首页 > 最新文献

Gut Microbes最新文献

英文 中文
The potential immunological mechanisms of gut microbiota dysbiosis caused by antibiotics exacerbate the lethality of influenza viruses. 抗生素引起的肠道菌群失调的潜在免疫学机制加剧了流感病毒的致命性。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-12-31 Epub Date: 2026-01-02 DOI: 10.1080/19490976.2025.2609451
Jianing Zhu, Zihang Huang, Ying Lin, Jie Zhu, Rui Min, Zibo Wan, Yuting Chen, Jianwen Zhu, Li Xing, Sheng Li, Chinasa Valerie Olovo, Xiaoquan Wang, Guocai Li, Pinghu Zhang

Background: Antibiotics are not recommended to treat influenza A virus (IAV). However, antibiotic misuse for IAV persists worldwide. How to scientifically use antibiotics for IAV-infected patients remains a considerable challenge.

Results: Here, we investigated the impact of antibiotics on viral pathogenicity, pulmonary-intestinal antiviral immunity, and antiviral drug efficacy. Our findings indicated that antibiotic intervention exacerbated IAV-caused mortality and lung injury in mice, manifested as increased mortality rates, shortened survival time, aggravated pulmonary injury, and excessive inflammatory responses. Furthermore, antibiotic pretreatment significantly diminished the efficacy of antivirals. Metagenomic sequencing revealed that antibiotics reduced the diversity and abundance of beneficial gut microbiota, including Lactobacillus and Bifidobacterium, while promoting the proliferation of pathogenic bacteria such as Klebsiella pneumoniae and Escherichia coli. Mechanistically, antibiotic intervention exacerbated IAV-caused excessive inflammatory responses by the blockage of pulmonary-intestinal antiviral immune pathways, which were caused by the upregulation of PKR, RIG-I, ISG15, and TRIM25 levels while downregulating IPS-1 mRNA levels. However, it is noteworthy that the combination of antibiotics and antiviral drugs effectively offset the adverse effects of antibiotic pretreatment on influenza mortality by upregulating IPS-1 levels and partially restoring pulmonary-intestinal immune homeostasis.

Conclusions: Pulmonary-intestinal immune homeostasis imbalance caused by antibiotic misuse can not only markedly exacerbate the lethality of IAV, but also significantly attenuate the efficacy of antiviral drugs. A mechanistic study confirmed that gut microbes dysbiosis caused by antibiotic pretreatment exacerbates the homeostasis imbalance of host antiviral immunity by blocking the RIG/MDA5/IPS-1 antiviral signaling pathway. However, combination therapy with antibiotics and antivirals effectively reversed the fatal outcome exacerbated by antibiotic pretreatment. Collectively, our findings not only provide a scientific explanation from the perspective of antiviral immunity as to why antibiotics should not be arbitrarily used to treat viral infections but also lay the scientific foundation for the rational clinical use of antivirals and antibiotics for treating influenza.

背景:不推荐抗生素治疗甲型流感病毒(IAV)。然而,在世界范围内,抗生素滥用仍然存在。如何科学地使用抗生素治疗iav感染患者仍然是一个相当大的挑战。结果:我们研究了抗生素对病毒致病性、肺-肠抗病毒免疫和抗病毒药物疗效的影响。我们的研究结果表明,抗生素干预加剧了iav引起的小鼠死亡率和肺损伤,表现为死亡率增加,生存时间缩短,肺损伤加重,炎症反应过度。此外,抗生素预处理显著降低了抗病毒药物的疗效。宏基因组测序显示,抗生素降低了有益肠道菌群的多样性和丰度,包括乳酸杆菌和双歧杆菌,同时促进了肺炎克雷伯菌和大肠杆菌等致病菌的增殖。机制上,抗生素干预通过阻断肺-肠抗病毒免疫通路加重了iav引起的过度炎症反应,这是由PKR、RIG-I、ISG15和TRIM25水平上调而下调IPS-1 mRNA水平引起的。然而,值得注意的是,抗生素和抗病毒药物联合使用可通过上调IPS-1水平,部分恢复肺-肠免疫稳态,有效抵消抗生素预处理对流感死亡率的不良影响。结论:抗生素滥用引起的肺-肠免疫平衡失衡不仅会显著加重IAV的致死性,而且会显著减弱抗病毒药物的疗效。一项机制研究证实,抗生素预处理引起的肠道微生物失调通过阻断RIG/MDA5/IPS-1抗病毒信号通路,加剧了宿主抗病毒免疫稳态失衡。然而,抗生素和抗病毒药物联合治疗有效地逆转了抗生素预处理加剧的致命结果。综上所述,我们的研究结果不仅从抗病毒免疫的角度对抗生素不应被任意使用治疗病毒感染提供了科学的解释,而且为临床合理使用抗病毒药物和抗生素治疗流感奠定了科学基础。
{"title":"The potential immunological mechanisms of gut microbiota dysbiosis caused by antibiotics exacerbate the lethality of influenza viruses.","authors":"Jianing Zhu, Zihang Huang, Ying Lin, Jie Zhu, Rui Min, Zibo Wan, Yuting Chen, Jianwen Zhu, Li Xing, Sheng Li, Chinasa Valerie Olovo, Xiaoquan Wang, Guocai Li, Pinghu Zhang","doi":"10.1080/19490976.2025.2609451","DOIUrl":"10.1080/19490976.2025.2609451","url":null,"abstract":"<p><strong>Background: </strong>Antibiotics are not recommended to treat influenza A virus (IAV). However, antibiotic misuse for IAV persists worldwide. How to scientifically use antibiotics for IAV-infected patients remains a considerable challenge.</p><p><strong>Results: </strong>Here, we investigated the impact of antibiotics on viral pathogenicity, pulmonary-intestinal antiviral immunity, and antiviral drug efficacy. Our findings indicated that antibiotic intervention exacerbated IAV-caused mortality and lung injury in mice, manifested as increased mortality rates, shortened survival time, aggravated pulmonary injury, and excessive inflammatory responses. Furthermore, antibiotic pretreatment significantly diminished the efficacy of antivirals. Metagenomic sequencing revealed that antibiotics reduced the diversity and abundance of beneficial gut microbiota, including <i>Lactobacillus</i> and <i>Bifidobacterium</i>, while promoting the proliferation of pathogenic bacteria such as <i>Klebsiella pneumoniae</i> and <i>Escherichia coli</i>. Mechanistically, antibiotic intervention exacerbated IAV-caused excessive inflammatory responses by the blockage of pulmonary-intestinal antiviral immune pathways, which were caused by the upregulation of PKR, RIG-I, ISG15, and TRIM25 levels while downregulating IPS-1 mRNA levels. However, it is noteworthy that the combination of antibiotics and antiviral drugs effectively offset the adverse effects of antibiotic pretreatment on influenza mortality by upregulating IPS-1 levels and partially restoring pulmonary-intestinal immune homeostasis.</p><p><strong>Conclusions: </strong>Pulmonary-intestinal immune homeostasis imbalance caused by antibiotic misuse can not only markedly exacerbate the lethality of IAV, but also significantly attenuate the efficacy of antiviral drugs. A mechanistic study confirmed that gut microbes dysbiosis caused by antibiotic pretreatment exacerbates the homeostasis imbalance of host antiviral immunity by blocking the RIG/MDA5/IPS-1 antiviral signaling pathway. However, combination therapy with antibiotics and antivirals effectively reversed the fatal outcome exacerbated by antibiotic pretreatment. Collectively, our findings not only provide a scientific explanation from the perspective of antiviral immunity as to why antibiotics should not be arbitrarily used to treat viral infections but also lay the scientific foundation for the rational clinical use of antivirals and antibiotics for treating influenza.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"18 1","pages":"2609451"},"PeriodicalIF":11.0,"publicationDate":"2026-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12773635/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145888308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tamoxifen induced hepatotoxicity via gut microbiota-mediated hyodeoxycholic acid depletion and Farnesoid X receptor signaling disruption. 他莫昔芬通过肠道菌群介导的羟脱氧胆酸耗竭和法尼塞德X受体信号中断诱导肝毒性。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-12-31 Epub Date: 2026-01-02 DOI: 10.1080/19490976.2025.2610077
Yuchun Chen, Haiyan Du, Wenxin Zhou, Meirong Qin, Meifang Li, Yibao Jin, Yaning Xu, Chong Ma, Jiaxuan Xia, Yongshi Mo, Ning Chen, Houshuang Huang, Hao Li, Zhiyong Xie, Ping Wang, Yanjun Hong

Tamoxifen (TAM) is a widely used estrogen receptor modulator for breast cancer treatment. However, TAM exhibits significant hepatotoxicity in the clinic, affecting nearly 50% of patients and thereby limiting its clinical utility. The specific mechanisms underlying TAM-induced liver injury remain poorly understood. In this study, we elucidated the mechanistic role of the gut microbiota in the hepatotoxicity associated with TAM. TAM administration induced substantial liver injury and gut microbiota dysbiosis in mice, characterized by an increased abundance of Escherichia and a reduction in Lachnospiraceae NK4A136 group. These microbial shifts resulted in decreased levels of total fecal bile acids (BA), particularly hyodeoxycholic acid (HDCA), which was inversely correlated with TAM-induced liver injury. Additionally, TAM disrupted BA homeostasis by enhancing intestinal Farnesoid X receptor (FXR) activity and concurrently stimulating hepatic BA synthesis through an alternative nonintestinal FXR mechanism. Notably, gut microbiota depletion reversed these effects, demonstrating the critical role of the microbiota in modulating the gut‒liver FXR axis in TAM-induced liver injury. Fecal microbiota transplantation (FMT) further confirmed that TAM directly stimulated hepatic BA synthesis through a microbiota-dependent mechanism. The disruption of the gut‒liver BA‒FXR axis impaired enterohepatic BA circulation, contributing to the liver toxicity associated with TAM administration. Importantly, HDCA supplementation restored the gut‒liver BA‒FXR axis and alleviated TAM-induced liver injury. These findings highlight the intricate relationship between TAM, gut microbiota, and BA metabolism, suggesting that targeting the gut-liver FXR axis with HDCA may serve as a promising therapeutic strategy for alleviating TAM-associated liver injury.

他莫昔芬(TAM)是一种广泛应用于乳腺癌治疗的雌激素受体调节剂。然而,TAM在临床上表现出明显的肝毒性,影响了近50%的患者,从而限制了其临床应用。tam诱导的肝损伤的具体机制尚不清楚。在这项研究中,我们阐明了肠道微生物群在TAM相关肝毒性中的机制作用。TAM在小鼠中引起了严重的肝损伤和肠道微生物群失调,其特征是埃希氏菌的丰度增加和毛螺科NK4A136组的减少。这些微生物变化导致总粪便胆汁酸(BA)水平下降,特别是羟脱氧胆酸(HDCA),这与tam诱导的肝损伤呈负相关。此外,TAM通过增强肠道Farnesoid X受体(FXR)活性,同时通过另一种非肠道FXR机制刺激肝脏BA合成,从而破坏BA稳态。值得注意的是,肠道微生物群的减少逆转了这些效应,证明了微生物群在tam诱导的肝损伤中调节肠-肝FXR轴的关键作用。粪便菌群移植(FMT)进一步证实TAM通过菌群依赖机制直接刺激肝脏BA合成。肠-肝BA - fxr轴的破坏破坏了肠-肝BA循环,导致与TAM给药相关的肝毒性。重要的是,补充HDCA恢复了肠-肝BA-FXR轴,减轻了tam诱导的肝损伤。这些发现强调了TAM、肠道微生物群和BA代谢之间的复杂关系,表明用HDCA靶向肠-肝FXR轴可能是缓解TAM相关肝损伤的一种有希望的治疗策略。
{"title":"Tamoxifen induced hepatotoxicity via gut microbiota-mediated hyodeoxycholic acid depletion and Farnesoid X receptor signaling disruption.","authors":"Yuchun Chen, Haiyan Du, Wenxin Zhou, Meirong Qin, Meifang Li, Yibao Jin, Yaning Xu, Chong Ma, Jiaxuan Xia, Yongshi Mo, Ning Chen, Houshuang Huang, Hao Li, Zhiyong Xie, Ping Wang, Yanjun Hong","doi":"10.1080/19490976.2025.2610077","DOIUrl":"10.1080/19490976.2025.2610077","url":null,"abstract":"<p><p>Tamoxifen (TAM) is a widely used estrogen receptor modulator for breast cancer treatment. However, TAM exhibits significant hepatotoxicity in the clinic, affecting nearly 50% of patients and thereby limiting its clinical utility. The specific mechanisms underlying TAM-induced liver injury remain poorly understood. In this study, we elucidated the mechanistic role of the gut microbiota in the hepatotoxicity associated with TAM. TAM administration induced substantial liver injury and gut microbiota dysbiosis in mice, characterized by an increased abundance of <i>Escherichia</i> and a reduction in <i>Lachnospiraceae NK4A136 group</i>. These microbial shifts resulted in decreased levels of total fecal bile acids (BA), particularly hyodeoxycholic acid (HDCA), which was inversely correlated with TAM-induced liver injury. Additionally, TAM disrupted BA homeostasis by enhancing intestinal Farnesoid X receptor (FXR) activity and concurrently stimulating hepatic BA synthesis through an alternative nonintestinal FXR mechanism. Notably, gut microbiota depletion reversed these effects, demonstrating the critical role of the microbiota in modulating the gut‒liver FXR axis in TAM-induced liver injury. Fecal microbiota transplantation (FMT) further confirmed that TAM directly stimulated hepatic BA synthesis through a microbiota-dependent mechanism. The disruption of the gut‒liver BA‒FXR axis impaired enterohepatic BA circulation, contributing to the liver toxicity associated with TAM administration. Importantly, HDCA supplementation restored the gut‒liver BA‒FXR axis and alleviated TAM-induced liver injury. These findings highlight the intricate relationship between TAM, gut microbiota, and BA metabolism, suggesting that targeting the gut-liver FXR axis with HDCA may serve as a promising therapeutic strategy for alleviating TAM-associated liver injury.