PUBLICATION

The AP-1 transcription factor component Fosl2 potentiates the rate of myocardial differentiation from the zebrafish second heart field

Authors
Jahangiri, L., Sharpe, M., Novikov, N., González-Rosa, J.M., Borikova, A., Nevis, K., Paffett-Lugassy, N., Zhao, L., Adams, M., Guner-Ataman, B., Burns, C.E., Burns, C.G.
ID
ZDB-PUB-160107-8
Date
2016
Source
Development (Cambridge, England)   143: 113-22 (Journal)
Registered Authors
Burns (Erter), Caroline, Burns, Geoff, Paffett-Lugassy, Noelle, Zhao, Long
Keywords
AP-1, Cardiac development, Fosl2, Heart, Second heart field, Zebrafish
MeSH Terms
  • Amino Acid Sequence
  • Animals
  • Animals, Genetically Modified
  • Cell Differentiation/physiology*
  • Cell Proliferation/genetics
  • Fos-Related Antigen-2/biosynthesis
  • Fos-Related Antigen-2/genetics
  • Fos-Related Antigen-2/metabolism*
  • Gene Expression Regulation, Developmental
  • Gene Knockout Techniques
  • Heart/embryology*
  • Heart Defects, Congenital/genetics
  • Myocardium/cytology
  • Myocytes, Cardiac/cytology*
  • Sequence Analysis, Protein
  • Transcription Factor AP-1/metabolism*
  • Zebrafish/embryology*
  • Zebrafish Proteins/genetics
  • Zebrafish Proteins/metabolism
PubMed
26732840 Full text @ Development
Abstract
The vertebrate heart forms through successive phases of cardiomyocyte differentiation. Initially, cardiomyocytes derived from first heart field (FHF) progenitors assemble the linear heart tube. Thereafter, second heart field (SHF) progenitors differentiate into cardiomyocytes that are accreted to the poles of the heart tube over a well-defined developmental window. Although heart tube elongation deficiencies lead to life-threatening congenital heart defects, the variables controlling the initiation, rate and duration of myocardial accretion remain obscure. Here, we demonstrate that the AP-1 transcription factor, Fos-like antigen 2 (Fosl2), potentiates the rate of myocardial accretion from the zebrafish SHF. fosl2 mutants initiate accretion appropriately, but cardiomyocyte production is sluggish, resulting in a ventricular deficit coupled with an accumulation of SHF progenitors. Surprisingly, mutant embryos eventually correct the myocardial deficit by extending the accretion window. Overexpression of Fosl2 also compromises production of SHF-derived ventricular cardiomyocytes, a phenotype that is consistent with precocious depletion of the progenitor pool. Our data implicate Fosl2 in promoting the progenitor to cardiomyocyte transition and uncover the existence of regulatory mechanisms to ensure appropriate SHF-mediated cardiomyocyte contribution irrespective of embryonic stage.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping