Loss of Amphiregulin drives inflammation and endothelial apoptosis in pulmonary hypertension

Author:

Florentin Jonathan1,Zhao Jingsi1,Tai Yi-Yin1,Sun Wei1,Ohayon Lee L1,O’Neil Scott P1,Arunkumar Anagha1,Zhang Xinyi1ORCID,Zhu Jianhui2ORCID,Al Aaraj Yassmin1,Watson Annie1,Sembrat John13,Rojas Mauricio13,Chan Stephen Y1ORCID,Dutta Partha14ORCID

Affiliation:

1. Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA

2. Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA

3. Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA

4. Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA

Abstract

Pulmonary hypertension (PH) is a vascular disease characterized by elevated pulmonary arterial pressure, leading to right ventricular failure and death. Pathogenic features of PH include endothelial apoptosis and vascular inflammation, which drive vascular remodeling and increased pulmonary arterial pressure. Re-analysis of the whole transcriptome sequencing comparing human pulmonary arterial endothelial cells (PAECs) isolated from PH and control patients identifiedAREG, which encodes Amphiregulin, as a key endothelial survival factor. PAECs from PH patients and mice exhibited down-regulation ofAREGand its receptor epidermal growth factor receptor (EGFR). Moreover, the deficiency ofAREGandEGFRin ECs in vivo and in vitro heightened inflammatory leukocyte recruitment, cytokine production, and endothelial apoptosis, as well as diminished angiogenesis. Correspondingly, hypoxic mice lackingEgfrin ECs (cdh5cre/+Egfrfl/fl) displayed elevated RVSP and pulmonary remodeling. Computational analysis identifiedNCOA6,PHB2, andRRP1Bas putative genes regulatingAREGin endothelial cells. The master transcription factor of hypoxia HIF-1⍺ binds to the promoter regions of these genes and up-regulates their expression in hypoxia. Silencing of these genes in cultured PAECs decreased inflammation and apoptosis, and increased angiogenesis in hypoxic conditions. Our pathway analysis and gene silencing experiments revealed that BCL2-associated agonist of cell death (BAD) is a downstream mediator ofAREG.BADsilencing in ECs lackingAREGmitigated inflammation and apoptosis, and suppressed tube formation. In conclusion, loss of Amphiregulin and its receptor EGFR in PH is a crucial step in the pathogenesis of PH, promoting pulmonary endothelial cell death, influx of inflammatory myeloid cells, and vascular remodeling.

Funder

National Institute of Health

AHA Transformational Project Award

AHA Innovative Project Award

ALA Innovation Project Award

National, Heart, Lung and Blood Institute

NIH

American Heart Association Established Investigator Award

Publisher

Life Science Alliance, LLC

Subject

Health, Toxicology and Mutagenesis,Plant Science,Biochemistry, Genetics and Molecular Biology (miscellaneous),Ecology

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