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"18 1","pages":"2610077"},"PeriodicalIF":11.0,"publicationDate":"2026-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12773634/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892328","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Formate as electron carrier in the gut acetogen Blautia luti: a model for electron transfer in the gut microbiome. 甲酸盐作为电子载体在肠道菌蓝藻中:肠道微生物组中电子转移的模型。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-12-31 Epub Date: 2026-01-02 DOI: 10.1080/19490976.2025.2609406
Raphael Trischler, Volker Müller

Species of the genus Blautia are commonly found in the human gut and are known to be beneficial for the human well-being. However, only little is known about the physiology and the specific role of Blautia species in the human gut. In this study, we investigated the heterotrophic metabolism of the formate dehydrogenase lacking gut acetogen Blautia luti. We identified acetate, succinate, lactate, formate, and hydrogen as end products of sugar fermentation. Interestingly, formate is produced by the pyruvate-formate lyase reaction and used as electron acceptor in the Wood-Ljungdahl pathway of CO2 fixation. Thus, formate connects the oxidative branch of glucose metabolism with the reductive branch. The use of formate as an intraspecies electron carrier seems to be common in gut acetogens. This study highlights the role of formate as electron carrier in the gut microbiome and improves our understanding of the physiology of Blautia species in the human gut. It also introduces B. luti as potential candidate for biotechnological applications due to the production of highly desired succinate.

Blautia属的物种通常在人类肠道中发现,并且已知对人类健康有益。然而,人们对蓝藻的生理和在人体肠道中的具体作用知之甚少。在这项研究中,我们研究了甲酸脱氢酶的异养代谢缺乏肠道醋酸蓝藻。我们确定醋酸盐、琥珀酸盐、乳酸盐、甲酸盐和氢是糖发酵的最终产物。有趣的是,甲酸是由丙酮酸-甲酸裂解酶反应产生的,并在Wood-Ljungdahl途径中作为电子受体固定CO2。因此,甲酸酯连接了葡萄糖代谢的氧化分支和还原分支。使用甲酸作为种内电子载体似乎是常见的肠道菌。这项研究强调了甲酸在肠道微生物组中作为电子载体的作用,并提高了我们对人体肠道中蓝藻物种生理的理解。它还介绍了卢氏杆菌作为生物技术应用的潜在候选者,由于生产高度期望的琥珀酸盐。
{"title":"Formate as electron carrier in the gut acetogen <i>Blautia luti</i>: a model for electron transfer in the gut microbiome.","authors":"Raphael Trischler, Volker Müller","doi":"10.1080/19490976.2025.2609406","DOIUrl":"10.1080/19490976.2025.2609406","url":null,"abstract":"<p><p>Species of the genus <i>Blautia</i> are commonly found in the human gut and are known to be beneficial for the human well-being. However, only little is known about the physiology and the specific role of <i>Blautia</i> species in the human gut. In this study, we investigated the heterotrophic metabolism of the formate dehydrogenase lacking gut acetogen <i>Blautia luti</i>. We identified acetate, succinate, lactate, formate, and hydrogen as end products of sugar fermentation. Interestingly, formate is produced by the pyruvate-formate lyase reaction and used as electron acceptor in the Wood-Ljungdahl pathway of CO<sub>2</sub> fixation. Thus, formate connects the oxidative branch of glucose metabolism with the reductive branch. The use of formate as an intraspecies electron carrier seems to be common in gut acetogens. This study highlights the role of formate as electron carrier in the gut microbiome and improves our understanding of the physiology of <i>Blautia</i> species in the human gut. It also introduces <i>B. luti</i> as potential candidate for biotechnological applications due to the production of highly desired succinate.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"18 1","pages":"2609406"},"PeriodicalIF":11.0,"publicationDate":"2026-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12773636/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The gut microbiota and host immunity synergistically orchestrate colonization resistance. 肠道菌群和宿主免疫协同协调定植抗性。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-12-31 Epub Date: 2026-01-03 DOI: 10.1080/19490976.2025.2611545
Na Li, Xiaohuan Guo

Colonization resistance is a fundamental host defense mechanism that relies on the synergistic interaction between the gut microbiota and the host immune system to prevent enteric pathogen colonization and infection. This review synthesizes current knowledge of the multifaceted mechanisms governing colonization resistance against intestinal pathogens. We examine how commensal microbes directly suppress pathogens through niche and nutrient competition, contact-dependent inhibition, and the production of antimicrobial compounds and metabolites. From the host perspective, we outline the essential roles of gut barriers, innate and adaptive immunity, and antimicrobial peptides in maintaining microbiota homeostasis while selectively restricting pathogen expansion. We also emphasize the role of IL-22 signaling and its regulation of epithelial glycosylation, which modulates nutrient availability and shapes microbial competitiveness. Finally, we discuss key challenges and future research directions in colonization resistance and related translational research, with the goal of informing novel strategies to prevent and treat intestinal infections and inflammatory diseases.

