Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Endocrinology ; 150(12): 5395-404, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837877

ABSTRACT

Although it is well known that catecholamines inhibit skeletal muscle protein degradation, the molecular underlying mechanism remains unclear. This study was undertaken to investigate the role of beta(2)-adrenoceptors (AR) and cAMP in regulating the ubiquitin-proteasome system (UPS) in skeletal muscle. We report that increased levels of cAMP in isolated muscles, promoted by the cAMP phosphodiesterase inhibitor isobutylmethylxanthine was accompanied by decreased activity of the UPS, levels of ubiquitin-protein conjugates, and expression of atrogin-1, a key ubiquitin-protein ligase involved in muscle atrophy. In cultured myotubes, atrogin-1 induction after dexamethasone treatment was completely prevented by isobutylmethylxanthine. Furthermore, administration of clenbuterol, a selective beta(2)-agonist, to mice increased muscle cAMP levels and suppressed the fasting-induced expression of atrogin-1 and MuRF-1, atrogin-1 mRNA being much more responsive to clenbuterol. Moreover, clenbuterol increased the phosphorylation of muscle Akt and Foxo3a in fasted rats. Similar responses were observed in muscles exposed to dibutyryl-cAMP. The stimulatory effect of clenbuterol on cAMP and Akt was abolished in muscles from beta(2)-AR knockout mice. The suppressive effect of beta(2)-agonist on atrogin-1 was not mediated by PGC-1alpha (peroxisome proliferator-activated receptor-gamma coactivator 1alpha known to be induced by beta(2)-agonists and previously shown to inhibit atrogin-1 expression), because food-deprived PGC-1alpha knockout mice were still sensitive to clenbuterol. These findings suggest that the cAMP increase induced by stimulation of beta(2)-AR in skeletal muscles from fasted mice is possibly the mechanism by which catecholamines suppress atrogin-1 and the UPS, this effect being mediated via phosphorylation of Akt and thus inactivation of Foxo3.


Subject(s)
Cyclic AMP/metabolism , Muscle, Skeletal/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adrenergic beta-2 Receptor Agonists , Animals , Blotting, Western , Cell Line , Clenbuterol/pharmacology , Dexamethasone/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Tripartite Motif Proteins , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
FASEB J ; 23(9): 2844-54, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19406843

ABSTRACT

Statins are widely used to treat hypercholesterolemia but can lead to a number of side effects in muscle, including rhabdomyolysis. Our recent findings implicated the induction of atrogin-1, a gene required for the development of muscle atrophy, in statin-induced muscle damage. Since statins inhibit many biochemical reactions besides cholesterol synthesis, we sought to define the statin-inhibited pathways responsible for atrogin-1 expression and muscle damage. We report here that lovastatin-induced atrogin-1 expression and muscle damage in cultured mouse myotubes and zebrafish can be prevented in the presence of geranylgeranol but not farnesol. Further, inhibitors of the transfer of geranylgeranyl isoprene units to protein targets cause statin muscle damage and atrogin-1 induction in cultured cells and in fish. These findings support the concept that dysfunction of small GTP-binding proteins lead to statin-induced muscle damage since these molecules require modification by geranylgeranyl moieties for their cellular localization and activity. Collectively, our animal and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be regulated by novel signaling pathways.


Subject(s)
F-Box Proteins/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscular Atrophy/chemically induced , Prenylation/genetics , SKP Cullin F-Box Protein Ligases/genetics , Zebrafish Proteins/genetics , Animals , Cells, Cultured , GTP-Binding Proteins , Lovastatin/adverse effects , Mice , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/etiology , Transcriptional Activation , Zebrafish
3.
J Clin Invest ; 117(12): 3940-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17992259

ABSTRACT

Statins inhibit HMG-CoA reductase, a key enzyme in cholesterol synthesis, and are widely used to treat hypercholesterolemia. These drugs can lead to a number of side effects in muscle, including muscle fiber breakdown; however, the mechanisms of muscle injury by statins are poorly understood. We report that lovastatin induced the expression of atrogin-1, a key gene involved in skeletal muscle atrophy, in humans with statin myopathy, in zebrafish embryos, and in vitro in murine skeletal muscle cells. In cultured mouse myotubes, atrogin-1 induction following lovastatin treatment was accompanied by distinct morphological changes, largely absent in atrogin-1 null cells. In zebrafish embryos, lovastatin promoted muscle fiber damage, an effect that was closely mimicked by knockdown of zebrafish HMG-CoA reductase. Moreover, atrogin-1 knockdown in zebrafish embryos prevented lovastatin-induced muscle injury. Finally, overexpression of PGC-1alpha, a transcriptional coactivator that induces mitochondrial biogenesis and protects against the development of muscle atrophy, dramatically prevented lovastatin-induced muscle damage and abrogated atrogin-1 induction both in fish and in cultured mouse myotubes. Collectively, our human, animal, and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be a critical mediator of the muscle damage induced by statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Lovastatin/adverse effects , Muscle Proteins/metabolism , Muscular Disorders, Atrophic/enzymology , SKP Cullin F-Box Protein Ligases/metabolism , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cholesterol/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Mice , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/chemically induced , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , SKP Cullin F-Box Protein Ligases/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R971-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17008454

ABSTRACT

The regulation of cell size depends on a delicate balance between protein synthesis and breakdown. Skeletal and cardiac muscle adapt to hormonal and neuronal stimuli and can rapidly hypertrophy and atrophy; however, the extent to which these processes occur in smooth muscle is less clear. Atrophy in striated muscle results from enhanced protein breakdown and is associated with a common transcriptional profile and activation of the ubiquitin-proteasome pathway, including induction of the muscle-specific ubiquitin protein ligases atrogin-1 and muscle ring-finger protein 1 (MuRF-1). Here we show that atrogin-1 is also expressed in smooth muscle, and that both atrogin-1 and MuRF-1 are upregulated in the uterus following delivery, as rapid involution occurs. While these two genes are similarly induced in all types of muscle during rapid loss of cell mass, other striated muscle atrophy-specific transcriptional changes are not observed during uterine involution, suggesting different underlying molecular mechanisms. These results raise the possibility that activation of atrogin-1 and MuRF-1 may be a common general adaptation in cells undergoing a rapid reduction in size.


Subject(s)
Muscle Proteins/metabolism , Muscle, Smooth/enzymology , Postpartum Period/physiology , Ubiquitin-Protein Ligases/metabolism , Uterine Diseases/enzymology , Uterine Diseases/pathology , Uterus/enzymology , Uterus/pathology , Animals , Atrophy , Blotting, Northern , Blotting, Western , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Pregnancy , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...