Distinct roles of HIF1A in endothelial adaptations to physiological and ambient oxygen

作者:Jiang, Yi-Zhou; Li, Yan; Wang, Kai; Dai, Cai-Feng; Huang, Shi-An; Chen, Dong-Bao; Zheng, Jing*
来源:Molecular and Cellular Endocrinology, 2014, 391(1-2): 60-67.
DOI:10.1016/j.mce.2014.04.008

摘要

Fetoplacental endothelial cells reside under physiological normoxic conditions (similar to 2-8% O-2) in vivo. Under such conditions, cells are believed to sense O-2 changes primarily via hypoxia inducible factor 1 alpha (HIF1A). However, little is known regarding the role of HIF1A in fetoplacental endothelial function under physiological normoxia. We recently reported that physiological chronic normoxia (PCN; 20-25 day, 3% O-2) enhanced FGF2- and VEGFA-stimulated proliferation and migration of human umbilical vein endothelial cells (HUVECs) via the MEK/ERK1/2 and PI3K/AKT1 pathways compared to standard cell culture normoxia (SCN; ambient O-2: similar to 21% O-2). Here, we investigated the action of HIF1A in regulating these cellular responses in HUVECs. HIF1A adenovirus infection in SCN-cells increased HIF1A protein expression, enhanced FGF2- and VEGFA-stimulated cell proliferation by 2.4 and 2.0-fold respectively, and promoted VEGFA-stimulated cell migration by 1.4-fold. HIF1A adenovirus infection in SCN-cells did not affect either basal or FGF2- and VEGFA-induced ERK1/2 activation, but it decreased basal AKT1 phosphorylation. Interestingly, HIF1A knockdown in PCN-cells via specific HIF1A siRNA transfection did not alter FGF2- and VEGFA-stimulated cell proliferation and migration, or ERK1/2 activation; however, it inhibited FGF2-induced ion activation by similar to 50%. These data indicate that HIF1A differentially regulates cell proliferation and migration, and ERK1/2 and AKT1 activation in PCN- and SCN-HUVECs. These data also suggest that HIF1A critically regulates cell proliferation and migration in SCN-, but not in PCN-HUVECs.