SOX17 Enhancer Variants Disrupt Transcription Factor Binding And Enhancer Inactivity Drives Pulmonary Hypertension

Author:

Walters Rachel1ORCID,Vasilaki Eleni1,Aman Jurjan12ORCID,Chen Chien-Nien1,Wu Yukyee1,Liang Olin D.3ORCID,Ashek Ali1,Dubois Olivier1,Zhao Lin1ORCID,Sabrin Farah1ORCID,Cebola Inês4ORCID,Ferrer Jorge456,Morrell Nicholas W.789ORCID,Klinger James R.10ORCID,Wilkins Martin R.19ORCID,Zhao Lan1ORCID,Rhodes Christopher J.19ORCID

Affiliation:

1. National Heart and Lung Institute, Hammersmith Hospital, Imperial College, London, United Kingdom (R.W., E.V., J.A., C.-N.C., Y.W., A.A., O.D., L.Z., F.S., M.R.W., L.Z., C.J.R.).

2. Department of Pulmonary Medicine, Amsterdam University Medical Center, The Netherlands (J.A.).

3. Division of Hematology/Oncology, Department of Medicine (O.D.L.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence.

4. Section of Genetics & Genomics, Department of Metabolism, Digestion & Reproduction, Hammersmith Hospital, Imperial College, London, United Kingdom (I.C., J.F.).

5. Computational Biology and Health Genomics Programme, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Spain (J.F.).

6. Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain (J.F.).

7. Department of Medicine, University of Cambridge, United Kingdom (N.W.M.).

8. NIHR BioResource for Translational Research, University of Cambridge, United Kingdom (N.W.M.).

9. On Behalf of the British Heart Foundation/Medical Research Council UK PAH Cohort Consortium (N.W.M., M.R.W., C.J.R.).

10. Division of Pulmonary, Sleep and Critical Care Medicine, Department of Medicine (J.R.K.), Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence.

Abstract

Background: Pulmonary arterial hypertension (PAH) is a rare disease characterized by remodeling of the pulmonary arteries, increased vascular resistance, and right-sided heart failure. Genome-wide association studies of idiopathic/heritable PAH established novel genetic risk variants, including conserved enhancers upstream of transcription factor (TF) SOX17 containing 2 independent signals. SOX17 is an important TF in embryonic development and in the homeostasis of pulmonary artery endothelial cells (hPAEC) in the adult. Rare pathogenic mutations in SOX17 cause heritable PAH. We hypothesized that PAH risk alleles in an enhancer region impair TF-binding upstream of SOX17 , which in turn reduces SOX17 expression and contributes to disturbed endothelial cell function and PAH development. Methods: CRISPR manipulation and siRNA were used to modulate SOX17 expression. Electromobility shift assays were used to confirm in silico predicted TF differential binding to the SOX17 variants. Functional assays in hPAECs were used to establish the biological consequences of SOX17 loss. In silico analysis with the connectivity map was used to predict compounds that rescue disturbed SOX17 signaling. Mice with deletion of the SOX17 -signal 1 enhancer region ( SOX17 -4593/enhKO) were phenotyped in response to chronic hypoxia and SU5416/hypoxia. Results: CRISPR inhibition of SOX17 -signal 2 and deletion of SOX17 -signal 1 specifically decreased SOX17 expression. Electromobility shift assays demonstrated differential binding of hPAEC nuclear proteins to the risk and nonrisk alleles from both SOX17 signals. Candidate TFs HOXA5 and ROR-α were identified through in silico analysis and antibody electromobility shift assays. Analysis of the hPAEC transcriptomes revealed alteration of PAH-relevant pathways on SOX17 silencing, including extracellular matrix regulation. SOX17 silencing in hPAECs resulted in increased apoptosis, proliferation, and disturbance of barrier function. With the use of the connectivity map, compounds were identified that reversed the SOX17-dysfunction transcriptomic signatures in hPAECs. SOX17 enhancer knockout in mice reduced lung SOX17 expression, resulting in more severe pulmonary vascular leak and hypoxia or SU5416/hypoxia-induced pulmonary hypertension. Conclusions: Common PAH risk variants upstream of the SOX17 promoter reduce endothelial SOX17 expression, at least in part, through differential binding of HOXA5 and ROR-α. Reduced SOX17 expression results in disturbed hPAEC function and PAH. Existing drug compounds can reverse the disturbed SOX17 pulmonary endothelial transcriptomic signature.

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Physiology (medical),Cardiology and Cardiovascular Medicine

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