Umbilical cord mesenchymal stem cell exosomes alleviate necrotizing enterocolitis in neonatal mice by regulating intestinal epithelial cells autophagy.

IF 3.6 3区 医学 Q3 CELL & TISSUE ENGINEERING
Lin Zhu, Lu He, Wu Duan, Bo Yang, Ning Li
{"title":"Umbilical cord mesenchymal stem cell exosomes alleviate necrotizing enterocolitis in neonatal mice by regulating intestinal epithelial cells autophagy.","authors":"Lin Zhu, Lu He, Wu Duan, Bo Yang, Ning Li","doi":"10.4252/wjsc.v16.i6.728","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease that affects premature infants. Although mounting evidence supports the therapeutic effect of exosomes on NEC, the underlying mechanisms remain unclear.</p><p><strong>Aim: </strong>To investigate the mechanisms underlying the regulation of inflammatory response and intestinal barrier function by umbilical cord mesenchymal stem cell (UCMSCs) exosomes, as well as their potential in alleviating NEC in neonatal mice.</p><p><strong>Methods: </strong>NEC was induced in 5-d-old C57BL/6 pups through hypoxia and gavage feeding of formula containing lipopolysaccharide (LPS), after which the mice received human UCMSC exosomes (hUCMSC-exos). The control mice were allowed to breastfeed with their dams. Ileal tissues were collected from the mice and analyzed by histopathology and immunoblotting. Colon tissues were collected from NEC neonates and analyzed by immunofluorescence. Molecular biology and cell culture approaches were employed to study the related mechanisms in intestinal epithelial cells.</p><p><strong>Results: </strong>We found that autophagy is overactivated in intestinal epithelial cells during NEC, resulting in reduced expression of tight junction proteins and an increased inflammatory response. The ability of hUCMSC-exos to ameliorate NEC in a mouse model was dependent on decreased intestinal autophagy. We also showed that hUCMSC-exos alleviate the inflammatory response and increase migration ability in intestinal epithelial cells induced by LPS.</p><p><strong>Conclusion: </strong>These results contribute to a better understanding of the protective mechanisms of hUCMSC-exos against NEC and provide a new theoretical and experimental foundation for NEC treatment. These findings also enhance our understanding of the role of the autophagy mechanism in NEC, offering potential avenues for identifying new therapeutic targets.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":null,"pages":null},"PeriodicalIF":3.6000,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11212546/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"World journal of stem cells","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.4252/wjsc.v16.i6.728","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"CELL & TISSUE ENGINEERING","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease that affects premature infants. Although mounting evidence supports the therapeutic effect of exosomes on NEC, the underlying mechanisms remain unclear.

Aim: To investigate the mechanisms underlying the regulation of inflammatory response and intestinal barrier function by umbilical cord mesenchymal stem cell (UCMSCs) exosomes, as well as their potential in alleviating NEC in neonatal mice.

Methods: NEC was induced in 5-d-old C57BL/6 pups through hypoxia and gavage feeding of formula containing lipopolysaccharide (LPS), after which the mice received human UCMSC exosomes (hUCMSC-exos). The control mice were allowed to breastfeed with their dams. Ileal tissues were collected from the mice and analyzed by histopathology and immunoblotting. Colon tissues were collected from NEC neonates and analyzed by immunofluorescence. Molecular biology and cell culture approaches were employed to study the related mechanisms in intestinal epithelial cells.

Results: We found that autophagy is overactivated in intestinal epithelial cells during NEC, resulting in reduced expression of tight junction proteins and an increased inflammatory response. The ability of hUCMSC-exos to ameliorate NEC in a mouse model was dependent on decreased intestinal autophagy. We also showed that hUCMSC-exos alleviate the inflammatory response and increase migration ability in intestinal epithelial cells induced by LPS.

Conclusion: These results contribute to a better understanding of the protective mechanisms of hUCMSC-exos against NEC and provide a new theoretical and experimental foundation for NEC treatment. These findings also enhance our understanding of the role of the autophagy mechanism in NEC, offering potential avenues for identifying new therapeutic targets.

脐带间充质干细胞外泌体通过调节肠上皮细胞自噬缓解新生小鼠的坏死性小肠结肠炎
背景:坏死性小肠结肠炎(NEC坏死性小肠结肠炎(NEC)是一种影响早产儿的严重胃肠道疾病。目的:研究脐带间充质干细胞(UCMSCs)外泌体调节炎症反应和肠屏障功能的机制,以及外泌体缓解新生小鼠坏死性小肠结肠炎的潜力:方法:通过缺氧和灌胃含有脂多糖(LPS)的配方奶诱导5天大的C57BL/6幼鼠发生NEC,然后让小鼠接受人UCMSC外泌体(hUCMSC-exos)。对照组小鼠与母鼠一起进行母乳喂养。收集小鼠的回肠组织并进行组织病理学和免疫印迹分析。收集 NEC 新生儿的结肠组织并进行免疫荧光分析。采用分子生物学和细胞培养方法研究肠上皮细胞的相关机制:结果:我们发现自噬在 NEC 期间在肠上皮细胞中被过度激活,导致紧密连接蛋白表达减少和炎症反应增加。hUCMSC-exos 在小鼠模型中改善 NEC 的能力取决于肠道自噬的减少。我们还发现,hUCMSC-exos 可减轻 LPS 诱导的肠上皮细胞炎症反应并提高其迁移能力:这些结果有助于更好地理解 hUCMSC-exos 对 NEC 的保护机制,并为 NEC 治疗提供了新的理论和实验基础。这些发现还加深了我们对自噬机制在 NEC 中作用的理解,为确定新的治疗靶点提供了潜在途径。
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 求助全文
来源期刊
World journal of stem cells
World journal of stem cells Biochemistry, Genetics and Molecular Biology-Molecular Biology
CiteScore
7.80
自引率
4.90%
发文量
750
期刊介绍: The World Journal of Stem Cells (WJSC) is a leading academic journal devoted to reporting the latest, cutting-edge research progress and findings of basic research and clinical practice in the field of stem cells. It was launched on December 31, 2009 and is published monthly (12 issues annually) by BPG, the world''s leading professional clinical medical journal publishing company.
×
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