Molecular Basis of Hypoxia Induced Excessive Erythrocytosis

Author:

Azad Priti1,Haddad Gabriel1

Affiliation:

1. Division of Respiratory Medicine, Department of Pediatrics University of California San Diego La Jolla CA

Abstract

ObjectiveSince polycythemia is a predominant trait in some high altitude dwellers (Chronic Mountain Sickness, CMS, or Monge's disease) but not others living at the same altitude in the Andes, we took advantage of this human “experiment in nature” and studied both populations (with CMS and without, non‐CMS). Although polycythemia could be advantageous at high altitude because it increases O2‐carrying capacity, this adaptive pattern to high altitude has deleterious effects since blood increases its viscosity and induces serious morbidities, such as myocardial infarction and stroke in young adults.MethodsIn order to understand the molecular basis for polycythemia of high altitude, we generated a disease in‐the dish‐model by re‐programming fibroblasts from CMS and non‐CMS subjects. In the past, we have done whole genome sequencing of subjects from this region (CMS and non‐CMS) to study selective sweeps and genetic basis of CMS. In this study, by manipulating expression of some of these genes in using our in‐vitro model system, we are delineating the functional basis and the molecular mechanism(s) linked to the pathology.ResultsAs compared to sea level controls and non‐CMS subjects who responded to hypoxia by increasing their RBCs modestly or not at all, CMS cells increased theirs remarkably (up to 60 fold) with a dose‐dependent response to graded hypoxia (1.5, 5, 10% O2). We knocked down SENP1 (a desumoylase) in CMS iPS cells using lentiviral constructs and observed a striking reduction (>90%) of the CMS excessive erythropoietic response to low O2. And, by over‐expressing SENP1 in non‐CMS iPS cells, the hypoxic response in these subjects increased enormously. By further analyzing RBC differentiation in hypoxia, we found that VEGF, GATA1 and Bcl‐xL increased their gene expression in the CMS erythroid cells, in contrast to their minimal expression in the other two populations. We demonstrate also that, by utilizing a SUMO‐GATA1 fused construct, GATA1 desumoylation, a target of SENP1, is critical for the CMS phenotype. Unlike GATA1, the over‐expression of the anti‐apoptotic gene Bcl‐xL (a GATA1 target), only partially rescued the blunted erythroid response to hypoxia in non‐CMS cells.ConclusionWe conclude that the increased erythropoietic sensitivity to hypoxia in CMS subjects is genetic in nature and depends on an increase in SENP1 expression and its desumoylation mediated activation of GATA1 under hypoxia. By combining the iPS technology with this unique Andean population that has adapted (or mal‐adaptated) to chronic hypoxia over thousands of years, we have built an important in‐vitro model. We believe that utility of this model lies not only for studying hypoxia‐induced polycythemia but also other hypoxia‐driven diseases experienced at sea level.Support or Funding InformationThis study is funded by National Institutes of Health (NIH) grants (1P01HL098053 and 5P01HD32573) to G.G. Haddad

Publisher

Wiley

Subject

Genetics,Molecular Biology,Biochemistry,Biotechnology

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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