ATF4-activated parkin induction contributes to deferasirox-mediated cytoprotection in Parkinson's disease.

IF 1.6 4区 医学 Q4 TOXICOLOGY Toxicological Research Pub Date : 2022-12-08 eCollection Date: 2023-04-01 DOI:10.1007/s43188-022-00157-x
Sangwoo Ham, Ji Hun Kim, Heejeong Kim, Jeong-Yong Shin, Yunjong Lee
{"title":"ATF4-activated parkin induction contributes to deferasirox-mediated cytoprotection in Parkinson's disease.","authors":"Sangwoo Ham, Ji Hun Kim, Heejeong Kim, Jeong-Yong Shin, Yunjong Lee","doi":"10.1007/s43188-022-00157-x","DOIUrl":null,"url":null,"abstract":"<p><p>The E3 ubiquitin ligase parkin plays neuroprotective functions in the brain and the deficits of parkin's ligase function in Parkinson's disease (PD) is associated with reduced survival of dopaminergic neurons. Thus, compounds enhancing parkin expression have been developed as potential neuroprotective agents that prevent ongoing neurodegeneration in PD environments. Besides, iron chelators have been shown to have neuroprotective effects in diverse neurological disorders including PD. Although repression of iron accumulation and oxidative stress in brains has been implicated in their marked neuroprotective potential, molecular mechanisms of iron chelator's neuroprotective function are largely unexplored. Here, we show that the iron chelator deferasirox provides cytoprotection against oxidative stress through enhancing parkin expression under basal conditions. Parkin expression is required for cytoprotection against oxidative stress in SH-SY5Y cells with deferasirox treatment as confirmed by abolished deferasirox's cytoprotective effect after parkin knockdown by shRNA. Similar to the previously reported parkin inducing compound diaminodiphenyl sulfone, deferasirox-mediated parkin expression was induced by activation of the PERK-ATF4 pathway, which is associated with and stimulated by mild endoplasmic reticulum stress. The translational potential of deferasirox for PD treatment was further evaluated in cultured mouse dopaminergic neurons. There was a robust ATF4 activation and parkin expression in response to deferasirox treatment in dopaminergic neurons under basal conditions. Consequently, the enhanced parkin expression by deferasirox provided substantial neuroprotection against 6-hydroxydopamine-induced oxidative stress. Taken together, our study results revealed a novel mechanism through which an iron chelator, deferasirox induces neuroprotection. Since parkin function in the brain is compromised in PD and during aging, maintenance of parkin expression through the iron chelator treatment could be beneficial by increasing dopaminergic neuronal survival.</p>","PeriodicalId":23181,"journal":{"name":"Toxicological Research","volume":null,"pages":null},"PeriodicalIF":1.6000,"publicationDate":"2022-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10050497/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Toxicological Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s43188-022-00157-x","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/4/1 0:00:00","PubModel":"eCollection","JCR":"Q4","JCRName":"TOXICOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

The E3 ubiquitin ligase parkin plays neuroprotective functions in the brain and the deficits of parkin's ligase function in Parkinson's disease (PD) is associated with reduced survival of dopaminergic neurons. Thus, compounds enhancing parkin expression have been developed as potential neuroprotective agents that prevent ongoing neurodegeneration in PD environments. Besides, iron chelators have been shown to have neuroprotective effects in diverse neurological disorders including PD. Although repression of iron accumulation and oxidative stress in brains has been implicated in their marked neuroprotective potential, molecular mechanisms of iron chelator's neuroprotective function are largely unexplored. Here, we show that the iron chelator deferasirox provides cytoprotection against oxidative stress through enhancing parkin expression under basal conditions. Parkin expression is required for cytoprotection against oxidative stress in SH-SY5Y cells with deferasirox treatment as confirmed by abolished deferasirox's cytoprotective effect after parkin knockdown by shRNA. Similar to the previously reported parkin inducing compound diaminodiphenyl sulfone, deferasirox-mediated parkin expression was induced by activation of the PERK-ATF4 pathway, which is associated with and stimulated by mild endoplasmic reticulum stress. The translational potential of deferasirox for PD treatment was further evaluated in cultured mouse dopaminergic neurons. There was a robust ATF4 activation and parkin expression in response to deferasirox treatment in dopaminergic neurons under basal conditions. Consequently, the enhanced parkin expression by deferasirox provided substantial neuroprotection against 6-hydroxydopamine-induced oxidative stress. Taken together, our study results revealed a novel mechanism through which an iron chelator, deferasirox induces neuroprotection. Since parkin function in the brain is compromised in PD and during aging, maintenance of parkin expression through the iron chelator treatment could be beneficial by increasing dopaminergic neuronal survival.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
ATF4 激活的帕金诱导有助于地拉罗司介导的帕金森病细胞保护。
E3 泛素连接酶 parkin 在大脑中发挥着神经保护功能,帕金森病(PD)中 parkin 连接酶功能的缺失与多巴胺能神经元存活率的降低有关。因此,提高 parkin 表达的化合物已被开发为潜在的神经保护剂,以防止帕金森病环境中持续的神经变性。此外,铁螯合剂已被证明对包括帕金森病在内的多种神经系统疾病具有神经保护作用。虽然铁螯合剂具有显著的神经保护潜能,但其抑制大脑中铁积累和氧化应激的作用与铁螯合剂的神经保护功能的分子机制有关。在这里,我们发现铁螯合剂地拉罗司在基础条件下通过增强帕金蛋白的表达来提供细胞保护,抵御氧化应激。Parkin的表达是去铁胺处理SH-SY5Y细胞抗氧化应激的细胞保护所必需的,这一点通过用shRNA敲除parkin后去铁胺的细胞保护作用被取消而得到证实。与之前报道的诱导parkin的化合物二氨基二苯砜相似,去铁酮介导的parkin表达是通过激活PERK-ATF4通路诱导的,而PERK-ATF4通路与轻微的内质网应激有关,并受到轻微内质网应激的刺激。在培养的小鼠多巴胺能神经元中进一步评估了去铁酮治疗帕金森病的转化潜力。在基础条件下,地拉罗司处理多巴胺能神经元后,ATF4会被强有力地激活,parkin也会随之表达。因此,地拉罗司增强的parkin表达对6-羟基多巴胺诱导的氧化应激具有实质性的神经保护作用。综上所述,我们的研究结果揭示了铁螯合剂地拉罗司诱导神经保护的新机制。由于帕金森病和衰老过程中大脑中的帕金功能会受到损害,因此通过铁螯合剂治疗维持帕金的表达可以提高多巴胺能神经元的存活率,从而使其获益。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
4.20
自引率
4.30%
发文量
39
期刊介绍: Toxicological Research is the official journal of the Korean Society of Toxicology. The journal covers all areas of Toxicological Research of chemicals, drugs and environmental agents affecting human and animals, which in turn impact public health. The journal’s mission is to disseminate scientific and technical information on diverse areas of toxicological research. Contributions by toxicologists, molecular biologists, geneticists, biochemists, pharmacologists, clinical researchers and epidemiologists with a global view on public health through toxicological research are welcome. Emphasis will be given to articles providing an understanding of the toxicological mechanisms affecting animal, human and public health. In the case of research articles using natural extracts, detailed information with respect to the origin, extraction method, chemical profiles, and characterization of standard compounds to ensure the reproducible pharmacological activity should be provided.
期刊最新文献
Hepatocyte-specific RIG-I loss attenuates metabolic dysfunction-associated steatotic liver disease in mice via changes in mitochondrial respiration and metabolite profiles. Correction: Upregulation of YPEL3 expression and induction of human breast cancer cell death by microRNAs. The impact of manganese on vascular endothelium. Therapeutic strategies for colorectal cancer: antitumor efficacy of dopamine D2 receptor antagonists. Adverse events associated with SARS-CoV-2 neutralizing monoclonal antibodies using the FDA adverse event reporting system database.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1