Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 23(3): 723-734, 2018 03.
Article in English | MEDLINE | ID: mdl-29257130

ABSTRACT

The development of the cerebral cortex is a tightly regulated process that relies on exquisitely coordinated actions of intrinsic and extrinsic cues. Here, we show that the communication between forebrain meninges and apical neural progenitor cells (aNPC) is essential to cortical development, and that the basal compartment of aNPC is key to this communication process. We found that Celsr1, a cadherin of the adhesion G protein coupled receptor family, controls branching of aNPC basal processes abutting the meninges and thereby regulates retinoic acid (RA)-dependent neurogenesis. Loss-of-function of Celsr1 results in a decreased number of endfeet, modifies RA-dependent transcriptional activity and biases aNPC commitment toward self-renewal at the expense of basal progenitor and neuron production. The mutant cortex has a reduced number of neurons, and Celsr1 mutant mice exhibit microcephaly and behavioral abnormalities. Our results uncover an important role for Celsr1 protein and for the basal compartment of neural progenitor cells in fate decision during the development of the cerebral cortex.


Subject(s)
Neural Stem Cells/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Animals , Cadherins , Cerebral Cortex/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Prosencephalon/embryology , Stem Cells/metabolism
2.
Pflugers Arch ; 466(7): 1377-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24162233

ABSTRACT

The Ca(2+)-dependent kinase myosin light chain kinase (MLCK) is the activator of smooth muscle contraction. In addition, it has been reported to be involved in Ca(2+) channel regulation in cultured cells, and we previously showed that the MLCK inhibitor ML-7 decreases arginine vasopressin (AVP)-induced Ca(2+) influx in rat aorta. This study was designed to investigate whether MLCK is involved in Ca(2+) regulation in resistance artery smooth muscle cell, which plays a major role in the control of blood pressure. As ML compounds were shown to have off-target effects, MLCK was downregulated by transfection with a small interfering RNA targeting MLCK (MLCK-siRNA) in rat small resistance mesenteric artery (RMA) and in the rat embryonic aortic cell line A7r5. Noradrenaline-induced contraction and Ca(2+) signal were significantly depressed in MLCK-siRNA compared to scramble-siRNA-transfected RMA. Contraction and Ca(2+) signal induced by high KCl and voltage-activated Ca(2+) current were also significantly decreased in MLCK-siRNA-transfected RMA, suggesting that MLCK depletion modifies voltage-operated Ca(2+) channels. KCl- and AVP-induced Ca(2+) signals and voltage-activated Ca(2+) current were decreased in MLCK-depleted A7r5 cells. Eventually, real-time quantitative PCR analysis indicated that in A7r5, MLCK controlled mRNA expression of CaV1.2 (L-type) and CaV3.1 (T-type) voltage-dependent Ca(2+) channels. Our results suggest that MLCK controls the transcription of voltage-dependent Ca(2+) channels in vascular smooth muscle cells.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Kinase/metabolism , Animals , Arginine Vasopressin/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/genetics , Cell Line , Male , Muscle Contraction , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Kinase/genetics , Norepinephrine/pharmacology , Potassium Chloride/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Vasoconstriction
3.
Int J Biochem Cell Biol ; 45(10): 2163-72, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23806868

ABSTRACT

Many pathological states characterized by muscle atrophy (e.g., sepsis, cachexia, starvation, metabolic acidosis and severe insulinopenia) are associated with an increase in circulating glucocorticoids (GC) levels, suggesting that GC could trigger the muscle atrophy observed in these conditions. GC-induced muscle atrophy is characterized by fast-twitch, glycolytic muscles atrophy illustrated by decreased fiber cross-sectional area and reduced myofibrillar protein content. GC-induced muscle atrophy results from increased protein breakdown and decreased protein synthesis. Increased muscle proteolysis, in particular through the activation of the ubiquitin proteasome and the lysosomal systems, is considered to play a major role in the catabolic action of GC. The stimulation by GC of these two proteolytic systems is mediated through the increased expression of several Atrogenes ("genes involved in atrophy"), such as FOXO, Atrogin-1, and MuRF-1. The inhibitory effect of GC on muscle protein synthesis is thought to result mainly from the inhibition of the mTOR/S6 kinase 1 pathway. These changes in muscle protein turnover could be explained by changes in the muscle production of two growth factors, namely Insulin-like Growth Factor (IGF)-I, a muscle anabolic growth factor and Myostatin, a muscle catabolic growth factor. This review will discuss the recent progress made in the understanding of the mechanisms involved in GC-induced muscle atrophy and consider the implications of these advancements in the development of new therapeutic approaches for treating GC-induced myopathy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.


Subject(s)
Glucocorticoids/adverse effects , Glucocorticoids/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Animals , Glucocorticoids/toxicity , Humans , Muscular Atrophy/pathology , Protein Biosynthesis , Signal Transduction
4.
Am J Physiol Endocrinol Metab ; 303(6): E729-39, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22739109

ABSTRACT

Several catabolic states (sepsis, cancer, etc.) associated with acute inflammation are characterized by a loss of skeletal muscle due to accelerated proteolysis. The main proteolytic systems involved are the autophagy and the ubiquitin-proteasome (UPS) pathways. Among the signaling pathways that could mediate proteolysis induced by acute inflammation, the transcription factor NF-κB, induced by TNFα, and the transcription factor forkhead box O (FOXO), induced by glucocorticoids (GC) and inhibited by IGF-I, are likely to play a key role. The aim of this study was to identify the nature of the molecular mediators responsible for the induction of these muscle proteolytic systems in response to acute inflammation caused by LPS injection. LPS injection robustly stimulated the expression of several components of the autophagy and the UPS pathways in the skeletal muscle. This induction was associated with a rapid increase of circulating levels of TNFα together with a muscular activation of NF-κB followed by a decrease in circulating and muscle levels of IGF-I. Neither restoration of circulating IGF-I nor restoration of muscle IGF-I levels prevented the activation of autophagy and UPS genes by LPS. The inhibition of TNFα production and muscle NF-κB activation, respectively by using pentoxifilline and a repressor of NF-κB, did not prevent the activation of autophagy and UPS genes by LPS. Finally, inhibition of GC action with RU-486 blunted completely the activation of these atrogenes by LPS. In conclusion, we show that increased GC production plays a more crucial role than decreased IGF-I and increased TNFα/NF-κB pathway for the induction of the proteolytic systems caused by acute inflammation.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Animals , Autophagy/drug effects , Glucocorticoids/adverse effects , Glucocorticoids/antagonists & inhibitors , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscular Atrophy/blood , Muscular Atrophy/immunology , Muscular Atrophy/prevention & control , NF-kappa B/antagonists & inhibitors , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
5.
Endocrinology ; 153(1): 241-53, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22087027

ABSTRACT

Myostatin inhibition by follistatin (FS) offers a new approach for muscle mass enhancement. The aim of the present study was to characterize the mediators responsible for the FS hypertrophic action on skeletal muscle in male mice. Because IGF-I and IGF-II, two crucial skeletal muscle growth factors, are induced by myostatin inhibition, we assessed their role in FS action. First, we tested whether type 1 IGF receptor (IGF-IR) is required for FS-induced hypertrophy. By using mice expressing a dominant-negative IGF-IR in skeletal muscle, we showed that IGF-IR inhibition blunted by 63% fiber hypertrophy caused by FS. Second, we showed that FS caused the same degree of fiber hypertrophy in wild-type and IGF-II knockout mice. We then tested the role of the signaling molecules stimulated by IGF-IR, in particular the Akt/mammalian target of rapamycin (mTOR)/70-kDa ribosomal protein S6 kinase (S6K) pathway. We investigated whether Akt phosphorylation is required for the FS action. By cotransfecting a dominant-negative form of Akt together with FS, we showed that Akt inhibition reduced by 65% fiber hypertrophy caused by FS. Second, we evaluated the role of mTOR in FS action. Fiber hypertrophy induced by FS was reduced by 36% in rapamycin-treated mice. Finally, because the activity of S6K is increased by FS, we tested its role in FS action. FS caused the same degree of fiber hypertrophy in wild-type and S6K1/2 knockout mice. In conclusion, the IGF-IR/Akt/mTOR pathway plays a critical role in FS-induced muscle hypertrophy. In contrast, induction of IGF-II expression and S6K activity by FS are not required for the hypertrophic action of FS.


Subject(s)
Follistatin/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Receptor, IGF Type 1/metabolism , Animals , Base Sequence , DNA Primers/genetics , Follistatin/genetics , Humans , Hypertrophy , Insulin-Like Growth Factor II/deficiency , Insulin-Like Growth Factor II/genetics , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transfection
6.
Gene Ther ; 17(3): 328-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20010628

ABSTRACT

Inhibiting myostatin (mstn) causes spectacular increase in muscle mass, spurring research for therapeutic approaches against neuromuscular disorders. Yet, possible functional deterioration and compromised force production have been reported in isolated muscle of null mstn(-/-) mice. We analyzed vascular and metabolic response to repeated electro-stimulated exercise in vivo in mstn(-/-) mice compared with FVB wild-type controls (WT), using interleaved multi-parametric functional nuclear magnetic resonance (NMR) imaging and spectroscopy. At steady-state exercise, specific force of plantar flexion, phosphocreatine consumption measured by phosphorus spectroscopy and maximum perfusion measured by arterial spin-labeled (ASL) NMR imaging were identical in both groups. After exercise, phosphorus spectroscopy revealed reduced oxidative mitochondrial capacity in mstn(-/-), whereas early recovery perfusion was identical and oxygen extraction, estimated from the blood oxygen level-dependent (BOLD) contrast, was decreased when compared with WT. Hyperemia was prolonged, indicating specific regulation of the perfusional response in mstn(-/-) mice. Histology showed an increased proportion of type IIb fibers in hypertrophied muscles, but the distribution of capillary contacts per fiber between oxidative and glycolytic fibers was unaltered in mstn(-/-) compared with WT. These integrated results formed coherent evidence of a congruous, non-pathologic shift toward a more glycolytic metabolism in this model of mstn(-/-).


Subject(s)
Muscle, Skeletal/physiology , Myostatin/deficiency , Animals , Exercise Test , Glycolysis/genetics , Hyperemia/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Knockout , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Myostatin/genetics , Phosphocreatine/metabolism
7.
Horm Res ; 72 Suppl 1: 36-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19940494

ABSTRACT

BACKGROUND: Many pathological states characterized by muscle atrophy (e.g., sepsis, cachexia, starvation, metabolic acidosis and severe insulinopenia) are associated with an increase in circulating glucocorticoid (GC) levels, suggesting that GC could trigger the muscle atrophy observed in these conditions. GC-induced muscle atrophy results from decreased protein synthesis and increased protein degradation. The inhibitory effect of GCs on protein synthesis is thought to result mainly from the inhibition of the p70 ribosomal S6 protein kinase. The stimulatory effect of GCs on muscle proteolysis results from the activation of two major cellular proteolytic systems: ubiquitin proteasome and lysosomal systems. The decrease in muscle production of insulin-like growth factor I (IGF-I), a muscle anabolic growth factor, could contribute to GC-induced muscle atrophy. By activating the phosphatidylinositol-3-kinase/Akt pathway, IGF-I overrides GC action to stunt muscle atrophy. Evidence also indicates that increased production of myostatin, a catabolic growth factor, could play a critical role in GC-induced muscle atrophy. CONCLUSIONS: Recent progress in understanding the role of growth factors in GC-induced muscle atrophy allows investigation into new therapies to minimize this myopathy.


Subject(s)
Glucocorticoids/adverse effects , Muscle Proteins/metabolism , Muscular Atrophy/chemically induced , Animals , Forkhead Transcription Factors/physiology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Like Growth Factor I/physiology , Intracellular Signaling Peptides and Proteins/physiology , Muscular Atrophy/pathology , Myostatin/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Ubiquitin-Protein Ligases/metabolism , beta Catenin/physiology
8.
Endocrinology ; 149(8): 3900-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467435

ABSTRACT

Decrease of muscle IGF-I plays a critical role in muscle atrophy caused by glucocorticoids (GCs) because IGF-I gene electrotransfer prevents muscle atrophy caused by GCs. The goal of the present study was to identify the intracellular mediators responsible for the IGF-I anti-atrophic action in GC-induced muscle atrophy. We first assessed the IGF-I transduction pathway alterations caused by GC administration and their reversibility by local IGF-I overexpression performed by electrotransfer. Muscle atrophy induced by dexamethasone (dexa) administration occurred with a decrease in Akt (-53%; P<0.01) phosphorylation together with a decrease in beta-catenin protein levels (-40%; P<0.001). Prevention of atrophy by IGF-I was associated with restoration of Akt phosphorylation and beta-catenin levels. We then investigated whether muscle overexpression of these intracellular mediators could mimic the IGF-I anti-atrophic effects. Overexpression of a constitutively active form of Akt induced a marked fiber hypertrophy in dexa-treated animals (+175% of cross-sectional area; P<0.001) and prevented dexa-induced atrophy. This hypertrophy was associated with an increase in phosphorylated GSK-3beta (+17%; P<0.05) and in beta-catenin content (+35%; P<0.05). Furthermore, overexpression of a dominant-negative GSK-3beta or a stable form of beta-catenin increased fiber cross-sectional area by, respectively, 23% (P<0.001) and 29% (P<0.001) in dexa-treated rats, preventing completely the atrophic effect of GC. In conclusion, this work indicates that Akt, GSK-3beta, and beta-catenin probably contribute together to the IGF-I anti-atrophic effect in GC-induced muscle atrophy.


Subject(s)
Cytoprotection/genetics , Glucocorticoids/adverse effects , Glycogen Synthase Kinase 3/physiology , Insulin-Like Growth Factor I/genetics , Muscular Atrophy/chemically induced , Proto-Oncogene Proteins c-akt/physiology , beta Catenin/physiology , Animals , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Male , Muscular Atrophy/genetics , Organ Specificity/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Signal Transduction/physiology , Transfection , beta Catenin/genetics
9.
J Endocrinol ; 197(1): 1-10, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18372227

ABSTRACT

Glucocorticoid-induced muscle atrophy is characterized by fast-twitch or type II muscle fiber atrophy illustrated by decreased fiber cross-sectional area and reduced myofibrillar protein content. Muscle proteolysis, in particular through the ubiquitin- proteasome system (UPS), is considered to play a major role in the catabolic action of glucocorticoids. The stimulation by glucocorticoids of the UPS is mediated through the increased expression of several atrogenes ('genes involved in atrophy'), such as atrogin-1 and MuRF-1, two ubiquitin ligases involved in the targeting of protein to be degraded by the proteasome machinery. Glucocorticoids also exert an anti-anabolic action by blunting muscle protein synthesis. These changes in protein turnover may result from changes in the production of two growth factors which control muscle mass, namely IGF-I and myostatin respectively anabolic and catabolic toward the skeletal muscle. The decreased production of IGF-I as well as the increased production of myostatin have been both demonstrated to contribute to the muscle atrophy caused by glucocorticoids. At the molecular level, IGF-I antagonizes the catabolic action of glucocorticoids by inhibiting, through the PI3-kinase/Akt pathway, the activity of the transcription factor FOXO, a major switch for the stimulation of several atrogenes. These recent progress in the understanding of the glucocorticoid-induced muscle atrophy should allow to define new therapies aiming to minimize this myopathy. Promising new therapeutic approaches for treating glucocorticoid-induced muscle atrophy are also presented in this review.


Subject(s)
Glucocorticoids/toxicity , Muscular Atrophy/chemically induced , Activating Transcription Factor 4/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Like Growth Factor I/physiology , Muscular Atrophy/prevention & control , Myostatin/physiology , Protein Kinases/physiology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases
10.
Endocrinology ; 148(1): 452-60, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17038559

ABSTRACT

Glucocorticoids mediate muscle atrophy in many catabolic states. Myostatin expression, a negative regulator of muscle growth, is increased by glucocorticoids and myostatin overexpression is associated with lower muscle mass. This suggests that myostatin is required for the catabolic effects of glucocorticoids. We therefore investigated whether myostatin gene disruption could prevent muscle atrophy caused by glucocorticoids. Male myostatin knockout (KO) and wild-type mice were subjected to dexamethasone treatment (1 mg/kg.d for 10 d or 5 mg/kg.d for 4 d). In wild-type mice, daily administration of low-dose dexamethasone for 10 d resulted in muscle atrophy (tibialis anterior: -15%; gastrocnemius: -13%; P < 0.01) due to 15% decrease in the muscle fiber cross-sectional area (1621 +/- 31 vs. 1918 +/- 64 microm(2), P < 0.01). In KO mice, there was no reduction of muscle mass nor fiber cross-sectional area after dexamethasone treatment. Muscle atrophy after 4 d of high-dose dexamethasone was associated with increased mRNA of enzymes involved in proteolytic pathways (atrogin-1, muscle ring finger 1, and cathepsin L) and increased chymotrypsin-like proteasomal activity. In contrast, the mRNA of these enzymes and the proteasomal activity were not significantly affected by dexamethasone in KO mice. Muscle IGF-I mRNA was paradoxically decreased in KO mice (-35%, P < 0.05); this was associated with a potentially compensatory increase of IGF-II expression in both saline and dexamethasone-treated KO mice (2-fold, P < 0.01). In conclusion, our results show that myostatin deletion prevents muscle atrophy in glucocorticoid-treated mice, by blunting the glucocorticoid-induced enhanced proteolysis, and suggest an important role of myostatin in muscle atrophy caused by glucocorticoids.


Subject(s)
Dexamethasone/pharmacology , Gene Deletion , Glucocorticoids/pharmacology , Muscular Atrophy/physiopathology , Transforming Growth Factor beta/genetics , Animals , Body Weight , Gene Expression Regulation, Enzymologic/drug effects , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Myofibrils/enzymology , Myofibrils/pathology , Myostatin , Organ Size , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
11.
Endocrinology ; 146(4): 1789-97, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15661854

ABSTRACT

Catabolic states caused by injury are characterized by a loss of skeletal muscle. The anabolic action of IGF-I on muscle and the reduction of its muscle content in response to injury suggest that restoration of muscle IGF-I content might prevent skeletal muscle loss caused by injury. We investigated whether local overexpression of IGF-I protein by gene transfer could prevent skeletal muscle atrophy induced by glucocorticoids, a crucial mediator of muscle atrophy in catabolic states. Localized overexpression of IGF-I in tibialis anterior (TA) muscle was performed by injection of IGF-I cDNA followed by electroporation 3 d before starting dexamethasone injections (0.1 mg/kg.d sc). A control plasmid was electroporated in the contralateral TA muscle. Dexamethasone induced atrophy of the TA muscle as illustrated by reduction in muscle mass (403 +/- 11 vs. 461 +/- 19 mg, P < 0.05) and fiber cross-sectional area (1759 +/- 131 vs. 2517 +/- 93 mum(2), P < 0.05). This muscle atrophy was paralleled by a decrease in the IGF-I muscle content (7.2 +/- 0.9 vs. 15.7 +/- 1.4 ng/g of muscle, P < 0.001). As the result of IGF-I gene transfer, the IGF-I muscle content increased 2-fold (15.8 +/- 1.2 vs. 7.2 +/- 0.9 ng/g of muscle, P < 0.001). In addition, the muscle mass (437 +/- 8 vs. 403 +/- 11 mg, P < 0.01) and the fiber cross-sectional area (2269 +/- 129 vs. 1759 +/- 131 mum(2), P < 0.05) were increased in the TA muscle electroporated with IGF-I DNA, compared with the contralateral muscle electroporated with a control plasmid. Our results show therefore that IGF-I gene transfer by electroporation prevents muscle atrophy in glucocorticoid-treated rats. Our observation supports the important role of decreased muscle IGF-I in the muscle atrophy caused by glucocorticoids.


Subject(s)
Dexamethasone/pharmacology , Genetic Therapy , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/therapy , Animals , Electroporation , Insulin-Like Growth Factor I/analysis , Male , Muscle Proteins/analysis , Myofibrils/pathology , Rats , Rats, Wistar , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...