首页 > 最新文献

Gut Microbes最新文献

英文 中文
Protection against experimental necrotizing enterocolitis by fecal filtrate transfer requires an active donor virome. 通过粪便滤液转移对实验性坏死性小肠结肠炎的保护需要一个活跃的供体病毒。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-04-10 DOI: 10.1080/19490976.2025.2486517
Malene Roed Spiegelhauer, Simone Margaard Offersen, Xiaotian Mao, Michela Gambino, Dennis Sandris Nielsen, Duc Ninh Nguyen, Anders Brunse

Necrotizing enterocolitis (NEC) remains a frequent catastrophic disease in preterm infants, and fecal filtrate transfer (FFT) has emerged as a promising prophylactic therapy. This study explored the role of virome viability for the protective effect of FFT. Using ultraviolet (UV) irradiation, we established a viral inactivation protocol and administered FFT, UV-inactivated FFT (iFFT) or sterile saline orally to preterm piglets at risk for experimental NEC. The gut pathology and barrier properties were assessed, while the microbiome was explored by 16S rRNA amplicon and metavirome sequencing. Like in prior studies, FFT reduced NEC severity and intestinal inflammation, while these effects were absent in the iFFT group. Unexpectedly, piglets receiving FFT exhibited mild side effects in the form of early-onset diarrhea. The FFT also converged the gut colonization by increased viral heterogeneity and a reduced abundance of pathobionts like Clostridium perfringens and Escherichia. In contrast, the gut microbiome of iFFT recipients diverged from both FFT and the controls. These findings highlight the clear distinction between the ability of active and inactivate viromes to modulate gut microbiota and decrease pathology. The efficacy of FFT may be driven by active bacteriophages, and loss of virome activity could have consequences for the treatment efficacy.

坏死性小肠结肠炎(NEC)仍然是早产儿常见的灾难性疾病,粪便滤液转移(FFT)已成为一种有希望的预防治疗方法。本研究探讨了病毒活力在FFT保护作用中的作用。我们利用紫外线(UV)照射建立了一种病毒灭活方案,并对有实验性NEC风险的早产仔猪口服FFT、UV灭活FFT或无菌生理盐水。评估肠道病理和屏障特性,同时通过16S rRNA扩增子和元病毒组测序探索微生物组。与先前的研究一样,FFT降低了NEC的严重程度和肠道炎症,而这些作用在iFFT组中不存在。出乎意料的是,接受FFT的仔猪表现出轻微的副作用,表现为早发性腹泻。FFT还通过增加病毒异质性和减少产气荚膜梭菌和埃希氏菌等病原体的丰度来聚集肠道定植。相比之下,iFFT接受者的肠道微生物组与FFT和对照组都存在差异。这些发现强调了活性和灭活病毒组调节肠道微生物群和减少病理的能力之间的明显区别。FFT的效果可能是由活性噬菌体驱动的,而病毒活性的丧失可能会对治疗效果产生影响。
{"title":"Protection against experimental necrotizing enterocolitis by fecal filtrate transfer requires an active donor virome.","authors":"Malene Roed Spiegelhauer, Simone Margaard Offersen, Xiaotian Mao, Michela Gambino, Dennis Sandris Nielsen, Duc Ninh Nguyen, Anders Brunse","doi":"10.1080/19490976.2025.2486517","DOIUrl":"https://doi.org/10.1080/19490976.2025.2486517","url":null,"abstract":"<p><p>Necrotizing enterocolitis (NEC) remains a frequent catastrophic disease in preterm infants, and fecal filtrate transfer (FFT) has emerged as a promising prophylactic therapy. This study explored the role of virome viability for the protective effect of FFT. Using ultraviolet (UV) irradiation, we established a viral inactivation protocol and administered FFT, UV-inactivated FFT (iFFT) or sterile saline orally to preterm piglets at risk for experimental NEC. The gut pathology and barrier properties were assessed, while the microbiome was explored by 16S rRNA amplicon and metavirome sequencing. Like in prior studies, FFT reduced NEC severity and intestinal inflammation, while these effects were absent in the iFFT group. Unexpectedly, piglets receiving FFT exhibited mild side effects in the form of early-onset diarrhea. The FFT also converged the gut colonization by increased viral heterogeneity and a reduced abundance of pathobionts like <i>Clostridium perfringens</i> and <i>Escherichia</i>. In contrast, the gut microbiome of iFFT recipients diverged from both FFT and the controls. These findings highlight the clear distinction between the ability of active and inactivate viromes to modulate gut microbiota and decrease pathology. The efficacy of FFT may be driven by active bacteriophages, and loss of virome activity could have consequences for the treatment efficacy.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2486517"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11988273/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144011434","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
Genomic and functional co-diversification imprint African Hominidae microbiomes to signal dietary and lifestyle adaptations. 基因组和功能共同多样化印记非洲人科微生物组表明饮食和生活方式适应。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-03-31 DOI: 10.1080/19490976.2025.2484385
Saria Otani, Marie Louise Jespersen, Christian Brinch, Frederik Duus Møller, Bo Pilgaard, Emilie Egholm Bruun Jensen, Pimlapas Leekitcharoenphon, Christina Aaby Svendsen, Amalie H Aarestrup, Tolbert Sonda, Teresa J Sylvina, Jeff Leach, Alexander Piel, Fiona Stewart, Panagiotis Sapountzis, Paul E Kazyoba, Happiness Kumburu, Frank M Aarestrup

In the diverse landscape of African hominids, the obligate relationship between the host and its microbiome narrates signals of adaptation and co-evolution. Sequencing 546 African hominid metagenomes, including those from indigenous Hadza and wild chimpanzees, identified similar bacterial richness and diversity surpassing those of westernized populations. While hominids share core bacterial communities, they also harbor distinct, population-specific bacterial taxa tailored to specific diets, ecology and lifestyles, differentiating non-indigenous and indigenous humans and chimpanzees. Even amongst shared bacterial communities, several core bacteria have co-diversified to fulfil unique dietary degradation functions within their host populations. These co-evolutionary trends extend to non-bacterial elements, such as mitochondrial DNA, antimicrobial resistance, and parasites. Our findings indicate that microbiome-host co-adaptations have led to both taxonomic and within taxa functional displacements to meet host physiological demands. The microbiome, in turn, transcends its taxonomic interchangeable role, reflecting the lifestyle, ecology and dietary history of its host.

在非洲原始人的多样化景观中,宿主与其微生物群之间的义务关系讲述了适应和共同进化的信号。对546个非洲原始人类宏基因组进行测序,包括来自当地哈扎人和野生黑猩猩的基因组,发现了类似的细菌丰富度和多样性,超过了西方化的人群。虽然原始人类共享核心细菌群落,但它们也拥有独特的、针对特定饮食、生态和生活方式量身定制的特定种群细菌分类群,从而区分非土著人类和土著人类以及黑猩猩。即使在共同的细菌群落中,一些核心细菌也共同多样化,以在其宿主群体中实现独特的饮食降解功能。这些共同进化趋势延伸到非细菌因素,如线粒体DNA、抗菌素耐药性和寄生虫。我们的研究结果表明,微生物组与宿主的共同适应导致了分类和分类群内部的功能位移,以满足宿主的生理需求。反过来,微生物组超越了其分类学上可互换的作用,反映了其宿主的生活方式、生态和饮食历史。
{"title":"Genomic and functional co-diversification imprint African Hominidae microbiomes to signal dietary and lifestyle adaptations.","authors":"Saria Otani, Marie Louise Jespersen, Christian Brinch, Frederik Duus Møller, Bo Pilgaard, Emilie Egholm Bruun Jensen, Pimlapas Leekitcharoenphon, Christina Aaby Svendsen, Amalie H Aarestrup, Tolbert Sonda, Teresa J Sylvina, Jeff Leach, Alexander Piel, Fiona Stewart, Panagiotis Sapountzis, Paul E Kazyoba, Happiness Kumburu, Frank M Aarestrup","doi":"10.1080/19490976.2025.2484385","DOIUrl":"10.1080/19490976.2025.2484385","url":null,"abstract":"<p><p>In the diverse landscape of African hominids, the obligate relationship between the host and its microbiome narrates signals of adaptation and co-evolution. Sequencing 546 African hominid metagenomes, including those from indigenous Hadza and wild chimpanzees, identified similar bacterial richness and diversity surpassing those of westernized populations. While hominids share core bacterial communities, they also harbor distinct, population-specific bacterial taxa tailored to specific diets, ecology and lifestyles, differentiating non-indigenous and indigenous humans and chimpanzees. Even amongst shared bacterial communities, several core bacteria have co-diversified to fulfil unique dietary degradation functions within their host populations. These co-evolutionary trends extend to non-bacterial elements, such as mitochondrial DNA, antimicrobial resistance, and parasites. Our findings indicate that microbiome-host co-adaptations have led to both taxonomic and within taxa functional displacements to meet host physiological demands. The microbiome, in turn, transcends its taxonomic interchangeable role, reflecting the lifestyle, ecology and dietary history of its host.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2484385"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11959905/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143752380","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
Microbiome signatures of virulence in the oral-gut-brain axis influence Parkinson's disease and cognitive decline pathophysiology. 口腔-肠-脑轴中毒力的微生物组特征影响帕金森病和认知能力下降的病理生理学。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-05-27 DOI: 10.1080/19490976.2025.2506843
Frederick Clasen, Suleyman Yildirim, Muzaffer Arıkan, Fernando Garcia-Guevara, Lűtfű Hanoğlu, Nesrin H Yılmaz, Aysu Şen, Handan Kaya Celik, Alagoz Aybala Neslihan, Tuǧçe Kahraman Demir, Zeynep Temel, Adil Mardinoglu, David L Moyes, Mathias Uhlen, Saeed Shoaie

The human microbiome is increasingly recognized for its crucial role in the development and progression of neurodegenerative diseases. While the gut-brain axis has been extensively studied, the contribution of the oral microbiome and gut-oral tropism in neurodegeneration has been largely overlooked. Cognitive impairment (CI) is common in neurodegenerative diseases and develops on a spectrum. In Parkinson's Disease (PD) patients, CI is one of the most common non-motor symptoms but its mechanistic development across the spectrum remains unclear, complicating early diagnosis of at-risk individuals. Here, we generated 228 shotgun metagenomics samples of the gut and oral microbiomes across PD patients with mild cognitive impairment (PD-MCI) or dementia (PDD), and a healthy cohort, to study the role of gut and oral microbiomes on CI in PD. In addition to revealing compositional and functional signatures, the role of pathobionts, and dysregulated metabolic pathways of the oral and gut microbiome in PD-MCI and PDD, we also revealed the importance of oral-gut translocation in increasing abundance of virulence factors in PD and CI. The oral-gut virulence was further integrated with saliva metaproteomics and demonstrated their potential role in dysfunction of host immunity and brain endothelial cells. Our findings highlight the significance of the oral-gut-brain axis and underscore its potential for discovering novel biomarkers for PD and CI.

