Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Physiol ; 9: 1336, 2018.
Article in English | MEDLINE | ID: mdl-30356878

ABSTRACT

The transcriptional coactivators peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and PGC-1ß are positive regulators of skeletal muscle mass and energy metabolism; however, whether they influence muscle growth and metabolic adaptations via increased protein synthesis is not clear. This study revealed PGC-1α or PGC-1ß overexpression in C2C12 myotubes increased protein synthesis and myotube diameter under basal conditions and attenuated the loss in protein synthesis following the treatment with the catabolic agent, dexamethasone. To investigate whether PGC-1α or PGC-1ß signal through the Akt/mTOR pathway to increase protein synthesis, treatment with the PI3K and mTOR inhibitors, LY294002 and rapamycin, respectively, was undertaken but found unable to block PGC-1α or PGC-1ß's promotion of protein synthesis. Furthermore, PGC-1α and PGC-1ß decreased phosphorylation of Akt and the Akt/mTOR substrate, p70S6K. In contrast to Akt/mTOR inhibition, the suppression of ERRα, a major effector of PGC-1α and PGC-1ß activity, attenuated the increase in protein synthesis and myotube diameter in the presence of PGC-1α or PGC-1ß overexpression. To characterize further the biological processes occurring, gene set enrichment analysis of genes commonly regulated by both PGC-1α and PGC-1ß was performed following a microarray screen. Genes were found enriched in metabolic and mitochondrial oxidative processes, in addition to protein translation and muscle development categories. This suggests concurrent responses involving both increased metabolism and myotube protein synthesis. Finally, based on their known function or unbiased identification through statistical selection, two sets of genes were investigated in a human exercise model of stimulated protein synthesis to characterize further the genes influenced by PGC-1α and PGC-1ß during physiological adaptive changes in skeletal muscle.

2.
Sci Transl Med ; 8(348): 348ra98, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27440729

ABSTRACT

Patients with advanced cancer often succumb to complications arising from striated muscle wasting associated with cachexia. Excessive activation of the type IIB activin receptor (ActRIIB) is considered an important mechanism underlying this wasting, where circulating procachectic factors bind ActRIIB and ultimately lead to the phosphorylation of SMAD2/3. Therapeutics that antagonize the binding of ActRIIB ligands are in clinical development, but concerns exist about achieving efficacy without off-target effects. To protect striated muscle from harmful ActRIIB signaling, and to reduce the risk of off-target effects, we developed an intervention using recombinant adeno-associated viral vectors (rAAV vectors) that increase expression of Smad7 in skeletal and cardiac muscles. SMAD7 acts as an intracellular negative regulator that prevents SMAD2/3 activation and promotes degradation of ActRIIB complexes. In mouse models of cachexia, rAAV:Smad7 prevented wasting of skeletal muscles and the heart independent of tumor burden and serum levels of procachectic ligands. Mechanistically, rAAV:Smad7 administration abolished SMAD2/3 signaling downstream of ActRIIB and inhibited expression of the atrophy-related ubiquitin ligases MuRF1 and MAFbx. These findings identify muscle-directed Smad7 gene delivery as a potential approach for preventing muscle wasting under conditions where excessive ActRIIB signaling occurs, such as cancer cachexia.


Subject(s)
Muscular Atrophy/metabolism , Muscular Atrophy/therapy , Neoplasms/physiopathology , Smad7 Protein/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Blotting, Western , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Myocardium/metabolism , Myocardium/pathology , Neoplasms/complications , Neoplasms/metabolism , Phosphorylation/genetics , Phosphorylation/physiology , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Smad7 Protein/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Sci Rep ; 5: 17535, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26657343

ABSTRACT

Follistatin is an inhibitor of TGF-ß superfamily ligands that repress skeletal muscle growth and promote muscle wasting. Accordingly, follistatin has emerged as a potential therapeutic to ameliorate the deleterious effects of muscle atrophy. However, it remains unclear whether the anabolic effects of follistatin are conserved across different modes of non-degenerative muscle wasting. In this study, the delivery of a recombinant adeno-associated viral vector expressing follistatin (rAAV:Fst) to the hind-limb musculature of mice two weeks prior to denervation or tenotomy promoted muscle hypertrophy that was sufficient to preserve muscle mass comparable to that of untreated sham-operated muscles. However, administration of rAAV:Fst to muscles at the time of denervation or tenotomy did not prevent subsequent muscle wasting. Administration of rAAV:Fst to innervated or denervated muscles increased protein synthesis, but markedly reduced protein degradation only in innervated muscles. Phosphorylation of the signalling proteins mTOR and S6RP, which are associated with protein synthesis, was increased in innervated muscles administered rAAV:Fst, but not in treated denervated muscles. These results demonstrate that the anabolic effects of follistatin are influenced by the interaction between muscle fibres and motor nerves. These findings have important implications for understanding the potential efficacy of follistatin-based therapies for non-degenerative muscle wasting.


