Programming to S1PR1 + Endothelial Cells Promotes Restoration of Vascular Integrity

Author:

Akhter Md Zahid1,Chandra Joshi Jagdish1ORCID,Balaji Ragunathrao Vijay Avin1ORCID,Maienschein-Cline Mark2ORCID,Proia Richard L.3,Malik Asrar B.1ORCID,Mehta Dolly1ORCID

Affiliation:

1. Pharmacology and Regenerative Medicine and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago (M.Z.A., J.C.J., V.A.B.R., A.B.M., D.M.).

2. Research Informatics Core, University of Illinois, Chicago (M.M.-C.).

3. Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD (R.L.P.).

Abstract

Rationale: Increased endothelial permeability and defective repair are the hallmarks of several vascular diseases, including acute lung injury. However, little is known about the intrinsic pathways activating the endothelial cell (EC) regenerative programs. Objective: Studies have invoked a crucial role of S1P (sphingosine-1-phosphate) in resolving endothelial hyperpermeability through the activation of the GPCR (G-protein–coupled receptor), S1PR1 (S1P receptor 1). Here, we addressed mechanisms of generation of a population of S1PR1 + EC and their pivotal role in restoring endothelial integrity. Methods and Results: Studies were made using inducible EC-S1PR1 −/− ( iEC-S1PR1 −/− ) mice and S1PR1-GFP (green fluorescent protein) reporter mice to trace the generation of S1PR1 + EC. We observed in a mouse model of endotoxemia that S1P generation induced the programming of S1PR1 lo to S1PR1 + EC, which eventually comprised 80% of the lung EC. The cell transition was required for reestablishing the endothelial junctional barrier. We observed that conditional deletion of S1PR1 in EC increased endothelial permeability. RNA-seq analysis of S1PR1 + EC showed enrichment of genes regulating S1P synthesis and transport, specifically SPHK1 (sphingosine kinase 1) and SPNS2 (sphingolipid transporter 2). Activation of transcription factors EGR1 (early growth response 1) and STAT3 (signal transducer and activator of transcription 3) was required for transcribing SPHK1 and SPNS2, respectively, and both served to increase S1P production and amplify S1PR1 + EC transition. Furthermore, transplantation of S1PR1 + EC population into injured lung vasculature restored endothelial integrity. Conclusions: Our findings show that generation of the S1PR1 + EC population activates the endothelial regenerative program to mediate endothelial repair. Results raise the possibility of harnessing this pathway to restore vascular homeostasis in inflammatory vascular injury states.

Funder

HHS | National Institutes of Health

American Heart Association

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Cardiology and Cardiovascular Medicine,Physiology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3