Genomics

Dataset Information

0

Endothelial Cell Cycle State Determines Propensity for Arterial-Venous Fate


ABSTRACT: Formation and maturation of a functional blood vascular system is required for the development and maintenance of all tissues in the body. During the process of blood vessel development, primordial endothelial cells are formed and 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. Specification of arterial and venous endothelial cells occurs in conjunction with suppression of endothelial cell cycle progression, and endothelial cell hyperproliferation is associated with potentially lethal arterial-venous malformations. However, the mechanistic role that cell cycle state plays in arterial-venous specification is unknown. Herein, studying vascular development in Cdh5-CreERT2;R26FUCCI2aR reporter mice, we find that venous and arterial endothelial cells exhibit a propensity for different cell cycle states during development and in adulthood. That is, venous endothelial cells are predominantly FUCCI-Negative, while arterial endothelial cells are enriched for the FUCCI-Red reporter. Single cell RNA sequencing analysis of developing retinal endothelial cells reveals that venous endothelial cells are enriched for the FUCCI-Negative state and BMP signaling, while arterial endothelial cells are enriched for the FUCCI-Red state and TGF-b signaling. Further transcriptional analyses and live imaging of cultured endothelial cells expressing the FUCCI reporter show that reporter-negative corresponds to an early G1 state and reporter-red corresponds to late G1 state. We find the early G1 state is essential for BMP4-induced venous gene expression, whereas late G1 state is essential for TGF-b1-induced arterial gene expression. In a mouse model of endothelial cell hyperproliferation and disrupted arterial-venous specification, pharmacological inhibition of endothelial cell cycle prevents the vascular defects. Collectively, our results show that endothelial cell cycle control plays a key role in arterial-venous network formation, and distinct cell cycle states provide distinct windows of opportunity for the molecular induction of arterial vs. venous specification.

ORGANISM(S): Mus musculus Homo sapiens

PROVIDER: GSE211658 | GEO | 2022/08/22

REPOSITORIES: GEO

Similar Datasets

2022-08-28 | GSE211958 | GEO
2022-08-22 | GSE169039 | GEO
2022-10-12 | PXD036326 | Pride
2023-12-31 | GSE245197 | GEO
2014-01-09 | GSE34940 | GEO
2014-01-09 | E-GEOD-34940 | biostudies-arrayexpress
2016-07-03 | E-GEOD-66073 | biostudies-arrayexpress
2016-01-01 | GSE66073 | GEO
2011-01-11 | E-MTAB-494 | biostudies-arrayexpress
2024-01-25 | GSE247450 | GEO