Sbno1 mediates cell-cell communication between neural stem cells and microglia through small extracellular vesicles.

IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Cell and Bioscience Pub Date : 2024-09-29 DOI:10.1186/s13578-024-01296-4
Yifan Zhang, Zhihan Zhu, Zhinuo Li, Jia Feng, Jun Long, Yushu Deng, Waqas Ahmed, Ahsan Ali Khan, Shiying Huang, Qingling Fu, Lukui Chen
{"title":"Sbno1 mediates cell-cell communication between neural stem cells and microglia through small extracellular vesicles.","authors":"Yifan Zhang, Zhihan Zhu, Zhinuo Li, Jia Feng, Jun Long, Yushu Deng, Waqas Ahmed, Ahsan Ali Khan, Shiying Huang, Qingling Fu, Lukui Chen","doi":"10.1186/s13578-024-01296-4","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Neural stem cells (NSCs) play a crucial role in the progress of ischemic stroke. Research on zebrafish embryonic demonstrates an association between Strawberry Notch 1 (Sbno1) and central nervous system development. However, the regulation and underlying mechanism of Sbno1 in NSCs have not been studied yet. Here, we investigated the role and the mechanism of Sbno1 in NSCs development and the potential therapeutic value of Sbno1 in ischemic stroke.</p><p><strong>Methods: </strong>Adeno-associated virus (AAV) was used for overexpression or knockdown of Sbno1 in vitro or in vivo. A mouse model of MCAO was established to evaluate the neuroprotective effects of AAV-Sbno1, including balance beam test, rotarod test, and strength evaluation. H&E and immunofluorescence assessed neuronal impairment. Western blot and RT-qPCR were used to detect the expression of Sbno1 and its downstream target genes. RNA-seq and western blot were performed to explore further molecular mechanisms by which Sbno1 promoted endogenous repair of NSCs and macrophages M2 polarization. CCK8 was conducted to assess the effects of Sbno1 on NSCs proliferation. The impact of Sbno1 on NSCs apoptosis was evaluated by flow cytometry. NSCs derived from small extracellular vesicles (sEV) were obtained using ultracentrifugation and identified through nanoparticle tracking analysis (NTA) and western blot analysis.</p><p><strong>Results: </strong>Our results showed that Sbno1 is highly expressed in the central nervous system, which plays a crucial role in regulating the proliferation of NSCs through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. In addition, with overexpression of Sbno1 in the hippocampus, post-stroke behavioral scores were superior to the wild-type mice, and immunofluorescence staining revealed an increased number of newly generated neurons. sEV released by NSCs overexpressing Sbno1 inhibited neuroinflammation, which mechanistically impaired the activation of the microglial NF-κB and MAPK signaling pathways.</p><p><strong>Conclusions: </strong>Our studies indicate that sbno1 promotes the proliferation of NSCs and enhances endogenous repairing through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. Additionally, NSCs overexpressing sbno1 improve ischemic stroke recovery and inhibit neuroinflammation after ischemia by sEV through the MAPK and NF-κB signaling pathways.</p>","PeriodicalId":49095,"journal":{"name":"Cell and Bioscience","volume":null,"pages":null},"PeriodicalIF":6.1000,"publicationDate":"2024-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441009/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell and Bioscience","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s13578-024-01296-4","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Neural stem cells (NSCs) play a crucial role in the progress of ischemic stroke. Research on zebrafish embryonic demonstrates an association between Strawberry Notch 1 (Sbno1) and central nervous system development. However, the regulation and underlying mechanism of Sbno1 in NSCs have not been studied yet. Here, we investigated the role and the mechanism of Sbno1 in NSCs development and the potential therapeutic value of Sbno1 in ischemic stroke.

Methods: Adeno-associated virus (AAV) was used for overexpression or knockdown of Sbno1 in vitro or in vivo. A mouse model of MCAO was established to evaluate the neuroprotective effects of AAV-Sbno1, including balance beam test, rotarod test, and strength evaluation. H&E and immunofluorescence assessed neuronal impairment. Western blot and RT-qPCR were used to detect the expression of Sbno1 and its downstream target genes. RNA-seq and western blot were performed to explore further molecular mechanisms by which Sbno1 promoted endogenous repair of NSCs and macrophages M2 polarization. CCK8 was conducted to assess the effects of Sbno1 on NSCs proliferation. The impact of Sbno1 on NSCs apoptosis was evaluated by flow cytometry. NSCs derived from small extracellular vesicles (sEV) were obtained using ultracentrifugation and identified through nanoparticle tracking analysis (NTA) and western blot analysis.

Results: Our results showed that Sbno1 is highly expressed in the central nervous system, which plays a crucial role in regulating the proliferation of NSCs through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. In addition, with overexpression of Sbno1 in the hippocampus, post-stroke behavioral scores were superior to the wild-type mice, and immunofluorescence staining revealed an increased number of newly generated neurons. sEV released by NSCs overexpressing Sbno1 inhibited neuroinflammation, which mechanistically impaired the activation of the microglial NF-κB and MAPK signaling pathways.

Conclusions: Our studies indicate that sbno1 promotes the proliferation of NSCs and enhances endogenous repairing through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. Additionally, NSCs overexpressing sbno1 improve ischemic stroke recovery and inhibit neuroinflammation after ischemia by sEV through the MAPK and NF-κB signaling pathways.

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
Sbno1 通过细胞外小泡介导神经干细胞和小胶质细胞之间的细胞间通信。
背景:神经干细胞(NSCs)在缺血性中风的进展过程中起着至关重要的作用。对斑马鱼胚胎的研究表明,草莓诺奇1(Sbno1)与中枢神经系统发育有关。然而,Sbno1 在 NSCs 中的调控及其内在机制尚未得到研究。在此,我们研究了Sbno1在NSCs发育中的作用和机制,以及Sbno1在缺血性脑卒中中的潜在治疗价值:方法:使用腺相关病毒(AAV)在体外或体内过表达或敲除 Sbno1。方法:利用腺相关病毒(AAV)在体外或体内过表达或敲除 Sbno1,建立 MCAO 小鼠模型,评估 AAV-Sbno1 的神经保护作用,包括平衡木测试、转体测试和力量评估。H&E和免疫荧光评估了神经元损伤情况。Western blot和RT-qPCR用于检测Sbno1及其下游靶基因的表达。通过RNA-seq和Western blot进一步探索Sbno1促进NSCs内源性修复和巨噬细胞M2极化的分子机制。CCK8评估了Sbno1对NSCs增殖的影响。流式细胞术评估了Sbno1对NSCs凋亡的影响。通过超速离心法获得来自小细胞外囊泡(sEV)的NSCs,并通过纳米颗粒追踪分析(NTA)和Western印迹分析进行鉴定:结果:我们的研究结果表明,Sbno1在中枢神经系统中高表达,它在通过PI3k-Akt-GSK3β-Wnt/β-catenin信号通路调节NSCs增殖中发挥着重要作用。此外,在海马中过表达Sbno1后,卒中后行为评分优于野生型小鼠,免疫荧光染色显示新生成的神经元数量增加。过表达Sbno1的NSCs释放的sEV抑制了神经炎症,从机制上损害了小胶质细胞NF-κB和MAPK信号通路的激活:我们的研究表明,sbno1可通过PI3k-Akt-GSK3β-Wnt/β-catenin信号通路促进NSCs增殖并增强内源性修复。此外,过表达 sbno1 的 NSCs 还能改善缺血性中风的恢复,并通过 MAPK 和 NF-κB 信号通路抑制缺血后 sEV 的神经炎症。
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
Cell and Bioscience
Cell and Bioscience BIOCHEMISTRY & MOLECULAR BIOLOGY-
CiteScore
10.70
自引率
0.00%
发文量
187
审稿时长
>12 weeks
期刊介绍: Cell and Bioscience, the official journal of the Society of Chinese Bioscientists in America, is an open access, peer-reviewed journal that encompasses all areas of life science research.
期刊最新文献
HPV16 integration regulates ferroptosis resistance via the c-Myc/miR-142-5p/HOXA5/SLC7A11 axis during cervical carcinogenesis. Galectin-3 inhibition reduces fibrotic scarring and promotes functional recovery after spinal cord injury in mice. CASTOR1 phosphorylation predicts poor survival in male patients with KRAS-mutated lung adenocarcinoma. Transcription and post-translational mechanisms: dual regulation of adiponectin-mediated Occludin expression in diabetes. Sbno1 mediates cell-cell communication between neural stem cells and microglia through small extracellular vesicles.
×
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