首页 > 最新文献

Gut Microbes最新文献

英文 中文
Incorporating metabolic activity, taxonomy and community structure to improve microbiome-based predictive models for host phenotype prediction. 结合代谢活动、分类和群落结构,改进基于微生物组的宿主表型预测模型。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-01-12 DOI: 10.1080/19490976.2024.2302076
Mahsa Monshizadeh, Yuzhen Ye

We developed MicroKPNN, a prior-knowledge guided interpretable neural network for microbiome-based human host phenotype prediction. The prior knowledge used in MicroKPNN includes the metabolic activities of different bacterial species, phylogenetic relationships, and bacterial community structure, all in a shallow neural network. Application of MicroKPNN to seven gut microbiome datasets (involving five different human diseases including inflammatory bowel disease, type 2 diabetes, liver cirrhosis, colorectal cancer, and obesity) shows that incorporation of the prior knowledge helped improve the microbiome-based host phenotype prediction. MicroKPNN outperformed fully connected neural network-based approaches in all seven cases, with the most improvement of accuracy in the prediction of type 2 diabetes. MicroKPNN outperformed a recently developed deep-learning based approach DeepMicro, which selects the best combination of autoencoder and machine learning approach to make predictions, in all of the seven cases. Importantly, we showed that MicroKPNN provides a way for interpretation of the predictive models. Using importance scores estimated for the hidden nodes, MicroKPNN could provide explanations for prior research findings by highlighting the roles of specific microbiome components in phenotype predictions. In addition, it may suggest potential future research directions for studying the impacts of microbiome on host health and diseases. MicroKPNN is publicly available at https://github.com/mgtools/MicroKPNN.

我们开发了 MicroKPNN,这是一种先验知识指导下的可解释神经网络,用于基于微生物组的人类宿主表型预测。MicroKPNN 使用的先验知识包括不同细菌物种的代谢活动、系统发育关系和细菌群落结构,所有这些都在一个浅层神经网络中。MicroKPNN 在七个肠道微生物组数据集(涉及五种不同的人类疾病,包括炎症性肠病、2 型糖尿病、肝硬化、结直肠癌和肥胖症)中的应用表明,先验知识的加入有助于改善基于微生物组的宿主表型预测。在所有七个案例中,MicroKPNN 的表现都优于基于全连接神经网络的方法,其中对 2 型糖尿病的预测准确率提高最多。MicroKPNN 在所有 7 个案例中的表现都优于最近开发的基于深度学习的 DeepMicro 方法,该方法选择自动编码器和机器学习方法的最佳组合来进行预测。重要的是,我们发现 MicroKPNN 提供了一种解释预测模型的方法。利用为隐藏节点估算的重要性分数,MicroKPNN 可以通过强调特定微生物组成分在表型预测中的作用,为先前的研究结果提供解释。此外,它还可以为研究微生物组对宿主健康和疾病的影响提出潜在的未来研究方向。MicroKPNN 可通过 https://github.com/mgtools/MicroKPNN 公开获取。
{"title":"Incorporating metabolic activity, taxonomy and community structure to improve microbiome-based predictive models for host phenotype prediction.","authors":"Mahsa Monshizadeh, Yuzhen Ye","doi":"10.1080/19490976.2024.2302076","DOIUrl":"10.1080/19490976.2024.2302076","url":null,"abstract":"<p><p>We developed MicroKPNN, a prior-knowledge guided interpretable neural network for microbiome-based human host phenotype prediction. The prior knowledge used in MicroKPNN includes the metabolic activities of different bacterial species, phylogenetic relationships, and bacterial community structure, all in a shallow neural network. Application of MicroKPNN to seven gut microbiome datasets (involving five different human diseases including inflammatory bowel disease, type 2 diabetes, liver cirrhosis, colorectal cancer, and obesity) shows that incorporation of the prior knowledge helped improve the microbiome-based host phenotype prediction. MicroKPNN outperformed fully connected neural network-based approaches in all seven cases, with the most improvement of accuracy in the prediction of type 2 diabetes. MicroKPNN outperformed a recently developed deep-learning based approach DeepMicro, which selects the best combination of autoencoder and machine learning approach to make predictions, in all of the seven cases. Importantly, we showed that MicroKPNN provides a way for interpretation of the predictive models. Using importance scores estimated for the hidden nodes, MicroKPNN could provide explanations for prior research findings by highlighting the roles of specific microbiome components in phenotype predictions. In addition, it may suggest potential future research directions for studying the impacts of microbiome on host health and diseases. MicroKPNN is publicly available at https://github.com/mgtools/MicroKPNN.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10793686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139424637","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
Disruption of gut barrier integrity and host-microbiome interactions underlie MASLD severity in patients with type-2 diabetes mellitus. 2 型糖尿病患者肠道屏障完整性和宿主微生物组相互作用的破坏是 MASLD 严重性的基础。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-01-18 DOI: 10.1080/19490976.2024.2304157
R Forlano, L Martinez-Gili, P Takis, J Miguens-Blanco, T Liu, E Triantafyllou, C Skinner, R Loomba, M Thursz, J R Marchesi, B H Mullish, P Manousou

Aberration of the "gut-liver axis" contributes to the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Here, we use multi-omics to analyze the gut microbiota composition and metabolic profile of patients with type-2 diabetes mellitus (T2DM). T2DM patients were screened for liver disease by blood tests, ultrasound, and liver stiffness measurements. Stool microbiota was analyzed by 16S rRNA gene sequencing; metabolomic profiling by Nuclear Magnetic Resonance spectroscopy and Ultra-High Performance-Mass Spectrometry. Microbiome and metabolic signatures were analyzed in the whole cohort and in matched subsets to identify signatures specific for steatosis (MASLD±) or fibrosis (Fibrosis±). Gut permeability was assessed in-vitro using monolayers of MDCK cells and trans-epithelial electric resistance (TEER). Cytokine profile was assessed in serum and stools.Overall, 285 patients were enrolled: 255 serum, 252 urine and 97 stool samples were analyzed. Anaeroplasma and Escherichia/Shigella ASVs were higher, while Butyricicoccus ASVs were lower in those with normal liver. In MASLD±, Butyricicoccus ASV was significantly higher in those with steatosis. In the Fibrosis±, Butyricicoccus ASV was significantly lower in those with fibrosis. Glycochenodeoxycholic acid-3-sulfate (G-UDCA-3S) appeared to be higher in MASLD with fibrosis. Fecal water from patients with MASLD and fibrosis caused the greatest drop in the TEER vs those with normal liver; this was reversed with protease inhibitors. Finally, fecal IL-13 was lower in MASLD with fibrosis. We identified microbiome signatures which were specific for steatosis and fibrosis and independent of other metabolic risk factors. Moreover, we conclude that protease-related gut permeability plays a role in those MASLD patients with fibrosis, and that disease progression is linked to a gut-liver axis which is at least partially independent of T2DM.

肠肝轴 "畸变是代谢功能障碍相关性脂肪性肝病(MASLD)发生和发展的原因之一。在这里,我们利用多组学分析了2型糖尿病(T2DM)患者的肠道微生物群组成和代谢概况。我们通过血液化验、超声波检查和肝脏硬度测量对 T2DM 患者进行了肝病筛查。粪便微生物群通过 16S rRNA 基因测序进行分析;代谢组学分析通过核磁共振波谱和超高效质谱仪进行。对整个队列和匹配子集的微生物组和代谢特征进行了分析,以确定脂肪变性(MASLD±)或纤维化(Fibrosis±)的特异性特征。利用 MDCK 细胞单层和跨上皮电阻(TEER)对体外肠道通透性进行了评估。对血清和粪便中的细胞因子谱进行了评估。总共有 285 名患者参与了研究:对 255 份血清样本、252 份尿液样本和 97 份粪便样本进行了分析。在肝功能正常的患者中,厌氧支原体和埃希氏/志贺氏菌 ASV 较高,而丁酸球菌 ASV 较低。在 MASLD±中,脂肪肝患者的丁酸球菌 ASV 明显较高。在纤维化±中,纤维化患者的丁酸球菌 ASV 明显较低。甘氨胆酸-3-硫酸盐(G-UDCA-3S)在伴有纤维化的 MASLD 中似乎更高。MASLD和肝纤维化患者的粪便水与正常肝脏患者的粪便水相比,TEER下降幅度最大;蛋白酶抑制剂可逆转这种情况。最后,纤维化的MASLD患者粪便中的IL-13含量较低。我们发现了特异于脂肪变性和肝纤维化的微生物组特征,并且与其他代谢风险因素无关。此外,我们还得出结论,蛋白酶相关的肠道通透性在纤维化的 MASLD 患者中起着一定的作用,而且疾病的进展与肠道-肝脏轴相关,至少部分与 T2DM 无关。
{"title":"Disruption of gut barrier integrity and host-microbiome interactions underlie MASLD severity in patients with type-2 diabetes mellitus.","authors":"R Forlano, L Martinez-Gili, P Takis, J Miguens-Blanco, T Liu, E Triantafyllou, C Skinner, R Loomba, M Thursz, J R Marchesi, B H Mullish, P Manousou","doi":"10.1080/19490976.2024.2304157","DOIUrl":"10.1080/19490976.2024.2304157","url":null,"abstract":"<p><p>Aberration of the \"gut-liver axis\" contributes to the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Here, we use multi-omics to analyze the gut microbiota composition and metabolic profile of patients with type-2 diabetes mellitus (T2DM). T2DM patients were screened for liver disease by blood tests, ultrasound, and liver stiffness measurements. Stool microbiota was analyzed by 16S rRNA gene sequencing; metabolomic profiling by Nuclear Magnetic Resonance spectroscopy and Ultra-High Performance-Mass Spectrometry. Microbiome and metabolic signatures were analyzed in the whole cohort and in matched subsets to identify signatures specific for steatosis (MASLD±) or fibrosis (Fibrosis±). Gut permeability was assessed <i>in-vitro</i> using monolayers of MDCK cells and trans-epithelial electric resistance (TEER). Cytokine profile was assessed in serum and stools.Overall, 285 patients were enrolled: 255 serum, 252 urine and 97 stool samples were analyzed. <i>Anaeroplasma</i> and <i>Escherichia/Shigella</i> ASVs were higher, while <i>Butyricicoccus</i> ASVs were lower in those with normal liver. In MASLD±, <i>Butyricicoccus</i> ASV was significantly higher in those with steatosis. In the Fibrosis±, <i>Butyricicoccus</i> ASV was significantly lower in those with fibrosis. Glycochenodeoxycholic acid-3-sulfate (G-UDCA-3S) appeared to be higher in MASLD with fibrosis. Fecal water from patients with MASLD and fibrosis caused the greatest drop in the TEER vs those with normal liver; this was reversed with protease inhibitors. Finally, fecal IL-13 was lower in MASLD with fibrosis. We identified microbiome signatures which were specific for steatosis and fibrosis and independent of other metabolic risk factors. Moreover, we conclude that protease-related gut permeability plays a role in those MASLD patients with fibrosis, and that disease progression is linked to a gut-liver axis which is at least partially independent of T2DM.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10798360/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139485497","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-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice. 肠道微生物群驱动的代谢改变揭示了阿尔茨海默氏症模型小鼠的肠脑交流。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-01-23 DOI: 10.1080/19490976.2024.2302310
Yijing Chen, Yinhu Li, Yingying Fan, Shuai Chen, Li Chen, Yuewen Chen, Yu Chen

The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the "microbiota - metabolite - brain axis" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched Dubosiella affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that Dubosiella enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria Erysipelatoclostridium and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.

肠道微生物群(GM)及其代谢产物会影响宿主的神经系统,并与各种神经系统疾病的病因有关。然而,在阿尔茨海默病(AD)的致病压力下,肠道微生物群的具体改变及其对 "微生物群-代谢物-脑轴 "的贡献仍不清楚。在这里,我们研究了 APP/PS1 小鼠和野生型(WT)小鼠的基因组和粪便、血清、皮质代谢组,在两组小鼠共享的无标度基因组网络中发现了 AD 小鼠中不同的枢纽细菌。此外,我们还在AD小鼠和WT小鼠之间发现了不同的外周-中心代谢景观,其特点是胆汁酸(如脱氧胆酸和异脱氧胆酸)和不饱和脂肪酸(如11Z-二十烯酸和棕榈油酸)。机器学习模型揭示了差异/枢纽细菌与从外周到大脑的这些代谢特征之间的关系。值得注意的是,富含AD的Dubosiella通过皮质棕榈油酸影响AD的发生,反之亦然。考虑到 AD 小鼠的转基因背景,我们认为富集杜博斯菌可通过棕榈油酸的合成阻碍 AD 的发展,而棕榈油酸对炎症和代谢紊乱具有保护作用。我们还发现了另一种关联,即由粪便脱氧胆酸介导的 AD 中枢细菌埃希氏菌与 AD 的发生之间的相互作用,人类脱氧胆酸水平与认知评分之间的相关性也证实了这一点。总之,这项研究阐明了转基因网络的改变、转基因对外周-中枢代谢景观的贡献以及代谢产物在转基因与AD发生之间的中介作用,从而揭示了细菌在AD发病机制和致病压力下肠道-大脑沟通中的关键作用。
{"title":"Gut microbiota-driven metabolic alterations reveal gut-brain communication in Alzheimer's disease model mice.","authors":"Yijing Chen, Yinhu Li, Yingying Fan, Shuai Chen, Li Chen, Yuewen Chen, Yu Chen","doi":"10.1080/19490976.2024.2302310","DOIUrl":"10.1080/19490976.2024.2302310","url":null,"abstract":"<p><p>The gut microbiota (GM) and its metabolites affect the host nervous system and are involved in the pathogeneses of various neurological diseases. However, the specific GM alterations under pathogenetic pressure and their contributions to the \"microbiota - metabolite - brain axis\" in Alzheimer's disease (AD) remain unclear. Here, we investigated the GM and the fecal, serum, cortical metabolomes in APP/PS1 and wild-type (WT) mice, revealing distinct hub bacteria in AD mice within scale-free GM networks shared by both groups. Moreover, we identified diverse peripheral - central metabolic landscapes between AD and WT mice that featured bile acids (e.g. deoxycholic and isodeoxycholic acid) and unsaturated fatty acids (e.g. 11Z-eicosenoic and palmitoleic acid). Machine-learning models revealed the relationships between the differential/hub bacteria and these metabolic signatures from the periphery to the brain. Notably, AD-enriched <i>Dubosiella</i> affected AD occurrence via cortical palmitoleic acid and vice versa. Considering the transgenic background of the AD mice, we propose that <i>Dubosiella</i> enrichment impedes AD progression via the synthesis of palmitoleic acid, which has protective properties against inflammation and metabolic disorders. We identified another association involving fecal deoxycholic acid-mediated interactions between the AD hub bacteria <i>Erysipelatoclostridium</i> and AD occurrence, which was corroborated by the correlation between deoxycholate levels and cognitive scores in humans. Overall, this study elucidated the GM network alterations, contributions of the GM to peripheral - central metabolic landscapes, and mediatory roles of metabolites between the GM and AD occurrence, thus revealing the critical roles of bacteria in AD pathogenesis and gut - brain communications under pathogenetic pressure.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10807476/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139542255","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
Anti-S-layer monoclonal antibodies impact Clostridioides difficile physiology. 抗 S 层单克隆抗体影响艰难梭菌的生理机能。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-01-30 DOI: 10.1080/19490976.2023.2301147
Lise Hunault, Emile Auria, Patrick England, Julien Deschamps, Romain Briandet, Vanessa Kremer, Bruno Iannascoli, Léo Vidal-Maison, Chunguang Guo, Lynn Macdonald, Séverine Péchiné, Cécile Denève-Larrazet, Bruno Dupuy, Guy Gorochov, Pierre Bruhns, Delphine Sterlin

Clostridioides difficile (C. difficile), a gram-positive anaerobic and spore-forming bacterium, is the leading cause of nosocomial antibiotic-associated diarrhea in adults which is characterized by high levels of recurrence and mortality. Surface (S)-layer Protein A (SlpA), the most abundantly expressed protein on the bacterial surface, plays a crucial role in the early stages of infection although the nature of its involvement in C. difficile physiology is yet to be fully understood. Anti-S-layer antibodies have been identified in the sera of convalescent patients and have been correlated with improved outcomes of C. difficile infection (CDI). However, the precise mechanisms by which anti-S-layer antibodies confer protection to the host remain unknown. In this study, we report the first monoclonal antibodies (mAbs) targeting the S-layer of reference strain 630. Characterization of these mAbs unraveled important roles for the S-layer protein in growth, toxin secretion, and biofilm formation by C. difficile, with differential and even opposite effects of various anti-SlpA mAbs on these functions. Moreover, one anti-SlpA mAb impaired C. difficile growth and conferred sensitivity to lysozyme-induced lysis. The results of this study show that anti-S-layer antibody responses can be beneficial or harmful for the course of CDI and provide important insights for the development of adequate S-layer-targeting therapeutics.

艰难梭状芽孢杆菌(C. difficile)是一种革兰氏阳性厌氧芽孢杆菌,是引起成人院内抗生素相关性腹泻的主要原因,其特点是复发率和死亡率高。表面(S)层蛋白 A(SlpA)是细菌表面表达最丰富的蛋白质,在感染的早期阶段起着至关重要的作用,但它参与艰难梭菌生理学的性质尚不完全清楚。在康复患者的血清中发现了抗 S 层抗体,这种抗体与艰难梭菌感染(CDI)疗效的改善有关。然而,抗 S 层抗体为宿主提供保护的确切机制仍然未知。在这项研究中,我们首次报道了针对参考菌株 630 的 S 层的单克隆抗体(mAbs)。这些 mAbs 的特性揭示了 S 层蛋白在艰难梭菌的生长、毒素分泌和生物膜形成中的重要作用,不同的抗 SlpA mAbs 对这些功能的影响不同,甚至相反。此外,一种抗 SlpA mAb 会阻碍艰难梭菌的生长,并使其对溶菌酶诱导的裂解敏感。这项研究结果表明,抗S层抗体反应对艰难梭菌感染的病程有利或有害,并为开发适当的S层靶向疗法提供了重要启示。
{"title":"Anti-S-layer monoclonal antibodies impact <i>Clostridioides difficile</i> physiology.","authors":"Lise Hunault, Emile Auria, Patrick England, Julien Deschamps, Romain Briandet, Vanessa Kremer, Bruno Iannascoli, Léo Vidal-Maison, Chunguang Guo, Lynn Macdonald, Séverine Péchiné, Cécile Denève-Larrazet, Bruno Dupuy, Guy Gorochov, Pierre Bruhns, Delphine Sterlin","doi":"10.1080/19490976.2023.2301147","DOIUrl":"10.1080/19490976.2023.2301147","url":null,"abstract":"<p><p><i>Clostridioides difficile</i> (<i>C. difficile</i>), a gram-positive anaerobic and spore-forming bacterium, is the leading cause of nosocomial antibiotic-associated diarrhea in adults which is characterized by high levels of recurrence and mortality. Surface (S)-layer Protein A (SlpA), the most abundantly expressed protein on the bacterial surface, plays a crucial role in the early stages of infection although the nature of its involvement in <i>C. difficile</i> physiology is yet to be fully understood. Anti-S-layer antibodies have been identified in the sera of convalescent patients and have been correlated with improved outcomes of <i>C. difficile</i> infection (CDI). However, the precise mechanisms by which anti-S-layer antibodies confer protection to the host remain unknown. In this study, we report the first monoclonal antibodies (mAbs) targeting the S-layer of reference strain 630. Characterization of these mAbs unraveled important roles for the S-layer protein in growth, toxin secretion, and biofilm formation by <i>C. difficile</i>, with differential and even opposite effects of various anti-SlpA mAbs on these functions. Moreover, one anti-SlpA mAb impaired <i>C. difficile</i> growth and conferred sensitivity to lysozyme-induced lysis. The results of this study show that anti-S-layer antibody responses can be beneficial or harmful for the course of CDI and provide important insights for the development of adequate S-layer-targeting therapeutics.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10829821/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575461","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
Phocaeicola vulgatus alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA. Phocaeicola vulgatus通过3-HPAA下调组蛋白乙酰化水平,缓解饮食诱导的代谢功能障碍相关脂肪性肝病的进展。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-02-05 DOI: 10.1080/19490976.2024.2309683
Shengxi Jin, Peng Chen, Jing Yang, Duguang Li, Xiaolong Liu, Yiyin Zhang, Qiming Xia, Yiling Li, Guoqiao Chen, Yixuan Li, Yifan Tong, Weihua Yu, Xiaoxiao Fan, Hui Lin

Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify Phocaeicola vulgatus (P. vulgatus) as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of P. vulgatus mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by P. vulgatus, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of P. vulgatus in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.

饮食引起的代谢功能障碍相关性脂肪肝(MASLD)是一种普遍存在的代谢性疾病,但有效的干预措施却很有限。解决这一问题的新方法是基于肠道微生物群的疗法。在我们的研究中,我们利用多组学分析鉴定了Phocaeicola vulgatus(P. vulgatus)作为治疗MASLD的潜在益生菌。我们在小鼠模型中的研究结果清楚地表明,补充 Phocaeicola vulgatus 可减轻 MASLD 的发展。这种益处部分归因于硫代蜗牛产生的代谢物 3-羟基苯乙酸(3-HPAA),它能降低 H3K27 的乙酰化水平,并下调角鲨烯环氧化物酶(SQLE)的转录,角鲨烯环氧化物酶是类固醇生物合成过程中的限速酶,能促进肝细胞中的脂质积累。这项研究强调了 P. vulgatus 在 MASLD 发病过程中的重要作用,以及其代谢产物 3-HPAA 在调节脂质平衡中的关键重要性。这些发现为MASLD的早期干预治疗提供了一个前景广阔的途径。
{"title":"<i>Phocaeicola vulgatus</i> alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA.","authors":"Shengxi Jin, Peng Chen, Jing Yang, Duguang Li, Xiaolong Liu, Yiyin Zhang, Qiming Xia, Yiling Li, Guoqiao Chen, Yixuan Li, Yifan Tong, Weihua Yu, Xiaoxiao Fan, Hui Lin","doi":"10.1080/19490976.2024.2309683","DOIUrl":"10.1080/19490976.2024.2309683","url":null,"abstract":"<p><p>Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify <i>Phocaeicola vulgatus (P. vulgatus)</i> as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of <i>P. vulgatus</i> mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by <i>P. vulgatus</i>, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of <i>P. vulgatus</i> in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10854360/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139681020","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
Repeated (S)-ketamine administration ameliorates the spatial working memory impairment in mice with chronic pain: role of the gut microbiota-brain axis. 重复服用(S)-氯胺酮可改善慢性疼痛小鼠的空间工作记忆障碍:肠道微生物群-大脑轴的作用。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-02-08 DOI: 10.1080/19490976.2024.2310603
Yubin Jiang, Xingming Wang, Jiawei Chen, Yibao Zhang, Kenji Hashimoto, Jian-Jun Yang, Zhiqiang Zhou

Chronic pain is commonly linked with diminished working memory. This study explores the impact of the anesthetic (S)-ketamine on spatial working memory in a chronic constriction injury (CCI) mouse model, focusing on gut microbiome. We found that multiple doses of (S)-ketamine, unlike a single dose, counteracted the reduced spontaneous alteration percentage (%SA) in the Y-maze spatial working memory test, without affecting mechanical or thermal pain sensitivity. Additionally, repeated (S)-ketamine treatments improved the abnormal composition of the gut microbiome (β-diversity), as indicated by fecal 16S rRNA analysis, and increased levels of butyrate, a key gut - brain axis mediator. Protein analysis showed that these treatments also corrected the upregulated histone deacetylase 2 (HDAC2) and downregulated brain-derived neurotrophic factor (BDNF) in the hippocampi of CCI mice. Remarkably, fecal microbiota transplantation from mice treated repeatedly with (S)-ketamine to CCI mice restored %SA and hippocampal BDNF levels in CCI mice. Butyrate supplementation alone also improved %SA, BDNF, and HDAC2 levels in CCI mice. Furthermore, the TrkB receptor antagonist ANA-12 negated the beneficial effects of repeated (S)-ketamine on spatial working memory impairment in CCI mice. These results indicate that repeated (S)-ketamine administration ameliorates spatial working memory impairment in CCI mice, mediated by a gut microbiota - brain axis, primarily through the enhancement of hippocampal BDNF - TrkB signaling by butyrate.

慢性疼痛通常与工作记忆减退有关。本研究探讨了麻醉剂(S)-氯胺酮对慢性收缩性损伤(CCI)小鼠模型空间工作记忆的影响,重点关注肠道微生物组。我们发现,与单剂量不同,多剂量(S)-氯胺酮能抵消Y-迷宫空间工作记忆测试中自发改变百分比(%SA)的降低,而不影响机械或热痛觉敏感性。此外,粪便 16S rRNA 分析显示,重复(S)-氯胺酮治疗可改善肠道微生物组(β-多样性)的异常组成,并提高丁酸盐(一种关键的肠道-大脑轴介质)的水平。蛋白质分析表明,这些处理方法还纠正了CCI小鼠海马中上调的组蛋白去乙酰化酶2(HDAC2)和下调的脑源性神经营养因子(BDNF)。值得注意的是,将反复接受(S)-氯胺酮治疗的小鼠的粪便微生物群移植到CCI小鼠体内,可恢复CCI小鼠的%SA和海马BDNF水平。单独补充丁酸盐也能改善CCI小鼠的%SA、BDNF和HDAC2水平。此外,TrkB受体拮抗剂ANA-12否定了重复(S)-氯胺酮对CCI小鼠空间工作记忆损伤的有益影响。这些结果表明,重复施用(S)-氯胺酮可改善CCI小鼠的空间工作记忆损伤,这是由肠道微生物群-大脑轴介导的,主要是通过丁酸盐增强海马BDNF-TrkB信号传导。
{"title":"Repeated (<i>S</i>)-ketamine administration ameliorates the spatial working memory impairment in mice with chronic pain: role of the gut microbiota-brain axis.","authors":"Yubin Jiang, Xingming Wang, Jiawei Chen, Yibao Zhang, Kenji Hashimoto, Jian-Jun Yang, Zhiqiang Zhou","doi":"10.1080/19490976.2024.2310603","DOIUrl":"10.1080/19490976.2024.2310603","url":null,"abstract":"<p><p>Chronic pain is commonly linked with diminished working memory. This study explores the impact of the anesthetic (<i>S</i>)-ketamine on spatial working memory in a chronic constriction injury (CCI) mouse model, focusing on gut microbiome. We found that multiple doses of (<i>S</i>)-ketamine, unlike a single dose, counteracted the reduced spontaneous alteration percentage (%SA) in the Y-maze spatial working memory test, without affecting mechanical or thermal pain sensitivity. Additionally, repeated (<i>S</i>)-ketamine treatments improved the abnormal composition of the gut microbiome (β-diversity), as indicated by fecal 16S rRNA analysis, and increased levels of butyrate, a key gut - brain axis mediator. Protein analysis showed that these treatments also corrected the upregulated histone deacetylase 2 (HDAC2) and downregulated brain-derived neurotrophic factor (BDNF) in the hippocampi of CCI mice. Remarkably, fecal microbiota transplantation from mice treated repeatedly with (<i>S</i>)-ketamine to CCI mice restored %SA and hippocampal BDNF levels in CCI mice. Butyrate supplementation alone also improved %SA, BDNF, and HDAC2 levels in CCI mice. Furthermore, the TrkB receptor antagonist ANA-12 negated the beneficial effects of repeated (<i>S</i>)-ketamine on spatial working memory impairment in CCI mice. These results indicate that repeated (<i>S</i>)-ketamine administration ameliorates spatial working memory impairment in CCI mice, mediated by a gut microbiota - brain axis, primarily through the enhancement of hippocampal BDNF - TrkB signaling by butyrate.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10860353/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139706573","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
Roles of microbiota in pancreatic cancer development and treatment. 微生物群在胰腺癌发展和治疗中的作用。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-02-27 DOI: 10.1080/19490976.2024.2320280
Mariana Santos Cruz, Joseph Tintelnot, Nicola Gagliani

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.

胰腺导管腺癌(PDAC)是一种侵袭性疾病,预后较差。这是因为大多数病例都是在晚期和姑息性疾病阶段才被确诊,而且耐药率很高。尽管人们一直在努力,但迄今为止,PDAC 的致癌机制及其对治疗的不良反应在很大程度上仍不清楚。随着对癌症微生物组研究的不断深入,越来越多的证据表明,细菌或真菌可能是 PDAC 癌症发生及其对化疗和免疫疗法耐药的关键因素,例如通过调节肿瘤微环境和重塑宿主免疫反应。在此,我们回顾了微生物群如何通过微生物衍生代谢产物直接或间接地发挥这些作用。最后,我们将进一步讨论调节微生物群组成作为 PDAC 治疗方法的潜力。
{"title":"Roles of microbiota in pancreatic cancer development and treatment.","authors":"Mariana Santos Cruz, Joseph Tintelnot, Nicola Gagliani","doi":"10.1080/19490976.2024.2320280","DOIUrl":"10.1080/19490976.2024.2320280","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10900280/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971702","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
A metatranscriptomics strategy for efficient characterization of the microbiome in human tissues with low microbial biomass. 一种元转录组学策略,用于高效描述微生物生物量较低的人体组织中的微生物组。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-02-29 DOI: 10.1080/19490976.2024.2323235
Joana Pereira-Marques, Rui M Ferreira, Ceu Figueiredo

The high background of host RNA poses a major challenge to metatranscriptome analysis of human samples. Hence, metatranscriptomics has been mainly applied to microbe-rich samples, while its application in human tissues with low ratio of microbial to host cells has yet to be explored. Since there is no computational workflow specifically designed for the taxonomic and functional analysis of this type of samples, we propose an effective metatranscriptomics strategy to accurately characterize the microbiome in human tissues with a low ratio of microbial to host content. We experimentally generated synthetic samples with well-characterized bacterial and host cell compositions, and mimicking human samples with high and low microbial loads. These synthetic samples were used for optimizing and establishing the workflow in a controlled setting. Our results show that the integration of the taxonomic analysis of optimized Kraken 2/Bracken with the functional analysis of HUMAnN 3 in samples with low microbial content, enables the accurate identification of a large number of microbial species with a low false-positive rate, while improving the detection of microbial functions. The effectiveness of our metatranscriptomics workflow was demonstrated in synthetic samples, simulated datasets, and most importantly, human gastric tissue specimens, thus providing a proof of concept for its applicability on mucosal tissues of the gastrointestinal tract. The use of an accurate and reliable metatranscriptomics approach for human tissues with low microbial content will expand our understanding of the functional activity of the mucosal microbiome, uncovering critical interactions between the microbiome and the host in health and disease.

宿主 RNA 的高背景对人体样本的元转录组分析构成了重大挑战。因此,元转录组学主要应用于富含微生物的样本,而在微生物与宿主细胞比例较低的人体组织中的应用还有待探索。由于目前还没有专门为这类样本的分类和功能分析而设计的计算工作流程,我们提出了一种有效的元转录组学策略,以准确表征微生物含量与宿主细胞含量比例较低的人体组织中的微生物组。我们通过实验生成了具有良好表征的细菌和宿主细胞组成的合成样本,并模拟了高微生物负荷和低微生物负荷的人体样本。这些合成样本用于在受控环境中优化和建立工作流程。我们的研究结果表明,在微生物含量较低的样本中,将优化的 Kraken 2/Bracken 分类分析与 HUMAnN 3 功能分析相结合,能准确识别大量微生物物种,且假阳性率较低,同时还能提高微生物功能的检测率。我们的元转录组学工作流程在合成样本、模拟数据集以及最重要的人体胃组织标本中都证明了其有效性,从而为其在胃肠道粘膜组织中的应用提供了概念验证。在微生物含量较低的人体组织中使用准确可靠的元转录组学方法,将扩大我们对粘膜微生物组功能活动的了解,揭示微生物组和宿主在健康和疾病中的重要相互作用。
{"title":"A metatranscriptomics strategy for efficient characterization of the microbiome in human tissues with low microbial biomass.","authors":"Joana Pereira-Marques, Rui M Ferreira, Ceu Figueiredo","doi":"10.1080/19490976.2024.2323235","DOIUrl":"10.1080/19490976.2024.2323235","url":null,"abstract":"<p><p>The high background of host RNA poses a major challenge to metatranscriptome analysis of human samples. Hence, metatranscriptomics has been mainly applied to microbe-rich samples, while its application in human tissues with low ratio of microbial to host cells has yet to be explored. Since there is no computational workflow specifically designed for the taxonomic and functional analysis of this type of samples, we propose an effective metatranscriptomics strategy to accurately characterize the microbiome in human tissues with a low ratio of microbial to host content. We experimentally generated synthetic samples with well-characterized bacterial and host cell compositions, and mimicking human samples with high and low microbial loads. These synthetic samples were used for optimizing and establishing the workflow in a controlled setting. Our results show that the integration of the taxonomic analysis of optimized Kraken 2/Bracken with the functional analysis of HUMAnN 3 in samples with low microbial content, enables the accurate identification of a large number of microbial species with a low false-positive rate, while improving the detection of microbial functions. The effectiveness of our metatranscriptomics workflow was demonstrated in synthetic samples, simulated datasets, and most importantly, human gastric tissue specimens, thus providing a proof of concept for its applicability on mucosal tissues of the gastrointestinal tract. The use of an accurate and reliable metatranscriptomics approach for human tissues with low microbial content will expand our understanding of the functional activity of the mucosal microbiome, uncovering critical interactions between the microbiome and the host in health and disease.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10913719/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139996082","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
Bile acid profiling as an effective biomarker for staging in pediatric inflammatory bowel disease. 胆汁酸分析作为小儿炎症性肠病分期的有效生物标记物。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-03-04 DOI: 10.1080/19490976.2024.2323231
Wei Chen, Daosheng Wang, Xing Deng, Hong Zhang, Danfeng Dong, Tongxuan Su, Qiuya Lu, Cen Jiang, Qi Ni, Yingchao Cui, Qianli Zhao, Xuefeng Wang, Yuan Xiao, Yibing Peng

Rapid and accurate clinical staging of pediatric patients with inflammatory bowel disease (IBD) is crucial to determine the appropriate therapeutic approach. This study aimed to identify effective, convenient biomarkers for staging IBD in pediatric patients. We recruited cohorts of pediatric patients with varying severities of IBD to compare the features of the intestinal microbiota and metabolites between the active and remitting disease stages. Metabolites with potential for staging were targeted for further assessment in both patients and colitis model mice. The performance of these markers was determined using machine learning and was validated in a separate patient cohort. Pediatric patients with IBD exhibited distinct gut microbiota structures at different stages of disease activity. The enterotypes of patients with remitting and active disease were Bacteroides-dominant and Escherichia-Shigella-dominant, respectively. The bile secretion pathway showed the most significant differences between the two stages. Fecal and serum bile acid (BA) levels were strongly related to disease activity in both children and mice. The ratio of primary BAs to secondary BAs in serum was developed as a novel comprehensive index, showing excellent diagnostic performance in stratifying IBD activity (0.84 area under the receiver operating characteristic curve in the primary cohort; 77% accuracy in the validation cohort). In conclusion, we report profound insights into the interactions between the gut microbiota and metabolites in pediatric IBD. Serum BAs have potential as biomarkers for classifying disease activity, and may facilitate the personalization of treatment for IBD.

对儿科炎症性肠病(IBD)患者进行快速、准确的临床分期对于确定适当的治疗方法至关重要。本研究旨在为儿科患者的 IBD 分期确定有效、便捷的生物标志物。我们招募了不同严重程度的 IBD 儿科患者,比较疾病活动期和缓解期的肠道微生物群和代谢物特征。在患者和结肠炎模型小鼠中,具有分期潜力的代谢物被列为进一步评估的目标。利用机器学习确定了这些标记物的性能,并在一个单独的患者队列中进行了验证。小儿 IBD 患者在疾病活动的不同阶段表现出不同的肠道微生物群结构。缓解期和活动期患者的肠道菌群分别以乳酸杆菌为主和埃希氏-志贺菌为主。胆汁分泌途径在两个阶段之间的差异最为显著。儿童和小鼠的粪便和血清胆汁酸(BA)水平与疾病活动性密切相关。血清中原发性胆汁酸与继发性胆汁酸的比值被开发为一种新的综合指标,在对 IBD 活动性进行分层方面显示出卓越的诊断性能(在原发性队列中,接收者操作特征曲线下面积为 0.84;在验证队列中,准确率为 77%)。总之,我们报告了对小儿 IBD 中肠道微生物群与代谢物之间相互作用的深刻见解。血清 BAs 有可能成为疾病活动性分类的生物标志物,并有助于对 IBD 进行个性化治疗。
{"title":"Bile acid profiling as an effective biomarker for staging in pediatric inflammatory bowel disease.","authors":"Wei Chen, Daosheng Wang, Xing Deng, Hong Zhang, Danfeng Dong, Tongxuan Su, Qiuya Lu, Cen Jiang, Qi Ni, Yingchao Cui, Qianli Zhao, Xuefeng Wang, Yuan Xiao, Yibing Peng","doi":"10.1080/19490976.2024.2323231","DOIUrl":"10.1080/19490976.2024.2323231","url":null,"abstract":"<p><p>Rapid and accurate clinical staging of pediatric patients with inflammatory bowel disease (IBD) is crucial to determine the appropriate therapeutic approach. This study aimed to identify effective, convenient biomarkers for staging IBD in pediatric patients. We recruited cohorts of pediatric patients with varying severities of IBD to compare the features of the intestinal microbiota and metabolites between the active and remitting disease stages. Metabolites with potential for staging were targeted for further assessment in both patients and colitis model mice. The performance of these markers was determined using machine learning and was validated in a separate patient cohort. Pediatric patients with IBD exhibited distinct gut microbiota structures at different stages of disease activity. The enterotypes of patients with remitting and active disease were <i>Bacteroides-dominant</i> and <i>Escherichia-Shigella-dominant</i>, respectively. The bile secretion pathway showed the most significant differences between the two stages. Fecal and serum bile acid (BA) levels were strongly related to disease activity in both children and mice. The ratio of primary BAs to secondary BAs in serum was developed as a novel comprehensive index, showing excellent diagnostic performance in stratifying IBD activity (0.84 area under the receiver operating characteristic curve in the primary cohort; 77% accuracy in the validation cohort). In conclusion, we report profound insights into the interactions between the gut microbiota and metabolites in pediatric IBD. Serum BAs have potential as biomarkers for classifying disease activity, and may facilitate the personalization of treatment for IBD.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10913721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140021579","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
mNFE: microbiome network flow entropy for detecting pre-disease states of type 1 diabetes. mNFE:用于检测 1 型糖尿病病前状态的微生物组网络流熵。
IF 12.2 1区 医学 Q1 GASTROENTEROLOGY & HEPATOLOGY Pub Date : 2024-01-01 Epub Date: 2024-03-21 DOI: 10.1080/19490976.2024.2327349
Rong Gao, Peiluan Li, Yueqiong Ni, Xueqing Peng, Jing Ren, Luonan Chen

In the development of Type 1 diabetes (T1D), there are critical states just before drastic changes, and identifying these pre-disease states may predict T1D or provide crucial early-warning signals. Unlike gene expression data, gut microbiome data can be collected noninvasively from stool samples. Gut microbiome sequencing data contain different levels of phylogenetic information that can be utilized to detect the tipping point or critical state in a reliable manner, thereby providing accurate and effective early-warning signals. However, it is still difficult to detect the critical state of T1D based on gut microbiome data due to generally non-significant differences between healthy and critical states. To address this problem, we proposed a new method - microbiome network flow entropy (mNFE) based on a single sample from each individual - for detecting the critical state before seroconversion and abrupt transitions of T1D at various taxonomic levels. The numerical simulation validated the robustness of mNFE under different noise levels. Furthermore, based on real datasets, mNFE successfully identified the critical states and their dynamic network biomarkers (DNBs) at different taxonomic levels. In addition, we found some high-frequency species, which are closely related to the unique clinical characteristics of autoantibodies at the four levels, and identified some non-differential 'dark species' play important roles during the T1D progression. mNFE can robustly and effectively detect the pre-disease states at various taxonomic levels and identify the corresponding DNBs with only a single sample for each individual. Therefore, our mNFE method provides a new approach not only for T1D pre-disease diagnosis or preventative treatment but also for preventative medicine of other diseases by gut microbiome.

在 1 型糖尿病(T1D)的发展过程中,在急剧变化之前会出现一些关键状态,识别这些疾病前状态可以预测 T1D 或提供重要的预警信号。与基因表达数据不同,肠道微生物组数据可以通过粪便样本无创收集。肠道微生物组测序数据包含不同层次的系统发育信息,可用于可靠地检测临界点或临界状态,从而提供准确有效的预警信号。然而,由于健康状态和临界状态之间的差异一般不显著,因此根据肠道微生物组数据检测 T1D 的临界状态仍然很困难。为解决这一问题,我们提出了一种新方法--基于每个个体单个样本的微生物组网络流熵(mNFE),用于检测不同分类水平的血清转换前临界状态和 T1D 的突然转变。数值模拟验证了 mNFE 在不同噪声水平下的稳健性。此外,基于真实数据集,mNFE 成功识别了不同分类水平的临界状态及其动态网络生物标志物(DNB)。此外,我们还发现了一些高频物种,它们与四个层次上自身抗体的独特临床特征密切相关,并确定了一些在 T1D 进展过程中发挥重要作用的非差异性 "暗物种"。因此,我们的 mNFE 方法不仅为 T1D 病前诊断或预防治疗提供了一种新方法,也为通过肠道微生物组预防其他疾病提供了一种新方法。
{"title":"mNFE: microbiome network flow entropy for detecting pre-disease states of type 1 diabetes.","authors":"Rong Gao, Peiluan Li, Yueqiong Ni, Xueqing Peng, Jing Ren, Luonan Chen","doi":"10.1080/19490976.2024.2327349","DOIUrl":"10.1080/19490976.2024.2327349","url":null,"abstract":"<p><p>In the development of Type 1 diabetes (T1D), there are critical states just before drastic changes, and identifying these pre-disease states may predict T1D or provide crucial early-warning signals. Unlike gene expression data, gut microbiome data can be collected noninvasively from stool samples. Gut microbiome sequencing data contain different levels of phylogenetic information that can be utilized to detect the tipping point or critical state in a reliable manner, thereby providing accurate and effective early-warning signals. However, it is still difficult to detect the critical state of T1D based on gut microbiome data due to generally non-significant differences between healthy and critical states. To address this problem, we proposed a new method - microbiome network flow entropy (mNFE) based on a single sample from each individual - for detecting the critical state before seroconversion and abrupt transitions of T1D at various taxonomic levels. The numerical simulation validated the robustness of mNFE under different noise levels. Furthermore, based on real datasets, mNFE successfully identified the critical states and their dynamic network biomarkers (DNBs) at different taxonomic levels. In addition, we found some high-frequency species, which are closely related to the unique clinical characteristics of autoantibodies at the four levels, and identified some non-differential 'dark species' play important roles during the T1D progression. mNFE can robustly and effectively detect the pre-disease states at various taxonomic levels and identify the corresponding DNBs with only a single sample for each individual. Therefore, our mNFE method provides a new approach not only for T1D pre-disease diagnosis or preventative treatment but also for preventative medicine of other diseases by gut microbiome.</p>","PeriodicalId":12909,"journal":{"name":"Gut Microbes","volume":null,"pages":null},"PeriodicalIF":12.2,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10962612/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140184257","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:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1