Suppr超能文献

肠道上皮Dicer1调节APP基因敲入小鼠的肠道微生物群和阿尔茨海默病病理。

Intestinal epithelial Dicer1 regulates gut microbiome and Alzheimer's pathology in App-knock-in mice.

作者信息

Hao Wenlin, Luo Qinghua, Szabo Ilona Magdalena, Gasparoni Gilles, Tierling Sascha, Quan Wenqiang, Chang Hsin-Fang, Wu Gang, Schulze-Hentrich Julia, Liu Yang

机构信息

Department of Neurology, Saarland University, Kirrberger Straße, 66421, Homburg/Saar, Germany.

German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany.

出版信息

Alzheimers Res Ther. 2025 Sep 9;17(1):202. doi: 10.1186/s13195-025-01849-w.

Abstract

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.

摘要

背景

阿尔茨海默病(AD)患者及动物模型表现出肠道微生物群改变,这与大脑的病理变化相关。肠道微小RNA(miRNA)进入细菌并调节细菌代谢和增殖。本研究旨在探究操纵miRNA是否能改变肠道微生物群及AD病理状态。

方法

通过条件性敲除肠道上皮细胞中的Dicer1基因,在淀粉样前体蛋白(App)基因敲入小鼠中删除产生miRNA的酶。对肠道和大脑进行16S核糖体DNA测序/微生物群分析。通过测量相关标记基因的转录本,分析肠道中的屏障完整性、炎症激活和T细胞分化。通过免疫组织化学、酶联免疫吸附测定(ELISA)、定量蛋白质免疫印迹、信使核糖核酸(mRNA)测序/转录组分析、实时聚合酶链反应(PCR)和行为测试,研究大脑中与AD相关的病理状态,包括淀粉样病理、神经炎症和突触损伤。为了研究控制淀粉样蛋白β(Aβ)水平、β-和γ-分泌酶活性的机制,分析了分离的血脑微血管中低密度脂蛋白受体相关蛋白1(LRP1)和ATP结合盒转运蛋白B1(ABCB1)的蛋白水平、Aβ沉积物周围的CD68免疫荧光以及大脑中脑啡肽酶和胰岛素降解酶(IDE)基因的转录。

结果

在App基因敲入小鼠的肠道上皮细胞中删除Dicer1可减少肠道和大脑中细菌的绝对数量并改变其组成,抑制肠道中的炎症激活,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-分泌酶活性并增加血脑屏障处LRP1和ABCB1的表达来降低大脑中的Aβ负荷。删除肠道Dicer1可增加脑组织中白细胞介素10(Il-10)的转录并降低趋化因子配体2(Ccl-2)的转录。转录组分析进一步表明,删除Dicer1可降低Ndufa2和Ndufa5基因的转录。在行为测试中,删除肠道Dicer1可诱导AD小鼠出现焦虑症状,但未改善其认知功能。

结论

在肠道上皮细胞中删除Dicer1可调节肠道和大脑中的微生物群以及App基因敲入小鼠大脑中的AD病理状态。未来的研究应聚焦于鉴定肠道中可用于治疗以改变肠道微生物群并预防AD进展的AD特异性miRNA。

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍。

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验