肠上皮Dicer1调控app敲入小鼠肠道微生物组和阿尔茨海默病病理。

IF 7.6 1区 医学 Q1 CLINICAL NEUROLOGY
Wenlin Hao, Qinghua Luo, Ilona Magdalena Szabo, Gilles Gasparoni, Sascha Tierling, Wenqiang Quan, Hsin-Fang Chang, Gang Wu, Julia Schulze-Hentrich, Yang Liu
{"title":"肠上皮Dicer1调控app敲入小鼠肠道微生物组和阿尔茨海默病病理。","authors":"Wenlin Hao, Qinghua Luo, Ilona Magdalena Szabo, Gilles Gasparoni, Sascha Tierling, Wenqiang Quan, Hsin-Fang Chang, Gang Wu, Julia Schulze-Hentrich, Yang Liu","doi":"10.1186/s13195-025-01849-w","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) patients and animal models exhibit an altered gut microbiome that is associated with pathological changes in the brain. Intestinal miRNA enters bacteria and regulates bacterial metabolism and proliferation. This study aimed to investigate whether the manipulation of miRNA could alter the gut microbiome and AD pathologies.</p><p><strong>Methods: </strong>The enzyme producing miRNA was deleted in App-knock-in mice by conditional knock-out of Dicer1 gene in intestinal epithelial cells. 16S rDNA sequencing/microbiome analysis was performed in both the gut and brain. Barrier integrity, inflammatory activation and T cell differentiation in the gut were analyzed by measuring transcripts of relevant marker genes. AD-associated pathologies in the brain, including amyloid pathology, neuroinflammation and synaptic impairment, were investigated by immunohistochemistry, ELISA, quantitative Western blot, mRNA-sequencing/transcriptomic analysis, real-time PCR and behavior tests. To investigate the mechanisms controlling Aβ level, β- and γ-secretase activities, protein levels of LRP1 and ABCB1 in isolated blood microvessels, CD68 immunofluorescence around Aβ deposits and transcription of neprilysin and IDE genes in the brain were analyzed.</p><p><strong>Results: </strong>Deletion of Dicer1 in intestinal epithelial cells of App-knock-in mice reduced the absolute number and altered the composition of bacteria in both the gut and brain, and inhibited inflammatory activation in the gut, but had no effect on the differentiation of CD4-positive T lymphocytes. It lowered Aβ load in the brain, possibly by inhibiting β-secretase activity, and increasing the expression of LRP1 and ABCB1 at the blood-brain barrier. Deletion of intestinal Dicer1 increased Il-10 transcription and decreased Ccl-2 transcription in the brain tissue. Transcriptomic analysis further showed that Dicer1 deletion reduced transcription of Ndufa2 and Ndufa5 genes. In behavior tests, deletion of intestinal Dicer1 induced anxiety symptoms without improving cognitive function in AD mice.</p><p><strong>Conclusions: </strong>Deletion of Dicer1 in intestinal epithelial cells modulates the microbiome in both the gut and brain, and AD pathologies in the brain of App-knock-in mice. Future studies should focus on the identification of AD-specific miRNAs in the gut that can be therapeutically utilized to alter the gut microbiome and prevent AD progression.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":"17 1","pages":"202"},"PeriodicalIF":7.6000,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12418706/pdf/","citationCount":"0","resultStr":"{\"title\":\"Intestinal epithelial Dicer1 regulates gut microbiome and Alzheimer's pathology in App-knock-in mice.\",\"authors\":\"Wenlin Hao, Qinghua Luo, Ilona Magdalena Szabo, Gilles Gasparoni, Sascha Tierling, Wenqiang Quan, Hsin-Fang Chang, Gang Wu, Julia Schulze-Hentrich, Yang Liu\",\"doi\":\"10.1186/s13195-025-01849-w\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Alzheimer's disease (AD) patients and animal models exhibit an altered gut microbiome that is associated with pathological changes in the brain. Intestinal miRNA enters bacteria and regulates bacterial metabolism and proliferation. This study aimed to investigate whether the manipulation of miRNA could alter the gut microbiome and AD pathologies.</p><p><strong>Methods: </strong>The enzyme producing miRNA was deleted in App-knock-in mice by conditional knock-out of Dicer1 gene in intestinal epithelial cells. 16S rDNA sequencing/microbiome analysis was performed in both the gut and brain. Barrier integrity, inflammatory activation and T cell differentiation in the gut were analyzed by measuring transcripts of relevant marker genes. AD-associated pathologies in the brain, including amyloid pathology, neuroinflammation and synaptic impairment, were investigated by immunohistochemistry, ELISA, quantitative Western blot, mRNA-sequencing/transcriptomic analysis, real-time PCR and behavior tests. To investigate the mechanisms controlling Aβ level, β- and γ-secretase activities, protein levels of LRP1 and ABCB1 in isolated blood microvessels, CD68 immunofluorescence around Aβ deposits and transcription of neprilysin and IDE genes in the brain were analyzed.</p><p><strong>Results: </strong>Deletion of Dicer1 in intestinal epithelial cells of App-knock-in mice reduced the absolute number and altered the composition of bacteria in both the gut and brain, and inhibited inflammatory activation in the gut, but had no effect on the differentiation of CD4-positive T lymphocytes. It lowered Aβ load in the brain, possibly by inhibiting β-secretase activity, and increasing the expression of LRP1 and ABCB1 at the blood-brain barrier. Deletion of intestinal Dicer1 increased Il-10 transcription and decreased Ccl-2 transcription in the brain tissue. Transcriptomic analysis further showed that Dicer1 deletion reduced transcription of Ndufa2 and Ndufa5 genes. In behavior tests, deletion of intestinal Dicer1 induced anxiety symptoms without improving cognitive function in AD mice.</p><p><strong>Conclusions: </strong>Deletion of Dicer1 in intestinal epithelial cells modulates the microbiome in both the gut and brain, and AD pathologies in the brain of App-knock-in mice. Future studies should focus on the identification of AD-specific miRNAs in the gut that can be therapeutically utilized to alter the gut microbiome and prevent AD progression.</p>\",\"PeriodicalId\":7516,\"journal\":{\"name\":\"Alzheimer's Research & Therapy\",\"volume\":\"17 1\",\"pages\":\"202\"},\"PeriodicalIF\":7.6000,\"publicationDate\":\"2025-09-09\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12418706/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Alzheimer's Research & Therapy\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s13195-025-01849-w\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CLINICAL NEUROLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Alzheimer's Research & Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13195-025-01849-w","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CLINICAL NEUROLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:阿尔茨海默病(AD)患者和动物模型表现出与大脑病理变化相关的肠道微生物组改变。肠道miRNA进入细菌,调节细菌代谢和增殖。本研究旨在探讨miRNA的操作是否可以改变肠道微生物组和AD病理。方法:通过条件敲除肠上皮细胞Dicer1基因,在app敲入小鼠中缺失产生酶的miRNA。在肠道和大脑中进行16S rDNA测序/微生物组分析。通过测量相关标记基因的转录本,分析肠道屏障完整性、炎症激活和T细胞分化。通过免疫组织化学、ELISA、定量Western blot、mrna测序/转录组学分析、实时荧光定量PCR和行为测试,研究ad在大脑中的相关病理,包括淀粉样蛋白病理、神经炎症和突触损伤。为探讨Aβ水平、β-和γ-分泌酶活性、离体血管LRP1和ABCB1蛋白水平、Aβ沉积物周围CD68免疫荧光以及脑内neprilysin和IDE基因转录的调控机制。结果:app敲入小鼠肠上皮细胞中Dicer1的缺失减少了肠道和脑内细菌的绝对数量,改变了细菌的组成,抑制了肠道内的炎症激活,但对cd4阳性T淋巴细胞的分化没有影响。它可能通过抑制β分泌酶活性,增加血脑屏障处LRP1和ABCB1的表达来降低脑内Aβ负荷。肠Dicer1的缺失增加了脑组织中Il-10的转录,降低了Ccl-2的转录。转录组学分析进一步表明,Dicer1缺失减少了Ndufa2和Ndufa5基因的转录。在行为测试中,肠道Dicer1的缺失会引起AD小鼠的焦虑症状,但不会改善认知功能。结论:肠上皮细胞中Dicer1的缺失调节了app敲入小鼠肠道和大脑的微生物组,以及大脑的AD病理。未来的研究应侧重于鉴定肠道中AD特异性mirna,这些mirna可用于治疗,以改变肠道微生物群并预防AD的进展。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Intestinal epithelial Dicer1 regulates gut microbiome and Alzheimer's pathology in App-knock-in mice.

