Long noncoding RNA KCNQ1OT1 aggravates cerebral infarction by regulating PTBT1/SIRT1 via miR-16-5p.

IF 3.2 3区 医学 Q2 CLINICAL NEUROLOGY Journal of Neuropathology and Experimental Neurology Pub Date : 2024-03-20 DOI:10.1093/jnen/nlae005
Yuanming Jiang, Chi Ma, Yuxiu Guan, Wenqi Yang, Jiaqi Yu, Hanfei Shi, Zihang Ding, Zhuobo Zhang
{"title":"Long noncoding RNA KCNQ1OT1 aggravates cerebral infarction by regulating PTBT1/SIRT1 via miR-16-5p.","authors":"Yuanming Jiang, Chi Ma, Yuxiu Guan, Wenqi Yang, Jiaqi Yu, Hanfei Shi, Zihang Ding, Zhuobo Zhang","doi":"10.1093/jnen/nlae005","DOIUrl":null,"url":null,"abstract":"<p><p>Cerebral infarction (CI) is one of the leading causes of disability and death. LncRNAs are key factors in CI progression. Herein, we studied the function of long noncoding RNA KCNQ1OT1 in CI patient plasma samples and in CI models. Quantitative real-time PCR and Western blotting tested gene and protein expressions. The interactions of KCNQ1OT1/PTBP1 and miR-16-5p were analyzed using dual-luciferase reporter and RNA immunoprecipitation assays; MTT assays measured cell viability. Cell migration and angiogenesis were tested by wound healing and tube formation assays. Pathological changes were analyzed by triphenyltetrazolium chloride and routine staining. We found that KCNQ1OT1 and PTBP1 were overexpressed and miR-16-5p was downregulated in CI patient plasma and in oxygen-glucose deprived (OGD) induced mouse brain microvascular endothelial (bEnd.3) cells. KCNQ1OT1 knockdown suppressed pro-inflammatory cytokine production and stimulated angiogenic responses in OGD-bEnd.3 cells. KCNQ1OT1 upregulated PTBP1 by sponging miR-16-5p. PTBP1 overexpression or miR-16-5p inhibition attenuated the effects of KCNQ1OT1 knockdown. PTBP1 silencing protected against OGD-bEnd.3 cell injury by enhancing SIRT1. KCNQ1OT1 silencing or miR-16-5p overexpression also alleviated ischemic injury in a mice middle cerebral artery occlusion model. Thus, KCNQ1OT1 silencing alleviates CI by regulating the miR-16-5p/PTBP1/SIRT1 pathway, providing a theoretical basis for novel therapeutic strategies targeting CI.</p>","PeriodicalId":16682,"journal":{"name":"Journal of Neuropathology and Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":3.2000,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Neuropathology and Experimental Neurology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1093/jnen/nlae005","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CLINICAL NEUROLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Cerebral infarction (CI) is one of the leading causes of disability and death. LncRNAs are key factors in CI progression. Herein, we studied the function of long noncoding RNA KCNQ1OT1 in CI patient plasma samples and in CI models. Quantitative real-time PCR and Western blotting tested gene and protein expressions. The interactions of KCNQ1OT1/PTBP1 and miR-16-5p were analyzed using dual-luciferase reporter and RNA immunoprecipitation assays; MTT assays measured cell viability. Cell migration and angiogenesis were tested by wound healing and tube formation assays. Pathological changes were analyzed by triphenyltetrazolium chloride and routine staining. We found that KCNQ1OT1 and PTBP1 were overexpressed and miR-16-5p was downregulated in CI patient plasma and in oxygen-glucose deprived (OGD) induced mouse brain microvascular endothelial (bEnd.3) cells. KCNQ1OT1 knockdown suppressed pro-inflammatory cytokine production and stimulated angiogenic responses in OGD-bEnd.3 cells. KCNQ1OT1 upregulated PTBP1 by sponging miR-16-5p. PTBP1 overexpression or miR-16-5p inhibition attenuated the effects of KCNQ1OT1 knockdown. PTBP1 silencing protected against OGD-bEnd.3 cell injury by enhancing SIRT1. KCNQ1OT1 silencing or miR-16-5p overexpression also alleviated ischemic injury in a mice middle cerebral artery occlusion model. Thus, KCNQ1OT1 silencing alleviates CI by regulating the miR-16-5p/PTBP1/SIRT1 pathway, providing a theoretical basis for novel therapeutic strategies targeting CI.

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
长非编码 RNA KCNQ1OT1 通过 miR-16-5p 调节 PTBT1/SIRT1 使脑梗塞恶化
脑梗塞(CI)是导致残疾和死亡的主要原因之一。LncRNA是CI进展的关键因素。在此,我们研究了长非编码 RNA KCNQ1OT1 在 CI 患者血浆样本和 CI 模型中的功能。定量实时 PCR 和 Western 印迹检测了基因和蛋白质的表达。使用双荧光素酶报告和 RNA 免疫沉淀试验分析了 KCNQ1OT1/PTBP1 和 miR-16-5p 的相互作用;MTT 试验测定了细胞活力。通过伤口愈合和管形成试验检测了细胞迁移和血管生成。病理变化通过三苯基氯化四氮唑和常规染色进行分析。我们发现,在 CI 患者血浆和氧-葡萄糖剥夺(OGD)诱导的小鼠脑微血管内皮细胞(bEnd.3)中,KCNQ1OT1 和 PTBP1 表达过高,miR-16-5p 下调。敲除 KCNQ1OT1 可抑制促炎细胞因子的产生,并刺激 OGD-bEnd.3 细胞的血管生成反应。KCNQ1OT1 通过疏导 miR-16-5p 上调 PTBP1。过表达 PTBP1 或抑制 miR-16-5p 可减轻 KCNQ1OT1 敲除的影响。沉默 PTBP1 可通过增强 SIRT1 保护 OGD-bEnd.3 细胞免受损伤。在小鼠大脑中动脉闭塞模型中,KCNQ1OT1沉默或miR-16-5p过表达也能减轻缺血性损伤。因此,KCNQ1OT1沉默可通过调节miR-16-5p/PTBP1/SIRT1通路缓解CI,为针对CI的新型治疗策略提供理论基础。
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
5.40
自引率
6.20%
发文量
118
审稿时长
6-12 weeks
期刊介绍: Journal of Neuropathology & Experimental Neurology is the official journal of the American Association of Neuropathologists, Inc. (AANP). The journal publishes peer-reviewed studies on neuropathology and experimental neuroscience, book reviews, letters, and Association news, covering a broad spectrum of fields in basic neuroscience with an emphasis on human neurological diseases. It is written by and for neuropathologists, neurologists, neurosurgeons, pathologists, psychiatrists, and basic neuroscientists from around the world. Publication has been continuous since 1942.
期刊最新文献
FH-mutant glioma displaying the epigenetic signature of IDH-mutant astrocytomas. Polymicrobial brain abscesses: A complex condition with diagnostic and therapeutic challenges. Cytoplasmic aggregation of TDP43 and topographic correlation with tau and α-synuclein accumulation in the rTg4510 mouse model of tauopathy. Primary CNS histiocytic sarcoma: Two case reports highlighting a novel MIGA2::BRAF gene fusion and genome-wide DNA methylation profiling results. Revealing the potential role of hsa-miR-663a in modulating the PI3K-Akt signaling pathway via miRNA microarray in spinal muscular atrophy patient fibroblast-derived iPSCs.
×
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