Ligand-free mitochondria-localized mutant AR-induced cytotoxicity in spinal bulbar muscular atrophy

Author:

Feng Xia123ORCID,Cheng Xiu-Tang4,Zheng Pengli2,Li Yan5,Hakim Jill3,Zhang Shirley Q6,Anderson Stacie M7,Linask Kaari8,Prestil Ryan3,Zou Jizhong8,Sheng Zu-Hang4,Blackstone Craig2910ORCID

Affiliation:

1. Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine , Baltimore, MD , USA

2. Cell Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA

3. Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA

4. Synaptic Function Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA

5. Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA

6. River Hill High School , Clarksville, MD , USA

7. Flow Cytometry Core, National Human Genome Research Institute, National Institute of Health , Bethesda, MD , USA

8. iPSC Core, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, MD , USA

9. MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital , Charlestown, MA , USA

10. Department of Neurology, Massachusetts General Hospital and Harvard Medical School , Boston, MA , USA

Abstract

Abstract Spinal bulbar muscular atrophy (SBMA), the first identified CAG-repeat expansion disorder, is an X-linked neuromuscular disorder involving CAG-repeat-expansion mutations in the androgen receptor (AR) gene. We utilized CRISPR-Cas9 gene editing to engineer novel isogenic human induced pluripotent stem cell (hiPSC) models, consisting of isogenic AR knockout, control and disease lines expressing mutant AR with distinct repeat lengths, as well as control and disease lines expressing FLAG-tagged wild-type and mutant AR, respectively. Adapting a small-molecule cocktail-directed approach, we differentiate the isogenic hiPSC models into motor neuron-like cells with a highly enriched population to uncover cell-type-specific mechanisms underlying SBMA and to distinguish gain- from loss-of-function properties of mutant AR in disease motor neurons. We demonstrate that ligand-free mutant AR causes drastic mitochondrial dysfunction in neurites of differentiated disease motor neurons due to gain-of-function mechanisms and such cytotoxicity can be amplified upon ligand (androgens) treatment. We further show that aberrant interaction between ligand-free, mitochondria-localized mutant AR and F-ATP synthase is associated with compromised mitochondrial respiration and multiple other mitochondrial impairments. These findings counter the established notion that androgens are requisite for mutant AR-induced cytotoxicity in SBMA, reveal a compelling mechanistic link between ligand-free mutant AR, F-ATP synthase and mitochondrial dysfunction, and provide innovative insights into motor neuron-specific therapeutic interventions for SBMA.

Funder

NINDS Competitive Postdoctoral Fellowship Award

Kennedy’s Disease Association Research Grant

Maryland Stem Cell Research Fund Launch Award

Intramural Research Program of the NINDS

National Institutes of Health

Publisher

Oxford University Press (OUP)

Subject

Neurology (clinical)

Cited by 4 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