GONUTS has been updated to MW1.31 Most things seem to be working but be sure to report problems.

Have any questions? Please email us at ecoliwiki@gmail.com

TableEdit

Jump to: navigation, search

PMID:23752591

You don't have sufficient rights on this wiki to edit tables. Perhaps you need to log in. Changes you make in the Table editor will not be saved back to the wiki

See Help for Help on this wiki. See the documentation for how to use the table editor

Citation

Proserpio, V, Fittipaldi, R, Ryall, JG, Sartorelli, V and Caretti, G (2013) The methyltransferase SMYD3 mediates the recruitment of transcriptional cofactors at the myostatin and c-Met genes and regulates skeletal muscle atrophy. Genes Dev. 27:1299-312

Abstract

Elucidating the epigenetic mechanisms underlying muscle mass determination and skeletal muscle wasting holds the potential of identifying molecular pathways that constitute possible drug targets. Here, we report that the methyltransferase SMYD3 modulates myostatin and c-Met transcription in primary skeletal muscle cells and C2C12 myogenic cells. SMYD3 targets the myostatin and c-Met genes and participates in the recruitment of the bromodomain protein BRD4 to their regulatory regions through protein-protein interaction. By recruiting BRD4, SMYD3 favors chromatin engagement of the pause-release factor p-TEFb (positive transcription elongation factor) and elongation of Ser2-phosphorylated RNA polymerase II (PolIISer2P). Reducing SMYD3 decreases myostatin and c-Met transcription, thus protecting from glucocorticoid-induced myotube atrophy. Supporting functional relevance of the SMYD3/BRD4 interaction, BRD4 pharmacological blockade by the small molecule JQ1 prevents dexamethasone-induced myostatin and atrogene up-regulation and spares myotube atrophy. Importantly, in a mouse model of dexamethasone-induced skeletal muscle atrophy, SMYD3 depletion prevents muscle loss and fiber size decrease. These findings reveal a mechanistic link between SMYD3/BRD4-dependent transcriptional regulation, muscle mass determination, and skeletal muscle atrophy and further encourage testing of small molecules targeting specific epigenetic regulators in animal models of muscle wasting.

Links

PubMed PMC3690402 Online version:10.1101/gad.217240.113

Keywords

Animals; Cell Line; Cyclin-Dependent Kinase 9/metabolism; Dexamethasone/pharmacology; Histone-Lysine N-Methyltransferase/chemistry; Histone-Lysine N-Methyltransferase/deficiency; Histone-Lysine N-Methyltransferase/genetics; Histone-Lysine N-Methyltransferase/metabolism; Mice; Muscle Fibers, Skeletal/drug effects; Muscle Fibers, Skeletal/pathology; Muscle Proteins/genetics; Muscle, Skeletal/drug effects; Muscle, Skeletal/metabolism; Muscle, Skeletal/pathology; Muscular Atrophy/chemically induced; Muscular Atrophy/genetics; Myostatin/genetics; Nuclear Proteins/chemistry; Nuclear Proteins/metabolism; Phosphorylation; Phosphoserine/metabolism; Positive Transcriptional Elongation Factor B/metabolism; Protein Binding; Proto-Oncogene Proteins c-met/genetics; RNA Polymerase II/chemistry; RNA Polymerase II/metabolism; SKP Cullin F-Box Protein Ligases/genetics; Transcription Factors/chemistry; Transcription Factors/metabolism; Transcription, Genetic

public



Cancel