Subject(s)
Denervation/adverse effects , Follistatin/genetics , Genetic Therapy , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Tenotomy/adverse effects , Animals , Dependovirus/genetics , Disease Models, Animal , Follistatin/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hypertrophy , Mice , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/therapy , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transduction, Genetic
4.
Clin Exp Pharmacol Physiol ; 42(1): 1-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25311629

ABSTRACT

Dysfunction and wasting of skeletal muscle as a consequence of illness decreases the length and quality of life. Currently, there are few, if any, effective treatments available to address these conditions. Hence, the existence of this unmet medical need has fuelled large scientific efforts. Fortunately, these efforts have shown many of the underlying mechanisms adversely affecting skeletal muscle health. With increased understanding have come breakthrough disease-specific and broad spectrum interventions, some progressing through clinical development. The present review focuses its attention on the role of the antagonistic process regulating skeletal muscle mass before branching into prospective promising therapeutic targets and interventions. Special attention is given to therapies in development against cancer cachexia and Duchenne muscular dystrophy before closing remarks on design and conceptualization of future therapies are presented to the reader.


Subject(s)
Body Composition/drug effects , Body Mass Index , Drug Delivery Systems/methods , Thinness/drug therapy , Androgens/administration & dosage , Animals , Body Composition/physiology , Drug Delivery Systems/trends , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development , Thinness/diagnosis , Thinness/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Wasting Syndrome/diagnosis , Wasting Syndrome/drug therapy , Wasting Syndrome/metabolism
5.
PLoS One ; 8(9): e73589, 2013.
Article in English | MEDLINE | ID: mdl-24023888

ABSTRACT

microRNAs regulate the development of myogenic progenitors, and the formation of skeletal muscle fibers. However, the role miRNAs play in controlling the growth and adaptation of post-mitotic musculature is less clear. Here, we show that inhibition of the established pro-myogenic regulator miR-206 can promote hypertrophy and increased protein synthesis in post-mitotic cells of the myogenic lineage. We have previously demonstrated that histone deacetylase 4 (HDAC4) is a target of miR-206 in the regulation of myogenic differentiation. We confirmed that inhibition of miR-206 de-repressed HDAC4 accumulation in cultured myotubes. Importantly, inhibition of HDAC4 activity by valproic acid or sodium butyrate prevented hypertrophy of myogenic cells otherwise induced by inhibition of miR-206. To test the significance of miRNA-206 as a regulator of skeletal muscle mass in vivo, we designed recombinant adeno-associated viral vectors (rAAV6 vectors) expressing miR-206, or a miR-206 "sponge," featuring repeats of a validated miR-206 target sequence. We observed that over-expression or inhibition of miR-206 in the muscles of mice decreased or increased endogenous HDAC4 levels respectively, but did not alter muscle mass or myofiber size. We subsequently manipulated miR-206 levels in muscles undergoing follistatin-induced hypertrophy or denervation-induced atrophy (models of muscle adaptation where endogenous miR-206 expression is altered). Vector-mediated manipulation of miR-206 activity in these models of cell growth and wasting did not alter gain or loss of muscle mass respectively. Our data demonstrate that although the miR-206/HDAC4 axis operates in skeletal muscle, the post-natal expression of miR-206 is not a key regulator of basal skeletal muscle mass or specific modes of muscle growth and wasting. These studies support a context-dependent role of miR-206 in regulating hypertrophy that may be dispensable for maintaining or modifying the adult skeletal muscle phenotype--an important consideration in relation to the development of therapeutics designed to manipulate microRNA activity in musculature.


Subject(s)
Histone Deacetylases/genetics , MicroRNAs/genetics , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/pathology , Animals , Cell Line , Dependovirus/genetics , Genetic Vectors/genetics , Hypertrophy/genetics , Mice , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/physiology , Muscular Atrophy/genetics
6.
J Cell Biol ; 197(7): 997-1008, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22711699

ABSTRACT

Follistatin is essential for skeletal muscle development and growth, but the intracellular signaling networks that regulate follistatin-mediated effects are not well defined. We show here that the administration of an adeno-associated viral vector expressing follistatin-288aa (rAAV6:Fst-288) markedly increased muscle mass and force-producing capacity concomitant with increased protein synthesis and mammalian target of rapamycin (mTOR) activation. These effects were attenuated by inhibition of mTOR or deletion of S6K1/2. Furthermore, we identify Smad3 as the critical intracellular link that mediates the effects of follistatin on mTOR signaling. Expression of constitutively active Smad3 not only markedly prevented skeletal muscle growth induced by follistatin but also potently suppressed follistatin-induced Akt/mTOR/S6K signaling. Importantly, the regulation of Smad3- and mTOR-dependent events by follistatin occurred independently of overexpression or knockout of myostatin, a key repressor of muscle development that can regulate Smad3 and mTOR signaling and that is itself inhibited by follistatin. These findings identify a critical role of Smad3/Akt/mTOR/S6K/S6RP signaling in follistatin-mediated muscle growth that operates independently of myostatin-driven mechanisms.


Subject(s)
Follistatin/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myostatin/metabolism , Smad3 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Follistatin/genetics , HEK293 Cells , Humans , Hypertrophy/metabolism , Mice , Mice, Inbred C57BL , Myostatin/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...