed myostatin inhibition in 21-mo-old mice reveals novel roles for myostatin signaling in skeletal muscle structure and function. FASEB J. 24(11), 4433–4442 (2010). doi:10.1096/fj.10-159608 PubMedCrossRef K.T. Murphy, R. Koopman, T. Naim, B. Léger, J. Trieu, C. Ibebunjo, G.S. Lynch, Antibody-directed myostatin inhibition in 21-mo-old mice reveals novel roles for myostatin signaling in skeletal muscle structure and function. FASEB J. 24(11), 4433–4442 (2010). doi:10.​1096/​fj.​10-159608 PubMedCrossRef
115.
go back to reference R.D. Cohn, C. van Erp, J.P. Habashi, A.A. Soleimani, E.C. Klein, M.T. Lisi, M. Gamradt, C.M. ap Rhys, T.M. Holm, B.L. Loeys, F. Ramirez, D.P. Judge, C.W. Ward, H.C. Dietz, Angiotensin II type 1 receptor blockade attenuates TGF-beta-induced failure of muscle regeneration in multiple myopathic states. Nat. Med. 13(2), 204–210 (2007). doi:10.1038/nm1536 PubMedCrossRef R.D. Cohn, C. van Erp, J.P. Habashi, A.A. Soleimani, E.C. Klein, M.T. Lisi, M. Gamradt, C.M. ap Rhys, T.M. Holm, B.L. Loeys, F. Ramirez, D.P. Judge, C.W. Ward, H.C. Dietz, Angiotensin II type 1 receptor blockade attenuates TGF-beta-induced failure of muscle regeneration in multiple myopathic states. Nat. Med. 13(2), 204–210 (2007). doi:10.​1038/​nm1536 PubMedCrossRef
116.
go back to reference T.N. Burks, E. Andres-Mateos, R. Marx, R. Mejias, C. van Erp, J.L. Simmers, J.D. Walston, C.W. Ward, R.D. Cohn, Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci. Transl. Med. 3(82), 82ra37 (2011). doi:10.1126/scitranslmed.3002227 PubMedCrossRef T.N. Burks, E. Andres-Mateos, R. Marx, R. Mejias, C. van Erp, J.L. Simmers, J.D. Walston, C.W. Ward, R.D. Cohn, Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci. Transl. Med. 3(82), 82ra37 (2011). doi:10.​1126/​scitranslmed.​3002227 PubMedCrossRef
Metadata
Title
Atrogin-1, MuRF-1, and sarcopenia
Authors
Jonathan P. Gumucio
Christopher L. Mendias
Publication date
01-02-2013
Publisher
Springer US
Published in
Endocrine / Issue 1/2013
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-012-9751-7

Other articles of this Issue 1/2013

Endocrine 1/2013 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.