人类微生物组在神经退行性疾病的发展和进展中发挥着至关重要的作用,这一点越来越得到人们的认可。虽然肠-脑轴已被广泛研究,但口腔微生物组和肠-口向性在神经变性中的作用在很大程度上被忽视了。认知障碍(CI)在神经退行性疾病中很常见,并呈谱系性发展。在帕金森病(PD)患者中,CI是最常见的非运动症状之一,但其在整个谱系中的机制发展尚不清楚,使高危个体的早期诊断复杂化。在这里,我们在患有轻度认知障碍(PD- mci)或痴呆症(PDD)的PD患者和健康队列中生成了228个肠道和口腔微生物组的霰弹枪宏基因组学样本,以研究肠道和口腔微生物组在PD患者CI中的作用。除了揭示PD- mci和PDD中口腔和肠道微生物组的组成和功能特征、病原体的作用以及失调的代谢途径外,我们还揭示了口腔肠道易位在PD和CI中增加毒力因子丰度的重要性。口腔-肠道毒力进一步与唾液宏蛋白质组学结合,并证明其在宿主免疫功能障碍和脑内皮细胞功能障碍中的潜在作用。我们的发现强调了口腔-肠-脑轴的重要性,并强调了它在发现PD和CI的新型生物标志物方面的潜力。
{"title":"Microbiome signatures of virulence in the oral-gut-brain axis influence Parkinson's disease and cognitive decline pathophysiology.","authors":"Frederick Clasen, Suleyman Yildirim, Muzaffer Arıkan, Fernando Garcia-Guevara, Lűtfű Hanoğlu, Nesrin H Yılmaz, Aysu Şen, Handan Kaya Celik, Alagoz Aybala Neslihan, Tuǧçe Kahraman Demir, Zeynep Temel, Adil Mardinoglu, David L Moyes, Mathias Uhlen, Saeed Shoaie","doi":"10.1080/19490976.2025.2506843","DOIUrl":"10.1080/19490976.2025.2506843","url":null,"abstract":"<p><p>The human microbiome is increasingly recognized for its crucial role in the development and progression of neurodegenerative diseases. While the gut-brain axis has been extensively studied, the contribution of the oral microbiome and gut-oral tropism in neurodegeneration has been largely overlooked. Cognitive impairment (CI) is common in neurodegenerative diseases and develops on a spectrum. In Parkinson's Disease (PD) patients, CI is one of the most common non-motor symptoms but its mechanistic development across the spectrum remains unclear, complicating early diagnosis of at-risk individuals. Here, we generated 228 shotgun metagenomics samples of the gut and oral microbiomes across PD patients with mild cognitive impairment (PD-MCI) or dementia (PDD), and a healthy cohort, to study the role of gut and oral microbiomes on CI in PD. In addition to revealing compositional and functional signatures, the role of pathobionts, and dysregulated metabolic pathways of the oral and gut microbiome in PD-MCI and PDD, we also revealed the importance of oral-gut translocation in increasing abundance of virulence factors in PD and CI. The oral-gut virulence was further integrated with saliva metaproteomics and demonstrated their potential role in dysfunction of host immunity and brain endothelial cells. Our findings highlight the significance of the oral-gut-brain axis and underscore its potential for discovering novel biomarkers for PD and CI.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2506843"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12118390/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144150222","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
Gut microbiota-mediated betaine regulates skeletal muscle fiber type transition by affecting m6A RNA methylation and Myh7 expression. 肠道菌群介导的甜菜碱通过影响m6A RNA甲基化和Myh7表达调节骨骼肌纤维类型转变。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-08-18 DOI: 10.1080/19490976.2025.2545434
Chao Yan, Yilong Yao, Zhaobo Zhang, Fanqinyu Li, Danyang Fan, Wen Liu, Xinhao Fan, Lingna Xu, Yanwen Liu, Shilong Wang, Mengling Hu, Yalan Yang, Zhonglin Tang

Skeletal muscle fiber composition is essential for maintaining muscle function and overall health. Growing evidence underscores the pivotal role of the gut-muscle axis in mediating the influence of gut microbiota on skeletal muscle development. However, the mechanisms underlying microbiota-mediated regulation of skeletal muscle fiber type remain unclear. Here, we employed multi-omics approaches, including RNA-seq, MeRIP-seq, 16S rRNA gene sequencing, and metabolomics, to investigate the causal relationship between the gut microbiota and skeletal muscle fiber transition. Our results demonstrate that the gut microbiota modulates skeletal muscle fiber transition by influencing N6-methyladenosine (m6A) methylation to regulate the expression of the slow-twitch fiber marker Myh7. Specifically, METTL3-dependent m6A methylation enhances Myh7 gene expression, leading to an increased proportion of slow-twitch fibers and a reduction in fast-twitch fibers. Furthermore, the microbiota-derived methyl donor betaine promotes Myh7 expression and Akkermansia muciniphila (AKK) abundance, and facilitates fast-to-slow fiber conversion via m6A modification. The transplantation of AKK significantly altered betaine levels and m6A modification, thereby promoting muscle fiber remodeling. In conclusion, these findings reveal that AKK-coordinated betaine drives skeletal muscle fiber conversion by modulating Myh7 mRNA expression. This study provides novel insights into the role of m6A RNA methylation in the gut-muscle crosstalk, highlighting potential therapeutic targets for muscle-related disorders.

骨骼肌纤维成分对维持肌肉功能和整体健康至关重要。越来越多的证据强调肠肌轴在调节肠道微生物群对骨骼肌发育的影响中的关键作用。然而,微生物群介导的骨骼肌纤维类型调节的机制尚不清楚。在这里,我们采用多组学方法,包括RNA-seq、MeRIP-seq、16S rRNA基因测序和代谢组学,来研究肠道微生物群与骨骼肌纤维转化之间的因果关系。我们的研究结果表明,肠道微生物群通过影响n6 -甲基腺苷(m6A)甲基化来调节慢肌纤维标记Myh7的表达,从而调节骨骼肌纤维的转变。具体来说,mettl3依赖的m6A甲基化增强了Myh7基因表达,导致慢肌纤维比例增加,快肌纤维比例减少。此外,微生物来源的甲基给体甜菜碱促进Myh7表达和Akkermansia muciniphila (AKK)丰度,并通过m6A修饰促进快到慢的纤维转化。AKK移植显著改变甜菜碱水平和m6A修饰,从而促进肌纤维重塑。综上所述,这些发现表明akk -协同甜菜碱通过调节Myh7 mRNA的表达来驱动骨骼肌纤维的转化。这项研究为m6A RNA甲基化在肠肌串扰中的作用提供了新的见解,突出了肌肉相关疾病的潜在治疗靶点。
{"title":"Gut microbiota-mediated betaine regulates skeletal muscle fiber type transition by affecting m<sup>6</sup>A RNA methylation and <i>Myh7</i> expression.","authors":"Chao Yan, Yilong Yao, Zhaobo Zhang, Fanqinyu Li, Danyang Fan, Wen Liu, Xinhao Fan, Lingna Xu, Yanwen Liu, Shilong Wang, Mengling Hu, Yalan Yang, Zhonglin Tang","doi":"10.1080/19490976.2025.2545434","DOIUrl":"10.1080/19490976.2025.2545434","url":null,"abstract":"<p><p>Skeletal muscle fiber composition is essential for maintaining muscle function and overall health. Growing evidence underscores the pivotal role of the gut-muscle axis in mediating the influence of gut microbiota on skeletal muscle development. However, the mechanisms underlying microbiota-mediated regulation of skeletal muscle fiber type remain unclear. Here, we employed multi-omics approaches, including RNA-seq, MeRIP-seq, 16S rRNA gene sequencing, and metabolomics, to investigate the causal relationship between the gut microbiota and skeletal muscle fiber transition. Our results demonstrate that the gut microbiota modulates skeletal muscle fiber transition by influencing N6-methyladenosine (m<sup>6</sup>A) methylation to regulate the expression of the slow-twitch fiber marker <i>Myh7</i>. Specifically, METTL3-dependent m<sup>6</sup>A methylation enhances <i>Myh7</i> gene expression, leading to an increased proportion of slow-twitch fibers and a reduction in fast-twitch fibers. Furthermore, the microbiota-derived methyl donor betaine promotes <i>Myh7</i> expression and <i>Akkermansia muciniphila</i> (<i>AKK</i>) abundance, and facilitates fast-to-slow fiber conversion via m<sup>6</sup>A modification. The transplantation of <i>AKK</i> significantly altered betaine levels and m<sup>6</sup>A modification, thereby promoting muscle fiber remodeling. In conclusion, these findings reveal that <i>AKK</i>-coordinated betaine drives skeletal muscle fiber conversion by modulating <i>Myh7</i> mRNA expression. This study provides novel insights into the role of m<sup>6</sup>A RNA methylation in the gut-muscle crosstalk, highlighting potential therapeutic targets for muscle-related disorders.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2545434"},"PeriodicalIF":11.0,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12363516/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144872879","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
Systematically-designed mixtures outperform single fibers for gut microbiota support. 系统设计的混合物在支持肠道微生物群方面优于单一纤维。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-20 DOI: 10.1080/19490976.2024.2442521
T M Cantu-Jungles, V Agamennone, T J Van den Broek, F H J Schuren, B Hamaker

Dietary fiber interventions to modulate the gut microbiota have largely relied on isolated fibers or specific fiber sources. We hypothesized that fibers systematically blended could promote more health-related bacterial groups. Initially, pooled in vitro fecal fermentations were used to design dietary fiber mixtures to support complementary microbial groups related to health. Then, microbial responses were compared for the designed mixtures versus their single fiber components in vitro using fecal samples from a separate cohort of 10 healthy adults. The designed fiber mixtures outperformed individual fibers in supporting bacterial taxa across donors resulting in superior alpha diversity and unexpected higher SCFA production. Moreover, unique shifts in community structure and specific taxa were observed for fiber mixtures that were not observed for single fibers, suggesting a synergistic effect when certain fibers are put together. Fiber mixture responses were remarkably more consistent than individual fibers across donors in promoting several taxa, especially butyrate producers from the Clostridium cluster XIVa. This is the first demonstration of synergistic fiber interactions for superior support of a diverse group of important beneficial microbes consistent across people, and unexpectedly high SCFA production. Overall, harnessing the synergistic potential of designed fiber mixtures represents a promising and more efficacious avenue for future prebiotic development.

调节肠道微生物群的膳食纤维干预措施主要依赖于孤立的纤维或特定的纤维来源。我们假设,系统地混合纤维可以促进更多与健康相关的细菌群。最初,我们利用体外粪便发酵来设计膳食纤维混合物,以支持与健康相关的互补微生物群。然后,利用 10 名健康成年人的粪便样本,比较了设计的混合物与单一纤维成分在体外的微生物反应。设计的纤维混合物在支持各供体细菌分类群方面的表现优于单个纤维,从而产生了卓越的α多样性和意想不到的更高SCFA产量。此外,在纤维混合物中观察到了群落结构和特定分类群的独特变化,而在单一纤维中没有观察到这种变化,这表明当某些纤维组合在一起时会产生协同效应。在促进多个分类群,特别是梭状芽孢杆菌 XIVa 群的丁酸盐生产者方面,不同供体的纤维混合物反应比单个纤维反应更加一致。这是首次展示纤维之间的协同作用,可为不同人群的多种重要有益微生物提供卓越支持,并产生意想不到的高 SCFA。总之,利用设计纤维混合物的协同潜力是未来益生元开发的一条前景广阔、更有效的途径。
{"title":"Systematically-designed mixtures outperform single fibers for gut microbiota support.","authors":"T M Cantu-Jungles, V Agamennone, T J Van den Broek, F H J Schuren, B Hamaker","doi":"10.1080/19490976.2024.2442521","DOIUrl":"https://doi.org/10.1080/19490976.2024.2442521","url":null,"abstract":"<p><p>Dietary fiber interventions to modulate the gut microbiota have largely relied on isolated fibers or specific fiber sources. We hypothesized that fibers systematically blended could promote more health-related bacterial groups. Initially, pooled <i>in vitro</i> fecal fermentations were used to design dietary fiber mixtures to support complementary microbial groups related to health. Then, microbial responses were compared for the designed mixtures versus their single fiber components <i>in vitro</i> using fecal samples from a separate cohort of 10 healthy adults. The designed fiber mixtures outperformed individual fibers in supporting bacterial taxa across donors resulting in superior alpha diversity and unexpected higher SCFA production. Moreover, unique shifts in community structure and specific taxa were observed for fiber mixtures that were not observed for single fibers, suggesting a synergistic effect when certain fibers are put together. Fiber mixture responses were remarkably more consistent than individual fibers across donors in promoting several taxa, especially butyrate producers from the <i>Clostridium</i> cluster XIVa. This is the first demonstration of synergistic fiber interactions for superior support of a diverse group of important beneficial microbes consistent across people, and unexpectedly high SCFA production. Overall, harnessing the synergistic potential of designed fiber mixtures represents a promising and more efficacious avenue for future prebiotic development.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2442521"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142863986","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
Desulfovibrio vulgaris flagellin exacerbates colorectal cancer through activating LRRC19/TRAF6/TAK1 pathway. 寻常Desulfovibrio vulgaris鞭毛蛋白通过激活LRRC19/TRAF6/TAK1通路加重结直肠癌。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-24 DOI: 10.1080/19490976.2024.2446376
Yue Dong, Fanyi Meng, Jingyi Wang, Jingge Wei, Kexin Zhang, Siqi Qin, Mengfan Li, Fucheng Wang, Bangmao Wang, Tianyu Liu, Weilong Zhong, Hailong Cao

The initiation and progression of colorectal cancer (CRC) are intimately associated with genetic, environmental and biological factors. Desulfovibrio vulgaris (DSV), a sulfate-reducing bacterium, has been found excessive growth in CRC patients, suggesting a potential role in carcinogenesis. However, the precise mechanisms underlying this association remain incompletely understood. We have found Desulfovibrio was abundant in high-fat diet-induced Apcmin/+ mice, and DSV, a member of Desulfovibrio, triggered colonocyte proliferation of germ-free mice. Furthermore, the level of DSV progressively rose from healthy individuals to CRC patients. Flagella are important accessory structures of bacteria, which can help them colonize and enhance their invasive ability. We found that D. vulgaris flagellin (DVF) drove the proliferation, migration, and invasion of CRC cells and fostered the growth of CRC xenografts. DVF enriched the epithelial-mesenchymal transition (EMT)-associated genes and characterized the facilitation of DVF on EMT. Mechanistically, DVF induced EMT through a functional transmembrane receptor called leucine-rich repeat containing 19 (LRRC19). DVF interacted with LRRC19 to modulate the ubiquitination of tumor necrosis factor receptor-associated factor (TRAF)6, rather than TRAF2. This interaction drove the ubiquitination of pivotal molecule TAK1, further enhancing its autophosphorylation and ultimately contributing to EMT. Collectively, DVF interacts with LRRC19 to activate the TRAF6/TAK1 signaling pathway, thereby promoting the EMT of CRC. These data shed new light on the role of gut microbiota in CRC and establish a potential clinical therapeutic target.

结直肠癌(CRC)的发生和发展与遗传、环境和生物学因素密切相关。寻常脱硫弧菌(Desulfovibrio vulgaris, DSV)是一种硫酸盐还原菌,在结直肠癌患者中被发现过度生长,提示其可能在致癌过程中起作用。然而,这种关联背后的确切机制仍不完全清楚。我们发现在高脂肪饮食诱导的Apcmin/+小鼠中大量存在Desulfovibrio,而作为Desulfovibrio成员的DSV可触发无菌小鼠的结肠细胞增殖。此外,从健康个体到结直肠癌患者,DSV水平逐渐升高。鞭毛是细菌的重要附属结构,可以帮助细菌定植,增强其侵袭能力。我们发现,D. vulgaris鞭毛蛋白(DVF)驱动结直肠癌细胞的增殖、迁移和侵袭,并促进结直肠癌异种移植物的生长。DVF丰富了上皮-间质转化(EMT)相关基因,并表征了DVF对EMT的促进作用。从机制上讲,DVF通过一种称为富含亮氨酸重复序列19 (LRRC19)的功能性跨膜受体诱导EMT。DVF与lrrrc19相互作用,调节肿瘤坏死因子受体相关因子(TRAF)6的泛素化,而不是TRAF2。这种相互作用驱动关键分子TAK1的泛素化,进一步增强其自磷酸化,最终导致EMT。总的来说,DVF与LRRC19相互作用激活TRAF6/TAK1信号通路,从而促进CRC的EMT。这些数据揭示了肠道菌群在结直肠癌中的作用,并建立了潜在的临床治疗靶点。
{"title":"<i>Desulfovibrio vulgaris</i> flagellin exacerbates colorectal cancer through activating LRRC19/TRAF6/TAK1 pathway.","authors":"Yue Dong, Fanyi Meng, Jingyi Wang, Jingge Wei, Kexin Zhang, Siqi Qin, Mengfan Li, Fucheng Wang, Bangmao Wang, Tianyu Liu, Weilong Zhong, Hailong Cao","doi":"10.1080/19490976.2024.2446376","DOIUrl":"https://doi.org/10.1080/19490976.2024.2446376","url":null,"abstract":"<p><p>The initiation and progression of colorectal cancer (CRC) are intimately associated with genetic, environmental and biological factors. <i>Desulfovibrio vulgaris</i> (DSV), a sulfate-reducing bacterium, has been found excessive growth in CRC patients, suggesting a potential role in carcinogenesis. However, the precise mechanisms underlying this association remain incompletely understood. We have found <i>Desulfovibrio</i> was abundant in high-fat diet-induced <i>Apc</i><sup><i>min/+</i></sup> mice, and DSV, a member of <i>Desulfovibrio</i>, triggered colonocyte proliferation of germ-free mice. Furthermore, the level of DSV progressively rose from healthy individuals to CRC patients. Flagella are important accessory structures of bacteria, which can help them colonize and enhance their invasive ability. We found that <i>D. vulgaris</i> flagellin (DVF) drove the proliferation, migration, and invasion of CRC cells and fostered the growth of CRC xenografts. DVF enriched the epithelial-mesenchymal transition (EMT)-associated genes and characterized the facilitation of DVF on EMT. Mechanistically, DVF induced EMT through a functional transmembrane receptor called leucine-rich repeat containing 19 (LRRC19). DVF interacted with LRRC19 to modulate the ubiquitination of tumor necrosis factor receptor-associated factor (TRAF)6, rather than TRAF2. This interaction drove the ubiquitination of pivotal molecule TAK1, further enhancing its autophosphorylation and ultimately contributing to EMT. Collectively, DVF interacts with LRRC19 to activate the TRAF6/TAK1 signaling pathway, thereby promoting the EMT of CRC. These data shed new light on the role of gut microbiota in CRC and establish a potential clinical therapeutic target.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2446376"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142881946","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 modelling framework to characterize the impact of antibiotics on the gut microbiota diversity. 描述抗生素对肠道微生物群多样性影响的建模框架。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-22 DOI: 10.1080/19490976.2024.2442523
Carlos Olivares, Etienne Ruppé, Stéphanie Ferreira, Tanguy Corbel, Antoine Andremont, Jean de Gunzburg, Jeremie Guedj, Charles Burdet

