Amyloid‐β exposed astrocytes induce iron transport from endothelial cells at the blood–brain barrier by altering the ratio of apo‐ and holo‐transferrin

Author:

Baringer Stephanie L.1ORCID,Lukacher Avraham S.1,Palsa Kondaiah1,Kim Hyosung2,Lippmann Ethan S.2ORCID,Spiegelman Vladimir S.3ORCID,Simpson Ian A.4,Connor James R.1ORCID

Affiliation:

1. Department of Neurosurgery Penn State College of Medicine Hershey Pennsylvania USA

2. Department of Chemical and Biomolecular Engineering Vanderbilt University Nashville Tennessee USA

3. Department of Pediatrics Penn State College of Medicine Hershey Pennsylvania USA

4. Department of Neural and Behavioral Sciences Penn State College of Medicine Hershey Pennsylvania USA

Abstract

AbstractExcessive brain iron accumulation is observed early in the onset of Alzheimer's disease, notably prior to widespread proteinopathy. These findings suggest that increases in brain iron levels are due to a dysregulation of the iron transport mechanism at the blood–brain barrier. Astrocytes release signals (apo‐ and holo‐transferrin) that communicate brain iron needs to endothelial cells in order to modulate iron transport. Here we use iPSC‐derived astrocytes and endothelial cells to investigate how early‐disease levels of amyloid‐β disrupt iron transport signals secreted by astrocytes to stimulate iron transport from endothelial cells. We demonstrate that conditioned media from astrocytes treated with amyloid‐β stimulates iron transport from endothelial cells and induces changes in iron transport pathway proteins. The mechanism underlying this response begins with increased iron uptake and mitochondrial activity by the astrocytes, which in turn increases levels of apo‐transferrin in the amyloid‐β conditioned astrocyte media leading to increased iron transport from endothelial cells. These novel findings offer a potential explanation for the initiation of excessive iron accumulation in early stages of Alzheimer's disease. What's more, these data provide the first example of how the mechanism of iron transport regulation by apo‐ and holo‐transferrin becomes misappropriated in disease that can lead to iron accumulation. The clinical benefit from understanding early dysregulation in brain iron transport in AD cannot be understated. If therapeutics can target this early process, they could possibly prevent the detrimental cascade that occurs with excessive iron accumulation.image

Funder

National Institutes of Health

Publisher

Wiley

Subject

Cellular and Molecular Neuroscience,Biochemistry

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