Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
J Biol Chem ; 289(9): 5784-98, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24425880

ABSTRACT

One of the features of uncontrolled type 1 diabetes is oxidative stress that induces DNA damage and cell death. Skeletal muscle atrophy is also considerable in type 1 diabetes, however, the signaling mechanisms that induce oxidative stress culminating in muscle atrophy are not fully known. Here, we show that in Streptozotocin-induced diabetic wild type mice, hypo-phosphorylation of Akt, resulted in activation of Foxa2 transcription factor in the muscle. Foxa2 transcriptionally up-regulated Myostatin, contributing to exaggerated oxidative stress leading to DNA damage via p63/REDD1 pathway in skeletal muscle of Streptozotocin-treated wild type mice. In Myostatin(-/-) mice however, Streptozotocin treatment did not reduce Akt phosphorylation despite reduced IRS-1 signaling. Moreover, Foxa2 levels remained unaltered in Myostatin(-/-) mice, while levels of p63/REDD1 were higher compared with wild type mice. Consistent with these results, relatively less DNA damage and muscle atrophy was observed in Myostatin(-/-) muscle in response to Streptozotocin treatment. Taken together, our results for the first time show the role of Foxa2 in Myostatin regulation in skeletal muscle in diabetic mice. Altogether, these results demonstrate the mechanism by which Myostatin contributes to DNA damage in skeletal muscle of the diabetic mice that would lead to myofiber degeneration.


Subject(s)
DNA Damage , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Muscle Fibers, Skeletal/metabolism , Myostatin/metabolism , Oxidative Stress , Animals , Cell Line , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Myostatin/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Exp Cell Res ; 315(12): 2012-21, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19406121

ABSTRACT

Akirin1 (Mighty) is a downstream target gene of myostatin and has been shown to be a promyogenic factor. Although expressed in many tissues, akirin1 is negatively regulated by myostatin specifically in skeletal muscle tissue. In this manuscript we have characterized the possible function of akirin1 in postnatal muscle growth. Molecular and immunohistological analyses indicated that while low levels of akirin1 are associated with quiescent satellite cells (SC), higher levels of akirin1 are detected in activated proliferating SC indicating that akirin1 could be associated with satellite cell activation. In addition to SC, macrophages also express akirin1, and increased expression of akirin1 resulted in more efficient chemotaxis of both macrophages and myoblasts. Akirin1 appears to regulate chemotaxis of both macrophages and myoblasts by reorganising actin cytoskeleton, leading to more efficient lamellipodia formation via a PI3 kinase dependent pathway. Expression analysis during muscle regeneration also indicated that akirin1 expression is detected very early (day 2) in regenerating muscle, and expression gradually peaks to coincide the nascent myotube formation stage of muscle regeneration. Based on these results we propose that akirin1 could be acting as a transducer of early signals of muscle regeneration. Thus, we speculate that myostatin regulates key steps of muscle regeneration including chemotaxis of inflammatory cells, SC activation and migration through akirin1.


Subject(s)
Chemotaxis/physiology , Muscle, Skeletal/physiology , Proteins/physiology , Regeneration/physiology , Animals , Cells, Cultured , Gene Expression Regulation , Macrophages/physiology , Mice , Mice, Knockout , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/growth & development , Myoblasts/physiology , Myostatin/genetics , Myostatin/metabolism , Nuclear Proteins , Repressor Proteins , Satellite Cells, Skeletal Muscle/physiology
4.
Dev Genes Evol ; 219(9-10): 497-508, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20052486

ABSTRACT

Myostatin (Mstn) is a negative regulator of skeletal muscle mass, and Mstn mutations are responsible for the double muscling phenotype observed in many animal species. Moreover, Mstn is a positive regulator of adult muscle stem cell (satellite cell) quiescence, and hence, Mstn is being targeted in therapeutic approaches to muscle diseases. In order to better understand the mechanisms underlying Mstn regulation, we searched for the gene's proximal enhancer and promoter elements, using an evolutionary approach. We identified a 260-bp-long, evolutionary conserved region upstream of tetrapod Mstn and teleost mstn b genes. This region contains binding sites for TATA binding protein, Meis1, NF-Y, and for CREB family members, suggesting the involvement of cAMP in Myostatin regulation. The conserved fragment was able to drive reporter gene expression in C2C12 cells in vitro and in chicken somites in vivo; both normally express Mstn. In contrast, the reporter construct remained silent in the avian neural tube that normally does not express Mstn. This suggests that the identified element serves as a minimal promoter, harboring some spatial specificity. Finally, using bioinformatic approaches, we identified additional genes in the human genome associated with sequences similar to the Mstn proximal promoter/enhancer. Among them are genes important for myogenesis. This suggests that Mstn and these genes may form a synexpression group, regulated by a common signaling pathway.


Subject(s)
Myostatin/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Binding Sites , Enhancer Elements, Genetic , Humans , Molecular Sequence Data , Transcription, Genetic
5.
Exp Cell Res ; 314(5): 1013-29, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18255059

ABSTRACT

Genetic analysis has revealed an important function in myogenesis for Myostatin, a member of the TGF-beta superfamily. However, the cascade of genes that responds to Myostatin signalling to regulate myogenesis is not well understood. Thus, a suppressive subtraction hybridization to identify such genes was undertaken and here we report the cloning and characterization of a novel gene, Mighty. Mighty is expressed in a variety of different tissues but appears to be specifically regulated by Myostatin in skeletal muscle. Overexpression of Mighty in C2C12 cells results in early withdrawal of myoblasts from the cell cycle, enhanced and accelerated differentiation and hypertrophy of myotubes. Most importantly, Mighty overexpression leads to increased and earlier expression of MyoD and increased secretion of another known differentiation inducing factor, IGF-II. Furthermore, viral expression of Mighty in mdx mice resulted in an increase in the number of larger healthy muscle fibers. Given its role in myogenesis, we propose that Mighty is a critical promyogenic factor which plays a key role in the signalling pathway downstream of Myostatin.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Muscle Development , Muscle Proteins/physiology , Muscle, Skeletal/growth & development , MyoD Protein/genetics , Myogenic Regulatory Factors/physiology , Animals , Cell Differentiation , Cloning, Molecular , Gene Expression Regulation , Mice , Muscle Fibers, Skeletal , Muscle Proteins/genetics , Myoblasts, Skeletal/cytology , Myogenic Regulatory Factors/genetics , Myostatin , Transforming Growth Factor beta/physiology
6.
Mol Ther ; 15(8): 1463-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17551508

ABSTRACT

A reduction in muscle mass and strength is often observed with aging, and this phenomenon is known as sarcopenia. This age-related atrophy frequently correlates with insufficient levels of muscle regeneration resulting from impairment of satellite cell involvement and myogenesis brought about by the aged environment. Using myostatin-null mice, we recently showed that negative regulators of muscle mass such as myostatin play an active role in the regulation of myogenesis during aging. The present study specifically tests the therapeutic value of a myostatin antagonist in sarcopenia. We report here that a short-term blockade of myostatin, through stage-specific administration of a myostatin antagonist, significantly enhanced muscle regeneration in aged mice after injury and during sarcopenia. Antagonism of myostatin led to satellite cell activation, increased Pax7 and MyoD protein levels, and greater myoblast and macrophage cell migration, resulting in enhanced muscle regeneration after notexin injury in aged mice. In addition, the antagonist demonstrated a high degree of efficacy, as only minimal doses during the critical period of regeneration after injury were sufficient to restore the myogenic and inflammatory responses in the aged environment. Thus, we propose that the antagonism of myostatin has significant therapeutic potential in the alleviation of sarcopenia.


Subject(s)
Aging/physiology , Muscles/physiology , Regeneration/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Movement , Cell Proliferation , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Muscles/injuries , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myostatin , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
7.
J Cell Physiol ; 209(3): 866-73, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16972257

ABSTRACT

Sarcopenia is a progressive age-related loss of skeletal muscle mass and strength. Parabiotic experiments show that circulating factors positively influence the proliferation and regenerative capacity of satellite cells in aged mice. In addition, we believe that negative regulators of muscle mass also serve to balance the signals that influence satellite cell activation and regeneration capacity with ageing. Myostatin, a negative regulator of pre- and postnatal myogenesis, inhibits satellite cell activation and muscle regeneration postnatally. To investigate the role of myostatin during age-related sarcopenia, we examined muscle mass and regeneration in young and old myostatin-null mice. Young myostatin-null muscle fibers were characterized by massive hypertrophy and hyperplasia and an increase in type IIB fibers, resulting in a more glycolytic muscle. With ageing, wild-type muscle became increasingly oxidative and fiber atrophy was prominent. In contrast no fiber type switching was observed and atrophy was minimal in aged myostatin-null muscle. The effect of ageing on satellite cell numbers appeared minimal, however, satellite cell activation declined significantly in both wild-type and myostatin-null muscles. In young mice, lack of myostatin resulted in increased satellite cell number and activation compared to wild-type, suggesting a greater propensity to undergo myogenesis, a difference maintained in the aged mice. In addition, muscle regeneration of myostatin-null muscle following notexin injury was accelerated and fiber hypertrophy and type were recovered with regeneration, unlike in wild-type muscle. In conclusion, a lack of myostatin appears to reduce age-related sarcopenia and loss of muscle regenerative capacity.


Subject(s)
Aging/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Muscular Atrophy/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Shape , Elapid Venoms/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/drug effects , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Myostatin , Regeneration/drug effects , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Transforming Growth Factor beta/genetics
8.
Am J Physiol Cell Physiol ; 287(4): C1031-40, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15189813

ABSTRACT

Myostatin is a negative regulator of muscle growth, and absence of the functional myostatin protein leads to the heavy muscle phenotype in both mouse and cattle. Although the role of myostatin in controlling muscle mass is established, little is known of the mechanisms regulating the expression of the myostatin gene. In this study, we have characterized the murine myostatin promoter in vivo. Various constructs of the murine myostatin promoter were injected into the quadriceps muscle of mice, and the reporter luciferase activity was analyzed. The results indicate that of the seven E-boxes present in the 2.5-kb fragment of the murine myostatin promoter, the E5 E-box plays an important role in the regulation of promoter activity in vivo. Furthermore, the in vitro studies demonstrated that MyoD preferentially binds and upregulates the murine myostatin promoter activity. We also analyzed the activity of the bovine and murine promoters in murine skeletal muscle and showed that, despite displaying comparable levels of activity in murine myoblast cultures, bovine myostatin promoter activity is much weaker than murine myostatin promoter in mice. Finally, we demonstrate that in vivo, the 2.5-kb region of the murine myostatin promoter is sufficient to drive the activity of the reporter gene in a fiber type-specific manner.


Subject(s)
Gene Expression Regulation , Muscle Fibers, Skeletal/physiology , Promoter Regions, Genetic , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Animals , Cattle , DNA Primers , E-Box Elements/genetics , Electrophoretic Mobility Shift Assay , Mice , Muscle, Skeletal/physiology , MyoD Protein/physiology , Myostatin , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...