定植抗性是宿主的一种基本防御机制,它依赖于肠道微生物群与宿主免疫系统之间的协同相互作用来阻止肠道病原体的定植和感染。这篇综述综合了目前对肠道病原体定植耐药的多方面机制的了解。我们研究了共生微生物如何通过生态位和营养竞争、接触依赖性抑制以及抗菌化合物和代谢物的产生直接抑制病原体。从宿主的角度来看,我们概述了肠道屏障、先天免疫和适应性免疫以及抗菌肽在维持微生物群稳态和选择性限制病原体扩张方面的重要作用。我们还强调了IL-22信号的作用及其对上皮糖基化的调节,这调节了营养物质的可用性和形成了微生物的竞争力。最后,我们讨论了定植抵抗和相关转化研究的关键挑战和未来研究方向,目的是为预防和治疗肠道感染和炎症性疾病提供新的策略。
{"title":"The gut microbiota and host immunity synergistically orchestrate colonization resistance.","authors":"Na Li, Xiaohuan Guo","doi":"10.1080/19490976.2025.2611545","DOIUrl":"10.1080/19490976.2025.2611545","url":null,"abstract":"<p><p>Colonization resistance is a fundamental host defense mechanism that relies on the synergistic interaction between the gut microbiota and the host immune system to prevent enteric pathogen colonization and infection. This review synthesizes current knowledge of the multifaceted mechanisms governing colonization resistance against intestinal pathogens. We examine how commensal microbes directly suppress pathogens through niche and nutrient competition, contact-dependent inhibition, and the production of antimicrobial compounds and metabolites. From the host perspective, we outline the essential roles of gut barriers, innate and adaptive immunity, and antimicrobial peptides in maintaining microbiota homeostasis while selectively restricting pathogen expansion. We also emphasize the role of IL-22 signaling and its regulation of epithelial glycosylation, which modulates nutrient availability and shapes microbial competitiveness. Finally, we discuss key challenges and future research directions in colonization resistance and related translational research, with the goal of informing novel strategies to prevent and treat intestinal infections and inflammatory diseases.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"18 1","pages":"2611545"},"PeriodicalIF":11.0,"publicationDate":"2026-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12773493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892457","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Limosilactobacillus mucosae attenuates hyperlipidemic periodontitis via the gut-oral axis. 粘膜乳酸杆菌通过肠道-口腔轴减轻高脂血症牙周炎。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-20 DOI: 10.1080/19490976.2026.2617699
Jing Xu,Ziyi Han,Qing Xue,Haoran Wang,Jiyu Song,Yutong Li,Yongfeng Zhang,Di Wang,Min Hu
The link between hyperlipidemia and periodontitis is well-established, but the underlying mechanisms remain incompletely understood. Here, we reveal a critical role for a 'gut-oral' axis in mediating this interaction. Integrating multi-omics analyses of clinical samples and mouse models, we identified that a significant reduction of intestinal Limosilactobacillus mucosae is a key feature of hyperlipidemic periodontitis (HPD). Fecal microbiota transplantation established a causal link between this gut dysbiosis and exacerbated periodontitis. Mechanistically, oral administration of live L. mucosae ameliorates HPD by restoring intestinal levels of the key metabolite, glycerophosphocholine (α-GPC). Notably, supplementation with α-GPC alone recapitulated this protective effect by upregulating the tight junction protein Claudin-1 (CLDN1) in periodontal tissue. This reinforcement of the epithelial barrier curtailed inflammatory infiltration and restored bone homeostasis. Our findings uncover a protective 'L. mucosae-α-GPC-CLDN1' axis, providing mechanistic insight into how gut microbiota mediates metabolism-associated inflammation and proposing a potential therapeutic strategy for HPD.
高脂血症和牙周炎之间的联系是公认的,但其潜在的机制仍然不完全清楚。在这里,我们揭示了“肠-口”轴在调解这种相互作用中的关键作用。综合临床样本和小鼠模型的多组学分析,我们发现肠道粘膜乳酸杆菌的显著减少是高脂血症牙周炎(HPD)的一个关键特征。粪便菌群移植建立了肠道生态失调和牙周炎加重之间的因果关系。从机制上说,口服活乳杆菌粘膜通过恢复肠道中关键代谢物甘油酰胆碱(α-GPC)的水平来改善HPD。值得注意的是,单独补充α-GPC通过上调牙周组织中的紧密连接蛋白CLDN1来重现这种保护作用。这种上皮屏障的强化减少了炎症浸润并恢复了骨稳态。我们的研究结果揭示了保护性的“L. mucosae-α-GPC-CLDN1”轴,为肠道微生物群如何介导代谢相关炎症提供了机制见解,并提出了HPD的潜在治疗策略。
{"title":"Limosilactobacillus mucosae attenuates hyperlipidemic periodontitis via the gut-oral axis.","authors":"Jing Xu,Ziyi Han,Qing Xue,Haoran Wang,Jiyu Song,Yutong Li,Yongfeng Zhang,Di Wang,Min Hu","doi":"10.1080/19490976.2026.2617699","DOIUrl":"https://doi.org/10.1080/19490976.2026.2617699","url":null,"abstract":"The link between hyperlipidemia and periodontitis is well-established, but the underlying mechanisms remain incompletely understood. Here, we reveal a critical role for a 'gut-oral' axis in mediating this interaction. Integrating multi-omics analyses of clinical samples and mouse models, we identified that a significant reduction of intestinal Limosilactobacillus mucosae is a key feature of hyperlipidemic periodontitis (HPD). Fecal microbiota transplantation established a causal link between this gut dysbiosis and exacerbated periodontitis. Mechanistically, oral administration of live L. mucosae ameliorates HPD by restoring intestinal levels of the key metabolite, glycerophosphocholine (α-GPC). Notably, supplementation with α-GPC alone recapitulated this protective effect by upregulating the tight junction protein Claudin-1 (CLDN1) in periodontal tissue. This reinforcement of the epithelial barrier curtailed inflammatory infiltration and restored bone homeostasis. Our findings uncover a protective 'L. mucosae-α-GPC-CLDN1' axis, providing mechanistic insight into how gut microbiota mediates metabolism-associated inflammation and proposing a potential therapeutic strategy for HPD.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"6 1","pages":"2617699"},"PeriodicalIF":12.2,"publicationDate":"2026-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"146005013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gut microbiome remodeling induced by microplastic exposure in humans. 人体微塑料暴露引起的肠道微生物群重塑。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-19 DOI: 10.1080/19490976.2026.2617696
Xin-Yue Yang,Zhong-Wei Zhang,Guang-Deng Chen,Shu Yuan
The impact of microplastics (MPs) on the diversity and composition of the gut microbiome has been extensively documented in animal studies, but evidence in humans remains limited. Recognizing the potential differences in MP effects between animal and human gut microbiomes, this review synthesizes current evidence concerning their impact on the human gut microbiota. Furthermore, the potential links between microplastic-induced dysbiosis and the pathogenesis of human diseases were analyzed. Cross-sectional studies have been conducted to explore microplastic exposures (such as in humans who consume hot foods served in disposable plastic tableware) and their associations with gut microbiome functionalities in infants, preschool children and adults. Exposure to MPs increased the abundance of Dethiosulfovibrionaceae, Enterobacteriaceae, Moraxellaceae, Actinomycetota, Pseudomonadota, and Veillonella. On the other hand, MPs decreased the abundances of Bacillota, Bacteroidota, Lactobacillales, Rikenellaceae, Parabacteroides, Roseburia, Coprococcus, Turicibacter, and Eubacterium coprostanoligenes. These changes were associated with a decrease in butyrate production and a decrease in short-chain fatty acid levels. However, for some other bacteria, both inductive (on Oscillospiraceae, Adlercreutzia, Phascolarctobacterium, and Collinsella) and repressive effects (on Streptococcus) have been documented. There are contradictory reports about MP-induced changes in Lachnospiraceae (including the Dorea genus), Alistipes and Faecalibacterium, which may be correlated with obesity, gastrointestinal dysfunction, some cancers, inflammatory bowel disease and Crohn's disease. Potential reasons for these discrepancies are proposed. This review also examines putative mechanisms, with a focus on biofilm formation on selective surfaces, and discusses the inherent limitations of current MP exposure assessments in human gut microbiota studies.
微塑料(MPs)对肠道微生物群多样性和组成的影响在动物研究中得到了广泛的记录,但在人类身上的证据仍然有限。认识到动物和人类肠道微生物群之间MP效应的潜在差异,本综述综合了目前关于它们对人类肠道微生物群影响的证据。此外,还分析了微塑诱导的生态失调与人类疾病发病机制之间的潜在联系。已经进行了横断面研究,以探索微塑料暴露(例如食用用一次性塑料餐具盛装的热食物的人类)及其与婴儿、学龄前儿童和成人肠道微生物群功能的关系。暴露于MPs增加了Dethiosulfovibrionaceae, enterobacteraceae, Moraxellaceae,放线菌,Pseudomonadota和Veillonella的丰度。另一方面,MPs降低了Bacillota、Bacteroidota、Lactobacillales、Rikenellaceae、Parabacteroides、Roseburia、Coprococcus、Turicibacter和Eubacterium coprostanoligene的丰度。这些变化与丁酸盐产量的减少和短链脂肪酸水平的降低有关。然而,对于其他一些细菌,诱导作用(对示波螺旋科,阿德勒克鲁茨亚菌,Phascolarctobacterium和Collinsella)和抑制作用(对链球菌)已被记录。关于mp诱导毛螺杆菌科(包括Dorea属)、Alistipes和Faecalibacterium发生变化的报道相互矛盾,这些变化可能与肥胖、胃肠功能障碍、某些癌症、炎症性肠病和克罗恩病有关。提出了造成这些差异的潜在原因。这篇综述还探讨了可能的机制,重点是在选择性表面上形成生物膜,并讨论了目前在人类肠道微生物群研究中MP暴露评估的固有局限性。
{"title":"Gut microbiome remodeling induced by microplastic exposure in humans.","authors":"Xin-Yue Yang,Zhong-Wei Zhang,Guang-Deng Chen,Shu Yuan","doi":"10.1080/19490976.2026.2617696","DOIUrl":"https://doi.org/10.1080/19490976.2026.2617696","url":null,"abstract":"The impact of microplastics (MPs) on the diversity and composition of the gut microbiome has been extensively documented in animal studies, but evidence in humans remains limited. Recognizing the potential differences in MP effects between animal and human gut microbiomes, this review synthesizes current evidence concerning their impact on the human gut microbiota. Furthermore, the potential links between microplastic-induced dysbiosis and the pathogenesis of human diseases were analyzed. Cross-sectional studies have been conducted to explore microplastic exposures (such as in humans who consume hot foods served in disposable plastic tableware) and their associations with gut microbiome functionalities in infants, preschool children and adults. Exposure to MPs increased the abundance of Dethiosulfovibrionaceae, Enterobacteriaceae, Moraxellaceae, Actinomycetota, Pseudomonadota, and Veillonella. On the other hand, MPs decreased the abundances of Bacillota, Bacteroidota, Lactobacillales, Rikenellaceae, Parabacteroides, Roseburia, Coprococcus, Turicibacter, and Eubacterium coprostanoligenes. These changes were associated with a decrease in butyrate production and a decrease in short-chain fatty acid levels. However, for some other bacteria, both inductive (on Oscillospiraceae, Adlercreutzia, Phascolarctobacterium, and Collinsella) and repressive effects (on Streptococcus) have been documented. There are contradictory reports about MP-induced changes in Lachnospiraceae (including the Dorea genus), Alistipes and Faecalibacterium, which may be correlated with obesity, gastrointestinal dysfunction, some cancers, inflammatory bowel disease and Crohn's disease. Potential reasons for these discrepancies are proposed. This review also examines putative mechanisms, with a focus on biofilm formation on selective surfaces, and discusses the inherent limitations of current MP exposure assessments in human gut microbiota studies.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"41 1","pages":"2617696"},"PeriodicalIF":12.2,"publicationDate":"2026-01-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145994918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Diet, gut microbiome, and type 1 diabetes: from risk to translational opportunity. 饮食、肠道微生物群和1型糖尿病:从风险到转化机会。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-15 DOI: 10.1080/19490976.2026.2614039
Khyati Girdhar,Sandra Dedrick,Lukas Rhodes,David Kim,Amaya Powis,Caitlin Mahon,Hannah Chapdelaine,Liane Obaid,Meghan McNamara,Emrah Altindis
The incidence of type 1 diabetes (T1D) has risen sharply in recent decades, implicating the role of environmental factors in disease pathogenesis. Diet, a primary driver of gut microbiome development and composition, along with other environmental exposures, has emerged as a potential modulator of T1D risk and progression. While nutrients, such as certain vitamins, may exert protective effects, the roles of other dietary factors (e.g., early exposure to dietary antigens) remain unclear. Importantly, diet shapes the gut microbiome, which produces immunomodulatory metabolites, including secondary bile acids, short-chain fatty acids (SCFAs), and others that directly influence immune responses. This review presents evidence on how specific dietary factors, including macronutrients (fats, carbohydrates, proteins, such as gluten and milk proteins), fibers, and breastfeeding, affect the gut microbiome and T1D. We also discuss the effects of microbiome-targeted interventions, including probiotics, prebiotics, and fecal microbiota transplantation, on T1D and their potential as future therapeutic strategies. Although animal studies provide compelling mechanistic insights, the results from human trials remain inconsistent, underscoring the urgent need for longitudinal and interventional studies to establish causality. Understanding the complex interplay between diet, the gut microbiome, and immune homeostasis is essential for developing personalized strategies to prevent and treat T1D and delay-related complications.
近几十年来,1型糖尿病(T1D)的发病率急剧上升,提示环境因素在疾病发病机制中的作用。饮食是肠道微生物群发育和组成的主要驱动因素,与其他环境暴露一起,已成为T1D风险和进展的潜在调节剂。虽然营养素(如某些维生素)可能发挥保护作用,但其他饮食因素(如早期接触饮食抗原)的作用仍不清楚。重要的是,饮食塑造了肠道微生物群,产生免疫调节代谢物,包括次级胆汁酸、短链脂肪酸(SCFAs)和其他直接影响免疫反应的代谢物。这篇综述提供了具体的饮食因素,包括常量营养素(脂肪、碳水化合物、蛋白质,如麸质和乳蛋白)、纤维和母乳喂养如何影响肠道微生物群和T1D的证据。我们还讨论了针对微生物群的干预措施,包括益生菌、益生元和粪便微生物群移植,对T1D的影响及其作为未来治疗策略的潜力。尽管动物研究提供了令人信服的机制见解,但人体试验的结果仍然不一致,强调迫切需要纵向和干入性研究来确定因果关系。了解饮食、肠道微生物群和免疫稳态之间复杂的相互作用,对于制定个性化策略来预防和治疗T1D和延迟相关并发症至关重要。
{"title":"Diet, gut microbiome, and type 1 diabetes: from risk to translational opportunity.","authors":"Khyati Girdhar,Sandra Dedrick,Lukas Rhodes,David Kim,Amaya Powis,Caitlin Mahon,Hannah Chapdelaine,Liane Obaid,Meghan McNamara,Emrah Altindis","doi":"10.1080/19490976.2026.2614039","DOIUrl":"https://doi.org/10.1080/19490976.2026.2614039","url":null,"abstract":"The incidence of type 1 diabetes (T1D) has risen sharply in recent decades, implicating the role of environmental factors in disease pathogenesis. Diet, a primary driver of gut microbiome development and composition, along with other environmental exposures, has emerged as a potential modulator of T1D risk and progression. While nutrients, such as certain vitamins, may exert protective effects, the roles of other dietary factors (e.g., early exposure to dietary antigens) remain unclear. Importantly, diet shapes the gut microbiome, which produces immunomodulatory metabolites, including secondary bile acids, short-chain fatty acids (SCFAs), and others that directly influence immune responses. This review presents evidence on how specific dietary factors, including macronutrients (fats, carbohydrates, proteins, such as gluten and milk proteins), fibers, and breastfeeding, affect the gut microbiome and T1D. We also discuss the effects of microbiome-targeted interventions, including probiotics, prebiotics, and fecal microbiota transplantation, on T1D and their potential as future therapeutic strategies. Although animal studies provide compelling mechanistic insights, the results from human trials remain inconsistent, underscoring the urgent need for longitudinal and interventional studies to establish causality. Understanding the complex interplay between diet, the gut microbiome, and immune homeostasis is essential for developing personalized strategies to prevent and treat T1D and delay-related complications.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"141 1","pages":"2614039"},"PeriodicalIF":12.2,"publicationDate":"2026-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145968388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
metaFun: An analysis pipeline for metagenomic big data with fast and unified functional searches. metaFun:提供快速统一功能搜索的宏基因组大数据分析管道。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-13 DOI: 10.1080/19490976.2025.2611544
Hyeon Gwon Lee,Ju Yeon Song,Jaekyung Yoon,Yusook Chung,Soon-Kyeong Kwon,Jihyun F Kim
Metagenomic approaches offer unprecedented opportunities to characterize microbial community structure and function, yet several challenges remain unresolved. Inconsistent genome quality impairs reliability of metagenome-assembled genomes, lack of unified taxonomic criteria limits cross-study comparability, and multi-step workflows involving numerous programs and parameters hinder reproducibility and accessibility. We benchmarked existing programs and parameters using simulated metagenomic data to identify optimal configurations. metaFun is an open-source, end-to-end pipeline that integrates quality control, taxonomic profiling, functional profiling, de novo assembly, binning, genome assessment, comparative genomic analysis, pangenome annotation, network analysis, and strain-level microdiversity analysis into a unified framework. Interactive modules support standardized data interpretation and exploratory visualization. The pipeline is implemented with Nextflow and containerized with Apptainer, ensuring environment reproducibility and scalability. Comprehensive documentation is available at https://metafun-doc.readthedocs.io/en/main. The pipeline was validated using a colorectal cancer cohort dataset. By addressing key methodological gaps, metaFun facilitates accessible and reproducible metagenomic analysis for the broader research community.
宏基因组方法为表征微生物群落结构和功能提供了前所未有的机会,但仍有一些挑战尚未解决。不一致的基因组质量损害了宏基因组组装基因组的可靠性,缺乏统一的分类标准限制了交叉研究的可比性,涉及众多程序和参数的多步骤工作流程阻碍了可重复性和可及性。我们使用模拟宏基因组数据对现有程序和参数进行基准测试,以确定最佳配置。metaFun是一个开源的端到端管道,它将质量控制、分类分析、功能分析、从头组装、分组、基因组评估、比较基因组分析、泛基因组注释、网络分析和品系级微多样性分析集成到一个统一的框架中。交互模块支持标准化的数据解释和探索性可视化。该管道由Nextflow实现,并由Apptainer容器化,确保了环境的可重复性和可扩展性。全面的文档可在https://metafun-doc.readthedocs.io/en/main上获得。该管道使用结直肠癌队列数据集进行验证。通过解决关键的方法差距,metaFun为更广泛的研究界提供了可访问和可重复的宏基因组分析。
{"title":"metaFun: An analysis pipeline for metagenomic big data with fast and unified functional searches.","authors":"Hyeon Gwon Lee,Ju Yeon Song,Jaekyung Yoon,Yusook Chung,Soon-Kyeong Kwon,Jihyun F Kim","doi":"10.1080/19490976.2025.2611544","DOIUrl":"https://doi.org/10.1080/19490976.2025.2611544","url":null,"abstract":"Metagenomic approaches offer unprecedented opportunities to characterize microbial community structure and function, yet several challenges remain unresolved. Inconsistent genome quality impairs reliability of metagenome-assembled genomes, lack of unified taxonomic criteria limits cross-study comparability, and multi-step workflows involving numerous programs and parameters hinder reproducibility and accessibility. We benchmarked existing programs and parameters using simulated metagenomic data to identify optimal configurations. metaFun is an open-source, end-to-end pipeline that integrates quality control, taxonomic profiling, functional profiling, de novo assembly, binning, genome assessment, comparative genomic analysis, pangenome annotation, network analysis, and strain-level microdiversity analysis into a unified framework. Interactive modules support standardized data interpretation and exploratory visualization. The pipeline is implemented with Nextflow and containerized with Apptainer, ensuring environment reproducibility and scalability. Comprehensive documentation is available at https://metafun-doc.readthedocs.io/en/main. The pipeline was validated using a colorectal cancer cohort dataset. By addressing key methodological gaps, metaFun facilitates accessible and reproducible metagenomic analysis for the broader research community.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"13 1","pages":"2611544"},"PeriodicalIF":12.2,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145961499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A novel Mediterranean diet-inspired supplement reduces hippocampal amyloid deposits and microglial activation through the modulation of the microbiota gut-brain axis in 5xFAD mice. 在5xFAD小鼠中,一种新的地中海饮食启发补充剂通过调节微生物群肠-脑轴减少海马淀粉样蛋白沉积和小胶质细胞激活。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-13 DOI: 10.1080/19490976.2026.2614030
Emily Connell,Gwénaëlle Le Gall,Simon McArthur,Leonie Lang,Bernadette Breeze,Marrium Liaquat,Matthew G Pontifex,Saber Sami,Line Pourtau,David Gaudout,Michael Müller,David Vauzour
BACKGROUNDAlzheimer's disease (AD) is projected to increase in prevalence, heightening the need for strategies to alleviate its neuropathological burden. The bioactive constituents of a Mediterranean-style diet are well-recognised for their neuroprotective properties. Due to their capacity to alter the gut microbiome composition, these benefits may involve modulation of the microbiota-gut-brain axis. In this study, we investigated whether a novel supplement enriched with key Mediterranean diet-derived bioactives (Neurosyn240) could reduce amyloid deposition and microglial activation in 5xFAD mice, a transgenic model of AD, through microbiota-mediated mechanisms.METHODSMale and female 5xFAD transgenic mice (n = 16 per sex) were randomly assigned to receive either a standard control diet or a diet supplemented with Neurosyn240 for 12 weeks. Employing a multi-omics approach, gut microbiota composition was profiled using 16S rRNA ampliconsequencing, serum metabolites were quantified via targeted metabolomics, and hippocampal gene expression was analysed through qPCR and RNA sequencing. Neuropathological markers, including amyloid-β deposition and microglial activation, were evaluated using immunofluorescence staining. Statistical analyses were performed using two-way ANOVA to examine the main effects of diet and sex and their interaction.RESULTSNeurosyn240 significantly shifted the gut microbiome composition, which was associated with increased circulatory serotonin levels and decreased kynurenine and bile acids (TCA, HDCA, TDCA, CDCA and LCA) concentrations. In the brain, Neurosyn240 consumption led to a significant reduction in hippocampal amyloid deposits and Iba-1 positive microglia (p<0.05), which were associated with decreased LCA and increased serotonin, respectively. Hippocampal RNA sequencing further highlighted the upregulation of genes involved in promoting amyloid beta clearance mechanisms.CONCLUSIONSTogether, these findings highlight novel neuroprotective effects of Neurosyn240 in modulating metabolite-mediated pathways of the microbiota-gut-brain axis, accentuating its therapeutic potential against AD progression.
背景:预计阿尔茨海默病(AD)的患病率将会增加,因此需要采取策略来减轻其神经病理负担。地中海式饮食的生物活性成分因其神经保护特性而得到广泛认可。由于其改变肠道微生物组成的能力,这些益处可能涉及微生物-肠-脑轴的调节。在这项研究中,我们研究了一种富含关键地中海饮食来源的生物活性物质(Neurosyn240)的新型补充剂是否能通过微生物介导的机制减少5xFAD小鼠(AD转基因模型)的淀粉样蛋白沉积和小胶质细胞激活。方法将5xFAD转基因小鼠雌雄各16只,随机分为两组,分别饲喂标准对照饮食和添加Neurosyn240的饮食,为期12周。采用多组学方法,通过16S rRNA扩增测序分析肠道微生物群组成,通过靶向代谢组学定量血清代谢物,通过qPCR和RNA测序分析海马基因表达。使用免疫荧光染色评估神经病理学标志物,包括淀粉样蛋白-β沉积和小胶质细胞活化。采用双向方差分析进行统计分析,以检验饮食和性别的主要影响及其相互作用。结果neurosyn240显著改变了肠道菌群组成,增加了循环血清素水平,降低了犬尿氨酸和胆汁酸(TCA、HDCA、TDCA、CDCA和LCA)浓度。在大脑中,Neurosyn240的摄入导致海马淀粉样蛋白沉积和Iba-1阳性小胶质细胞的显著减少(p<0.05),这分别与LCA的降低和血清素的增加有关。海马RNA测序进一步强调了参与促进淀粉样蛋白清除机制的基因的上调。综上所述,这些发现突出了Neurosyn240在调节微生物-肠-脑轴代谢物介导的通路中的新的神经保护作用,强调了其治疗AD进展的潜力。
{"title":"A novel Mediterranean diet-inspired supplement reduces hippocampal amyloid deposits and microglial activation through the modulation of the microbiota gut-brain axis in 5xFAD mice.","authors":"Emily Connell,Gwénaëlle Le Gall,Simon McArthur,Leonie Lang,Bernadette Breeze,Marrium Liaquat,Matthew G Pontifex,Saber Sami,Line Pourtau,David Gaudout,Michael Müller,David Vauzour","doi":"10.1080/19490976.2026.2614030","DOIUrl":"https://doi.org/10.1080/19490976.2026.2614030","url":null,"abstract":"BACKGROUNDAlzheimer's disease (AD) is projected to increase in prevalence, heightening the need for strategies to alleviate its neuropathological burden. The bioactive constituents of a Mediterranean-style diet are well-recognised for their neuroprotective properties. Due to their capacity to alter the gut microbiome composition, these benefits may involve modulation of the microbiota-gut-brain axis. In this study, we investigated whether a novel supplement enriched with key Mediterranean diet-derived bioactives (Neurosyn240) could reduce amyloid deposition and microglial activation in 5xFAD mice, a transgenic model of AD, through microbiota-mediated mechanisms.METHODSMale and female 5xFAD transgenic mice (n = 16 per sex) were randomly assigned to receive either a standard control diet or a diet supplemented with Neurosyn240 for 12 weeks. Employing a multi-omics approach, gut microbiota composition was profiled using 16S rRNA ampliconsequencing, serum metabolites were quantified via targeted metabolomics, and hippocampal gene expression was analysed through qPCR and RNA sequencing. Neuropathological markers, including amyloid-β deposition and microglial activation, were evaluated using immunofluorescence staining. Statistical analyses were performed using two-way ANOVA to examine the main effects of diet and sex and their interaction.RESULTSNeurosyn240 significantly shifted the gut microbiome composition, which was associated with increased circulatory serotonin levels and decreased kynurenine and bile acids (TCA, HDCA, TDCA, CDCA and LCA) concentrations. In the brain, Neurosyn240 consumption led to a significant reduction in hippocampal amyloid deposits and Iba-1 positive microglia (p<0.05), which were associated with decreased LCA and increased serotonin, respectively. Hippocampal RNA sequencing further highlighted the upregulation of genes involved in promoting amyloid beta clearance mechanisms.CONCLUSIONSTogether, these findings highlight novel neuroprotective effects of Neurosyn240 in modulating metabolite-mediated pathways of the microbiota-gut-brain axis, accentuating its therapeutic potential against AD progression.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"385 1","pages":"2614030"},"PeriodicalIF":12.2,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145956206","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Maternal prenatal co-exposure to air pollution and psychological distress shapes the neonatal gut: microbiota-mediated pathways to early neurodevelopment. 母亲产前共同暴露于空气污染和心理困扰塑造新生儿肠道:微生物介导的早期神经发育途径。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2026-01-13 DOI: 10.1080/19490976.2026.2614451
Pu Yang,Yifei Pei,Yongqi Huang,Mengyuan Dong,Fangming Cui,Shuang Nie,Xuan Zhang,Fenglin Cao
Early life gut microbiota function as biological sensors for maternal prenatal exposure and play a crucial role in infant neurodevelopment. During pregnancy, air pollution and psychological distress are regarded as general and specific external exposures, respectively; however, the joint influence of these two domains on shaping early life gut microbiota remains unexplored. In this study, 309 mother-infant pairs were recruited from the obstetrics departments of two tertiary hospitals. We collected data on maternal prenatal air pollution exposure and psychological distress, obtained meconium samples within 48 h after birth, and assessed infant neurodevelopmental outcomes using the Ages and Stages Questionnaire-3 at 1, 3, and 6 months postpartum. Maternal prenatal air pollution-psychological distress exposure patterns were identified using a self-organizing map (SOM). The differential features of the meconium microbiota in relation to co-exposure patterns were assessed using multivariate association of linear models. Finally, the mediating role of the meconium microbiota in co-exposure patterns and infant neurodevelopment was analyzed using mediation analysis. We observed that the meconium microbiota at both the phylum and genus levels differed among the three patterns. Ruminococcus mediated the relationship between co-exposure patterns and infant neurodevelopment at 3 months of age (IE = 0.181-0.261, pFDR < 0.001). These findings support the inclusion of infant gut microbiota within frameworks assessing the risks of maternal prenatal co-exposure to environmental pollution and psychological distress, providing a scientific basis for policymakers to identify intervention targets for high-risk populations.
生命早期肠道微生物群作为母体产前暴露的生物传感器,在婴儿神经发育中起着至关重要的作用。在怀孕期间,空气污染和心理困扰分别被视为一般和特定的外部暴露;然而,这两个领域对塑造早期生命肠道微生物群的共同影响仍未被探索。本研究从两家三级医院的产科招募了309对母婴。我们收集了产妇产前空气污染暴露和心理困扰的数据,在出生后48小时内获得胎粪样本,并在产后1、3和6个月使用年龄和阶段问卷-3评估婴儿神经发育结局。使用自组织图(SOM)确定了产妇产前空气污染-心理困扰暴露模式。使用多元线性关联模型评估胎粪微生物群与共暴露模式的差异特征。最后,利用中介分析分析了粪菌群在共暴露模式和婴儿神经发育中的中介作用。我们观察到,在门和属水平上,三种模式的粪菌群存在差异。瘤胃球菌介导了共暴露模式与3月龄婴儿神经发育之间的关系(IE = 0.181-0.261, pFDR < 0.001)。这些发现支持将婴儿肠道菌群纳入评估母体产前环境污染和心理困扰风险的框架,为决策者确定高危人群的干预目标提供科学依据。
{"title":"Maternal prenatal co-exposure to air pollution and psychological distress shapes the neonatal gut: microbiota-mediated pathways to early neurodevelopment.","authors":"Pu Yang,Yifei Pei,Yongqi Huang,Mengyuan Dong,Fangming Cui,Shuang Nie,Xuan Zhang,Fenglin Cao","doi":"10.1080/19490976.2026.2614451","DOIUrl":"https://doi.org/10.1080/19490976.2026.2614451","url":null,"abstract":"Early life gut microbiota function as biological sensors for maternal prenatal exposure and play a crucial role in infant neurodevelopment. During pregnancy, air pollution and psychological distress are regarded as general and specific external exposures, respectively; however, the joint influence of these two domains on shaping early life gut microbiota remains unexplored. In this study, 309 mother-infant pairs were recruited from the obstetrics departments of two tertiary hospitals. We collected data on maternal prenatal air pollution exposure and psychological distress, obtained meconium samples within 48 h after birth, and assessed infant neurodevelopmental outcomes using the Ages and Stages Questionnaire-3 at 1, 3, and 6 months postpartum. Maternal prenatal air pollution-psychological distress exposure patterns were identified using a self-organizing map (SOM). The differential features of the meconium microbiota in relation to co-exposure patterns were assessed using multivariate association of linear models. Finally, the mediating role of the meconium microbiota in co-exposure patterns and infant neurodevelopment was analyzed using mediation analysis. We observed that the meconium microbiota at both the phylum and genus levels differed among the three patterns. Ruminococcus mediated the relationship between co-exposure patterns and infant neurodevelopment at 3 months of age (IE = 0.181-0.261, pFDR < 0.001). These findings support the inclusion of infant gut microbiota within frameworks assessing the risks of maternal prenatal co-exposure to environmental pollution and psychological distress, providing a scientific basis for policymakers to identify intervention targets for high-risk populations.","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"35 1","pages":"2614451"},"PeriodicalIF":12.2,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145961498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Gut Microbes
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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