PUBLICATION

Dephosphorylation of HDAC4 by PP2A-Bδ unravels a new role for the HDAC4/MEF2 axis in myoblast fusion

Authors
Veloso, A., Martin, M., Bruyr, J., O'Grady, T., Deroanne, C., Mottet, D., Twizere, J.C., Cherrier, T., Dequiedt, F.
ID
ZDB-PUB-190706-6
Date
2019
Source
Cell Death & Disease   10: 512 (Journal)
Registered Authors
Keywords
none
MeSH Terms
  • Animals
  • Cell Fusion
  • Cell Line
  • Cytoskeleton/metabolism
  • Embryo, Nonmammalian/metabolism
  • Histone Deacetylases/metabolism*
  • Holoenzymes/metabolism
  • MEF2 Transcription Factors/metabolism*
  • Mice
  • Morphogenesis
  • Muscle Development
  • Muscle Fibers, Skeletal/cytology
  • Myoblasts/cytology*
  • Myoblasts/metabolism*
  • Phenotype
  • Phosphorylation
  • RNA, Messenger/genetics
  • RNA, Messenger/metabolism
  • Signal Transduction
  • Transcription, Genetic
  • Zebrafish/embryology
  • rac1 GTP-Binding Protein/metabolism
PubMed
31273193 Full text @ Cell Death Dis.
Abstract
Muscle formation is controlled by a number of key myogenic transcriptional regulators that govern stage-specific gene expression programs and act as terminal effectors of intracellular signaling pathways. To date, the role of phosphatases in the signaling cascades instructing muscle development remains poorly understood. Here, we show that a specific PP2A-B55δ holoenzyme is necessary for skeletal myogenesis. The primary role of PP2A-B55δ is to dephosphorylate histone deacetylase 4 (HDAC4) following myocyte differentiation and ensure repression of Myocyte enhancer factor 2D (MEF2D)-dependent gene expression programs during myogenic fusion. As a crucial HDAC4/MEF2D target gene that governs myocyte fusion, we identify ArgBP2, an upstream inhibitor of Abl, which itself is a repressor of CrkII signaling. Consequently, cells lacking PP2A-B55δ show upregulation of ArgBP2 and hyperactivation of CrkII downstream effectors, including Rac1 and FAK, precluding cytoskeletal and membrane rearrangements associated with myoblast fusion. Both in vitro and in zebrafish, loss-of-function of PP2A-B55δ severely impairs fusion of myocytes and formation of multinucleated muscle fibers, without affecting myoblast differentiation. Taken together, our results establish PP2A-B55δ as the first protein phosphatase to be involved in myoblast fusion and suggest that reversible phosphorylation of HDAC4 may coordinate differentiation and fusion events during myogenesis.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping