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










Publication year range
1.
Steroids ; 87: 59-66, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24928725

ABSTRACT

Androgens regulate body composition and skeletal muscle mass in males, but the molecular mechanisms are not fully understood. Recently, we demonstrated that trenbolone (a potent synthetic testosterone analogue that is not a substrate for 5-alpha reductase or for aromatase) induces myotrophic effects in skeletal muscle without causing prostate enlargement, which is in contrast to the known prostate enlarging effects of testosterone. These previous results suggest that the 5α-reduction of testosterone is not required for myotrophic action. We now report differential gene expression in response to testosterone versus trenbolone in the highly androgen-sensitive levator ani/bulbocavernosus (LABC) muscle complex of the adult rat after 6weeks of orchiectomy (ORX), using real time PCR. The ORX-induced expression of atrogenes (Muscle RING-finger protein-1 [MuRF1] and atrogin-1) was suppressed by both androgens, with trenbolone producing a greater suppression of atrogin-1 mRNA compared to testosterone. Both androgens elevated expression of anabolic genes (insulin-like growth factor-1 and mechano-growth factor) after ORX. ORX-induced increases in expression of glucocorticoid receptor (GR) mRNA were suppressed by trenbolone treatment, but not testosterone. In ORX animals, testosterone promoted WNT1-inducible-signaling pathway protein 2 (WISP-2) gene expression while trenbolone did not. Testosterone and trenbolone equally enhanced muscle regeneration as shown by increases in LABC mass and in protein expression of embryonic myosin by western blotting. In addition, testosterone increased WISP-2 protein levels. Together, these findings identify specific mechanisms by which testosterone and trenbolone may regulate skeletal muscle maintenance and growth.


Subject(s)
Androgens/pharmacology , Gene Expression Regulation/drug effects , Muscles/drug effects , Muscles/metabolism , Testosterone/pharmacology , Transcription, Genetic/drug effects , Trenbolone Acetate/pharmacology , Animals , Body Weight/drug effects , CCN Intercellular Signaling Proteins/metabolism , Male , Muscles/pathology , Muscles/physiology , Muscular Atrophy/genetics , Myosins/metabolism , Orchiectomy , Organ Size/drug effects , Rats , Receptors, Androgen/genetics , Receptors, Glucocorticoid/genetics , Regeneration/drug effects , Repressor Proteins/metabolism , Time Factors
2.
J Cell Sci ; 127(Pt 7): 1441-53, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24463822

ABSTRACT

The Forkhead box O (FoxO) transcription factors are activated, and necessary for the muscle atrophy, in several pathophysiological conditions, including muscle disuse and cancer cachexia. However, the mechanisms that lead to FoxO activation are not well defined. Recent data from our laboratory and others indicate that the activity of FoxO is repressed under basal conditions via reversible lysine acetylation, which becomes compromised during catabolic conditions. Therefore, we aimed to determine how histone deacetylase (HDAC) proteins contribute to activation of FoxO and induction of the muscle atrophy program. Through the use of various pharmacological inhibitors to block HDAC activity, we demonstrate that class I HDACs are key regulators of FoxO and the muscle-atrophy program during both nutrient deprivation and skeletal muscle disuse. Furthermore, we demonstrate, through the use of wild-type and dominant-negative HDAC1 expression plasmids, that HDAC1 is sufficient to activate FoxO and induce muscle fiber atrophy in vivo and is necessary for the atrophy of muscle fibers that is associated with muscle disuse. The ability of HDAC1 to cause muscle atrophy required its deacetylase activity and was linked to the induction of several atrophy genes by HDAC1, including atrogin-1, which required deacetylation of FoxO3a. Moreover, pharmacological inhibition of class I HDACs during muscle disuse, using MS-275, significantly attenuated both disuse muscle fiber atrophy and contractile dysfunction. Together, these data solidify the importance of class I HDACs in the muscle atrophy program and indicate that class I HDAC inhibitors are feasible countermeasures to impede muscle atrophy and weakness.


Subject(s)
Forkhead Transcription Factors/metabolism , Histone Deacetylase 1/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Acetylation , Animals , Histone Deacetylase 1/genetics , Humans , Male , Mice , Muscle, Skeletal/pathology , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Signal Transduction
3.
Front Physiol ; 4: 330, 2013.
Article in English | MEDLINE | ID: mdl-24273516

ABSTRACT

The stress-inducible 70-kDa heat shock protein (HSP70) is a highly conserved protein with diverse intracellular and extracellular functions. In skeletal muscle, HSP70 is rapidly induced in response to both non-damaging and damaging stress stimuli including exercise and acute muscle injuries. This upregulation of HSP70 contributes to the maintenance of muscle fiber integrity and facilitates muscle regeneration and recovery. Conversely, HSP70 expression is decreased during muscle inactivity and aging, and evidence supports the loss of HSP70 as a key mechanism which may drive muscle atrophy, contractile dysfunction and reduced regenerative capacity associated with these conditions. To date, the therapeutic benefit of HSP70 upregulation in skeletal muscle has been established in rodent models of muscle injury, muscle atrophy, modified muscle use, aging, and muscular dystrophy, which highlights HSP70 as a key therapeutic target for the treatment of various conditions which negatively affect skeletal muscle mass and function. This article will review these important findings and provide perspective on the unanswered questions related to HSP70 and skeletal muscle plasticity which require further investigation.

4.
PLoS One ; 8(4): e62687, 2013.
Article in English | MEDLINE | ID: mdl-23626847

ABSTRACT

Skeletal muscle regeneration following injury is a highly coordinated process that involves transient muscle inflammation, removal of necrotic cellular debris and subsequent replacement of damaged myofibers through secondary myogenesis. However, the molecular mechanisms which coordinate these events are only beginning to be defined. In the current study we demonstrate that Heat shock protein 70 (Hsp70) is increased following muscle injury, and is necessary for the normal sequence of events following severe injury induced by cardiotoxin, and physiological injury induced by modified muscle use. Indeed, Hsp70 ablated mice showed a significantly delayed inflammatory response to muscle injury induced by cardiotoxin, with nearly undetected levels of both neutrophil and macrophage markers 24 hours post-injury. At later time points, Hsp70 ablated mice showed sustained muscle inflammation and necrosis, calcium deposition and impaired fiber regeneration that persisted several weeks post-injury. Through rescue experiments reintroducing Hsp70 intracellular expression plasmids into muscles of Hsp70 ablated mice either prior to injury or post-injury, we confirm that Hsp70 optimally promotes muscle regeneration when expressed during both the inflammatory phase that predominates in the first four days following severe injury and the regenerative phase that predominates thereafter. Additional rescue experiments reintroducing Hsp70 protein into the extracellular microenvironment of injured muscles at the onset of injury provides further evidence that Hsp70 released from damaged muscle may drive the early inflammatory response to injury. Importantly, following induction of physiological injury through muscle reloading following a period of muscle disuse, reduced inflammation in 3-day reloaded muscles of Hsp70 ablated mice was associated with preservation of myofibers, and increased muscle force production at later time points compared to WT. Collectively our findings indicate that depending on the nature and severity of muscle injury, therapeutics which differentially target both intracellular and extracellular localized Hsp70 may optimally preserve muscle tissue and promote muscle functional recovery.


Subject(s)
HSP70 Heat-Shock Proteins/genetics , Inflammation/genetics , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Regeneration/genetics , Animals , Calcinosis/genetics , Calcinosis/pathology , HSP70 Heat-Shock Proteins/metabolism , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Time Factors
5.
Methods Mol Biol ; 798: 461-72, 2012.
Article in English | MEDLINE | ID: mdl-22130854

ABSTRACT

The use of nonviral (plasmid DNA) gene delivery into skeletal muscle has increased significantly in recent years. The procedure is used to overexpress wild-type proteins, express mutant proteins, or knock down endogenous proteins. These manipulations can identify the role of a specific protein in muscle cell biology and physiology. The same procedure of plasmid DNA gene delivery can be used to introduce a gene promoter reporter construct. Such constructs contain a defined sequence of a gene promoter that regulates the expression of a "reporter." This reporter is easily measured and reflects the in vivo transcriptional activity of the gene promoter sequence under study. The gene promoter can be mutated at known transcription factor-binding sites, truncated to identify specific regions of the gene promoter that are required for transcription, or introduced into skeletal muscle with an expression plasmid for a protein believed to regulate the gene's transcription. Therefore, the use of such gene promoter reporters allows for an in-depth physiological assessment of the gene's transcriptional regulation.


Subject(s)
Genes, Reporter , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Animals , Electroporation/methods , Gene Expression Regulation , Luciferases/genetics , Luciferases/metabolism , Plasmids/genetics , Rats
6.
FASEB J ; 26(3): 987-1000, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22102632

ABSTRACT

Cachexia is characterized by inexorable muscle wasting that significantly affects patient prognosis and increases mortality. Therefore, understanding the molecular basis of this muscle wasting is of significant importance. Recent work showed that components of the forkhead box O (FoxO) pathway are increased in skeletal muscle during cachexia. In the current study, we tested the physiological significance of FoxO activation in the progression of muscle atrophy associated with cachexia. FoxO-DNA binding dependent transcription was blocked in the muscles of mice through injection of a dominant negative (DN) FoxO expression plasmid prior to inoculation with Lewis lung carcinoma cells or the induction of sepsis. Expression of DN FoxO inhibited the increased mRNA levels of atrogin-1, MuRF1, cathepsin L, and/or Bnip3 and inhibited muscle fiber atrophy during cancer cachexia and sepsis. Interestingly, during control conditions, expression of DN FoxO decreased myostatin expression, increased MyoD expression and satellite cell proliferation, and induced fiber hypertrophy, which required de novo protein synthesis. Collectively, these data show that FoxO-DNA binding-dependent transcription is necessary for normal muscle fiber atrophy during cancer cachexia and sepsis, and further suggest that basal levels of FoxO play an important role during normal conditions to depress satellite cell activation and limit muscle growth.


Subject(s)
Cachexia/metabolism , Forkhead Transcription Factors/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Animals , Cachexia/genetics , Cachexia/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line , Cell Line, Tumor , Cycloheximide/pharmacology , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Hypertrophy , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Myostatin/genetics , Myostatin/metabolism , Plasmids/genetics , Protein Synthesis Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/genetics , Sepsis/metabolism , Sepsis/pathology , Transfection , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
Am J Physiol Cell Physiol ; 300(6): C1490-501, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21389279

ABSTRACT

The Forkhead Box O (FOXO) transcription factors regulate diverse cellular processes, and in skeletal muscle are both necessary and sufficient for muscle atrophy. Although the regulation of FOXO by Akt is well evidenced in skeletal muscle, the current study demonstrates that FOXO is also regulated in muscle via the histone acetyltransferase (HAT) activities of p300/CREB-binding protein (CBP). Transfection of rat soleus muscle with a dominant-negative p300, which lacks HAT activity and inhibits endogenous p300 HAT activity, increased FOXO reporter activity and induced transcription from the promoter of a bona fide FOXO target gene, atrogin-1. Conversely, increased HAT activity via transfection of either wild-type (WT) p300 or WT CBP repressed FOXO activation in vivo in response to muscle disuse, and in C2C12 cells in response to dexamethasone and acute starvation. Importantly, manipulation of HAT activity differentially regulated the expression of various FOXO target genes. Cotransfection of FOXO1, FOXO3a, or FOXO4 with the p300 constructs further identified p300 HAT activity to also differentially regulate the activity of the FOXO homologues. Markedly, decreased HAT activity strongly increased FOXO3a transcriptional activity, while increased HAT activity repressed FOXO3a activity and prevented its nuclear localization in response to nutrient deprivation. In contrast, p300 increased FOXO1 nuclear localization. In summary, this study provides the first evidence to support the acetyltransferase activities of p300/CBP in regulating FOXO signaling in skeletal muscle and suggests that acetylation may be an important mechanism to differentially regulate the FOXO homologues and dictate which FOXO target genes are activated in response to varying atrophic stimuli.


Subject(s)
Forkhead Transcription Factors/metabolism , Muscle, Skeletal/enzymology , p300-CBP Transcription Factors/metabolism , Animals , Cell Line , Forkhead Transcription Factors/genetics , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transcriptional Activation , p300-CBP Transcription Factors/genetics
8.
Muscle Nerve ; 41(1): 110-3, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19813194

ABSTRACT

We examined reactive oxygen species as upstream activators of nuclear factor kappaB; (NF-kappaB) and forkhead box O (Foxo) in skeletal muscle during disuse atrophy. Catalase, an enzyme that degrades H2O2, was overexpressed in soleus muscles via plasmid injection prior to 7 days of hindlimb immobilization. The increased catalase activity abolished immobilization-induced transactivation of both NF-kappaB and Foxo and attenuated the loss of muscle mass. Thus, H2O2 may be an important initiator of these signaling pathways that lead to muscle atrophy.


Subject(s)
Enzyme Activation/physiology , Forkhead Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Muscular Disorders, Atrophic/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/pharmacology , Animals , Blotting, Western , Cell Line , Disease Models, Animal , Follow-Up Studies , Forkhead Transcription Factors/drug effects , Male , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/pathology , NF-kappa B/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction
9.
Am J Physiol Cell Physiol ; 298(1): C38-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864323

ABSTRACT

The purpose of the current study was to determine whether heat shock protein 70 (Hsp70) directly regulates forkhead box O (FOXO) signaling in skeletal muscle. This aim stems from previous work demonstrating that Hsp70 overexpression inhibits disuse-induced FOXO transactivation and prevents muscle fiber atrophy. However, although FOXO is sufficient to cause muscle wasting, no data currently exist on the requirement of FOXO signaling in the progression of physiological muscle wasting, in vivo. In the current study we show that specific inhibition of FOXO, via expression of a dominant-negative FOXO3a, in rat soleus muscle during disuse prevented >40% of muscle fiber atrophy, demonstrating that FOXO signaling is required for disuse muscle atrophy. Subsequent experiments determined whether Hsp70 directly regulates FOXO3a signaling when independently activated in skeletal muscle, via transfection of FOXO3a. We show that Hsp70 inhibits FOXO3a-dependent transcription in a gene-specific manner. Specifically, Hsp70 inhibited FOXO3a-induced promoter activation of atrogin-1, but not MuRF1. Further studies showed that a FOXO3a DNA-binding mutant can activate MuRF1, but not atrogin-1, suggesting that FOXO3a activates these two genes through differential mechanisms. In summary, FOXO signaling is required for physiological muscle atrophy and is directly inhibited by Hsp70.


Subject(s)
Forkhead Transcription Factors/physiology , HSP70 Heat-Shock Proteins/physiology , Muscular Atrophy/metabolism , Animals , DNA, Complementary/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Genes, Reporter , Hindlimb/physiology , Homeostasis , Male , Muscular Atrophy/genetics , Muscular Atrophy/physiopathology , Plasmids , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Signal Transduction , Weight-Bearing
10.
FASEB J ; 23(10): 3415-23, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19528257

ABSTRACT

Heat shock protein 25/27 (Hsp25/27) is a cytoprotective protein that is ubiquitously expressed in most cells, and is up-regulated in response to cellular stress. Previous work, in nonmuscle cells, has shown that Hsp27 inhibits TNF-alpha-induced NF-kappaB activation. During skeletal muscle disuse, Hsp25/27 levels are decreased and NF-kappaB activity increased, and this increase in NF-kappaB activity is required for disuse muscle atrophy. Therefore, the purpose of the current study was to determine whether electrotransfer of Hsp27 into the soleus muscle of rats, prior to skeletal muscle disuse, is sufficient to inhibit skeletal muscle disuse atrophy and NF-kappaB activation. The 35% disuse muscle-fiber atrophy observed in nontransfected fibers was attenuated by 50% in fibers transfected with Hsp27. Hsp27 also inhibited the disuse-induced increase in MuRF1 and atrogin-1 transcription by 82 and 40%, respectively. Furthermore, disuse- and IKKbeta-induced NF-kappaB transactivation were abolished by Hsp27. In contrast, Hsp27 had no effect on Foxo transactivation. In conclusion, Hsp27 is a negative regulator of NF-kappaB in skeletal muscle, in vivo, and is sufficient to inhibit MuRF1 and atrogin-1 and attenuate skeletal muscle disuse atrophy.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , I-kappa B Kinase/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , NF-kappa B/antagonists & inhibitors , Animals , Atrophy/genetics , Atrophy/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Male , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , Transcription, Genetic , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
11.
FASEB J ; 22(11): 3836-45, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18644837

ABSTRACT

Heat shock protein 70 (Hsp70) is a highly conserved and ubiquitous protein that is reported to provide cytoprotection in various cell types and tissues. However, the importance of Hsp70 expression during skeletal muscle atrophy, when Hsp70 levels are significantly decreased, is not known. The current study aimed to determine whether plasmid-mediated overexpression of Hsp70, in the soleus muscle of rats, was sufficient to regulate specific atrophy signaling pathways and attenuate skeletal muscle disuse atrophy. We found that Hsp70 overexpression prevented disuse muscle fiber atrophy and inhibited the increased promoter activities of atrogin-1 and MuRF1. Importantly, the transcriptional activities of Foxo3a and NF-kappaB, which are implicated in the regulation of atrogin-1 and MuRF1, were abolished by Hsp70. These data suggest that Hsp70 may regulate key atrophy genes through inhibiting Foxo3a and NF-kappaB activities during disuse. Indeed, we show that specific inhibition of Foxo3a prevented the increases in both atrogin-1 and MuRF1 promoter activities during disuse. However, inhibition of NF-kappaB did not affect the activation of either promoter, suggesting its requirement for disuse atrophy is through its regulation of other atrophy genes. We conclude that overexpression of Hsp70 is sufficient to inhibit key atrophy signaling pathways and prevent skeletal muscle atrophy.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , Muscular Atrophy/metabolism , NF-kappa B/metabolism , Transcription, Genetic , Animals , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/genetics , HSP70 Heat-Shock Proteins/genetics , Male , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscular Atrophy/genetics , Muscular Atrophy/prevention & control , NF-kappa B/genetics , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/biosynthesis , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...