抑制microRNA-320可通过靶向HMGB1/NF-kappaB轴诱导脑缺血再灌注损伤的保护作用

IF 1.9 4区 医学 Q3 PHYSIOLOGY Physiological research Pub Date : 2024-03-11
S Liang, W Cao, Y Zhuang, D Zhang, S Du, H Shi
{"title":"抑制microRNA-320可通过靶向HMGB1/NF-kappaB轴诱导脑缺血再灌注损伤的保护作用","authors":"S Liang, W Cao, Y Zhuang, D Zhang, S Du, H Shi","doi":"","DOIUrl":null,"url":null,"abstract":"<p><p>MicroRNAs have been shown to potentially function in cerebral ischemia/reperfusion (IR) injury. This study aimed to examine the expression of microRNA-320 (miR-320) in cerebral IR injury and its involvement in cerebral mitochondrial function, oxidative stress, and inflammatory responses by targeting the HMGB1/NF-kappaB axis. Sprague-Dawley rats were subjected to middle cerebral artery occlusion to simulate cerebral IR injury. The cerebral expression of miR-320 was assessed using qRT-PCR. Neurological function, cerebral infarct volume, mitochondrial function, oxidative stress, and inflammatory cytokines were evaluated using relevant methods, including staining, fluorometry, and ELISA. HMGB1 expression was analyzed through Western blotting. The levels of miR-320, HMGB1, neurological deficits, and cerebral infarction were significantly higher after IR induction. Intracerebral overexpression of miR-320 resulted in substantial neurological deficits, increased infarct volume, elevated levels of 8-isoprostane, NF-kappaBp65, TNF-alpha, IL-1beta, ICAM-1, VCAM-1, and HMGB1 expression. It also promoted the loss of mitochondrial membrane potential and ROS levels while reducing MnSOD and GSH levels. Downregulation of miR-320 and inhibition of HMGB1 activity significantly reversed the outcomes of cerebral IR injury. MiR-320 plays a negative role in regulating cerebral inflammatory/oxidative reactions induced by IR injury by enhancing HMGB1 activity and modulating mitochondrial function.</p>","PeriodicalId":20235,"journal":{"name":"Physiological research","volume":null,"pages":null},"PeriodicalIF":1.9000,"publicationDate":"2024-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Suppression of microRNA-320 Induces Cerebral Protection Against Ischemia/Reperfusion Injury by Targeting HMGB1/NF-kappaB Axis.\",\"authors\":\"S Liang, W Cao, Y Zhuang, D Zhang, S Du, H Shi\",\"doi\":\"\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>MicroRNAs have been shown to potentially function in cerebral ischemia/reperfusion (IR) injury. This study aimed to examine the expression of microRNA-320 (miR-320) in cerebral IR injury and its involvement in cerebral mitochondrial function, oxidative stress, and inflammatory responses by targeting the HMGB1/NF-kappaB axis. Sprague-Dawley rats were subjected to middle cerebral artery occlusion to simulate cerebral IR injury. The cerebral expression of miR-320 was assessed using qRT-PCR. Neurological function, cerebral infarct volume, mitochondrial function, oxidative stress, and inflammatory cytokines were evaluated using relevant methods, including staining, fluorometry, and ELISA. HMGB1 expression was analyzed through Western blotting. The levels of miR-320, HMGB1, neurological deficits, and cerebral infarction were significantly higher after IR induction. Intracerebral overexpression of miR-320 resulted in substantial neurological deficits, increased infarct volume, elevated levels of 8-isoprostane, NF-kappaBp65, TNF-alpha, IL-1beta, ICAM-1, VCAM-1, and HMGB1 expression. It also promoted the loss of mitochondrial membrane potential and ROS levels while reducing MnSOD and GSH levels. Downregulation of miR-320 and inhibition of HMGB1 activity significantly reversed the outcomes of cerebral IR injury. MiR-320 plays a negative role in regulating cerebral inflammatory/oxidative reactions induced by IR injury by enhancing HMGB1 activity and modulating mitochondrial function.</p>\",\"PeriodicalId\":20235,\"journal\":{\"name\":\"Physiological research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":1.9000,\"publicationDate\":\"2024-03-11\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Physiological research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"PHYSIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Physiological research","FirstCategoryId":"3","ListUrlMain":"","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"PHYSIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

已有研究表明,微RNA可能在脑缺血再灌注(IR)损伤中发挥作用。本研究旨在通过靶向HMGB1/NF-kappaB轴研究microRNA-320(miR-320)在脑IR损伤中的表达及其在脑线粒体功能、氧化应激和炎症反应中的参与。对 Sprague-Dawley 大鼠进行大脑中动脉闭塞以模拟脑 IR 损伤。采用 qRT-PCR 技术评估 miR-320 在大脑中的表达。采用染色法、荧光测定法和酶联免疫吸附法等相关方法评估神经功能、脑梗塞体积、线粒体功能、氧化应激和炎症细胞因子。通过 Western 印迹分析了 HMGB1 的表达。IR诱导后,miR-320、HMGB1、神经功能缺损和脑梗死的水平显著升高。脑内过表达 miR-320 会导致严重的神经功能缺损、脑梗塞体积增大、8-异前列腺素、NF-kappaBp65、TNF-α、IL-1beta、ICAM-1、VCAM-1 和 HMGB1 表达水平升高。它还会促进线粒体膜电位和 ROS 水平的丧失,同时降低 MnSOD 和 GSH 水平。下调 miR-320 和抑制 HMGB1 的活性可明显逆转脑 IR 损伤的结果。MiR-320通过增强HMGB1活性和调节线粒体功能,在调节红外损伤诱导的脑部炎症/氧化反应中发挥负面作用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Suppression of microRNA-320 Induces Cerebral Protection Against Ischemia/Reperfusion Injury by Targeting HMGB1/NF-kappaB Axis.

MicroRNAs have been shown to potentially function in cerebral ischemia/reperfusion (IR) injury. This study aimed to examine the expression of microRNA-320 (miR-320) in cerebral IR injury and its involvement in cerebral mitochondrial function, oxidative stress, and inflammatory responses by targeting the HMGB1/NF-kappaB axis. Sprague-Dawley rats were subjected to middle cerebral artery occlusion to simulate cerebral IR injury. The cerebral expression of miR-320 was assessed using qRT-PCR. Neurological function, cerebral infarct volume, mitochondrial function, oxidative stress, and inflammatory cytokines were evaluated using relevant methods, including staining, fluorometry, and ELISA. HMGB1 expression was analyzed through Western blotting. The levels of miR-320, HMGB1, neurological deficits, and cerebral infarction were significantly higher after IR induction. Intracerebral overexpression of miR-320 resulted in substantial neurological deficits, increased infarct volume, elevated levels of 8-isoprostane, NF-kappaBp65, TNF-alpha, IL-1beta, ICAM-1, VCAM-1, and HMGB1 expression. It also promoted the loss of mitochondrial membrane potential and ROS levels while reducing MnSOD and GSH levels. Downregulation of miR-320 and inhibition of HMGB1 activity significantly reversed the outcomes of cerebral IR injury. MiR-320 plays a negative role in regulating cerebral inflammatory/oxidative reactions induced by IR injury by enhancing HMGB1 activity and modulating mitochondrial function.

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
Physiological research
Physiological research 医学-生理学
CiteScore
4.00
自引率
4.80%
发文量
108
审稿时长
3 months
期刊介绍: Physiological Research is a peer reviewed Open Access journal that publishes articles on normal and pathological physiology, biochemistry, biophysics, and pharmacology. Authors can submit original, previously unpublished research articles, review articles, rapid or short communications. Instructions for Authors - Respect the instructions carefully when submitting your manuscript. Submitted manuscripts or revised manuscripts that do not follow these Instructions will not be included into the peer-review process. The articles are available in full versions as pdf files beginning with volume 40, 1991. The journal publishes the online Ahead of Print /Pre-Press version of the articles that are searchable in Medline and can be cited.
期刊最新文献
Influence of Lipid Class Used for Omega-3 Fatty Acid Supplementation on Liver Fat Accumulation in MASLD. Research on Experimental Hypertension in Prague (1966-2009). Variability of Clinical Phenotypes Caused by Isolated Defects of Mitochondrial ATP Synthase. 3-N-Butylphthalide Confers Antiarrhythmic Features in Ischemia/Reperfusion Injury of Diabetic Heart by Targeting Mitochondria-Endoplasmic Reticulum Network and Inhibiting Oxidative Stress and Inflammation. Baicalin Ameliorates Cartilage Injury in Rats With Osteoarthritis via Modulating miR-766-3p/AIFM1 Axis.
×
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