Citation

  • Authors: Chen, X. L., Nam, J. O., Jean, C., Lawson, C., Walsh, C. T., Goka, E., Lim, S. T., Tomar, A., Tancioni, I., Uryu, S., Guan, J. L., Acevedo, L. M., Weis, S. M., Cheresh, D. A., Schlaepfer, D. D.
  • Year: 2012
  • Journal: Dev Cell 22 146-57
  • Applications: in vitro / DNA / jetPEI-HUVEC
  • Cell type: HUVEC
    Description: Human umbilical vein endothelial cells

Abstract

Endothelial cells (ECs) form cell-cell adhesive junctional structures maintaining vascular integrity. This barrier is dynamically regulated by vascular endothelial growth factor (VEGF) receptor signaling. We created an inducible knockin mouse model to study the contribution of the integrin-associated focal adhesion tyrosine kinase (FAK) signaling on vascular function. Here we show that genetic or pharmacological FAK inhibition in ECs prevents VEGF-stimulated permeability downstream of VEGF receptor or Src tyrosine kinase activation in vivo. VEGF promotes tension-independent FAK activation, rapid FAK localization to cell-cell junctions, binding of the FAK FERM domain to the vascular endothelial cadherin (VE-cadherin) cytoplasmic tail, and direct FAK phosphorylation of beta-catenin at tyrosine-142 (Y142) facilitating VE-cadherin-beta-catenin dissociation and EC junctional breakdown. Kinase inhibited FAK is in a closed conformation that prevents VE-cadherin association and limits VEGF-stimulated beta-catenin Y142 phosphorylation. Our studies establish a role for FAK as an essential signaling switch within ECs regulating adherens junction dynamics.

Go to