Cellular and molecular mechanisms of muscle atrophy

Skeletal muscle is a plastic organ that is maintained by multiple pathways regulating cell and protein turnover. During muscle atrophy, proteolytic systems are activated, and contractile proteins and organelles are removed, resulting in the shrinkage of muscle fibers. Excessive loss of muscle mass is associated with poor prognosis in several diseases, including myopathies and muscular dystrophies, as well as in systemic disorders such as cancer, diabetes, sepsis and heart failure. Muscle loss also occurs during aging. In this paper, we review the key mechanisms that regulate the turnover of contractile proteins and organelles in muscle tissue, and discuss how impairments in these mechanisms can contribute to muscle atrophy. We also discuss how protein synthesis and degradation are coordinately regulated by signaling pathways that are influenced by mechanical stress, physical activity, and the availability of nutrients and growth factors. Understanding how these pathways regulate muscle mass will provide new therapeutic targets for the prevention and treatment of muscle atrophy in metabolic and neuromuscular diseases.

[1]  I. Nonaka,et al.  Autophagic degradation of nuclear components in mammalian cells , 2009, Autophagy.

[2]  D. Freyssenet,et al.  Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. , 2009, Endocrinology.

[3]  D. Guttridge,et al.  NF-κB signaling in skeletal muscle health and disease. , 2011, Current topics in developmental biology.

[4]  S. Wing,et al.  USP19-deubiquitinating enzyme regulates levels of major myofibrillar proteins in L6 muscle cells. , 2009, American journal of physiology. Endocrinology and metabolism.

[5]  Da-Zhi Wang,et al.  Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex. , 2004, The Journal of clinical investigation.

[6]  C. Reggiani,et al.  Inducible activation of Akt increases skeletal muscle mass and force without satellite cell activation , 2009, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[7]  S. Bhatnagar,et al.  The TWEAK–Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice , 2010, The Journal of cell biology.

[8]  V. Baracos,et al.  USP19 is a ubiquitin-specific protease regulated in rat skeletal muscle during catabolic states. , 2005, American journal of physiology. Endocrinology and metabolism.

[9]  Se-Jin Lee,et al.  Double muscling in cattle due to mutations in the myostatin gene. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[10]  Jennifer Skeen,et al.  Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. , 2003, Genes & development.

[11]  Keiichi Fukuda,et al.  Crosstalk between glucocorticoid receptor and nutritional sensor mTOR in skeletal muscle. , 2011, Cell metabolism.

[12]  Mi-Sung Kim,et al.  Myosin accumulation and striated muscle myopathy result from the loss of muscle RING finger 1 and 3. , 2007, The Journal of clinical investigation.

[13]  R. Youle,et al.  Mechanisms of mitophagy , 2010, Nature Reviews Molecular Cell Biology.

[14]  Daniel J. Klionsky,et al.  Autophagy fights disease through cellular self-digestion , 2008, Nature.

[15]  C. Dogra,et al.  TNF‐related weak inducer of apoptosis (TWEAK) is a potent skeletal muscle‐wasting cytokine , 2007, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[16]  Kenneth S. Campbell,et al.  Effective fiber hypertrophy in satellite cell-depleted skeletal muscle , 2011, Development.

[17]  M. Lorenzo,et al.  Tumor necrosis factor alpha produces insulin resistance in skeletal muscle by activation of inhibitor kappaB kinase in a p38 MAPK-dependent manner. , 2004, The Journal of biological chemistry.

[18]  A. Goldberg,et al.  Role of different proteolytic systems in the degradation of muscle proteins during denervation atrophy. , 1990, The Journal of biological chemistry.

[19]  E. Olson,et al.  Myogenin and Class II HDACs Control Neurogenic Muscle Atrophy by Inducing E3 Ubiquitin Ligases , 2010, Cell.

[20]  Hiroyuki Aburatani,et al.  Skeletal Muscle FOXO1 (FKHR) Transgenic Mice Have Less Skeletal Muscle Mass, Down-regulated Type I (Slow Twitch/Red Muscle) Fiber Genes, and Impaired Glycemic Control*[boxs] , 2004, Journal of Biological Chemistry.

[21]  A. Brunet,et al.  The FoxO code , 2008, Oncogene.

[22]  R. Youle,et al.  Targeting mitochondrial dysfunction: role for PINK1 and Parkin in mitochondrial quality control. , 2011, Antioxidants & redox signaling.

[23]  S. Schiaffino,et al.  Studies on the effect of denervation in developing muscle , 1969, Zeitschrift für Zellforschung und Mikroskopische Anatomie.

[24]  M. Komatsu,et al.  A role for NBR1 in autophagosomal degradation of ubiquitinated substrates. , 2009, Molecular cell.

[25]  G. Yancopoulos,et al.  The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. , 2004, Molecular cell.

[26]  Wei Wei,et al.  Role of glucocorticoids in the molecular regulation of muscle wasting , 2007, Critical care medicine.

[27]  A. Goldberg,et al.  Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[28]  M. Georges,et al.  Modulating skeletal muscle mass by postnatal, muscle‐specific inactivation of the myostatin gene , 2003, Genesis.

[29]  F. Trensz,et al.  The proteasome inhibitor MG132 reduces immobilization-induced skeletal muscle atrophy in mice , 2011, BMC musculoskeletal disorders.

[30]  Richard T. Lee,et al.  Transgenic Overexpression of Locally Acting Insulin-Like Growth Factor-1 Inhibits Ubiquitin-Mediated Muscle Atrophy in Chronic Left-Ventricular Dysfunction , 2005, Circulation research.

[31]  S. Bhatnagar,et al.  TWEAK and TRAF6 regulate skeletal muscle atrophy , 2012, Current opinion in clinical nutrition and metabolic care.

[32]  T. Luedde,et al.  Targeted ablation of IKK2 improves skeletal muscle strength, maintains mass, and promotes regeneration. , 2006, The Journal of clinical investigation.

[33]  Jiandie D. Lin,et al.  PGC-1α protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription , 2006, Proceedings of the National Academy of Sciences.

[34]  C. Leeuwenburgh,et al.  Skeletal muscle autophagy and apoptosis during aging: Effects of calorie restriction and life-long exercise , 2010, Experimental Gerontology.

[35]  N. Maraldi,et al.  Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration , 2010, Nature Medicine.

[36]  T. Braun,et al.  Myostatin mutation associated with gross muscle hypertrophy in a child. , 2004, The New England journal of medicine.

[37]  S. Kandarian,et al.  Role for IkappaBalpha, but not c-Rel, in skeletal muscle atrophy. , 2007, American journal of physiology. Cell physiology.

[38]  S. Bodine,et al.  The glucocorticoid receptor and FOXO1 synergistically activate the skeletal muscle atrophy-associated MuRF1 gene. , 2008, American journal of physiology. Endocrinology and metabolism.

[39]  D. Allen,et al.  Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. , 2006, American journal of physiology. Cell physiology.

[40]  C. Matsumura,et al.  Autophagy is increased in laminin α2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A. , 2011, Human molecular genetics.

[41]  E. Casanova,et al.  Skeletal muscle-specific ablation of raptor, but not of rictor, causes metabolic changes and results in muscle dystrophy. , 2008, Cell metabolism.

[42]  A. Cuervo Autophagy and aging: keeping that old broom working. , 2008, Trends in genetics : TIG.

[43]  R. E. Olson MECHANISMS CONTROLLING THE , 1971 .

[44]  K. Ikeda,et al.  A novel ubiquitin‐binding protein ZNF216 functioning in muscle atrophy , 2006, The EMBO journal.

[45]  T. Takenawa,et al.  Nebulin and N-WASP Cooperate to Cause IGF-1–Induced Sarcomeric Actin Filament Formation , 2010, Science.

[46]  D. Calderwood,et al.  The E3 ubiquitin ligase specificity subunit ASB2β is a novel regulator of muscle differentiation that targets filamin B to proteasomal degradation , 2009, Cell Death and Differentiation.

[47]  V. Sartorelli,et al.  Molecular and Cellular Determinants of Skeletal Muscle Atrophy and Hypertrophy , 2004, Science's STKE.

[48]  T. Zimmers,et al.  Induction of Cachexia in Mice by Systemically Administered Myostatin , 2002, Science.

[49]  S. Cook,et al.  Myostatin Regulates Cardiomyocyte Growth Through Modulation of Akt Signaling , 2006, Circulation research.

[50]  E. Ralston,et al.  Fiber Type Conversion by PGC-1α Activates Lysosomal and Autophagosomal Biogenesis in Both Unaffected and Pompe Skeletal Muscle , 2010, PloS one.

[51]  Jerry Donovan,et al.  Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. , 2003, Nature.

[52]  John L Cleveland,et al.  Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes , 2008, Autophagy.

[53]  W. Frontera,et al.  IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. , 2004, Cell.

[54]  Herman I. May,et al.  Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis , 2012, Nature.

[55]  W. Mitch,et al.  Regulation of muscle protein degradation: coordinated control of apoptotic and ubiquitin-proteasome systems by phosphatidylinositol 3 kinase. , 2004, Journal of the American Society of Nephrology : JASN.

[56]  M. Sandri,et al.  Autophagy inhibition induces atrophy and myopathy in adult skeletal muscles , 2010, Autophagy.

[57]  R. DePinho,et al.  A Foxo/Notch pathway controls myogenic differentiation and fiber type specification. , 2007, The Journal of clinical investigation.

[58]  S. Gygi,et al.  Ubiquitylation by Trim32 causes coupled loss of desmin, Z-bands, and thin filaments in muscle atrophy , 2012, The Journal of cell biology.

[59]  D J Glass,et al.  Identification of Ubiquitin Ligases Required for Skeletal Muscle Atrophy , 2001, Science.

[60]  Se-Jin Lee,et al.  Regulation of myostatin activity and muscle growth , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[61]  Nicholas Ling,et al.  Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF‐κB‐independent, FoxO1‐dependent mechanism , 2006 .

[62]  S. Wing,et al.  USP 19-deubiquitinating enzyme regulates levels of major myofibrillar proteins in L 6 muscle cells , 2009 .

[63]  S. Kandarian,et al.  Role for IκBα, but not c-Rel, in skeletal muscle atrophy , 2007 .

[64]  Chad E. Grueter,et al.  Histone deacetylases 1 and 2 regulate autophagy flux and skeletal muscle homeostasis in mice , 2012, Proceedings of the National Academy of Sciences.

[65]  Se-Jin Lee,et al.  Quadrupling Muscle Mass in Mice by Targeting TGF-ß Signaling Pathways , 2007, PloS one.

[66]  P. Bonaldo,et al.  Dysfunction of Mitochondria and Sarcoplasmic Reticulum in the Pathogenesis of Collagen VI Muscular Dystrophies , 2008, Annals of the New York Academy of Sciences.

[67]  Masaaki Komatsu,et al.  Homeostatic Levels of p62 Control Cytoplasmic Inclusion Body Formation in Autophagy-Deficient Mice , 2007, Cell.

[68]  R. Farrar,et al.  Muscle-specific inactivation of the IGF-I receptor induces compensatory hyperplasia in skeletal muscle. , 2002, The Journal of clinical investigation.

[69]  S. Bodine,et al.  Rapamycin inhibits the growth and muscle-sparing effects of clenbuterol. , 2007, Journal of applied physiology.

[70]  D. Attaix,et al.  Identification of cathepsin L as a differentially expressed message associated with skeletal muscle wasting. , 2001, The Biochemical journal.

[71]  V. Sirri,et al.  Degradation of MyoD Mediated by the SCF (MAFbx) Ubiquitin Ligase* , 2005, Journal of Biological Chemistry.

[72]  P. Tien,et al.  Effect of RNA oligonucleotide targeting Foxo-1 on muscle growth in normal and cancer cachexia mice , 2007, Cancer Gene Therapy.

[73]  A. Goldberg,et al.  Proteins containing peptide sequences related to Lys-Phe-Glu-Arg-Gln are selectively depleted in liver and heart, but not skeletal muscle, of fasted rats. , 1991, The Biochemical journal.

[74]  E. Froesch,et al.  Insulin and Insulin-Like Growth Factor I , 1987 .

[75]  G. Yancopoulos,et al.  Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo , 2001, Nature Cell Biology.

[76]  A. Goldberg,et al.  FoxO3 controls autophagy in skeletal muscle in vivo. , 2007, Cell metabolism.

[77]  T. Levine,et al.  Insulin and Insulin‐Like Growth Factor , 2012 .

[78]  J. Rilstone,et al.  RETRACTED: VMA21 Deficiency Causes an Autophagic Myopathy by Compromising V-ATPase Activity and Lysosomal Acidification , 2009, Cell.

[79]  Zhen Yan,et al.  PGC-1 (cid:1) Promotes Nitric Oxide Antioxidant Defenses and Inhibits FOXO Signaling Against Cardiac Cachexia in Mice , 2011 .

[80]  Susan C Kandarian,et al.  Disruption of either the Nfkb1 or the Bcl3 gene inhibits skeletal muscle atrophy. , 2004, The Journal of clinical investigation.

[81]  L. Deldicque,et al.  Prevention of muscle disuse atrophy by MG132 proteasome inhibitor , 2011, Muscle & nerve.

[82]  C. Mammucari,et al.  Smad2 and 3 transcription factors control muscle mass in adulthood. , 2009, American journal of physiology. Cell physiology.

[83]  M. Matsui,et al.  In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. , 2003, Molecular biology of the cell.

[84]  C. Mammucari,et al.  Regulation of skeletal muscle growth by the IGF1-Akt/PKB pathway: insights from genetic models , 2011, Skeletal Muscle.

[85]  Ivan Dikic,et al.  Nix is a selective autophagy receptor for mitochondrial clearance , 2010, EMBO reports.

[86]  M. Lorenzo,et al.  Tumor Necrosis Factor α Produces Insulin Resistance in Skeletal Muscle by Activation of Inhibitor κB Kinase in a p38 MAPK-dependent Manner* , 2004, Journal of Biological Chemistry.

[87]  S. Walkley,et al.  The Lysosomal System: Physiology and Pathology , 2012 .

[88]  M. Sandri,et al.  Mitochondrial Biogenesis and Fragmentation as Regulators of Muscle Protein Degradation , 2010, Current hypertension reports.

[89]  K. Nakashima,et al.  AMPK Activation Stimulates Myofibrillar Protein Degradation and Expression of Atrophy-Related Ubiquitin Ligases by Increasing FOXO Transcription Factors in C2C12 Myotubes , 2007, Bioscience, biotechnology, and biochemistry.

[90]  Marco Sandri,et al.  Foxo Transcription Factors Induce the Atrophy-Related Ubiquitin Ligase Atrogin-1 and Cause Skeletal Muscle Atrophy , 2004, Cell.

[91]  M. Fornaro,et al.  Gαi2 Signaling Promotes Skeletal Muscle Hypertrophy, Myoblast Differentiation, and Muscle Regeneration , 2011, Science Signaling.

[92]  M. Sandri,et al.  Physical exercise stimulates autophagy in normal skeletal muscles but is detrimental for collagen VI-deficient muscles , 2011, Autophagy.

[93]  Michael Karin,et al.  A central role for JNK in obesity and insulin resistance , 2002, Nature.

[94]  Antonio Musarò,et al.  Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle , 2001, Nature Genetics.

[95]  Hideyuki Okano,et al.  Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice , 2006, Nature.

[96]  E. Ralston,et al.  Autophagy and Lysosomes in Pompe Disease , 2006, Autophagy.

[97]  A. Musarò,et al.  Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. , 2008, Cell metabolism.

[98]  G. Yancopoulos,et al.  Conditional Activation of Akt in Adult Skeletal Muscle Induces Rapid Hypertrophy , 2004, Molecular and Cellular Biology.

[99]  D. Freyssenet,et al.  Ectopic expression of myostatin induces atrophy of adult skeletal muscle by decreasing muscle gene expression. , 2007, Endocrinology.

[100]  D. Sabatini,et al.  mTOR Signaling in Growth Control and Disease , 2012, Cell.

[101]  A. Goldberg,et al.  Atrogin1/MAFbx: what atrophy, hypertrophy, and cardiac failure have in common. , 2011, Circulation research.

[102]  D. Metzger,et al.  Autophagy is required to maintain muscle mass. , 2009, Cell metabolism.

[103]  J. Rilstone,et al.  VMA21 Deficiency Causes an Autophagic Myopathy by Compromising V-ATPase Activity and Lysosomal Acidification (Retracted article. See vol. 142, pg. 984, 2010) , 2009 .

[104]  Yonghong Xiao,et al.  FoxOs Are Lineage-Restricted Redundant Tumor Suppressors and Regulate Endothelial Cell Homeostasis , 2007, Cell.

[105]  S. Reed,et al.  Inhibition of FoxO transcriptional activity prevents muscle fiber atrophy during cachexia and induces hypertrophy , 2012, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[106]  M. Sandri,et al.  Autophagy induction rescues muscular dystrophy , 2011, Autophagy.

[107]  J. Haspel,et al.  Selective expression of Cre recombinase in skeletal muscle fibers , 2000, Genesis.

[108]  S. Kandarian,et al.  Disruption of either the Nfkb 1 or the Bcl 3 gene inhibits skeletal muscle atrophy , 2004 .

[109]  Y. Marchand-Brustel,et al.  Insulin and Insulin-like Growth Factor I: Effects on Protein Synthesis in Isolated Muscles from Lean and Goldthioglucose-Obese Mice , 1983, Diabetes.

[110]  G. Bjørkøy,et al.  p62/SQSTM1 Binds Directly to Atg8/LC3 to Facilitate Degradation of Ubiquitinated Protein Aggregates by Autophagy* , 2007, Journal of Biological Chemistry.

[111]  S. Hatakeyama,et al.  Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. , 2009, American journal of physiology. Cell physiology.

[112]  E. Kudryashova,et al.  Satellite cell senescence underlies myopathy in a mouse model of limb-girdle muscular dystrophy 2H. , 2012, The Journal of clinical investigation.

[113]  M. Sandri,et al.  S6 kinase inactivation impairs growth and translational target phosphorylation in muscle cells maintaining proper regulation of protein turnover. , 2007, American journal of physiology. Cell physiology.

[114]  S. Gygi,et al.  An AMPK-FOXO Pathway Mediates Longevity Induced by a Novel Method of Dietary Restriction in C. elegans , 2007, Current Biology.

[115]  W. Frontera,et al.  IKKβ/NF-κB Activation Causes Severe Muscle Wasting in Mice , 2004, Cell.

[116]  A. Goldberg,et al.  Protein degradation by the ubiquitin-proteasome pathway in normal and disease states. , 2006, Journal of the American Society of Nephrology : JASN.

[117]  J. Massagué,et al.  Smad transcription factors. , 2005, Genes & development.

[118]  A. Cuervo,et al.  Chaperone‐mediated autophagy in health and disease , 2010, FEBS letters.

[119]  Marcus D. Goncalves,et al.  Akt Deficiency Attenuates Muscle Size and Function but Not the Response to ActRIIB Inhibition , 2010, PloS one.

[120]  R. Rizzuto,et al.  Bcl-2-associated autophagy regulator Naf-1 required for maintenance of skeletal muscle. , 2012, Human molecular genetics.

[121]  T. Proikas-Cezanne,et al.  Control of autophagy initiation by phosphoinositide 3‐phosphatase jumpy , 2009, The EMBO journal.

[122]  E. Ralston,et al.  Dysfunction of endocytic and autophagic pathways in a lysosomal storage disease , 2006, Annals of neurology.

[123]  N. Perrimon,et al.  FOXO/4E-BP Signaling in Drosophila Muscles Regulates Organism-wide Proteostasis during Aging , 2010, Cell.

[124]  Luca Scorrano,et al.  Mitochondrial fission and remodelling contributes to muscle atrophy , 2010, The EMBO journal.

[125]  S. Bhatnagar,et al.  The E3 Ubiquitin Ligase TRAF6 Intercedes in Starvation-Induced Skeletal Muscle Atrophy through Multiple Mechanisms , 2012, Molecular and Cellular Biology.

[126]  J. Thissen,et al.  Mechanisms of glucocorticoid-induced myopathy. , 2008, The Journal of endocrinology.

[127]  Cam Patterson,et al.  Muscle-specific RING finger 1 is a bona fide ubiquitin ligase that degrades cardiac troponin I , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[128]  A. Goldberg,et al.  Peroxisome Proliferator-activated Receptor γ Coactivator 1α or 1β Overexpression Inhibits Muscle Protein Degradation, Induction of Ubiquitin Ligases, and Disuse Atrophy* , 2010, The Journal of Biological Chemistry.

[129]  A. Goldberg,et al.  Rapid disuse and denervation atrophy involve transcriptional changes similar to those of muscle wasting during systemic diseases , 2007, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[130]  Orciuolo Enrico,et al.  Unexpected cardiotoxicity in haematological bortezomib treated patients. , 2007, British journal of haematology.

[131]  Wei He,et al.  A FoxO–Smad synexpression group in human keratinocytes , 2006, Proceedings of the National Academy of Sciences.

[132]  G. Lanfranchi,et al.  JunB transcription factor maintains skeletal muscle mass and promotes hypertrophy , 2010, The Journal of cell biology.

[133]  A. Goldberg,et al.  IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. , 2004, American journal of physiology. Endocrinology and metabolism.

[134]  D. Glass,et al.  The SCF-Fbxo40 complex induces IRS1 ubiquitination in skeletal muscle, limiting IGF1 signaling. , 2011, Developmental cell.

[135]  N. Rosenthal,et al.  Muscle-specific expression of IGF-1 blocks angiotensin II-induced skeletal muscle wasting. , 2005, The Journal of clinical investigation.

[136]  Y. Itoyama,et al.  NO production results in suspension-induced muscle atrophy through dislocation of neuronal NOS. , 2007, The Journal of clinical investigation.

[137]  S. Schiaffino,et al.  Studies on the effect of denervation in developing muscle. II. The lysosomal system. , 1972, Journal of ultrastructure research.

[138]  M. Febbraio,et al.  FOXO1 Regulates the Expression of 4E-BP1 and Inhibits mTOR Signaling in Mammalian Skeletal Muscle* , 2007, Journal of Biological Chemistry.

[139]  D. Tindall,et al.  Dynamic FoxO transcription factors , 2007, Journal of Cell Science.

[140]  A. Poupon,et al.  The Translation Regulatory Subunit eIF3f Controls the Kinase-Dependent mTOR Signaling Required for Muscle Differentiation and Hypertrophy in Mouse , 2010, PloS one.

[141]  S. Rikka,et al.  Microtubule-associated Protein 1 Light Chain 3 (LC3) Interacts with Bnip3 Protein to Selectively Remove Endoplasmic Reticulum and Mitochondria via Autophagy* , 2012, The Journal of Biological Chemistry.

[142]  S. Gygi,et al.  The Energy Sensor AMP-activated Protein Kinase Directly Regulates the Mammalian FOXO3 Transcription Factor* , 2007, Journal of Biological Chemistry.

[143]  S. Rakhilin,et al.  The E3 Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle. , 2007, Cell metabolism.

[144]  Florian Caiment,et al.  A mutation creating a potential illegitimate microRNA target site in the myostatin gene affects muscularity in sheep , 2006, Nature Genetics.

[145]  P. Tien,et al.  Inhibition of atrogin-1/MAFbx expression by adenovirus-delivered small hairpin RNAs attenuates muscle atrophy in fasting mice. , 2011, Human gene therapy.

[146]  R. Lüllmann-Rauch,et al.  Accumulation of autophagic vacuoles and cardiomyopathy in LAMP-2-deficient mice , 2000, Nature.

[147]  M. Sandri,et al.  Posttranslational modifications control FoxO3 activity during denervation. , 2012, American journal of physiology. Cell physiology.

[148]  V. Hill,et al.  Suppression of autophagy in skeletal muscle uncovers the accumulation of ubiquitinated proteins and their potential role in muscle damage in Pompe disease. , 2008, Human molecular genetics.

[149]  S. Bodine,et al.  Muscle sparing in muscle RING finger 1 null mice: response to synthetic glucocorticoids , 2011, The Journal of physiology.

[150]  Bruce M. Spiegelman,et al.  Insulin-regulated hepatic gluconeogenesis through FOXO1–PGC-1α interaction , 2003, Nature.

[151]  Se-Jin Lee,et al.  Myostatin and the control of skeletal muscle mass. , 1999, Current opinion in genetics & development.

[152]  L. Schaeffer,et al.  Muscle inactivation of mTOR causes metabolic and dystrophin defects leading to severe myopathy , 2009, The Journal of cell biology.

[153]  S. Bhatnagar,et al.  Targeted ablation of TRAF6 inhibits skeletal muscle wasting in mice , 2010, The Journal of cell biology.

[154]  S. Gygi,et al.  During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation , 2009, The Journal of cell biology.

[155]  S. Powers,et al.  Rapid disuse atrophy of diaphragm fibers in mechanically ventilated humans. , 2008, The New England journal of medicine.

[156]  Christian C Witt,et al.  Cooperative control of striated muscle mass and metabolism by MuRF1 and MuRF2 , 2007, The EMBO journal.

[157]  V. Mootha,et al.  Mechanisms Controlling Mitochondrial Biogenesis and Respiration through the Thermogenic Coactivator PGC-1 , 1999, Cell.

[158]  T. Hornberger,et al.  Faculty Opinions recommendation of Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. , 2014 .

[159]  D. Lacey,et al.  Reversal of Cancer Cachexia and Muscle Wasting by ActRIIB Antagonism Leads to Prolonged Survival , 2010, Cell.

[160]  B. Spiegelman,et al.  Increased muscle PGC-1α expression protects from sarcopenia and metabolic disease during aging , 2009, Proceedings of the National Academy of Sciences.

[161]  G. Supinski,et al.  Effect of proteasome inhibitors on endotoxin-induced diaphragm dysfunction. , 2009, American journal of physiology. Lung cellular and molecular physiology.

[162]  M. Hoch,et al.  Chaperone-Assisted Selective Autophagy Is Essential for Muscle Maintenance , 2010, Current Biology.

[163]  N. LeBrasseur,et al.  Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. , 2008, Cell metabolism.

[164]  C. Rommel,et al.  Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways , 2001, Nature Cell Biology.

[165]  Mark H. Ellisman,et al.  Sestrin as a Feedback Inhibitor of TOR That Prevents Age-Related Pathologies , 2010, Science.

[166]  L. Schaeffer,et al.  DHPR α1S subunit controls skeletal muscle mass and morphogenesis , 2010, The EMBO journal.

[167]  T. Fielder,et al.  Lower skeletal muscle mass in male transgenic mice with muscle-specific overexpression of myostatin. , 2003, American journal of physiology. Endocrinology and metabolism.

[168]  S. Sze,et al.  Myostatin induces degradation of sarcomeric proteins through a Smad3 signaling mechanism during skeletal muscle wasting. , 2011, Molecular endocrinology.

[169]  S. Reed,et al.  p300 acetyltransferase activity differentially regulates the localization and activity of the FOXO homologues in skeletal muscle , 2011, American journal of physiology. Cell physiology.

[170]  I. Nonaka,et al.  Lysosomal myopathies: An excessive build-up in autophagosomes is too much to handle , 2008, Neuromuscular Disorders.

[171]  Se-Jin Lee Regulation of muscle mass by myostatin. , 2004, Annual review of cell and developmental biology.

[172]  K. Patel,et al.  Muscle hypertrophy driven by myostatin blockade does not require stem/precursor-cell activity , 2009, Proceedings of the National Academy of Sciences.