[HTML][HTML] Arhgef15 promotes retinal angiogenesis by mediating VEGF-induced Cdc42 activation and potentiating RhoJ inactivation in endothelial cells

S Kusuhara, Y Fukushima, S Fukuhara, LM Jakt… - 2012 - journals.plos.org
S Kusuhara, Y Fukushima, S Fukuhara, LM Jakt, M Okada, Y Shimizu, M Hata, K Nishida…
2012journals.plos.org
Background Drugs inhibiting vascular endothelial growth factor (VEGF) signaling are
globally administered to suppress deregulated angiogenesis in a variety of eye diseases.
However, anti-VEGF therapy potentially affects the normal functions of retinal neurons and
glias which constitutively express VEGF receptor 2. Thus, it is desirable to identify novel drug
targets which are exclusively expressed in endothelial cells (ECs). Here we attempted to
identify an EC-specific Rho guanine nucleotide exchange factor (GEF) and evaluate its role …
Background
Drugs inhibiting vascular endothelial growth factor (VEGF) signaling are globally administered to suppress deregulated angiogenesis in a variety of eye diseases. However, anti-VEGF therapy potentially affects the normal functions of retinal neurons and glias which constitutively express VEGF receptor 2. Thus, it is desirable to identify novel drug targets which are exclusively expressed in endothelial cells (ECs). Here we attempted to identify an EC-specific Rho guanine nucleotide exchange factor (GEF) and evaluate its role in retinal angiogenesis.
Methodology/Principal Findings
By exploiting fluorescence-activated cell sorting and microarray analyses in conjunction with in silico bioinformatics analyses, we comprehensively identified endothelial genes in angiogenic retinal vessels of postnatal mice. Of 9 RhoGEFs which were highly expressed in retinal ECs, we show that Arhgef15 acted as an EC-specific GEF to mediate VEGF-induced Cdc42 activation and potentiated RhoJ inactivation, thereby promoting actin polymerization and cell motility. Disruption of the Arhgef15 gene led to delayed extension of vascular networks and subsequent reduction of total vessel areas in postnatal mouse retinas.
Conclusions/Significance
Our study provides information useful to the development of new means of selectively manipulating angiogenesis without affecting homeostasis in un-targeted tissues; not only in eyes but also in various disease settings such as cancer.
PLOS