Background: Alzheimer's disease (AD) patients and animal models exhibit an altered gut microbiome that is associated with pathological changes in the brain. Intestinal miRNA enters bacteria and regulates bacterial metabolism and proliferation. This study aimed to investigate whether the manipulation of miRNA could alter the gut microbiome and AD pathologies.

Methods: The enzyme producing miRNA was deleted in App-knock-in mice by conditional knock-out of Dicer1 gene in intestinal epithelial cells. 16S rDNA sequencing/microbiome analysis was performed in both the gut and brain. Barrier integrity, inflammatory activation and T cell differentiation in the gut were analyzed by measuring transcripts of relevant marker genes. AD-associated pathologies in the brain, including amyloid pathology, neuroinflammation and synaptic impairment, were investigated by immunohistochemistry, ELISA, quantitative Western blot, mRNA-sequencing/transcriptomic analysis, real-time PCR and behavior tests. To investigate the mechanisms controlling Aβ level, β- and γ-secretase activities, protein levels of LRP1 and ABCB1 in isolated blood microvessels, CD68 immunofluorescence around Aβ deposits and transcription of neprilysin and IDE genes in the brain were analyzed.

Results: Deletion of Dicer1 in intestinal epithelial cells of App-knock-in mice reduced the absolute number and altered the composition of bacteria in both the gut and brain, and inhibited inflammatory activation in the gut, but had no effect on the differentiation of CD4-positive T lymphocytes. It lowered Aβ load in the brain, possibly by inhibiting β-secretase activity, and increasing the expression of LRP1 and ABCB1 at the blood-brain barrier. Deletion of intestinal Dicer1 increased Il-10 transcription and decreased Ccl-2 transcription in the brain tissue. Transcriptomic analysis further showed that Dicer1 deletion reduced transcription of Ndufa2 and Ndufa5 genes. In behavior tests, deletion of intestinal Dicer1 induced anxiety symptoms without improving cognitive function in AD mice.

Conclusions: Deletion of Dicer1 in intestinal epithelial cells modulates the microbiome in both the gut and brain, and AD pathologies in the brain of App-knock-in mice. Future studies should focus on the identification of AD-specific miRNAs in the gut that can be therapeutically utilized to alter the gut microbiome and prevent AD progression.

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
Alzheimer's Research & Therapy
Alzheimer's Research & Therapy 医学-神经病学
CiteScore
13.10
自引率
3.30%
发文量
172
审稿时长
>12 weeks
期刊介绍: Alzheimer's Research & Therapy is an international peer-reviewed journal that focuses on translational research into Alzheimer's disease and other neurodegenerative diseases. It publishes open-access basic research, clinical trials, drug discovery and development studies, and epidemiologic studies. The journal also includes reviews, viewpoints, commentaries, debates, and reports. All articles published in Alzheimer's Research & Therapy are included in several reputable databases such as CAS, Current contents, DOAJ, Embase, Journal Citation Reports/Science Edition, MEDLINE, PubMed, PubMed Central, Science Citation Index Expanded (Web of Science) and Scopus.
×
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:604180095
Book学术官方微信