PUBLICATION

The receptor protein-tyrosine phosphatase, Dep1, acts in arterial/venous cell fate decisions in zebrafish development

Authors
Rodriguez, F., Vacaru, A., Overvoorde, J., and den Hertog, J.
ID
ZDB-PUB-081008-14
Date
2008
Source
Developmental Biology   324(1): 122-130 (Journal)
Registered Authors
den Hertog, Jeroen, Overvoorde, John, Rodriguez, Fiona
Keywords
Dep1, protein-tyrosine phosphatase, arterial, venous, cell specification, zebrafish, morpholino
MeSH Terms
  • Animals
  • Arteries/cytology
  • Arteries/embryology*
  • Basic Helix-Loop-Helix Transcription Factors/metabolism
  • Cell Differentiation/physiology
  • Embryo, Nonmammalian/physiology
  • Endothelial Cells/cytology*
  • Endothelial Cells/metabolism
  • Endothelium, Vascular/cytology
  • Phosphatidylinositol 3-Kinases/metabolism
  • Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism*
  • Receptors, Notch/metabolism
  • Signal Transduction/physiology
  • Veins/cytology
  • Veins/embryology*
  • Zebrafish/embryology*
  • Zebrafish/metabolism
  • Zebrafish Proteins/metabolism
PubMed
18835554 Full text @ Dev. Biol.
Abstract
Dep1 is a transmembrane protein-tyrosine phosphatase (PTP) that is expressed in vascular endothelial cells and has tumor suppressor activity. Mouse models with gene targeted Dep1 either show vascular defects, or do not show any defects at all. We used the zebrafish to investigate the role of Dep1 in early development. The zebrafish genome encodes two highly homologous Dep1 genes, Dep1a and Dep1b. Morpholinos specific for Dep1a and Dep1b induced defects in vasculature, resulting in defective blood circulation. However, Green Fluorescent Protein expression in fli1a::gfp1 transgenic embryos and cdh5 expression, markers of vascular endothelial cells, were normal upon Dep1a- and Dep1b-MO injection. Molecular markers indicated that arterial specification was reduced and venous markers were expanded in Dep1 morphants. Moreover, the Dep1a/Dep1b knockdowns were rescued by inhibition of Phosphatidylinositol-3 kinase (PI3K) and by expression of active Notch and Grl/Hey2. Our results suggest a model in which Dep1 acts upstream in a signaling pathway inhibiting PI3K, resulting in expression of Notch and Grl, thus regulating arterial specification in development.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping