首页 > 最新文献

Gut Microbes最新文献

英文 中文
Reutericyclin, a specialized metabolite of Limosilactobacillus reuteri, mitigates risperidone-induced weight gain in mice. 罗伊霉素是罗伊氏乳酸杆菌的一种特殊代谢物,可以减轻利培酮引起的小鼠体重增加。
IF 11 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-04-07 DOI: 10.1080/19490976.2025.2477819
Fatima A Aboulalazm, Alexis B Kazen, Orlando deLeon, Susanne Müller, Fatima L Saravia, Valery Lozada-Fernandez, Matthew A Hadiono, Robert F Keyes, Brian C Smith, Stephanie L Kellogg, Justin L Grobe, Tammy L Kindel, John R Kirby

The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including Limosilactobacillus reuteri as a candidate for further study. Oral supplementation with L. reuteri protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and L. reuteri restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by L. reuteri as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.

外来微生物对微生物群的破坏作用、相应的生态失调以及与宿主代谢疾病的潜在联系至关重要。在这项研究中,我们使用了一种广泛使用的抗精神病药物利培酮(已知会影响人类体重增加)来诱导C57BL/6J雌性小鼠体重增加。我们假设,在利培酮治疗后,维持肠道内稳态和能量平衡所必需的微生物将被耗尽,导致相对于对照组的体重增加。因此,我们对粪便样本进行宏基因组分析,以识别在利培酮治疗动物中被排除但在对照组中仍然存在的微生物。我们确定了包括罗伊氏乳酸杆菌在内的多个分类群作为进一步研究的候选者。口服补充罗伊氏乳杆菌可以防止利培酮诱导的体重增加(RIWG),并依赖于一种特殊代谢物——罗伊霉素的细胞生产。此外,合成的reutericycle素足以减轻RIWG。合成的罗伊霉素和罗伊氏乳杆菌在利培酮的存在下都能恢复能量平衡,以减轻体重增加,并诱导与瘦相关的微生物群的变化。总之,我们的研究结果表明,罗伊氏乳杆菌产生的罗伊氏环素是一种潜在的益生菌,可以恢复利培酮引起的能量平衡,并促进苗条。
{"title":"Reutericyclin, a specialized metabolite of <i>Limosilactobacillus reuteri</i>, mitigates risperidone-induced weight gain in mice.","authors":"Fatima A Aboulalazm, Alexis B Kazen, Orlando deLeon, Susanne Müller, Fatima L Saravia, Valery Lozada-Fernandez, Matthew A Hadiono, Robert F Keyes, Brian C Smith, Stephanie L Kellogg, Justin L Grobe, Tammy L Kindel, John R Kirby","doi":"10.1080/19490976.2025.2477819","DOIUrl":"10.1080/19490976.2025.2477819","url":null,"abstract":"<p><p>The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including <i>Limosilactobacillus reuteri</i> as a candidate for further study. Oral supplementation with <i>L. reuteri</i> protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and <i>L. reuteri</i> restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by <i>L. reuteri</i> as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2477819"},"PeriodicalIF":11.0,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11980487/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795245","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
Identifying the location-dependent adipose tissue bacterial DNA signatures in obese patients that predict body weight loss. 确定肥胖患者预测体重减轻的位置依赖性脂肪组织细菌DNA特征。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2024-12-23 DOI: 10.1080/19490976.2024.2439105
Matthieu Minty, Alberic Germain, Jiuwen Sun, Gracia Kaglan, Florence Servant, Benjamin Lelouvier, Emiri Misselis, Radu Mircea Neagoe, Menghini Rossella, Marina Cardellini, Rémy Burcelin, Massimo Federici, José Manuel Fernandez-Real, Vincent Blasco-Baque

Recent sets of evidence have described profiles of 16S rDNA sequences in host tissues, notably in fat pads that are significantly overrepresented and can serve as signatures of metabolic disease. However, these recent and original observations need to be further detailed and functionally defined. Here, using state-of-the-art targeted DNA sequencing and discriminant predictive approaches, we describe, from the longitudinal FLORINASH cohort of patients who underwent bariatric surgery, visceral, and subcutaneous fat pad-specific bacterial 16SrRNA signatures. The corresponding Porphyromonadaceae, Campylobacteraceae, Prevotellaceae, Actimomycetaceae, Veillonellaceae, Anaerivoracaceae, Fusobacteriaceae, and the Clostridium family XI 16SrRNA DNA segment profiles are signatures of the subcutaneous adipose depot while Pseudomonadaceae and Micrococcacecae, 16SrRNA DNA sequence profiles characterize the visceral adipose depot. In addition, we have further identified that a specific pre-bariatric surgery adipose tissue bacterial DNA signature predicts the efficacy of body weight loss in obese patients 5-10 years after the surgery. 16SrRNA signatures discriminate (ROC ~ 1) the patients who did not maintain bodyweight loss and those who did. Second, from the 16SrRNA sequences we infer potential pathways suggestive of catabolic biochemical activities that could be signatures of subcutaneous adipose depots that predict body weight loss.

最近的一系列证据描述了宿主组织中16S rDNA序列的概况,特别是在脂肪垫中,脂肪垫的代表性明显过高,可以作为代谢性疾病的标志。然而,这些最近和原始的观察结果需要进一步详细和功能定义。在这里,我们使用最先进的靶向DNA测序和判别预测方法,从纵向FLORINASH队列中描述了接受减肥手术的患者,内脏和皮下脂肪垫特异性细菌16SrRNA特征。Porphyromonadaceae、Campylobacteraceae、Prevotellaceae、Actimomycetaceae、Veillonellaceae、Anaerivoracaceae、Fusobacteriaceae和Clostridium family XI 16SrRNA DNA序列是皮下脂肪库的特征,而Pseudomonadaceae和Micrococcacecae的16SrRNA DNA序列是内脏脂肪库的特征。此外,我们进一步确定了一种特定的减肥手术前脂肪组织细菌DNA标记可以预测手术后5-10年肥胖患者的体重减轻效果。16SrRNA特征区分(ROC ~ 1)未维持体重减轻和维持体重减轻的患者。其次,从16SrRNA序列中,我们推断出可能提示分解代谢生化活动的潜在途径,这些途径可能是预测体重减轻的皮下脂肪库的标志。
{"title":"Identifying the location-dependent adipose tissue bacterial DNA signatures in obese patients that predict body weight loss.","authors":"Matthieu Minty, Alberic Germain, Jiuwen Sun, Gracia Kaglan, Florence Servant, Benjamin Lelouvier, Emiri Misselis, Radu Mircea Neagoe, Menghini Rossella, Marina Cardellini, Rémy Burcelin, Massimo Federici, José Manuel Fernandez-Real, Vincent Blasco-Baque","doi":"10.1080/19490976.2024.2439105","DOIUrl":"https://doi.org/10.1080/19490976.2024.2439105","url":null,"abstract":"<p><p>Recent sets of evidence have described profiles of 16S rDNA sequences in host tissues, notably in fat pads that are significantly overrepresented and can serve as signatures of metabolic disease. However, these recent and original observations need to be further detailed and functionally defined. Here, using state-of-the-art targeted DNA sequencing and discriminant predictive approaches, we describe, from the longitudinal FLORINASH cohort of patients who underwent bariatric surgery, visceral, and subcutaneous fat pad-specific bacterial 16SrRNA signatures. The corresponding <i>Porphyromonadaceae</i>, <i>Campylobacteraceae</i>, <i>Prevotellaceae</i>, <i>Actimomycetaceae</i>, <i>Veillonellaceae</i>, <i>Anaerivoracaceae</i>, <i>Fusobacteriaceae</i>, and the <i>Clostridium family XI</i> 16SrRNA DNA segment profiles are signatures of the subcutaneous adipose depot while <i>Pseudomonadaceae</i> and <i>Micrococcacecae</i>, 16SrRNA DNA sequence profiles characterize the visceral adipose depot. In addition, we have further identified that a specific pre-bariatric surgery adipose tissue bacterial DNA signature predicts the efficacy of body weight loss in obese patients 5-10 years after the surgery. 16SrRNA signatures discriminate (ROC ~ 1) the patients who did not maintain bodyweight loss and those who did. Second, from the 16SrRNA sequences we infer potential pathways suggestive of catabolic biochemical activities that could be signatures of subcutaneous adipose depots that predict body weight loss.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2439105"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142876909","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
Engineering non-pathogenic bacteria for auto-transporter-driven secretion of functional interferon. 对非致病细菌进行工程改造,使其在自身转运体的驱动下分泌功能性干扰素。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-03-03 DOI: 10.1080/19490976.2025.2474146
May Tfilin Samuel, Irina Rostovsky, Alona Kuzmina, Ran Taube, Neta Sal-Man

In recent years, various strategies have been developed to enable the oral administration of protein-based drugs (biologics) with the aim of overcoming the degradation and inactivation of these drugs that can occur as they traverse the gastrointestinal tract (GIT). In this study, we investigated bacteria as a delivery vehicle for biologics, harnessing their ability to withstand the harsh gastric environment and deliver therapeutic drugs directly to the intestine. Specifically, we explored using the type 5 secretion system (T5SS) to secrete therapeutic cargoes under simulated gut conditions. Our research focused on EspC, a T5SS protein from enteropathogenic Escherichia coli, and its potential to secrete interferon-α (IFNα), a cytokine with immunomodulatory and antiviral properties widely used in the clinic. We demonstrated that EspC can facilitate the secretion of IFNα variant when expressed in nonpathogenic bacteria. Moreover, this EspC-secreted IFN was able to activate the JAK-STAT pathway, upregulate IFN-stimulated genes, and induce a robust antiviral response in cells. Collectively, these findings provide proof of concept supporting the utilization of the EspC protein as a novel delivery platform for protein-based therapeutics.

近年来,为了克服这些药物在通过胃肠道(GIT)时可能发生的降解和失活,已经开发了各种策略来使口服基于蛋白质的药物(生物制剂)成为可能。在这项研究中,我们研究了细菌作为生物制剂的递送载体,利用它们承受恶劣胃环境的能力,将治疗药物直接递送到肠道。具体来说,我们探索了在模拟肠道条件下使用5型分泌系统(T5SS)分泌治疗物质。我们的研究重点是肠致病性大肠杆菌的T5SS蛋白EspC及其分泌干扰素-α (IFNα)的潜力,IFNα是一种具有免疫调节和抗病毒特性的细胞因子,广泛应用于临床。我们证明了EspC在非致病性细菌中表达时可以促进IFNα变体的分泌。此外,这种espc分泌的IFN能够激活JAK-STAT通路,上调IFN刺激的基因,并在细胞中诱导强大的抗病毒反应。总的来说,这些发现为支持EspC蛋白作为基于蛋白质的疗法的新型递送平台的利用提供了概念证明。
{"title":"Engineering non-pathogenic bacteria for auto-transporter-driven secretion of functional interferon.","authors":"May Tfilin Samuel, Irina Rostovsky, Alona Kuzmina, Ran Taube, Neta Sal-Man","doi":"10.1080/19490976.2025.2474146","DOIUrl":"10.1080/19490976.2025.2474146","url":null,"abstract":"<p><p>In recent years, various strategies have been developed to enable the oral administration of protein-based drugs (biologics) with the aim of overcoming the degradation and inactivation of these drugs that can occur as they traverse the gastrointestinal tract (GIT). In this study, we investigated bacteria as a delivery vehicle for biologics, harnessing their ability to withstand the harsh gastric environment and deliver therapeutic drugs directly to the intestine. Specifically, we explored using the type 5 secretion system (T5SS) to secrete therapeutic cargoes under simulated gut conditions. Our research focused on EspC, a T5SS protein from enteropathogenic <i>Escherichia coli</i>, and its potential to secrete interferon-α (IFNα), a cytokine with immunomodulatory and antiviral properties widely used in the clinic. We demonstrated that EspC can facilitate the secretion of IFNα variant when expressed in nonpathogenic bacteria. Moreover, this EspC-secreted IFN was able to activate the JAK-STAT pathway, upregulate IFN-stimulated genes, and induce a robust antiviral response in cells. Collectively, these findings provide proof of concept supporting the utilization of the EspC protein as a novel delivery platform for protein-based therapeutics.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2474146"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11881866/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143541426","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
Microbiota transplant therapy in inflammatory bowel disease: advances and mechanistic insights. 微生物群移植治疗炎症性肠病:进展和机制见解。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-03-10 DOI: 10.1080/19490976.2025.2477255
Daphne Moutsoglou, Pavithra Ramakrishnan, Byron P Vaughn

Microbiota transplant therapy is an emerging therapy for inflammatory bowel disease, but factors influencing its efficacy and mechanism remain poorly understood. In this narrative review, we outline key elements affecting therapeutic outcomes, including donor factors (such as age and patient relationship), recipient factors, control selection, and elements impacting engraftment and its correlation with clinical response. We also examine potential mechanisms through inflammatory bowel disease trials, focusing on the interplay between the microbiota, host, and immune system. Finally, we briefly explore potential future directions for microbiota transplant therapy and promising emerging treatments.

微生物群移植治疗是炎症性肠病的一种新兴治疗方法,但影响其疗效的因素和机制尚不清楚。在这篇叙述性综述中,我们概述了影响治疗结果的关键因素,包括供体因素(如年龄和患者关系)、受体因素、对照选择、影响移植及其与临床反应的因素。我们还通过炎症性肠病试验研究了潜在的机制,重点关注微生物群、宿主和免疫系统之间的相互作用。最后,我们简要探讨了微生物群移植治疗的潜在未来方向和有希望的新兴治疗方法。
{"title":"Microbiota transplant therapy in inflammatory bowel disease: advances and mechanistic insights.","authors":"Daphne Moutsoglou, Pavithra Ramakrishnan, Byron P Vaughn","doi":"10.1080/19490976.2025.2477255","DOIUrl":"10.1080/19490976.2025.2477255","url":null,"abstract":"<p><p>Microbiota transplant therapy is an emerging therapy for inflammatory bowel disease, but factors influencing its efficacy and mechanism remain poorly understood. In this narrative review, we outline key elements affecting therapeutic outcomes, including donor factors (such as age and patient relationship), recipient factors, control selection, and elements impacting engraftment and its correlation with clinical response. We also examine potential mechanisms through inflammatory bowel disease trials, focusing on the interplay between the microbiota, host, and immune system. Finally, we briefly explore potential future directions for microbiota transplant therapy and promising emerging treatments.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2477255"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11901402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143585382","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
Profiling the gut microbiota to assess infection risk in Klebsiella pneumoniae-colonized patients. 分析肺炎克雷伯菌定殖患者的肠道微生物群以评估感染风险。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-02-18 DOI: 10.1080/19490976.2025.2468358
Flavio De Maio, Delia Mercedes Bianco, Giulia Santarelli, Roberto Rosato, Francesca Romana Monzo, Barbara Fiori, Maurizio Sanguinetti, Brunella Posteraro

Vornhagen et al. introduced a model combining gut microbiota structure and Klebsiella pneumoniae genotype to assess infection risk in K. pneumoniae-colonized patients. Building on their findings, we investigated the gut microbiota composition and K. pneumoniae genotype in 16 colonized patients, five of whom had bloodstream infections at the time of fecal sampling. Importantly, we did not apply the original machine learning model due to the small sample size of our cohort. Instead, we explored the distribution of key antimicrobial resistance and stress resistance genes and analyzed gut community structure based on amplicon sequence variants (ASVs) of the V3-V4 16S rRNA region. Notably, distinct gene profiles were observed in both infected and non-infected patients, and three patients without bloodstream infections showed no detectable Klebsiella ASVs despite microbiological confirmation of colonization. These findings highlight the need to integrate gut microbiota composition data into infection risk assessment and address limitations in taxonomic resolution and sample size. Future studies should aim to develop streamlined tools for clinical application in K. pneumoniae-colonized patients.

Vornhagen等人引入了一种结合肠道菌群结构和肺炎克雷伯菌基因型的模型来评估肺炎克雷伯菌定殖患者的感染风险。在他们的发现的基础上,我们调查了16名定植患者的肠道微生物群组成和肺炎克雷伯菌基因型,其中5人在粪便取样时有血液感染。重要的是,由于我们的队列样本量小,我们没有应用原始的机器学习模型。因此,我们基于V3-V4 16S rRNA区域扩增子序列变异(amplicon sequence variant, asv)分析了关键抗微生物药物耐药性和抗逆性基因的分布,并分析了肠道群落结构。值得注意的是,在感染和未感染的患者中观察到不同的基因谱,尽管微生物学证实了克雷伯菌的定植,但没有血液感染的3例患者未检测到克雷伯菌asv。这些发现强调需要将肠道微生物群组成数据整合到感染风险评估中,并解决分类学分辨率和样本量的限制。未来的研究应旨在开发简化的工具,用于肺炎克雷伯菌定殖患者的临床应用。
{"title":"Profiling the gut microbiota to assess infection risk in <i>Klebsiella pneumoniae</i>-colonized patients.","authors":"Flavio De Maio, Delia Mercedes Bianco, Giulia Santarelli, Roberto Rosato, Francesca Romana Monzo, Barbara Fiori, Maurizio Sanguinetti, Brunella Posteraro","doi":"10.1080/19490976.2025.2468358","DOIUrl":"10.1080/19490976.2025.2468358","url":null,"abstract":"<p><p>Vornhagen et al. introduced a model combining gut microbiota structure and <i>Klebsiella pneumoniae</i> genotype to assess infection risk in <i>K. pneumoniae</i>-colonized patients. Building on their findings, we investigated the gut microbiota composition and <i>K. pneumoniae</i> genotype in 16 colonized patients, five of whom had bloodstream infections at the time of fecal sampling. Importantly, we did not apply the original machine learning model due to the small sample size of our cohort. Instead, we explored the distribution of key antimicrobial resistance and stress resistance genes and analyzed gut community structure based on amplicon sequence variants (ASVs) of the V3-V4 16S rRNA region. Notably, distinct gene profiles were observed in both infected and non-infected patients, and three patients without bloodstream infections showed no detectable <i>Klebsiella</i> ASVs despite microbiological confirmation of colonization. These findings highlight the need to integrate gut microbiota composition data into infection risk assessment and address limitations in taxonomic resolution and sample size. Future studies should aim to develop streamlined tools for clinical application in <i>K. pneumoniae</i>-colonized patients.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2468358"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11845061/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440726","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
Interbacterial warfare in the human gut: insights from Bacteroidales' perspective. 人类肠道细菌间的战争:从拟杆菌的角度的见解。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-03-04 DOI: 10.1080/19490976.2025.2473522
Kun Jiang, Xinxin Pang, Weixun Li, Xiaoning Xu, Yan Yang, Chengbin Shang, Xiang Gao

Competition and cooperation are fundamental to the stability and evolution of ecological communities. The human gut microbiota, a dense and complex microbial ecosystem, plays a critical role in the host's health and disease, with competitive interactions being particularly significant. As a dominant and extensively studied group in the human gut, Bacteroidales serves as a successful model system for understanding these intricate dynamic processes. This review summarizes recent advances in our understanding of the intricate antagonism mechanisms among gut Bacteroidales at the biochemical or molecular-genetic levels, focusing on interference and exploitation competition. We also discuss unresolved questions and suggest strategies for studying the competitive mechanisms of Bacteroidales. The review presented here offers valuable insights into the molecular basis of bacterial antagonism in the human gut and may inform strategies for manipulating the microbiome to benefit human health.

竞争与合作是生态群落稳定与发展的基础。人类肠道菌群是一个密集而复杂的微生物生态系统,在宿主的健康和疾病中起着至关重要的作用,其中竞争相互作用尤为重要。作为人类肠道中一个被广泛研究的主要群体,拟杆菌门作为一个成功的模型系统来理解这些复杂的动态过程。本文综述了近年来在生物化学和分子遗传学水平上对肠道拟杆菌群之间复杂的拮抗机制的理解,重点介绍了干扰和利用竞争。我们还讨论了尚未解决的问题,并提出了研究拟杆菌科竞争机制的策略。本文综述为人类肠道细菌拮抗作用的分子基础提供了有价值的见解,并可能为操纵微生物组以造福人类健康提供策略。
{"title":"Interbacterial warfare in the human gut: insights from Bacteroidales' perspective.","authors":"Kun Jiang, Xinxin Pang, Weixun Li, Xiaoning Xu, Yan Yang, Chengbin Shang, Xiang Gao","doi":"10.1080/19490976.2025.2473522","DOIUrl":"10.1080/19490976.2025.2473522","url":null,"abstract":"<p><p>Competition and cooperation are fundamental to the stability and evolution of ecological communities. The human gut microbiota, a dense and complex microbial ecosystem, plays a critical role in the host's health and disease, with competitive interactions being particularly significant. As a dominant and extensively studied group in the human gut, Bacteroidales serves as a successful model system for understanding these intricate dynamic processes. This review summarizes recent advances in our understanding of the intricate antagonism mechanisms among gut Bacteroidales at the biochemical or molecular-genetic levels, focusing on interference and exploitation competition. We also discuss unresolved questions and suggest strategies for studying the competitive mechanisms of Bacteroidales. The review presented here offers valuable insights into the molecular basis of bacterial antagonism in the human gut and may inform strategies for manipulating the microbiome to benefit human health.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2473522"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11901371/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143556597","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
Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. 细菌细胞外囊泡在动脉粥样硬化的发生、发展和治疗中的作用。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-01-22 DOI: 10.1080/19490976.2025.2452229
Yuling Lin, Jingyu Wang, Fan Bu, Ruyi Zhang, Junhui Wang, Yubing Wang, Mei Huang, Yiyi Huang, Lei Zheng, Qian Wang, Xiumei Hu

Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.

动脉粥样硬化是心脑血管疾病的主要原因。然而,目前的抗动脉粥样硬化药物显示出相互矛盾的治疗结果,从而刺激了对新的有效治疗方法的研究。最近的研究表明口腔和胃肠道微生物群在动脉粥样硬化中起着至关重要的作用。虽然肠道微生物代谢产物,如胆碱衍生物,已经被广泛研究和回顾,但新出现的证据表明,细菌细胞外囊泡(BEVs),一种由细菌分泌的膜源性脂质双分子层,在这一过程中也起着重要作用。然而,bev在宿主-微生物群相互作用中的作用仍然没有得到充分的探索。本文旨在阐明bev沿肠-心轴介导的复杂通讯。在这篇综述中,我们总结了目前关于bev的知识,特别关注病原体来源的bev如何促进动脉粥样硬化,以及来自肠道共生体和益生菌的bev如何减缓其进展。我们还探讨了工程bev在预防和治疗动脉粥样硬化方面的潜力和挑战。最后,我们讨论了bev在动脉粥样硬化诊断和治疗中的益处和挑战,并提出了解决这些问题的未来研究方向。
{"title":"Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis.","authors":"Yuling Lin, Jingyu Wang, Fan Bu, Ruyi Zhang, Junhui Wang, Yubing Wang, Mei Huang, Yiyi Huang, Lei Zheng, Qian Wang, Xiumei Hu","doi":"10.1080/19490976.2025.2452229","DOIUrl":"10.1080/19490976.2025.2452229","url":null,"abstract":"<p><p>Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2452229"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143004537","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
Novel role of FTO in regulation of gut-brain communication via Desulfovibrio fairfieldensis-produced hydrogen sulfide under arsenic exposure. FTO在砷暴露下通过fairfield脱硫弧菌产生的硫化氢调节肠-脑通讯中的新作用。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-01-24 DOI: 10.1080/19490976.2024.2438471
Ruonan Chen, Xiaoqin Chai, Yunxiao Zhang, Tianxiu Zhou, Yinyin Xia, Xuejun Jiang, Bo Lv, Jun Zhang, Lixiao Zhou, Xin Tian, Ruonan Wang, Lejiao Mao, Feng Zhao, Hongyang Zhang, Jun Hu, Jingfu Qiu, Zhen Zou, Chengzhi Chen

Fat mass and obesity-associated protein (FTO) is the key demethylase that reverses the abnormally altered N6-methyladenosine (m6A) modification in eukaryotic cells under environmental pollutants exposure. Arsenic is an environmental metalloid and can cause severe symptoms in human mainly through drinking water. However, there is no specific treatment for its toxic effects due to the uncovered mechanisms. We previously revealed that exposure to arsenic increased the level of m6A via down-regulation of FTO, which might serve as a potential target for intervention against arsenic-related disorders. In this study, our results demonstrated that chronic exposure to arsenic significantly disrupted the intestinal barrier and microenvironment. Also, this administration resulted in the enhancement of m6A modification and the reduction of FTO expression in the intestine. By using both CRISPR/Cas9-based FTO knock-in strategy and adeno-associated virus (AAV)-mediated overexpression of FTO in the intestine, we established for the first time that up-regulation of FTO remarkably ameliorated arsenic-induced disruption of intestinal barriers and altered microenvironment of mice. We also firstly identified a dominant gut microbial species, Desulfovibrio fairfieldensis, which was sharply reduced in arsenic-exposed mice, was able to proceed arsenic-induced neurobehavioral impairments by declining the levels of its major metabolite hydrogen sulfide. Administration of Desulfovibrio fairfieldensis could significantly alleviate the neurotoxicity of arsenic. Intriguingly, the beneficial effects of FTO against arsenic neurotoxicity possibly occurred through a novel gut-brain communication via Desulfovibrio fairfieldensis and its produced hydrogen sulfide. Collectively, these findings will provide new ideas for understanding the mechanisms of arsenic-induced toxic effects from a gut-brain communication perspective, and will assist the development of explicit intervention strategy via regulation of a new potential target FTO for prevention and treatment against arsenic-related both intestinal and neurological disorders.

脂肪质量和肥胖相关蛋白(FTO)是在环境污染物暴露下真核细胞中逆转异常改变的n6 -甲基腺苷(m6A)修饰的关键去甲基化酶。砷是一种环境类金属,主要通过饮水引起人体严重症状。然而,由于其机制尚未发现,目前尚无针对其毒性作用的特异性治疗方法。我们之前发现,暴露于砷会通过下调FTO来增加m6A水平,这可能是干预砷相关疾病的潜在靶点。在这项研究中,我们的研究结果表明,长期暴露于砷显著破坏肠道屏障和微环境。此外,这种给药可以增强肠道中m6A的修饰和降低FTO的表达。通过使用基于CRISPR/ cas9的FTO敲入策略和腺相关病毒(AAV)介导的FTO在肠道中的过表达,我们首次发现FTO的上调显著改善了砷诱导的肠道屏障破坏和小鼠微环境的改变。我们还首次发现了一种优势的肠道微生物物种,fairfield densis,在砷暴露的小鼠中急剧减少,能够通过降低其主要代谢物硫化氢的水平来进行砷诱导的神经行为损伤。施用脱硫弧菌可显著减轻砷的神经毒性。有趣的是,FTO对砷神经毒性的有益作用可能是通过fairfield Desulfovibrio densis及其产生的硫化氢的新型肠-脑通讯发生的。总的来说,这些发现将从肠脑通讯的角度为理解砷诱导毒性作用的机制提供新的思路,并将通过调节新的潜在靶点FTO来帮助制定明确的干预策略,以预防和治疗砷相关的肠道和神经系统疾病。
{"title":"Novel role of FTO in regulation of gut-brain communication via <i>Desulfovibrio fairfieldensis</i>-produced hydrogen sulfide under arsenic exposure.","authors":"Ruonan Chen, Xiaoqin Chai, Yunxiao Zhang, Tianxiu Zhou, Yinyin Xia, Xuejun Jiang, Bo Lv, Jun Zhang, Lixiao Zhou, Xin Tian, Ruonan Wang, Lejiao Mao, Feng Zhao, Hongyang Zhang, Jun Hu, Jingfu Qiu, Zhen Zou, Chengzhi Chen","doi":"10.1080/19490976.2024.2438471","DOIUrl":"10.1080/19490976.2024.2438471","url":null,"abstract":"<p><p>Fat mass and obesity-associated protein (FTO) is the key demethylase that reverses the abnormally altered N6-methyladenosine (m6A) modification in eukaryotic cells under environmental pollutants exposure. Arsenic is an environmental metalloid and can cause severe symptoms in human mainly through drinking water. However, there is no specific treatment for its toxic effects due to the uncovered mechanisms. We previously revealed that exposure to arsenic increased the level of m6A via down-regulation of FTO, which might serve as a potential target for intervention against arsenic-related disorders. In this study, our results demonstrated that chronic exposure to arsenic significantly disrupted the intestinal barrier and microenvironment. Also, this administration resulted in the enhancement of m6A modification and the reduction of FTO expression in the intestine. By using both CRISPR/Cas9-based FTO knock-in strategy and adeno-associated virus (AAV)-mediated overexpression of FTO in the intestine, we established for the first time that up-regulation of FTO remarkably ameliorated arsenic-induced disruption of intestinal barriers and altered microenvironment of mice. We also firstly identified a dominant gut microbial species, <i>Desulfovibrio fairfieldensis</i>, which was sharply reduced in arsenic-exposed mice, was able to proceed arsenic-induced neurobehavioral impairments by declining the levels of its major metabolite hydrogen sulfide. Administration of <i>Desulfovibrio fairfieldensis</i> could significantly alleviate the neurotoxicity of arsenic. Intriguingly, the beneficial effects of FTO against arsenic neurotoxicity possibly occurred through a novel gut-brain communication via <i>Desulfovibrio fairfieldensis</i> and its produced hydrogen sulfide. Collectively, these findings will provide new ideas for understanding the mechanisms of arsenic-induced toxic effects from a gut-brain communication perspective, and will assist the development of explicit intervention strategy via regulation of a new potential target FTO for prevention and treatment against arsenic-related both intestinal and neurological disorders.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2438471"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11776478/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143033093","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
Anxiety-like behavior during protracted morphine withdrawal is driven by gut microbial dysbiosis and attenuated with probiotic treatment. 长期吗啡戒断期间的焦虑样行为是由肠道微生物生态失调驱动的,并通过益生菌治疗减弱。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-06-15 DOI: 10.1080/19490976.2025.2517838
Mark Oppenheimer, Junyi Tao, Shamsudheen Moidunny, Sabita Roy

The development of anxiety during protracted opioid withdrawal heightens the risk of relapse into the cycle of addiction. Understanding the mechanisms driving anxiety during opioid withdrawal could facilitate the development of therapeutics to prevent negative affect and promote continued abstinence. Our lab has previously established the gut microbiome as a driver of various side effects of opioid use, including analgesic tolerance and somatic withdrawal symptoms. We therefore hypothesized that the gut microbiome contributes to the development of anxiety-like behavior during protracted opioid withdrawal. In this study, we first established a mouse model of protracted morphine withdrawal, characterized by anxiety-like behavior and gut microbial dysbiosis. Next, we used fecal microbiota transplantation (FMT) to show that gut dysbiosis alone is sufficient to induce anxiety-like behavior. We further demonstrated that probiotic therapy during morphine withdrawal attenuated the onset of anxiety-like behavior, highlighting its therapeutic potential. Lastly, we examined transcriptional changes in the amygdala of morphine-withdrawn mice treated with probiotics to explore mechanisms by which the gut-brain axis mediates anxiety-like behavior. Our results support the use of probiotics as a promising therapeutic strategy to prevent gut dysbiosis and associated anxiety during opioid withdrawal, with potential implications for improving treatment outcomes in opioid recovery programs.

在阿片类药物长期戒断期间,焦虑的发展增加了再次进入成瘾循环的风险。了解阿片类药物戒断期间驱动焦虑的机制可以促进治疗方法的发展,以防止负面影响和促进持续戒断。我们的实验室之前已经确定肠道微生物组是阿片类药物使用的各种副作用的驱动因素,包括镇痛耐受性和躯体戒断症状。因此,我们假设肠道微生物群有助于阿片类药物长期戒断期间焦虑样行为的发展。在本研究中,我们首先建立了以焦虑样行为和肠道微生物失调为特征的长期吗啡戒断小鼠模型。接下来,我们使用粪便微生物群移植(FMT)来证明肠道生态失调本身足以诱导焦虑样行为。我们进一步证明,在吗啡戒断期间益生菌治疗减轻了焦虑样行为的发作,突出了其治疗潜力。最后,我们研究了用益生菌治疗吗啡戒断小鼠杏仁核的转录变化,以探索肠-脑轴介导焦虑样行为的机制。我们的研究结果支持使用益生菌作为一种有希望的治疗策略,以防止阿片类药物戒断期间肠道生态失调和相关焦虑,并对改善阿片类药物恢复计划的治疗结果具有潜在的意义。
{"title":"Anxiety-like behavior during protracted morphine withdrawal is driven by gut microbial dysbiosis and attenuated with probiotic treatment.","authors":"Mark Oppenheimer, Junyi Tao, Shamsudheen Moidunny, Sabita Roy","doi":"10.1080/19490976.2025.2517838","DOIUrl":"10.1080/19490976.2025.2517838","url":null,"abstract":"<p><p>The development of anxiety during protracted opioid withdrawal heightens the risk of relapse into the cycle of addiction. Understanding the mechanisms driving anxiety during opioid withdrawal could facilitate the development of therapeutics to prevent negative affect and promote continued abstinence. Our lab has previously established the gut microbiome as a driver of various side effects of opioid use, including analgesic tolerance and somatic withdrawal symptoms. We therefore hypothesized that the gut microbiome contributes to the development of anxiety-like behavior during protracted opioid withdrawal. In this study, we first established a mouse model of protracted morphine withdrawal, characterized by anxiety-like behavior and gut microbial dysbiosis. Next, we used fecal microbiota transplantation (FMT) to show that gut dysbiosis alone is sufficient to induce anxiety-like behavior. We further demonstrated that probiotic therapy during morphine withdrawal attenuated the onset of anxiety-like behavior, highlighting its therapeutic potential. Lastly, we examined transcriptional changes in the amygdala of morphine-withdrawn mice treated with probiotics to explore mechanisms by which the gut-brain axis mediates anxiety-like behavior. Our results support the use of probiotics as a promising therapeutic strategy to prevent gut dysbiosis and associated anxiety during opioid withdrawal, with potential implications for improving treatment outcomes in opioid recovery programs.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2517838"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12169037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144301922","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
Lactulose selectively stimulates members of the gut microbiota, as determined by multi-modal activity-based sorting. 乳果糖选择性地刺激肠道微生物群的成员,由多模态活性为基础的分类确定。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2025-12-01 Epub Date: 2025-06-27 DOI: 10.1080/19490976.2025.2525482
Hamid Rasoulimehrabani, Alessandra Riva, Deniz Inan, Adnan Hodžić, Bela Hausmann, Georgi Nikolov, Sanaz Khadem, Norbert Hieger, Julia Wiesenbauer, Christina Kaiser, Verena Filz, Thomas Böttcher, David Berry

There is much interest in the development of dietary supplements that selectively promote the growth of beneficial gut bacteria. The selectivity of many candidate prebiotics has, however, not been thoroughly investigated. Here, we evaluated stimulation of the human gut microbiota by the disaccharide lactulose using an ex vivo multimodal activity-based cell sorting approach. Incubation of human donor stool with lactulose resulted in growth or stimulation of a restricted diversity of bacterial genera, most prominently Bifidobacterium, Collinsella, and Lactococcus. Physiological analysis of lactulose-responsive strains isolated by Raman activated cell sorting revealed that most were capable of lactulose degradation. Among these isolates, Lactococcus lactis could not degrade lactulose, but its growth was boosted by co-cultivation with lactulose degraders. This suggests that inter-species facilitation contributes to the lactulose degradation niche. Moreover, we observed that lactulose selectively activates metabolically important taxa, including health-associated genera such as Faecalibacterium and Gemmiger1,2,3, potentially indicating broader functional effects beyond compositional changes. These results provide novel insights into the physiology and ecology of lactulose utilization by the human gut microbiota and underscore the potential of lactulose as a prebiotic dietary supplement.

人们对有选择地促进有益肠道细菌生长的膳食补充剂的开发非常感兴趣。然而,许多候选益生元的选择性尚未得到彻底的研究。在这里,我们使用基于离体多模式活性的细胞分选方法评估了双糖乳果糖对人体肠道微生物群的刺激。将人供体粪便与乳果糖孵育,导致有限种类细菌属的生长或刺激,最突出的是双歧杆菌、Collinsella和乳球菌。通过拉曼活化细胞分选对乳果糖敏感的菌株进行生理分析,发现大多数菌株能够降解乳果糖。在这些分离株中,乳酸乳球菌不能降解乳果糖,但与乳果糖降解剂共同培养可促进其生长。这表明种间促进有助于乳果糖降解生态位。此外,我们观察到乳果糖选择性地激活代谢重要的分类群,包括与健康相关的属,如Faecalibacterium和Gemmiger1,2,3,这可能表明除了成分变化之外,还有更广泛的功能影响。这些结果为人类肠道菌群利用乳果糖的生理学和生态学提供了新的见解,并强调了乳果糖作为益生元膳食补充剂的潜力。
{"title":"Lactulose selectively stimulates members of the gut microbiota, as determined by multi-modal activity-based sorting.","authors":"Hamid Rasoulimehrabani, Alessandra Riva, Deniz Inan, Adnan Hodžić, Bela Hausmann, Georgi Nikolov, Sanaz Khadem, Norbert Hieger, Julia Wiesenbauer, Christina Kaiser, Verena Filz, Thomas Böttcher, David Berry","doi":"10.1080/19490976.2025.2525482","DOIUrl":"https://doi.org/10.1080/19490976.2025.2525482","url":null,"abstract":"<p><p>There is much interest in the development of dietary supplements that selectively promote the growth of beneficial gut bacteria. The selectivity of many candidate prebiotics has, however, not been thoroughly investigated. Here, we evaluated stimulation of the human gut microbiota by the disaccharide lactulose using an <i>ex vivo</i> multimodal activity-based cell sorting approach. Incubation of human donor stool with lactulose resulted in growth or stimulation of a restricted diversity of bacterial genera, most prominently <i>Bifidobacterium</i>, <i>Collinsella</i>, and <i>Lactococcus</i>. Physiological analysis of lactulose-responsive strains isolated by Raman activated cell sorting revealed that most were capable of lactulose degradation. Among these isolates, <i>Lactococcus lactis</i> could not degrade lactulose, but its growth was boosted by co-cultivation with lactulose degraders. This suggests that inter-species facilitation contributes to the lactulose degradation niche. Moreover, we observed that lactulose selectively activates metabolically important taxa, including health-associated genera such as <i>Faecalibacterium</i> and <i>Gemmiger</i><sup>1,2,3</sup>, potentially indicating broader functional effects beyond compositional changes. These results provide novel insights into the physiology and ecology of lactulose utilization by the human gut microbiota and underscore the potential of lactulose as a prebiotic dietary supplement.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":"17 1","pages":"2525482"},"PeriodicalIF":12.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144505551","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