PUBLICATION

Rasip1 regulates vertebrate vascular endothelial junction stability through Epac1-Rap1 signaling

Authors
Wilson, C.W., Parker, L.H., Hall, C.J., Smyczek, T., Mak, J., Crow, A., Posthuma, G., De Mazière, A., Sagolla, M., Chalouni, C., Vitorino, P., Roose-Girma, M., Warming, S., Klumperman, J., Crosier, P.S., and Ye, W.
ID
ZDB-PUB-130807-5
Date
2013
Source
Blood   122(22): 3678-90 (Journal)
Registered Authors
Crosier, Phil, Hall, Chris, Parker, Leon
Keywords
none
MeSH Terms
  • Actins/metabolism
  • Animals
  • Animals, Genetically Modified
  • Carrier Proteins/antagonists & inhibitors
  • Carrier Proteins/genetics
  • Carrier Proteins/physiology*
  • Endothelium, Vascular/embryology*
  • Endothelium, Vascular/physiology*
  • Female
  • Guanine Nucleotide Exchange Factors/metabolism*
  • Human Umbilical Vein Endothelial Cells
  • Humans
  • Intercellular Junctions/physiology
  • Mice
  • Mice, Knockout
  • Monomeric GTP-Binding Proteins/metabolism
  • Neovascularization, Physiologic
  • Pregnancy
  • RNA Interference
  • Signal Transduction
  • Zebrafish
  • Zebrafish Proteins/antagonists & inhibitors
  • Zebrafish Proteins/genetics
  • Zebrafish Proteins/metabolism
  • Zebrafish Proteins/physiology
  • rap1 GTP-Binding Proteins/metabolism*
PubMed
23886837 Full text @ Blood
Abstract

Establishment and stabilization of endothelial tubes with patent lumens is vital during vertebrate development. Ras-interacting protein 1 (RASIP1) has been described as an essential regulator of de novo lumenogenesis through modulation of endothelial cell (EC) adhesion to extracellular matrix (ECM). Here, we show that in mouse and zebrafish embryos, Rasip1-deficient vessels transition from an angioblast cord to a hollow tube, permit circulation of primitive erythrocytes, but ultimately collapse, leading to hemorrhage and embryonic lethality. Knockdown of RASIP1 does not alter EC-ECM adhesion, but causes cell-cell detachment and increases permeability of EC monolayers in vitro. We also found that endogenous RASIP1 in ECs binds RAP1, but not RHOA or CDC42. Using an EPAC1-RAP1 dependent model of nascent junction formation, we demonstrate that a fraction of the RASIP1 protein pool localizes to cell-cell contacts. Loss of RASIP1 phenocopies loss of RAP1 or EPAC1 in ECs by altering junctional actin organization, localization of the actin-bundling protein non-muscle myosin heavy chain IIB, and junction remodeling. Our data show that RASIP1 regulates the integrity of newly formed blood vessels as an effector of EPAC1-RAP1 signaling.

Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping