1
|
Su F, Su M, Wei W, Wu J, Chen L, Sun X, Liu M, Sun S, Mao R, Bourgonje AR, Hu S. Integrating multi-omics data to reveal the host-microbiota interactome in inflammatory bowel disease. Gut Microbes 2025; 17:2476570. [PMID: 40063366 PMCID: PMC11901428 DOI: 10.1080/19490976.2025.2476570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Numerous studies have accelerated the knowledge expansion on the role of gut microbiota in inflammatory bowel disease (IBD). However, the precise mechanisms behind host-microbe cross-talk remain largely undefined, due to the complexity of the human intestinal ecosystem and multiple external factors. In this review, we introduce the interactome concept to systematically summarize how intestinal dysbiosis is involved in IBD pathogenesis in terms of microbial composition, functionality, genomic structure, transcriptional activity, and downstream proteins and metabolites. Meanwhile, this review also aims to present an updated overview of the relevant mechanisms, high-throughput multi-omics methodologies, different types of multi-omics cohort resources, and computational methods used to understand host-microbiota interactions in the context of IBD. Finally, we discuss the challenges pertaining to the integration of multi-omics data in order to reveal host-microbiota cross-talk and offer insights into relevant future research directions.
Collapse
Affiliation(s)
- Fengyuan Su
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meng Su
- The First Clinical Medical School, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenting Wei
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiayun Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Leyan Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiqiao Sun
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Moyan Liu
- Amsterdam UMC location Academic Medical Center, Department of Experimental Vascular Medicine, Amsterdam, The Netherlands
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
2
|
Jori C, Ahmad A, Kumar A, Kumar B, Ali A, Ali N, Tabassum H, Khan R. Bioactive chitosan-BSA Maillard-derived chrysin-loaded nanoparticles: A gastroprotective, biomucoadhesive approach for enhanced oral therapy in ulcerative colitis. Carbohydr Polym 2025; 359:123537. [PMID: 40306769 DOI: 10.1016/j.carbpol.2025.123537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
The current limitations of oral nanomedicines such as aminosalicylates, immunosuppressants, corticosteroids, and antibiotics include the toxic byproducts from nanocarrier synthesis, poor targeting and retention within the inflamed colon, delayed release at inflammation sites, susceptibility to gastric degradation, reduced efficacy under hypoxic conditions, MUC2 homeostasis disruption, and insufficiently addressing the disease's root causes. This research presents an innovative approach of using non-toxic, biodegradable, and biocompatible Maillard reaction-based nanoparticles (MPs) for targeted oral drug delivery in IBD therapy. Through the development of mucoadhevise chitosan-bovine serum albumin Maillard nanoparticles shielded with biocompatible, non-toxic, non-immunogenic, gastroprotective pectin (P@CMPs) encapsulating with chrysin, a flavonoid with anti-inflammatory and hyperoxia properties whose bioavailability is negatively affected by gastric degradation. P@CMPs had a spherical, uniform 300 nm hydrodynamic diameter, confirmed by TEM and FESEM. Chrysin encapsulation efficiency and loading capacity were ∼96 % and 16 %, respectively, demonstrating effective nanoparticle formulation The P@CMPs is designed to withstand the gastrointestinal environment, ensuring targeted delivery and prolonged retention in inflamed colonic regions. In a dextran sodium sulfate-induced colitis mouse model, P@CMPs markedly mitigated inflammation, suppressed proinflammatory cytokine levels, and augmented the expression of MUC2, a crucial factor for maintaining the integrity of the gut barrier. By employing non-toxic, biocompatible and biodegradable materials, our P@CMPs approach offers a promising avenue for advancing IBD treatment, addressing various challenges and precise oral delivery within the gastrointestinal system.
Collapse
Affiliation(s)
- Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110029, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| |
Collapse
|
3
|
Schoonakker MP, van Peet PG, van den Burg EL, Numans ME, Ducarmon QR, Pijl H, Wiese M. Impact of dietary carbohydrate, fat or protein restriction on the human gut microbiome: a systematic review. Nutr Res Rev 2025; 38:238-255. [PMID: 38602133 DOI: 10.1017/s0954422424000131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Restriction of dietary carbohydrates, fat and/or protein is often used to reduce body weight and/or treat (metabolic) diseases. Since diet is a key modulator of the human gut microbiome, which plays an important role in health and disease, this review aims to provide an overview of current knowledge of the effects of macronutrient-restricted diets on gut microbial composition and metabolites. A structured search strategy was performed in several databases. After screening for inclusion and exclusion criteria, thirty-six articles could be included. Data are included in the results only when supported by at least three independent studies to enhance the reliability of our conclusions. Low-carbohydrate (<30 energy%) diets tended to induce a decrease in the relative abundance of several health-promoting bacteria, including Bifidobacterium, as well as a reduction in short-chain fatty acid (SCFA) levels in faeces. In contrast, low-fat diets (<30 energy%) increased alpha diversity, faecal SCFA levels and abundance of some beneficial bacteria, including Faecalibacterium prausnitzii. There were insufficient data to draw conclusions concerning the effects of low-protein (<10 energy%) diets on gut microbiota. Although the data of included studies unveil possible benefits of low-fat and potential drawbacks of low-carbohydrate diets for human gut microbiota, the diversity in study designs made it difficult to draw firm conclusions. Using a more uniform methodology in design, sample processing and sharing raw sequence data could foster our understanding of the effects of macronutrient restriction on gut microbiota composition and metabolic dynamics relevant to health. This systematic review was registered at https://www.crd.york.ac.uk/prospero as CRD42020156929.
