Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Am J Physiol Endocrinol Metab ; 300(2): E327-40, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21045173

ABSTRACT

Declines in skeletal muscle size and strength, often seen with chronic wasting diseases, prolonged or high-dose glucocorticoid therapy, and the natural aging process in mammals, are usually associated with reduced physical activity and testosterone levels. However, it is not clear whether the decline in testosterone and activity are causally related. Using a mouse model, we found that removal of endogenous testosterone by orchidectomy results in an almost complete cessation in voluntary wheel running but only a small decline in muscle mass. Testosterone replacement restored running behavior and muscle mass to normal levels. Orchidectomy also suppressed the IGF-I/Akt pathway, activated the atrophy-inducing E3 ligases MuRF1 and MAFBx, and suppressed several energy metabolism pathways, and all of these effects were reversed by testosterone replacement. The study also delineated a distinct, previously unidentified set of genes that is inversely regulated by orchidectomy and testosterone treatment. These data demonstrate the necessity of testosterone for both speed and endurance of voluntary wheel running in mice and suggest a potential mechanism for declined activity in humans where androgens are deficient.


Subject(s)
Gene Expression/physiology , Motor Activity/physiology , Muscle, Skeletal/metabolism , Orchiectomy , Running/physiology , Signal Transduction/physiology , Testosterone/pharmacology , Anatomy, Cross-Sectional , Animals , Blotting, Western , Body Weight/physiology , Eating , Energy Metabolism/drug effects , Energy Metabolism/physiology , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/cytology , Organ Size/physiology , Physical Endurance/physiology , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/blood
2.
Am J Pathol ; 176(5): 2425-34, 2010 May.
Article in English | MEDLINE | ID: mdl-20363926

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by progressive skeletal muscle wasting and weakness, leading to premature death from respiratory and/or cardiac failure. A clinically relevant question is whether myostatin inhibition can improve function of the diaphragm, which exhibits a severe and progressive pathology comparable with that in DMD. We hypothesized that antibody-directed myostatin inhibition would improve the pathophysiology of diaphragm muscle strips from young mdx mice (when the pathology is mild) and adult mdx mice (when the pathology is quite marked). Five weeks treatment with a mouse chimera of anti-human myostatin antibody (PF-354, 10 mg/kg/week) increased muscle mass (P < 0.05) and increased diaphragm median fiber cross-sectional area (CSA, P < 0.05) in young C57BL/10 and mdx mice, compared with saline-treated controls. PF-354 had no effect on specific force (sPo, maximum force normalized to muscle CSA) of diaphragm muscle strips from young C57BL/10 mice, but increased sPo by 84% (P < 0.05) in young mdx mice. In contrast, 8 weeks of PF-354 treatment did not improve muscle mass, median fiber CSA, collagen infiltration, or sPo of diaphragm muscle strips from adult mdx mice. PF-354 antibody-directed myostatin inhibition completely restored the functional capacity of diaphragm strips to control levels when treatment was initiated early, but not in the later stages of disease progression, suggesting that such therapies may only have a limited window of efficacy for DMD and related conditions.


Subject(s)
Aging , Diaphragm/pathology , Muscular Dystrophy, Animal/metabolism , Myostatin/chemistry , Animals , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myostatin/antagonists & inhibitors , Myostatin/metabolism , Time Factors
3.
FASEB J ; 24(11): 4433-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624929

ABSTRACT

Sarcopenia is the progressive loss of skeletal muscle mass and function with advancing age, leading to reduced mobility and quality of life. We tested the hypothesis that antibody-directed myostatin inhibition would attenuate the decline in mass and function of muscles of aged mice and that apoptosis would be reduced. Eighteen-month-old C57BL/6 mice were treated for 14 wk with a once-weekly injection of saline (control, n=9) or a mouse chimera of anti-human myostatin antibody (PF-354, 10 mg/kg; n=12). PF-354 prevented the age-related reduction in body mass and increased soleus, gastrocnemius, and quadriceps muscle mass (P<0.05). PF-354 increased fiber cross-sectional area by 12% and enhanced maximum in situ force of tibialis anterior (TA) muscles by 35% (P<0.05). PF-354 increased the proportion of type IIa fibers by 114% (P<0.01) and enhanced activity of oxidative enzymes (SDH) by 39% (P<0.01). PF-354 reduced markers of apoptosis in TA muscle cross-sections by 56% (P<0.03) and reduced caspase3 mRNA by 65% (P<0.04). Antibody-directed myostatin inhibition attenuated the decline in mass and function of muscles of aging mice, in part, by reducing apoptosis. These observations identify novel roles for myostatin in regulation of muscle mass and highlight the therapeutic potential of antibody-directed myostatin inhibition for sarcopenia.


Subject(s)
Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Myostatin/antagonists & inhibitors , Signal Transduction , Aging/pathology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Apoptosis , Humans , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Oxidoreductases/metabolism
4.
Mech Ageing Dev ; 197: 111510, 2021 07.
Article in English | MEDLINE | ID: mdl-34019916

ABSTRACT

Progressive loss of muscle mass and function due to muscle fiber atrophy and loss in the elderly and chronically ill is now defined as sarcopenia. It is a major contributor to loss of independence, disability, need of long-term care as well as overall mortality. Sarcopenia is a heterogenous disease and underlying mechanisms are not completely understood. Here, we newly identified and used Tmem158, alongside Cdkn1a, as relevant senescence and denervation markers (SDMs), associated with muscle fiber atrophy. Subsequent application of laser capture microdissection (LCM) and RNA analyses revealed age- and disease-associated differences in gene expression and alternative splicing patterns in a rodent sarcopenia model. Of note, genes exhibiting such differential alternative splicing (DAS) are mainly involved in the contractile function of the muscle. Many of these splicing events are also found in a mouse model for myotonic dystrophy type 1 (DM1), underscoring the premature aging phenotype of this disease. We propose to add differential alternative splicing to the hallmarks of aging.


Subject(s)
Aging/metabolism , Alternative Splicing , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Receptors, Cell Surface/biosynthesis , Sarcopenia/metabolism , Aging/pathology , Animals , Cellular Senescence , Disease Models, Animal , Male , Muscle, Skeletal/pathology , Rats , Rats, Sprague-Dawley
5.
Cell Rep ; 29(3): 749-763.e12, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618641

ABSTRACT

HDAC4, a class IIa histone deacetylase, is upregulated in skeletal muscle in response to denervation-induced atrophy. When HDAC4 is deleted postnatally, mice are partially protected from denervation. Despite the name "histone" deacetylase, HDAC4 demonstrably deacetylates cytosolic and non-histone nuclear proteins. We developed potent and selective class IIa HDAC inhibitors. Using these tools and genetic knockdown, we identified three previously unidentified substrates of HDAC4: myosin heavy chain, peroxisome proliferator-activated receptor gamma co-activator 1alpha (PGC-1α), and heat shock cognate 71 kDa protein (Hsc70). HDAC4 inhibition almost completely prevented denervation-induced loss of myosin heavy chain isoforms and blocked the action of their E3 ligase, MuRF1. PGC-1α directly interacts with class IIa HDACs; selective inhibitors increased PGC-1α protein in muscles. Hsc70 deacetylation by HDAC4 affects its chaperone activity. Through these endogenous HDAC4 substrates, we identified several muscle metabolic pathways that are regulated by class IIa HDACs, opening up new therapeutic options to treat skeletal muscle disorders and potentially other disease where these specific pathways are affected.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Acetylation , Animals , Cells, Cultured , Female , Gene Expression , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Male , Mice , Mice, Knockout , Muscle Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myosin Heavy Chains/genetics , Protein Binding , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Glucocorticoid/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
6.
Nat Commun ; 10(1): 3187, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320633

ABSTRACT

Loss of innervation of skeletal muscle is a determinant event in several muscle diseases. Although several effectors have been identified, the pathways controlling the integrated muscle response to denervation remain largely unknown. Here, we demonstrate that PKB/Akt and mTORC1 play important roles in regulating muscle homeostasis and maintaining neuromuscular endplates after nerve injury. To allow dynamic changes in autophagy, mTORC1 activation must be tightly balanced following denervation. Acutely activating or inhibiting mTORC1 impairs autophagy regulation and alters homeostasis in denervated muscle. Importantly, PKB/Akt inhibition, conferred by sustained mTORC1 activation, abrogates denervation-induced synaptic remodeling and causes neuromuscular endplate degeneration. We establish that PKB/Akt activation promotes the nuclear import of HDAC4 and is thereby required for epigenetic changes and synaptic gene up-regulation upon denervation. Hence, our study unveils yet-unknown functions of PKB/Akt-mTORC1 signaling in the muscle response to nerve injury, with important implications for neuromuscular integrity in various pathological conditions.


Subject(s)
Autophagy/physiology , Histone Deacetylases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscle Denervation , Muscle, Skeletal/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Motor Endplate/pathology , Muscular Atrophy/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics
7.
Mol Cell Biol ; 34(4): 606-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24298022

ABSTRACT

The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.


Subject(s)
Activin Receptors, Type II/immunology , Activins/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Hypertrophy/metabolism , Myoblasts, Skeletal/metabolism , Activin Receptors, Type II/metabolism , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Atrophy/immunology , Atrophy/metabolism , Cell Differentiation/physiology , Humans , Hypertrophy/pathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/immunology , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism
8.
Mol Cell Biol ; 33(2): 194-212, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109432

ABSTRACT

Molecular mechanisms underlying sarcopenia, the age-related loss of skeletal muscle mass and function, remain unclear. To identify molecular changes that correlated best with sarcopenia and might contribute to its pathogenesis, we determined global gene expression profiles in muscles of rats aged 6, 12, 18, 21, 24, and 27 months. These rats exhibit sarcopenia beginning at 21 months. Correlation of the gene expression versus muscle mass or age changes, and functional annotation analysis identified gene signatures of sarcopenia distinct from gene signatures of aging. Specifically, mitochondrial energy metabolism (e.g., tricarboxylic acid cycle and oxidative phosphorylation) pathway genes were the most downregulated and most significantly correlated with sarcopenia. Also, perturbed were genes/pathways associated with neuromuscular junction patency (providing molecular evidence of sarcopenia-related functional denervation and neuromuscular junction remodeling), protein degradation, and inflammation. Proteomic analysis of samples at 6, 18, and 27 months confirmed the depletion of mitochondrial energy metabolism proteins and neuromuscular junction proteins. Together, these findings suggest that therapeutic approaches that simultaneously stimulate mitochondrogenesis and reduce muscle proteolysis and inflammation have potential for treating sarcopenia.


Subject(s)
Aging/genetics , Mitochondria/metabolism , Neuromuscular Junction/pathology , Proteome/analysis , Sarcopenia/pathology , Transcriptome , Aging/metabolism , Animals , DNA, Mitochondrial/genetics , Energy Metabolism , Gene Expression Profiling , Immunohistochemistry , Linear Models , Male , Microarray Analysis , Mitochondria/genetics , Mitochondria/pathology , Muscle Strength/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Postmortem Changes , Proteomics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation
9.
Sci Signal ; 5(211): ra14, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22337810

ABSTRACT

Skeletal muscle mass is regulated by activity, metabolism, and the availability of nutrients. During muscle atrophy, MNK2 expression increases. We found that MNK2 (mitogen-activated protein kinase-interacting kinase 2), but not MNK1, inhibited proteins involved in promoting protein synthesis, including eukaryotic translation initiation factor 4G (eIF4G) and mammalian target of rapamycin (mTOR). Phosphorylation at serine 1108 (Ser¹¹°8) of eIF4G, which is associated with enhanced protein translation, is promoted by insulin-like growth factor 1 and inhibited by rapamycin or starvation, suggesting that phosphorylation of this residue is regulated by mTOR. In cultured myotubes, small interfering RNA (siRNA) knockdown of MNK2 increased eIF4G Ser¹¹°8 phosphorylation and overcame rapamycin's inhibitory effect on this phosphorylation event. Phosphorylation of Ser¹¹°8 in eIF4G, in gastrocnemius muscle, was increased in mice lacking MNK2, but not those lacking MNK1, and this increased phosphorylation was maintained in MNK2-null animals under atrophy conditions and upon starvation. Conversely, overexpression of MNK2 decreased eIF4G Ser¹¹°8 phosphorylation. An siRNA screen revealed that serine-arginine-rich protein kinases linked increased MNK2 activity to decreased eIF4G phosphorylation. In addition, we found that MNK2 interacted with mTOR and inhibited phosphorylation of the mTOR target, the ribosomal kinase p70S6K (70-kD ribosomal protein S6 kinase), through a mechanism independent of the kinase activity of MNK2. These data indicate that MNK2 plays a unique role, not shared by its closest paralog MNK1, in limiting protein translation through its negative effect on eIF4G Ser¹¹°8 phosphorylation and p70S6K activation.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Arginine/metabolism , Blotting, Western , Cell Line , Dexamethasone/toxicity , Eukaryotic Initiation Factor-4G/genetics , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Serine-Threonine Kinases/genetics , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serine/genetics , Serine/metabolism , Sirolimus/pharmacology , Starvation/complications , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
10.
J Appl Physiol (1985) ; 110(4): 1065-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270350

ABSTRACT

Counteracting the atrophy of skeletal muscle associated with disuse has significant implications for minimizing the wasting and weakness in plaster casting, joint immobilization, and other forms of limb unloading, with relevance to orthopedics, sports medicine, and plastic and reconstructive surgery. We tested the hypothesis that antibody-directed myostatin inhibition would attenuate the loss of muscle mass and functional capacity in mice during 14 or 21 days of unilateral hindlimb casting. Twelve-week-old C57BL/10 mice were subjected to unilateral hindlimb plaster casting or served as controls. Mice received subcutaneous injections of saline or a mouse chimera of anti-human myostatin antibody (PF-354, 10 mg/kg; n = 6-9) on days 0 and 7 and were tested for muscle function on day 14, or were treated on days 0, 7, and 14 and tested for muscle function on day 21. Hindlimb casting reduced muscle mass, fiber size, and function of isolated soleus and extensor digitorum longus (EDL) muscles (P < 0.05). PF-354 attenuated the loss of muscle mass, fiber size, and function with greater effects after 14 days than after 21 days of casting, when wasting and weakness had plateaued (P < 0.05). Antibody-directed myostatin inhibition therefore attenuated the atrophy and loss of functional capacity in muscles from mice subjected to unilateral hindlimb casting with reductions in muscle size and strength being most apparent during the first 14 days of disuse. These findings highlight the therapeutic potential of antibody-directed myostatin inhibition for disuse atrophy especially within the first 2 wk of disuse.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myostatin/antagonists & inhibitors , Analysis of Variance , Animals , Hindlimb Suspension , Mice , Muscle Contraction/physiology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Atrophy/immunology , Muscular Atrophy/pathology , Myostatin/immunology , Myostatin/metabolism , Organ Size , Reverse Transcriptase Polymerase Chain Reaction
11.
Dev Cell ; 21(5): 835-47, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22033112

ABSTRACT

Insulin-like growth factor 1 (IGF1) induces skeletal muscle hypertrophy by activating the IGF1R/IRS1/PI3K/Akt pathway. However the effect of IGF1 in differentiated muscle is limited by IRS1 ubiquitination and proteasome-mediated breakdown. In skeletal muscle, IGF1R activation sensitizes IRS1 to degradation, and a screen for the responsible E3 ligase identified Fbxo40 as mediating this rapid turnover of IRS1, since IRS1 loss can be rescued by knockdown of Fbxo40. In biochemical assays, an SCF E3 ligase complex containing Fbxo40 directly ubiquitinates IRS1, and this activity is enhanced by increased tyrosine phosphorylation of IRS1. Fbxo40 is muscle specific in expression and is upregulated during differentiation. Knockdown of Fbxo40 induces dramatic hypertrophy of myofibers. Mice null for Fbxo40 have increased levels of IRS1 and demonstrate enhanced body and muscle size during the growth phase associated with elevated IGF1 levels. These findings establish an important means of restraining IGF1's effects on skeletal muscle.


Subject(s)
F-Box Proteins/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Cell Line , Mice , Mice, Inbred C57BL , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL