Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(24): 17654-62, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23637228

ABSTRACT

Sumoylation is an important post-translational modification that alters the activity of many transcription factors. However, the mechanisms that link sumoylation to alterations in chromatin structure, which culminate in tissue specific gene expression, are not fully understood. In this study, we demonstrate that SUMO modification of the transcription factor Sharp-1 is required for its full transcriptional repression activity and function as an inhibitor of skeletal muscle differentiation. Sharp-1 is modified by sumoylation at two conserved lysine residues 240 and 255. Mutation of these SUMO acceptor sites in Sharp-1 does not impact its subcellular localization but attenuates its ability to act as a transcriptional repressor and inhibit myogenic differentiation. Consistently, co-expression of the SUMO protease SENP1 with wild type Sharp-1 abrogates Sharp-1-dependent inhibition of myogenesis. Interestingly, sumoylation acts as a signal for recruitment of the co-repressor G9a. Thus, enrichment of G9a, and histone H3 lysine 9 dimethylation (H3K9me2), a signature of G9a activity, is dramatically reduced at muscle promoters in cells expressing sumoylation-defective Sharp-1. Our findings demonstrate how sumoylation of Sharp-1 exerts an impact on chromatin structure and transcriptional repression of muscle gene expression through recruitment of G9a.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Sumoylation , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Conserved Sequence , Cysteine Endopeptidases , Endopeptidases/metabolism , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Muscle Development , MyoD Protein/metabolism , Protein Binding , Protein Transport , Transcription, Genetic
2.
Subcell Biochem ; 61: 139-50, 2013.
Article in English | MEDLINE | ID: mdl-23150250

ABSTRACT

Skeletal muscle cells have served as a paradigm for understanding mechanisms leading to cellular differentiation. Formation of skeletal muscle involves a series of steps in which cells are committed towards the myogenic lineage, undergo expansion to give rise to myoblasts that differentiate into multinucleated myotubes, and mature to form adult muscle fibers. The commitment, proliferation, and differentiation of progenitor cells involve both genetic and epigenetic changes that culminate in alterations in gene expression. Members of the Myogenic regulatory factor (MRF), as well as the Myocyte Enhancer Factor (MEF2) families control distinct steps of skeletal muscle proliferation and differentiation. In addition, -growing evidence indicates that chromatin modifying enzymes and remodeling complexes epigenetically reprogram muscle promoters at various stages that preclude or promote MRF and MEF2 activites. Among these, histone deacetylases (HDACs), histone acetyltransferases (HATs), histone methyltransferases (HMTs) and SWI/SNF complexes alter chromatin structure through post-translational modifications to impact MRF and MEF2 activities. With such new and emerging knowledge, we are beginning to develop a true molecular understanding of the mechanisms by which skeletal muscle development and differentiation is regulated. Elucidation of the mechanisms by which epigenetic regulators control myogenesis will likely provide a new foundation for the development of novel therapeutic drugs for muscle dystrophies, ageing-related regeneration defects that occur due to altered proliferation and differentiation, and other malignancies.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Muscle Development/genetics , Muscle, Skeletal/growth & development , Myoblasts, Skeletal/physiology , Animals , Cell Differentiation/drug effects , Cell Lineage/genetics , Cell Proliferation , Chromatin Assembly and Disassembly , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Histone Deacetylase Inhibitors/therapeutic use , Humans , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophies/drug therapy , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
3.
Mol Biol Cell ; 23(24): 4778-85, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23087213

ABSTRACT

Sharp-1, a basic helix-loop-helix transcription factor, is a potent repressor of skeletal muscle differentiation and is dysregulated in muscle pathologies. However, the mechanisms by which it inhibits myogenesis are not fully understood. Here we show that G9a, a lysine methyltransferase, is involved in Sharp-1-mediated inhibition of muscle differentiation. We demonstrate that G9a directly interacts with Sharp-1 and enhances its ability to transcriptionally repress the myogenin promoter. Concomitant with a differentiation block, G9a-dependent histone H3 lysine 9 dimethylation (H3K9me2) and MyoD methylation are apparent upon Sharp-1 overexpression in muscle cells. RNA interference-mediated reduction of G9a or pharmacological inhibition of its activity erases these repressive marks and rescues the differentiation defect imposed by Sharp-1. Our findings provide new insights into Sharp-1-dependent regulation of myogenesis and identify epigenetic mechanisms that could be targeted in myopathies characterized by elevated Sharp-1 levels.


Subject(s)
Cell Differentiation , Histone-Lysine N-Methyltransferase/metabolism , Muscle, Skeletal/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line , Gene Expression Regulation , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Lysine/metabolism , Methylation , Mice , Microscopy, Fluorescence , Muscle, Skeletal/cytology , Mutation , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Transcription Factors/genetics
4.
PLoS One ; 7(8): e43137, 2012.
Article in English | MEDLINE | ID: mdl-22905217

ABSTRACT

Stra13, a basic helix-loop-helix (bHLH) transcription factor is involved in myriad biological functions including cellular growth arrest, differentiation and senescence. However, the mechanisms by which its transcriptional activity and function are regulated remain unclear. In this study, we provide evidence that post-translational modification of Stra13 by Small Ubiquitin-like Modifier (SUMO) dramatically potentiates its ability to transcriptionally repress cyclin D1 and mediate G(1) cell cycle arrest in fibroblast cells. Mutation of SUMO acceptor lysines 159 and 279 located in the C-terminal repression domain has no impact on nuclear localization; however, it abrogates association with the co-repressor histone deacetylase 1 (HDAC1), attenuates repression of cyclin D1, and prevents Stra13-mediated growth suppression. HDAC1, which promotes cellular proliferation and cell cycle progression, antagonizes Stra13 sumoylation-dependent growth arrest. Our results uncover an unidentified regulatory axis between Stra13 and HDAC1 in progression through the G(1)/S phase of the cell cycle, and provide new mechanistic insights into regulation of Stra13-mediated transcriptional repression by sumoylation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclin D1/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , SUMO-1 Protein/metabolism , Animals , COS Cells , Cell Cycle , Cell Survival , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Molecular Chaperones/metabolism , Mutation , NIH 3T3 Cells , Protein Inhibitors of Activated STAT/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary
5.
Proc Natl Acad Sci U S A ; 109(3): 841-6, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22215600

ABSTRACT

Skeletal muscle cells have served as a paradigm for understanding mechanisms leading to cellular differentiation. The proliferation and differentiation of muscle precursor cells require the concerted activity of myogenic regulatory factors including MyoD. In addition, chromatin modifiers mediate dynamic modifications of histone tails that are vital to reprogramming cells toward terminal differentiation. Here, we provide evidence for a unique dimension to epigenetic regulation of skeletal myogenesis. We demonstrate that the lysine methyltransferase G9a is dynamically expressed in myoblasts and impedes differentiation in a methyltransferase activity-dependent manner. In addition to mediating histone H3 lysine-9 di-methylation (H3K9me2) on MyoD target promoters, endogenous G9a interacts with MyoD in precursor cells and directly methylates it at lysine 104 (K104) to constrain its transcriptional activity. Mutation of K104 renders MyoD refractory to inhibition by G9a and enhances its myogenic activity. Interestingly, MyoD methylation is critical for G9a-mediated inhibition of myogenesis. These findings provide evidence of an unanticipated role for methyltransferases in cellular differentiation states by direct posttranslational modification of a transcription factor.


Subject(s)
Cell Differentiation , Histone-Lysine N-Methyltransferase/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , MyoD Protein/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Lysine/metabolism , Methylation , Mice , Molecular Sequence Data , Muscle Development , MyoD Protein/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...