Abstract

Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer. In human umbilical vein endothelial cells (HUVECs), knockdown of protein kinase D-1 (PKD1) or PKD2 down-regulates VEGFR-2 expression and inhibits VEGF-induced cell proliferation and migration. However, how PKD regulates VEGF signaling is unclear. Previous bioinformatics analyses have identified binding sites for the transcription factor activating enhancer-binding protein 2 (AP2) in the VEGFR-2 promoter. Using ChIP analyses, here we found that PKD knockdown in HUVECs increases binding of AP2β to the VEGFR-2 promoter. Luciferase reporter assays with serial deletions of AP2-binding sites within the VEGFR-2 promoter revealed that its transcriptional activity negatively correlates with the number of these sites. Next we demonstrated that AP2β up-regulation decreases VEGFR-2 expression and that loss of AP2β enhances VEGFR-2 expression in HUVECs. In vivo experiments confirmed increased VEGFR-2 immunostaining in the spinal cord of AP2β knockout mouse embryos. Mechanistically, we observed that PKD phosphorylates AP2β at Ser258 and Ser277 and suppresses its nuclear accumulation. Inhibition of PKD activity with a pan-PKD inhibitor increased AP2β nuclear localization, and overexpression of both WT and constitutively active PKD1 or PKD2 reduced AP2β nuclear localization through a Ser258- and Ser277-dependent mechanism. Furthermore, substitution of Ser277 in AP2β increased its binding to the VEGFR-2 promoter. Our findings uncover evidence of a molecular pathway that regulates VEGFR-2 expression, insights that may shed light on the etiology of diseases associated with aberrant VEGF/VEGFR signaling.

Highlights

  • Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer

  • PKD2 in human umbilical vein endothelial cells (HUVECs) increased the expression of VEGFR-2, among which protein kinase D-1 (PKD1).CA exhibited the greatest effect on VEGFR-2 up-regulation (Fig. 1D)

  • These results suggest that endogenous PKD1 and PKD2 control expression of VEGFR-2 and positively regulate the proliferation and migration of endothelial cells as well as angiogenesis in vivo

Read more

Summary

Results

PKD regulates VEGFR-2 expression, proliferation, and migration of endothelial cells and angiogenesis. Our results showed that inhibition of PKD activity with CRT suppressed VEGF-induced angiogenesis in vivo Taken together, these results suggest that endogenous PKD1 and PKD2 control expression of VEGFR-2 and positively regulate the proliferation and migration of endothelial cells as well as angiogenesis in vivo. Increased AP2␤ bound the VEGFR-2 promoter upon PKD1 knockdown, and this effect was most pronounced in PKD2-ablated HUVECs, as evidenced by the CHIP assay (Fig. 2A). Overexpression of PKD1.CA in HUVECs was shown to decrease the nuclear AP2␤ protein level (Fig. 4I), suggesting that active PKD promotes cytoplasmic retention of AP2␤. AP2␤ with a Ser277 substitution showed significantly increased binding to the VEGFR-2 promoter (Fig. 4J) These results indicate that PKD-mediated phosphorylation of AP2␤, especially Ser277, controls the nuclear localization and DNA binding of AP2␤

Discussion
Cells and reagents
Proliferation assay
ChIP assay
Matrigel plug assay
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call