Metagenomic sequencing deepened our knowledge about the role of the intestinal microbiota in human health, and several studies with various methodologies explored its dynamics during antibiotic treatments. We compared the impact of four widely used antibiotics on the gut bacterial diversity. We used plasma and fecal samples collected during and after treatment from healthy volunteers assigned to a 5-day treatment either by ceftriaxone (1 g every 24 h through IV route), ceftazidime/avibactam (2 g/500 mg every 8 h through IV route), piperacillin/tazobactam (1 g/500 mg every 8 h through IV route) or moxifloxacin (400 mg every 24 h through oral route). Antibiotic concentrations were measured in plasma and feces, and bacterial diversity was assessed by the Shannon index from 16S rRNA gene profiling. The relationship between the evolutions of antibiotic fecal exposure and bacterial diversity was modeled using non-linear mixed effects models. We compared the impact of antibiotics on gut microbiota diversity by simulation, using various reconstructed pharmacodynamic indices. Piperacillin/tazobactam was characterized by the highest impact in terms of intensity of perturbation (maximal [IQR] loss of diversity of 27.3% [1.9; 40.0]), while moxifloxacin had the longest duration of perturbation, with a time to return to 95% of baseline value after the last administration of 13.2 d [8.3; 19.1]. Overall, moxifloxacin exhibited the highest global impact, followed by piperacillin/tazobactam, ceftazidime/avibactam and ceftriaxone. Their AUC between day 0 and day 42 of the change of diversity indices from day 0 were, respectively, -13.2 Shannon unit.day [-20.4; -7.9], -10.9 Shannon unit.day [-20.4; -0.6] and -10.1 Shannon unit.day [-18.3; -4.6]. We conclude that antibiotics alter the intestinal diversity to varying degrees, both within and between antibiotics families. Such studies are needed to help antibiotic stewardship in using the antibiotics with the lowest impact on the intestinal microbiota.

宏基因组测序加深了我们对肠道微生物群在人类健康中的作用的认识,一些采用不同方法的研究探索了抗生素治疗期间肠道微生物群的动态。我们比较了四种广泛使用的抗生素对肠道细菌多样性的影响。我们使用了健康志愿者在治疗期间和治疗后收集的血浆和粪便样本,这些志愿者被分配到5天的治疗中,其中包括头孢曲松(每24小时1 g通过静脉注射)、头孢他啶/阿维巴坦(每8小时2 g/500 mg通过静脉注射)、哌拉西林/他唑巴坦(每8小时1 g/500 mg通过静脉注射)或莫西沙星(每24小时400 mg通过口服)。测定血浆和粪便中的抗生素浓度,并通过16S rRNA基因谱的Shannon指数评估细菌多样性。采用非线性混合效应模型模拟抗生素粪便暴露与细菌多样性之间的关系。我们利用各种重建的药效学指标,模拟比较了抗生素对肠道菌群多样性的影响。在扰动强度方面,哌拉西林/他唑巴坦的影响最大(最大[IQR]多样性损失27.3% [1.9;40.0]),而莫西沙星的扰动持续时间最长,末次给药后恢复到基线值95%的时间为13.2 d [8.3;19.1]。总体而言,莫西沙星的全球影响最大,其次是哌拉西林/他唑巴坦、头孢他啶/阿维巴坦和头孢曲松。从第0天到第42天的多样性指数变化AUC分别为-13.2 Shannon单位。天(-20.4;-7.9], -10.9香农单位。天(-20.4;-0.6]和-10.1香农单位。天(-18.3;-4.6]。我们得出结论,抗生素在不同程度上改变了肠道多样性,无论是在抗生素家族内部还是在抗生素家族之间。需要这样的研究来帮助抗生素管理人员使用对肠道微生物群影响最小的抗生素。
{"title":"A modelling framework to characterize the impact of antibiotics on the gut microbiota diversity.","authors":"Carlos Olivares, Etienne Ruppé, Stéphanie Ferreira, Tanguy Corbel, Antoine Andremont, Jean de Gunzburg, Jeremie Guedj, Charles Burdet","doi":"10.1080/19490976.2024.2442523","DOIUrl":"https://doi.org/10.1080/19490976.2024.2442523","url":null,"abstract":"<p><p>Metagenomic sequencing deepened our knowledge about the role of the intestinal microbiota in human health, and several studies with various methodologies explored its dynamics during antibiotic treatments. We compared the impact of four widely used antibiotics on the gut bacterial diversity. We used plasma and fecal samples collected during and after treatment from healthy volunteers assigned to a 5-day treatment either by ceftriaxone (1 g every 24 h through IV route), ceftazidime/avibactam (2 g/500 mg every 8 h through IV route), piperacillin/tazobactam (1 g/500 mg every 8 h through IV route) or moxifloxacin (400 mg every 24 h through oral route). Antibiotic concentrations were measured in plasma and feces, and bacterial diversity was assessed by the Shannon index from 16S rRNA gene profiling. The relationship between the evolutions of antibiotic fecal exposure and bacterial diversity was modeled using non-linear mixed effects models. We compared the impact of antibiotics on gut microbiota diversity by simulation, using various reconstructed pharmacodynamic indices. Piperacillin/tazobactam was characterized by the highest impact in terms of intensity of perturbation (maximal [IQR] loss of diversity of 27.3% [1.9; 40.0]), while moxifloxacin had the longest duration of perturbation, with a time to return to 95% of baseline value after the last administration of 13.2 d [8.3; 19.1]. Overall, moxifloxacin exhibited the highest global impact, followed by piperacillin/tazobactam, ceftazidime/avibactam and ceftriaxone. Their AUC between day 0 and day 42 of the change of diversity indices from day 0 were, respectively, -13.2 Shannon unit.day [-20.4; -7.9], -10.9 Shannon unit.day [-20.4; -0.6] and -10.1 Shannon unit.day [-18.3; -4.6]. We conclude that antibiotics alter the intestinal diversity to varying degrees, both within and between antibiotics families. Such studies are needed to help antibiotic stewardship in using the antibiotics with the lowest impact on the intestinal microbiota.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2442523"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142876887","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
Development of a Caco-2-based intestinal mucosal model to study intestinal barrier properties and bacteria-mucus interactions. 建立基于caco -2的肠粘膜模型,研究肠屏障特性和细菌-粘液相互作用。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-23 DOI: 10.1080/19490976.2024.2434685
Evelien Floor, Jinyi Su, Maitrayee Chatterjee, Elise S Kuipers, Noortje IJssennagger, Faranak Heidari, Laura Giordano, Richard W Wubbolts, Silvia M Mihăilă, Daphne A C Stapels, Yvonne Vercoulen, Karin Strijbis

The intestinal mucosal barrier is a dynamic system that allows nutrient uptake, stimulates healthy microbe-host interactions, and prevents invasion by pathogens. The mucosa consists of epithelial cells connected by cellular junctions that regulate the passage of nutrients covered by a mucus layer that plays an important role in host-microbiome interactions. Mimicking the intestinal mucosa for in vitro assays, particularly the generation of a mucus layer, has proven to be challenging. The intestinal cell-line Caco-2 is widely used in academic and industrial laboratories due to its capacity to polarize, form an apical brush border, and reproducibly grow into confluent cell layers in different culture systems. However, under normal culture conditions, Caco-2 cultures lack a mucus layer. Here, we demonstrate for the first time that Caco-2 cultures can form a robust mucus layer when cultured under air-liquid interface (ALI) conditions on Transwell inserts with addition of vasointestinal peptide (VIP) in the basolateral compartment. We demonstrate that unique gene clusters are regulated in response to ALI and VIP single stimuli, but the ALI-VIP combination treatment resulted in a significant upregulation of multiple mucin genes and proteins, including secreted MUC2 and transmembrane mucins MUC13 and MUC17. Expression of tight junction proteins was significantly altered in the ALI-VIP condition, leading to increased permeability to small molecules. Commensal Lactiplantibacillus plantarum bacteria closely associated with the Caco-2 mucus layer and differentially colonized the surface of the ALI cultures. Pathogenic Salmonella enterica were capable of invading beyond the mucus layer and brush border. In conclusion, Caco-2 ALI-VIP cultures provide an accessible and straightforward way to culture an in vitro intestinal mucosal model with improved biomimetic features. This novel in vitro intestinal model can facilitate studies into mucus and epithelial barrier functions and in-depth molecular characterization of pathogenic and commensal microbe-mucus interactions.

肠粘膜屏障是一个动态系统,允许营养摄取,刺激健康的微生物-宿主相互作用,并防止病原体的入侵。粘膜由细胞连接连接的上皮细胞组成,这些细胞连接调节黏液层覆盖的营养物质的通过,黏液层在宿主-微生物组相互作用中起重要作用。在体外实验中模拟肠粘膜,特别是黏液层的产生,已被证明是具有挑战性的。肠道细胞系Caco-2在不同的培养体系中具有极化、形成顶端刷状边界、可重复生长成融合细胞层的能力,被广泛应用于学术和工业实验室。然而,在正常培养条件下,Caco-2培养物缺乏黏液层。在这里,我们首次证明Caco-2培养物在气液界面(ALI)条件下在Transwell插入物上在基底外侧隔室中添加血管肠肽(VIP)时可以形成坚固的黏液层。我们发现,在ALI和VIP的单一刺激下,独特的基因簇受到调控,但ALI-VIP联合治疗导致多种粘蛋白基因和蛋白的显著上调,包括分泌的MUC2和跨膜粘蛋白MUC13和MUC17。在ALI-VIP条件下,紧密连接蛋白的表达显著改变,导致对小分子的通透性增加。共生植物乳杆菌与Caco-2黏液层密切相关,并在ALI培养物表面有差异定植。致病性肠炎沙门氏菌能够越过黏液层和毛刷边界侵入。综上所述,Caco-2 ALI-VIP培养物为体外培养具有较好仿生特性的肠粘膜模型提供了一种简便易行的方法。这种新型的体外肠道模型有助于研究黏液和上皮屏障功能,以及病原和共生微生物-黏液相互作用的深入分子表征。
{"title":"Development of a Caco-2-based intestinal mucosal model to study intestinal barrier properties and bacteria-mucus interactions.","authors":"Evelien Floor, Jinyi Su, Maitrayee Chatterjee, Elise S Kuipers, Noortje IJssennagger, Faranak Heidari, Laura Giordano, Richard W Wubbolts, Silvia M Mihăilă, Daphne A C Stapels, Yvonne Vercoulen, Karin Strijbis","doi":"10.1080/19490976.2024.2434685","DOIUrl":"https://doi.org/10.1080/19490976.2024.2434685","url":null,"abstract":"<p><p>The intestinal mucosal barrier is a dynamic system that allows nutrient uptake, stimulates healthy microbe-host interactions, and prevents invasion by pathogens. The mucosa consists of epithelial cells connected by cellular junctions that regulate the passage of nutrients covered by a mucus layer that plays an important role in host-microbiome interactions. Mimicking the intestinal mucosa for <i>in vitro</i> assays, particularly the generation of a mucus layer, has proven to be challenging. The intestinal cell-line Caco-2 is widely used in academic and industrial laboratories due to its capacity to polarize, form an apical brush border, and reproducibly grow into confluent cell layers in different culture systems. However, under normal culture conditions, Caco-2 cultures lack a mucus layer. Here, we demonstrate for the first time that Caco-2 cultures can form a robust mucus layer when cultured under air-liquid interface (ALI) conditions on Transwell inserts with addition of vasointestinal peptide (VIP) in the basolateral compartment. We demonstrate that unique gene clusters are regulated in response to ALI and VIP single stimuli, but the ALI-VIP combination treatment resulted in a significant upregulation of multiple mucin genes and proteins, including secreted MUC2 and transmembrane mucins MUC13 and MUC17. Expression of tight junction proteins was significantly altered in the ALI-VIP condition, leading to increased permeability to small molecules. Commensal <i>Lactiplantibacillus plantarum</i> bacteria closely associated with the Caco-2 mucus layer and differentially colonized the surface of the ALI cultures. Pathogenic <i>Salmonella enterica</i> were capable of invading beyond the mucus layer and brush border. In conclusion, Caco-2 ALI-VIP cultures provide an accessible and straightforward way to culture an <i>in vitro</i> intestinal mucosal model with improved biomimetic features. This novel <i>in vitro</i> intestinal model can facilitate studies into mucus and epithelial barrier functions and in-depth molecular characterization of pathogenic and commensal microbe-mucus interactions.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2434685"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142876894","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
The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia. 微生物来源的胆汁酸牛甲去氧胆酸可改善患有癌症恶病质小鼠的肝脏胆固醇水平。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-01-08 DOI: 10.1080/19490976.2025.2449586
Morgane M Thibaut, Martin Roumain, Edwige Piron, Justine Gillard, Axelle Loriot, Audrey M Neyrinck, Julie Rodriguez, Isabelle Massart, Jean-Paul Thissen, Joshua R Huot, Fabrizio Pin, Andrea Bonetto, Nathalie M Delzenne, Giulio G Muccioli, Laure B Bindels

Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo. In vitro, TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.

胆汁酸谱和途径的改变有助于癌症恶病质中的肝脏炎症,这是一种恶化癌症患者预后的综合征。肠道菌群通过胆汁酸影响宿主代谢,本研究旨在探讨肠道菌群失调对癌症恶病质中胆汁酸代谢失调及相关疾病的功能贡献。使用三种癌症恶病质小鼠模型(C26, MC38和HCT116模型),我们证明了几种次级胆汁酸的肝脏水平降低,主要是牛黄脱氧胆酸(TDCA)。肝TDCA的减少发生在恶病质出现之前。肠道微生物群的纵向分析确定了一种ASV,鉴定为啮齿木杆菌,作为一种细菌可能参与减少TDCA的产生。同时,基于稳定同位素的实验显示,在病毒质小鼠中,微生物7α-去羟基化(7α-DH)活性明显下降,而胆汁盐水解酶(BSH)活性没有变化。该方法还发现,微生物7α-羟基类固醇脱氢酶(7α-HSDH)和12α-羟基类固醇脱氢酶(12α-HSDH)活性降低。在体外和体内研究了TDCA的低产量对癌症恶病质的贡献。在体外,TDCA可以防止肌管萎缩,而在体内,肝脏全转录组分析显示,TDCA给药可以改善未折叠蛋白反应和胆固醇稳态途径。同时,TDCA可以逆转这些小鼠的肝脏胆固醇积累。总之,这项工作强调了肠道微生物群对胆汁酸代谢失调的贡献,以及次级胆汁酸TDCA在癌症恶病质背景下对肝脏胆固醇稳态的治疗意义。这一发现可能有助于理解其他以微生物生态失调为特征的代谢性疾病。更广泛地说,我们的工作证明了使用稳定同位素测量微生物活性的兴趣和相关性,这种方法目前在微生物组领域未得到充分利用。
{"title":"The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia.","authors":"Morgane M Thibaut, Martin Roumain, Edwige Piron, Justine Gillard, Axelle Loriot, Audrey M Neyrinck, Julie Rodriguez, Isabelle Massart, Jean-Paul Thissen, Joshua R Huot, Fabrizio Pin, Andrea Bonetto, Nathalie M Delzenne, Giulio G Muccioli, Laure B Bindels","doi":"10.1080/19490976.2025.2449586","DOIUrl":"10.1080/19490976.2025.2449586","url":null,"abstract":"<p><p>Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as <i>Xylanibacter rodentium</i>, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored <i>in vitro</i> and <i>in vivo</i>. <i>In vitro</i>, TDCA prevented myotube atrophy, whereas <i>in vivo</i> hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2449586"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730681/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142947812","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 crystal structure of the toxin EspC from enteropathogenic Escherichia coli reveals the mechanism that governs host cell entry and cytotoxicity. 肠致病性大肠杆菌毒素EspC的晶体结构揭示了控制宿主细胞进入和细胞毒性的机制。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-03-31 DOI: 10.1080/19490976.2025.2483777
Akila U Pilapitiya, Lilian Hor, Jing Pan, Lakshmi C Wijeyewickrema, Robert N Pike, Denisse L Leyton, Jason J Paxman, Begoña Heras

Enteropathogenic E. coli (EPEC) is a significant cause of diarrhea, leading to high infant mortality rates. A key toxin produced by EPEC is the EspC autotransporter, which is regulated alongside genes from the locus of enterocyte effacement (LEE), which collectively result in the characteristic attaching and effacing lesions on the intestinal epithelium. In this study, we present the crystal structure of the EspC passenger domain (αEspC) revealing a toxin comprised a serine protease attached to a large β-helix with additional subdomains. Using various modified EspC expression constructs, alongside type III secretion system-mediated cell internalization assays, we dissect how the αEspC structural features enable toxin entry into the intestinal epithelium to cause cell cytotoxicity.

肠致病性大肠杆菌(EPEC)是腹泻的重要原因,导致婴儿死亡率高。EPEC产生的一种关键毒素是EspC自转运蛋白,它与肠细胞消退(LEE)位点的基因一起受到调节,这些基因共同导致肠上皮上的特征性附着和消退病变。在这项研究中,我们展示了EspC乘客结构域(αEspC)的晶体结构,揭示了一种毒素由丝氨酸蛋白酶组成,该蛋白酶附着在具有附加子结构域的大β-螺旋上。利用各种改良的EspC表达构建,以及III型分泌系统介导的细胞内化实验,我们剖析了αEspC的结构特征是如何使毒素进入肠上皮引起细胞毒性的。
{"title":"The crystal structure of the toxin EspC from enteropathogenic <i>Escherichia coli</i> reveals the mechanism that governs host cell entry and cytotoxicity.","authors":"Akila U Pilapitiya, Lilian Hor, Jing Pan, Lakshmi C Wijeyewickrema, Robert N Pike, Denisse L Leyton, Jason J Paxman, Begoña Heras","doi":"10.1080/19490976.2025.2483777","DOIUrl":"10.1080/19490976.2025.2483777","url":null,"abstract":"<p><p>Enteropathogenic <i>E. coli</i> (EPEC) is a significant cause of diarrhea, leading to high infant mortality rates. A key toxin produced by EPEC is the EspC autotransporter, which is regulated alongside genes from the locus of enterocyte effacement (LEE), which collectively result in the characteristic attaching and effacing lesions on the intestinal epithelium. In this study, we present the crystal structure of the EspC passenger domain (α<sup>EspC</sup>) revealing a toxin comprised a serine protease attached to a large β-helix with additional subdomains. Using various modified EspC expression constructs, alongside type III secretion system-mediated cell internalization assays, we dissect how the α<sup>EspC</sup> structural features enable toxin entry into the intestinal epithelium to cause cell cytotoxicity.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2483777"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11970781/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143752385","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
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