PUBLICATION

Temporally distinct transcriptional regulation of myocyte dedifferentiation and Myofiber growth during muscle regeneration

Authors
Louie, K.W., Saera-Vila, A., Kish, P.E., Colacino, J.A., Kahana, A.
ID
ZDB-PUB-171111-9
Date
2017
Source
BMC Genomics   18: 854 (Journal)
Registered Authors
Kahana, Alon, Kish, Phillip
Keywords
Cell reprogramming, Chromatin, Epigenetic, Fibronectin, Polycomb, RNA-sequencing, Stem cell, Transcriptome, Zebrafish
Datasets
GEO:GSE92489
MeSH Terms
  • Animals
  • Cell Dedifferentiation/genetics*
  • Gene Expression Profiling*
  • Gene Ontology
  • Muscle Cells/cytology*
  • Muscle Development/genetics*
  • Muscles/physiology*
  • Regeneration/genetics*
  • Time Factors
  • Transcription, Genetic*
  • Zebrafish
PubMed
29121865 Full text @ BMC Genomics
Abstract
Tissue regeneration requires a series of steps, beginning with generation of the necessary cell mass, followed by cell migration into damaged area, and ending with differentiation and integration with surrounding tissues. Temporal regulation of these steps lies at the heart of the regenerative process, yet its basis is not well understood. The ability of zebrafish to dedifferentiate mature "post-mitotic" myocytes into proliferating myoblasts that in turn regenerate lost muscle tissue provides an opportunity to probe the molecular mechanisms of regeneration.
Following subtotal excision of adult zebrafish lateral rectus muscle, dedifferentiating residual myocytes were collected at two time points prior to cell cycle reentry and compared to uninjured muscles using RNA-seq. Functional annotation (GAGE or K-means clustering followed by GO enrichment) revealed a coordinated response encompassing epigenetic regulation of transcription, RNA processing, and DNA replication and repair, along with protein degradation and translation that would rewire the cellular proteome and metabolome. Selected candidate genes were phenotypically validated in vivo by morpholino knockdown. Rapidly induced gene products, such as the Polycomb group factors Ezh2 and Suz12a, were necessary for both efficient dedifferentiation (i.e. cell reprogramming leading to cell cycle reentry) and complete anatomic regeneration. In contrast, the late activated gene fibronectin was important for efficient anatomic muscle regeneration but not for the early step of myocyte cell cycle reentry.
Reprogramming of a "post-mitotic" myocyte into a dedifferentiated myoblast requires a complex coordinated effort that reshapes the cellular proteome and rewires metabolic pathways mediated by heritable yet nuanced epigenetic alterations and molecular switches, including transcription factors and non-coding RNAs. Our studies show that temporal regulation of gene expression is programmatically linked to distinct steps in the regeneration process, with immediate early expression driving dedifferentiation and reprogramming, and later expression facilitating anatomical regeneration.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping