[HTML][HTML] Endothelial cell cycle state determines propensity for arterial-venous fate

NW Chavkin, G Genet, M Poulet, ED Jeffery… - Nature …, 2022 - nature.com
NW Chavkin, G Genet, M Poulet, ED Jeffery, C Marziano, N Genet, H Vasavada, EA Nelson…
Nature communications, 2022nature.com
During blood vessel development, endothelial cells become specified toward arterial or
venous fates to generate a circulatory network that provides nutrients and oxygen to, and
removes metabolic waste from, all tissues. Arterial-venous specification occurs in
conjunction with suppression of endothelial cell cycle progression; however, the mechanistic
role of cell cycle state is unknown. Herein, using Cdh5-CreERT2; R26FUCCI2aR reporter
mice, we find that venous endothelial cells are enriched for the FUCCI-Negative state (early …
Abstract
During blood vessel development, endothelial cells become specified toward arterial or venous fates to generate a circulatory network that provides nutrients and oxygen to, and removes metabolic waste from, all tissues. Arterial-venous specification occurs in conjunction with suppression of endothelial cell cycle progression; however, the mechanistic role of cell cycle state is unknown. Herein, using Cdh5-CreERT2;R26FUCCI2aR reporter mice, we find that venous endothelial cells are enriched for the FUCCI-Negative state (early G1) and BMP signaling, while arterial endothelial cells are enriched for the FUCCI-Red state (late G1) and TGF-β signaling. Furthermore, early G1 state is essential for BMP4-induced venous gene expression, whereas late G1 state is essential for TGF-β1-induced arterial gene expression. Pharmacologically induced cell cycle arrest prevents arterial-venous specification defects in mice with endothelial hyperproliferation. Collectively, our results show that distinct endothelial cell cycle states provide distinct windows of opportunity for the molecular induction of arterial vs. venous fate.
nature.com