PGC-1α上调可减轻氧糖剥夺诱导的海马神经元损伤。

IF 3 4区 医学 Q2 NEUROSCIENCES
Neural Plasticity Pub Date : 2022-06-09 eCollection Date: 2022-01-01 DOI:10.1155/2022/9682999
Bin Han, Hui Zhao, Xingji Gong, Jinping Sun, Song Chi, Tao Liu, Anmu Xie
{"title":"PGC-1α上调可减轻氧糖剥夺诱导的海马神经元损伤。","authors":"Bin Han,&nbsp;Hui Zhao,&nbsp;Xingji Gong,&nbsp;Jinping Sun,&nbsp;Song Chi,&nbsp;Tao Liu,&nbsp;Anmu Xie","doi":"10.1155/2022/9682999","DOIUrl":null,"url":null,"abstract":"<p><p>Hippocampal neuronal damage likely underlies cognitive impairment in vascular dementia (VaD). PPAR<i>γ</i> coactivator-1<i>α</i> (PGC-1<i>α</i>) is a master regulator of mitochondrial biogenesis. However, the role and the precise mechanism of how PGC-1<i>α</i> alleviates hippocampal neuronal injury remain unknown. To address this question, HT-22 cells, an immortalized hippocampal neuron cell line, with or without PGC-1<i>α</i> overexpression were subjected to oxygen-glucose deprivation (OGD), which mimics the circumstance of chronic cerebral hypoperfusion in VaD. After OGD, cell viability was assessed using the MTS assay. The mitochondrial function and reactive oxygen species (ROS) were both detected. ChIP-Seq analysis was employed to discover the underlying molecular mechanism of PGC-1<i>α</i>-mediated neuroprotective effects. Our results showed that mitochondrial membrane potentials were increased and ROS production was decreased in PGC-1<i>α</i> overexpressing cells, which increased cell viability. The further bioinformatics analysis from ChIP-Seq data indicated that PGC-1<i>α</i> may participate in the regulation of apoptosis, autophagy, and mitophagy pathways in HT-22 cells. We found that PGC-1<i>α</i> promoted the LC3-II formation and reduced the neuronal apoptosis determined by TUNEL staining. In addition, PGC-1<i>α</i> upregulated the expressions of mitochondrial antioxidants, including SOD2, Trx2, and Prx3. In summary, our findings indicate that PGC-1<i>α</i> may attenuate OGD-induced hippocampal neuronal damage by regulating multiple mechanisms, like autophagy and mitochondrial function. Thus, PGC-1<i>α</i> may be a potential therapeutic target for hippocampal damage associated with cognitive impairment.</p>","PeriodicalId":51299,"journal":{"name":"Neural Plasticity","volume":null,"pages":null},"PeriodicalIF":3.0000,"publicationDate":"2022-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9203239/pdf/","citationCount":"3","resultStr":"{\"title\":\"Upregulation of PGC-1<i>α</i> Attenuates Oxygen-Glucose Deprivation-Induced Hippocampal Neuronal Injury.\",\"authors\":\"Bin Han,&nbsp;Hui Zhao,&nbsp;Xingji Gong,&nbsp;Jinping Sun,&nbsp;Song Chi,&nbsp;Tao Liu,&nbsp;Anmu Xie\",\"doi\":\"10.1155/2022/9682999\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Hippocampal neuronal damage likely underlies cognitive impairment in vascular dementia (VaD). PPAR<i>γ</i> coactivator-1<i>α</i> (PGC-1<i>α</i>) is a master regulator of mitochondrial biogenesis. However, the role and the precise mechanism of how PGC-1<i>α</i> alleviates hippocampal neuronal injury remain unknown. To address this question, HT-22 cells, an immortalized hippocampal neuron cell line, with or without PGC-1<i>α</i> overexpression were subjected to oxygen-glucose deprivation (OGD), which mimics the circumstance of chronic cerebral hypoperfusion in VaD. After OGD, cell viability was assessed using the MTS assay. The mitochondrial function and reactive oxygen species (ROS) were both detected. ChIP-Seq analysis was employed to discover the underlying molecular mechanism of PGC-1<i>α</i>-mediated neuroprotective effects. Our results showed that mitochondrial membrane potentials were increased and ROS production was decreased in PGC-1<i>α</i> overexpressing cells, which increased cell viability. The further bioinformatics analysis from ChIP-Seq data indicated that PGC-1<i>α</i> may participate in the regulation of apoptosis, autophagy, and mitophagy pathways in HT-22 cells. We found that PGC-1<i>α</i> promoted the LC3-II formation and reduced the neuronal apoptosis determined by TUNEL staining. In addition, PGC-1<i>α</i> upregulated the expressions of mitochondrial antioxidants, including SOD2, Trx2, and Prx3. In summary, our findings indicate that PGC-1<i>α</i> may attenuate OGD-induced hippocampal neuronal damage by regulating multiple mechanisms, like autophagy and mitochondrial function. Thus, PGC-1<i>α</i> may be a potential therapeutic target for hippocampal damage associated with cognitive impairment.</p>\",\"PeriodicalId\":51299,\"journal\":{\"name\":\"Neural Plasticity\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.0000,\"publicationDate\":\"2022-06-09\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9203239/pdf/\",\"citationCount\":\"3\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Neural Plasticity\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1155/2022/9682999\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2022/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q2\",\"JCRName\":\"NEUROSCIENCES\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Neural Plasticity","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1155/2022/9682999","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2022/1/1 0:00:00","PubModel":"eCollection","JCR":"Q2","JCRName":"NEUROSCIENCES","Score":null,"Total":0}
引用次数: 3

摘要

海马神经元损伤可能是血管性痴呆(VaD)认知障碍的基础。PGC-1α是线粒体生物发生的主要调控因子。然而,PGC-1α减轻海马神经元损伤的作用和确切机制尚不清楚。为了解决这个问题,我们将具有或不具有PGC-1α过表达的永生化海马神经元细胞系HT-22细胞进行氧糖剥夺(OGD),模拟VaD中慢性脑灌注不足的情况。OGD后,使用MTS法评估细胞活力。检测线粒体功能和活性氧(ROS)。通过ChIP-Seq分析发现pgc -1α-介导的神经保护作用的潜在分子机制。结果表明,过表达PGC-1α的细胞线粒体膜电位升高,ROS生成减少,细胞活力增加。进一步的ChIP-Seq数据生物信息学分析表明PGC-1α可能参与HT-22细胞凋亡、自噬和有丝自噬途径的调控。TUNEL染色发现PGC-1α促进LC3-II的形成,减少神经元凋亡。此外,PGC-1α上调线粒体抗氧化剂SOD2、Trx2和Prx3的表达。综上所述,我们的研究结果表明PGC-1α可能通过调节自噬和线粒体功能等多种机制来减轻ogd诱导的海马神经元损伤。因此,PGC-1α可能是认知障碍相关海马损伤的潜在治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Upregulation of PGC-1α Attenuates Oxygen-Glucose Deprivation-Induced Hippocampal Neuronal Injury.

Hippocampal neuronal damage likely underlies cognitive impairment in vascular dementia (VaD). PPARγ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis. However, the role and the precise mechanism of how PGC-1α alleviates hippocampal neuronal injury remain unknown. To address this question, HT-22 cells, an immortalized hippocampal neuron cell line, with or without PGC-1α overexpression were subjected to oxygen-glucose deprivation (OGD), which mimics the circumstance of chronic cerebral hypoperfusion in VaD. After OGD, cell viability was assessed using the MTS assay. The mitochondrial function and reactive oxygen species (ROS) were both detected. ChIP-Seq analysis was employed to discover the underlying molecular mechanism of PGC-1α-mediated neuroprotective effects. Our results showed that mitochondrial membrane potentials were increased and ROS production was decreased in PGC-1α overexpressing cells, which increased cell viability. The further bioinformatics analysis from ChIP-Seq data indicated that PGC-1α may participate in the regulation of apoptosis, autophagy, and mitophagy pathways in HT-22 cells. We found that PGC-1α promoted the LC3-II formation and reduced the neuronal apoptosis determined by TUNEL staining. In addition, PGC-1α upregulated the expressions of mitochondrial antioxidants, including SOD2, Trx2, and Prx3. In summary, our findings indicate that PGC-1α may attenuate OGD-induced hippocampal neuronal damage by regulating multiple mechanisms, like autophagy and mitochondrial function. Thus, PGC-1α may be a potential therapeutic target for hippocampal damage associated with cognitive impairment.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Neural Plasticity
Neural Plasticity NEUROSCIENCES-
CiteScore
6.80
自引率
0.00%
发文量
77
审稿时长
16 weeks
期刊介绍: Neural Plasticity is an international, interdisciplinary journal dedicated to the publication of articles related to all aspects of neural plasticity, with special emphasis on its functional significance as reflected in behavior and in psychopathology. Neural Plasticity publishes research and review articles from the entire range of relevant disciplines, including basic neuroscience, behavioral neuroscience, cognitive neuroscience, biological psychology, and biological psychiatry.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信