Reconstituting gut microbiota-colonocyte interactions reverses diet-induced cognitive deficits: The beneficial of eucommiae cortex polysaccharides.

IF 12.4 1区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Theranostics Pub Date : 2024-08-01 eCollection Date: 2024-01-01 DOI:10.7150/thno.99468
Mengli Wang, Penghao Sun, Xuejun Chai, Yong-Xin Liu, Luqi Li, Wei Zheng, Shulin Chen, Xiaoyan Zhu, Shanting Zhao
{"title":"Reconstituting gut microbiota-colonocyte interactions reverses diet-induced cognitive deficits: The beneficial of <i>eucommiae cortex</i> polysaccharides.","authors":"Mengli Wang, Penghao Sun, Xuejun Chai, Yong-Xin Liu, Luqi Li, Wei Zheng, Shulin Chen, Xiaoyan Zhu, Shanting Zhao","doi":"10.7150/thno.99468","DOIUrl":null,"url":null,"abstract":"<p><p><b>Rationale:</b> Consumption of a high-fat diet (HFD) has been implicated in cognitive deficits and gastrointestinal dysfunction in humans, with the gut microbiota emerging as a pivotal mediator of these diet-associated pathologies. The introduction of plant-based polysaccharides into the diet as a therapeutic strategy to alleviate such conditions is gaining attention. Nevertheless, the mechanistic paradigm by which polysaccharides modulate the gut microbiota remains largely undefined. This study investigated the mechanisms of action of <i>Eucommiae cortex</i> polysaccharides (EPs) in mitigating gut dysbiosis and examined their contribution to rectifying diet-related cognitive decline. <b>Methods:</b> Initially, we employed fecal microbiota transplantation (FMT) and gut microbiota depletion to verify the causative role of changes in the gut microbiota induced by HFD in synapse engulfment-dependent cognitive impairments. Subsequently, colonization of the gut of chow-fed mice with <i>Escherichia coli</i> (<i>E. coli</i>) from HFD mice confirmed that inhibition of <i>Proteobacteria</i> by EPs was a necessary prerequisite for alleviating HFD-induced cognitive impairments. Finally, supplementation of HFD mice with butyrate and treatment of EPs mice with GW9662 demonstrated that EPs inhibited the expansion of <i>Proteobacteria</i> in the colon of HFD mice by reshaping the interactions between the gut microbiota and colonocytes. <b>Results:</b> Findings from FMT and antibiotic treatments demonstrated that HFD-induced cognitive impairments pertaining to neuronal spine loss were contingent on gut microbial composition. Association analysis revealed strong associations between bacterial taxa belonging to the phylum <i>Proteobacteria</i> and cognitive performance in mice. Further, introducing <i>E. coli</i> from HFD-fed mice into standard diet-fed mice underscored the integral role of <i>Proteobacteria</i> proliferation in triggering excessive synaptic engulfment-related cognitive deficits in HFD mice. Crucially, EPs effectively counteracted the bloom of <i>Proteobacteria</i> and subsequent neuroinflammatory responses mediated by microglia, essential for cognitive improvement in HFD-fed mice. Mechanistic insights revealed that EPs promoted the production of bacteria-derived butyrate, thereby ameliorating HFD-induced colonic mitochondrial dysfunction and reshaping colonocyte metabolism. This adjustment curtailed the availability of growth substrates for facultative anaerobes, which in turn limited the uncontrolled expansion of <i>Proteobacteria</i>. <b>Conclusions:</b> Our study elucidates that colonocyte metabolic disturbances, which promote <i>Proteobacteria</i> overgrowth, are a likely cause of HFD-induced cognitive deficits. Furthermore, dietary supplementation with EPs can rectify behavioral dysfunctions associated with HFD by modifying gut microbiota-colonocyte interactions. These insights contribute to the broader understanding of the modulatory effects of plant prebiotics on the microbiota-gut-brain axis and suggest a potential therapeutic avenue for diet-associated cognitive dysfunction.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":null,"pages":null},"PeriodicalIF":12.4000,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11373620/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Theranostics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/thno.99468","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Rationale: Consumption of a high-fat diet (HFD) has been implicated in cognitive deficits and gastrointestinal dysfunction in humans, with the gut microbiota emerging as a pivotal mediator of these diet-associated pathologies. The introduction of plant-based polysaccharides into the diet as a therapeutic strategy to alleviate such conditions is gaining attention. Nevertheless, the mechanistic paradigm by which polysaccharides modulate the gut microbiota remains largely undefined. This study investigated the mechanisms of action of Eucommiae cortex polysaccharides (EPs) in mitigating gut dysbiosis and examined their contribution to rectifying diet-related cognitive decline. Methods: Initially, we employed fecal microbiota transplantation (FMT) and gut microbiota depletion to verify the causative role of changes in the gut microbiota induced by HFD in synapse engulfment-dependent cognitive impairments. Subsequently, colonization of the gut of chow-fed mice with Escherichia coli (E. coli) from HFD mice confirmed that inhibition of Proteobacteria by EPs was a necessary prerequisite for alleviating HFD-induced cognitive impairments. Finally, supplementation of HFD mice with butyrate and treatment of EPs mice with GW9662 demonstrated that EPs inhibited the expansion of Proteobacteria in the colon of HFD mice by reshaping the interactions between the gut microbiota and colonocytes. Results: Findings from FMT and antibiotic treatments demonstrated that HFD-induced cognitive impairments pertaining to neuronal spine loss were contingent on gut microbial composition. Association analysis revealed strong associations between bacterial taxa belonging to the phylum Proteobacteria and cognitive performance in mice. Further, introducing E. coli from HFD-fed mice into standard diet-fed mice underscored the integral role of Proteobacteria proliferation in triggering excessive synaptic engulfment-related cognitive deficits in HFD mice. Crucially, EPs effectively counteracted the bloom of Proteobacteria and subsequent neuroinflammatory responses mediated by microglia, essential for cognitive improvement in HFD-fed mice. Mechanistic insights revealed that EPs promoted the production of bacteria-derived butyrate, thereby ameliorating HFD-induced colonic mitochondrial dysfunction and reshaping colonocyte metabolism. This adjustment curtailed the availability of growth substrates for facultative anaerobes, which in turn limited the uncontrolled expansion of Proteobacteria. Conclusions: Our study elucidates that colonocyte metabolic disturbances, which promote Proteobacteria overgrowth, are a likely cause of HFD-induced cognitive deficits. Furthermore, dietary supplementation with EPs can rectify behavioral dysfunctions associated with HFD by modifying gut microbiota-colonocyte interactions. These insights contribute to the broader understanding of the modulatory effects of plant prebiotics on the microbiota-gut-brain axis and suggest a potential therapeutic avenue for diet-associated cognitive dysfunction.

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
重建肠道微生物群与结肠细胞的相互作用可逆转饮食引起的认知缺陷:杜仲皮多糖的益处
理由摄入高脂肪饮食(HFD)与人类的认知缺陷和胃肠功能紊乱有关,而肠道微生物群则是这些饮食相关病症的关键介质。在饮食中引入植物多糖作为缓解这些病症的治疗策略正受到越来越多的关注。然而,多糖调节肠道微生物群的机理模式在很大程度上仍未确定。本研究调查了杜仲皮多糖(Eucommiae cortex polysaccharides,EPs)在缓解肠道菌群失调方面的作用机制,并研究了它们在纠正与饮食相关的认知能力下降方面的贡献。研究方法:首先,我们采用粪便微生物群移植(FMT)和肠道微生物群耗竭法来验证高氟日粮引起的肠道微生物群变化在突触吞噬依赖性认知障碍中的致病作用。随后,用来自 HFD 小鼠的大肠埃希氏菌(E. coli)定植喂食饲料的小鼠肠道,证实了 EPs 对蛋白菌的抑制是缓解 HFD 引起的认知障碍的必要前提。最后,给 HFD 小鼠补充丁酸盐以及用 GW9662 处理 EPs 小鼠证明,EPs 通过重塑肠道微生物群和结肠细胞之间的相互作用,抑制了 Proteobacteria 在 HFD 小鼠结肠中的扩张。结果FMT和抗生素治疗的结果表明,HFD诱导的认知障碍与神经元脊柱丧失有关,取决于肠道微生物的组成。关联分析表明,属于变形菌门的细菌类群与小鼠的认知能力有很强的关联。此外,将高纤维食物喂养小鼠体内的大肠杆菌引入标准饮食喂养小鼠体内,凸显了变形杆菌的增殖在引发高纤维食物喂养小鼠与突触吞噬相关的认知缺陷中的重要作用。最重要的是,EPs能有效抵消变形杆菌的繁殖以及随后由小胶质细胞介导的神经炎症反应,这对改善高纤维食物喂养小鼠的认知能力至关重要。机理研究发现,EPs 促进了细菌衍生丁酸盐的产生,从而改善了高氟酸诱导的结肠线粒体功能障碍,并重塑了结肠细胞的新陈代谢。这种调整减少了兼性厌氧菌的生长底物供应,进而限制了变形杆菌的无节制扩张。结论我们的研究阐明,促进变形杆菌过度生长的结肠细胞代谢紊乱很可能是高氟酸膳食诱发认知缺陷的原因。此外,通过改变肠道微生物群与结肠细胞之间的相互作用,膳食中补充 EPs 可纠正与 HFD 相关的行为功能障碍。这些见解有助于人们更广泛地了解植物益生元对微生物群-肠道-大脑轴的调节作用,并为饮食相关的认知功能障碍提供了一种潜在的治疗途径。
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 去求助
来源期刊
Theranostics
Theranostics MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
25.40
自引率
1.60%
发文量
433
审稿时长
1 months
期刊介绍: Theranostics serves as a pivotal platform for the exchange of clinical and scientific insights within the diagnostic and therapeutic molecular and nanomedicine community, along with allied professions engaged in integrating molecular imaging and therapy. As a multidisciplinary journal, Theranostics showcases innovative research articles spanning fields such as in vitro diagnostics and prognostics, in vivo molecular imaging, molecular therapeutics, image-guided therapy, biosensor technology, nanobiosensors, bioelectronics, system biology, translational medicine, point-of-care applications, and personalized medicine. Encouraging a broad spectrum of biomedical research with potential theranostic applications, the journal rigorously peer-reviews primary research, alongside publishing reviews, news, and commentary that aim to bridge the gap between the laboratory, clinic, and biotechnology industries.
期刊最新文献
A hierarchically acidity-unlocking nanoSTING stimulant enables cascaded STING activation for potent innate and adaptive antitumor immunity. Hypoxanthine is a metabolic biomarker for inducing GSDME-dependent pyroptosis of endothelial cells during ischemic stroke. Multifunctionally disordered TiO2 nanoneedles prevent periprosthetic infection and enhance osteointegration by killing bacteria and modulating the osteoimmune microenvironment. Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation. p-STAT3-elevated E3 ubiquitin ligase DTX4 confers the stability of HBV cccDNA by ubiquitinating APOBEC3B in liver.
×
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