The role of microRNAs in neural stem cell-supported endothelial morphogenesis.

Q4 Neuroscience Vascular Cell Pub Date : 2011-11-09 DOI:10.1186/2045-824X-3-25
Tamara Roitbak, Olga Bragina, Jamie L Padilla, Gavin G Pickett
{"title":"The role of microRNAs in neural stem cell-supported endothelial morphogenesis.","authors":"Tamara Roitbak,&nbsp;Olga Bragina,&nbsp;Jamie L Padilla,&nbsp;Gavin G Pickett","doi":"10.1186/2045-824X-3-25","DOIUrl":null,"url":null,"abstract":"<p><p> Functional signaling between neural stem/progenitor cells (NSPCs) and brain endothelial cells (ECs) is essential to the coordination of organized responses during initial embryonic development and also during tissue repair, which occurs following brain injury. In this study, we investigated the molecular mechanisms underlying this functional signaling, using primary mouse brain ECs and NSPCs from embryonic mouse brain. EC/NSPC co-culture experiments have revealed that neural progenitors secrete factors supporting angiogenesis, which induce noticeable changes in endothelial morphology. We demonstrate that NSPCs influence the expression of mTOR and TGF-β signaling pathway components implicated in the regulation of angiogenesis. Endothelial morphogenesis, an essential component of vascular development, is a complex process involving gene activation and the upregulation of specific cell signaling pathways. Recently identified small molecules, called microRNAs (miRNAs), regulate the expression of genes and proteins in many tissues, including brain and vasculature. We found that NSPCs induced considerable changes in the expression of at least 24 miRNAs and 13 genes in ECs. Three NSPC-regulated EC miRNAs were identified as the potential primary mediators of this NSPC/EC interaction. We found that the specific inhibition, or overexpression, of miRNAs miR-155, miR-100, and miR-let-7i subsequently altered the expression of major components of the mTOR, TGF-β and IGF-1R signaling pathways in ECs. Overexpression of these miRNAs in ECs suppressed, while inhibition activated, the in vitro formation of capillary-like structures, a process representative of EC morphogenesis. In addition, we demonstrate that inhibition of FGF, VEGF, and TGF-β receptor signaling abolished NSPC-promoted changes in the endothelial miRNA profiles. Our findings demonstrate that NSPCs induce changes in the miRNA expression of ECs, which are capable of activating angiogenesis by modulating distinct cell signaling pathways.</p>","PeriodicalId":23948,"journal":{"name":"Vascular Cell","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2011-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1186/2045-824X-3-25","citationCount":"26","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Vascular Cell","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/2045-824X-3-25","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q4","JCRName":"Neuroscience","Score":null,"Total":0}
引用次数: 26

Abstract

Functional signaling between neural stem/progenitor cells (NSPCs) and brain endothelial cells (ECs) is essential to the coordination of organized responses during initial embryonic development and also during tissue repair, which occurs following brain injury. In this study, we investigated the molecular mechanisms underlying this functional signaling, using primary mouse brain ECs and NSPCs from embryonic mouse brain. EC/NSPC co-culture experiments have revealed that neural progenitors secrete factors supporting angiogenesis, which induce noticeable changes in endothelial morphology. We demonstrate that NSPCs influence the expression of mTOR and TGF-β signaling pathway components implicated in the regulation of angiogenesis. Endothelial morphogenesis, an essential component of vascular development, is a complex process involving gene activation and the upregulation of specific cell signaling pathways. Recently identified small molecules, called microRNAs (miRNAs), regulate the expression of genes and proteins in many tissues, including brain and vasculature. We found that NSPCs induced considerable changes in the expression of at least 24 miRNAs and 13 genes in ECs. Three NSPC-regulated EC miRNAs were identified as the potential primary mediators of this NSPC/EC interaction. We found that the specific inhibition, or overexpression, of miRNAs miR-155, miR-100, and miR-let-7i subsequently altered the expression of major components of the mTOR, TGF-β and IGF-1R signaling pathways in ECs. Overexpression of these miRNAs in ECs suppressed, while inhibition activated, the in vitro formation of capillary-like structures, a process representative of EC morphogenesis. In addition, we demonstrate that inhibition of FGF, VEGF, and TGF-β receptor signaling abolished NSPC-promoted changes in the endothelial miRNA profiles. Our findings demonstrate that NSPCs induce changes in the miRNA expression of ECs, which are capable of activating angiogenesis by modulating distinct cell signaling pathways.

Abstract Image

Abstract Image

Abstract Image

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
microrna在神经干细胞支持的内皮形态发生中的作用。
神经干细胞/祖细胞(NSPCs)和脑内皮细胞(ECs)之间的功能信号传导对于胚胎早期发育和脑损伤后组织修复过程中的有组织反应协调至关重要。在这项研究中,我们利用小鼠胚胎脑的原代脑ECs和NSPCs研究了这种功能信号传导的分子机制。EC/NSPC共培养实验显示,神经祖细胞分泌支持血管生成的因子,诱导内皮细胞形态发生明显变化。我们证明NSPCs影响mTOR和TGF-β信号通路成分的表达,这些成分与血管生成的调节有关。内皮细胞形态发生是血管发育的重要组成部分,是一个涉及基因激活和特定细胞信号通路上调的复杂过程。最近发现的小分子,称为microRNAs (miRNAs),调节许多组织中基因和蛋白质的表达,包括大脑和脉管系统。我们发现NSPCs诱导了ECs中至少24种mirna和13种基因的表达发生了相当大的变化。三个NSPC调控的EC mirna被确定为NSPC/EC相互作用的潜在主要介质。我们发现,mirna miR-155、miR-100和miR-let-7i的特异性抑制或过表达随后改变了ECs中mTOR、TGF-β和IGF-1R信号通路主要成分的表达。这些mirna在EC中的过度表达抑制了毛细血管样结构的体外形成,而抑制作用则激活了毛细血管样结构的体外形成,这是一个代表EC形态发生的过程。此外,我们证明抑制FGF、VEGF和TGF-β受体信号通路可消除nspc促进的内皮miRNA谱的变化。我们的研究结果表明,NSPCs诱导内皮细胞miRNA表达的变化,内皮细胞能够通过调节不同的细胞信号通路来激活血管生成。
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
Vascular Cell
Vascular Cell Neuroscience-Neurology
CiteScore
0.70
自引率
0.00%
发文量
0
期刊最新文献
A 3-Dimensional Hypothesis of Oxidative Phosphorylation The long and winding road: detecting and quantifying Notch activation in endothelial cells Vascular Tumors Result from Adeno-Associated Virus-9 Angiogenic Gene Therapy of Bone Allografts A clinically relevant model of stroke using aged rats The SARS-CoV2 - ACE2 link: a physiopathological analysis
×
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