Collapse
Affiliation(s)
- Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Maria Wiese
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
4
|
Tan C, Xiang Z, Wang S, He H, Li X, Xu M, Guo X, Pu Y, Zhen J, Dong W. Diosmin alleviates colitis by inhibiting PANoptosis of intestinal epithelial cells and regulating gut microbiota and metabolites. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156671. [PMID: 40138774 DOI: 10.1016/j.phymed.2025.156671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Inflammatory bowel disease (IBD), particularly ulcerative colitis (UC), is a chronic inflammatory disorder characterized by an unclear etiology, often linked to gut microbiota dysbiosis and immune dysregulation. Existing UC therapies are constrained by suboptimal efficacy and adverse effects, underscoring the necessity for novel therapeutic strategies. Diosmin (DIO), a naturally occurring flavonoid, has demonstrated anti-inflammatory and antioxidant potential, yet its precise mechanisms and therapeutic role in colitis remain poorly understood. PURPOSE This study aimed to investigate the therapeutic efficacy and mechanistic underpinnings of DIO in dextran sulfate sodium (DSS)-induced colitis, with a focus on its effects on intestinal epithelial cell PANoptosis, gut microbiota composition, fecal metabolites, and an in vitro inflammatory model using human colonic epithelial cells. STUDY DESIGN A controlled experimental design was employed, utilizing a DSS-induced murine colitis model and an LPS-induced inflammatory model in human colonic epithelial cells (NCM460). Mice were allocated into four groups: normal control, DSS-induced colitis, low-dose DIO (DIO-L, 100 mg/kg), and high-dose DIO (DIO-H, 200 mg/kg). In vitro experiments involved treating NCM460 cells with varying DIO concentrations post-LPS stimulation to assess its impact on inflammation and epithelial barrier integrity. METHODS Mice were administered DIO orally at 100 mg/kg or 200 mg/kg daily. Therapeutic outcomes were evaluated through body weight monitoring, Disease Activity Index (DAI) scoring, and histopathological examination. Gut microbiota composition was analyzed via 16S rRNA sequencing, while untargeted metabolomics was employed to profile fecal metabolites. Data integration was performed using O2PLS and WGCNA to identify microbiota-metabolite correlations. In vitro, immunofluorescence staining and Western blotting were utilized to evaluate the expression of tight junction proteins (ZO-1, E-cadherin, and Occludin). RESULTS DIO administration significantly ameliorated colitis symptoms in mice, as evidenced by attenuated weight loss, reduced DAI scores, and preserved colon length. Histopathological analysis confirmed diminished inflammation and tissue damage in DIO-treated groups. Mechanistically, DIO suppressed the expression of PANoptosis-associated genes and proteins, including ZBP1 and Caspase-1, while maintaining epithelial barrier integrity in vitro. Furthermore, DIO modulated gut microbiota composition, promoting beneficial taxa such as Ruminococcus and reducing pathogenic Proteobacteria. Metabolomic profiling revealed alterations in key metabolic pathways, including flavonoid and steroid hormone biosynthesis, which correlated with microbiota changes. CONCLUSION DIO effectively mitigates DSS-induced colitis by inhibiting intestinal epithelial cell PANoptosis, preserving barrier function, and modulating gut microbiota and metabolite profiles. These findings highlight DIO's potential as a therapeutic agent for IBD and warrant further exploration of its clinical applications.
Collapse
Affiliation(s)
- Cheng Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zixuan Xiang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shuo Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiangyun Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Miao Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xingzhou Guo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Pu
- Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Junhai Zhen
- Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
5
|
Rytter H, Sturgeon H, Chassaing B. Diet-pathobiont interplay in health and inflammatory bowel disease. Trends Microbiol 2025:S0966-842X(25)00112-X. [PMID: 40379577 DOI: 10.1016/j.tim.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/19/2025]
Abstract
The intestinal microbiota plays a crucial role in maintaining host health by participating in various beneficial functions. However, under certain conditions, it can contribute to the development of inflammatory bowel disease (IBD) and other chronic inflammatory conditions. Importantly, not all commensal microbiota members are drivers of inflammation. A specific subset of commensal bacteria, known as pathobionts, can exhibit pathogenic potential under specific circumstances. Their inflammatory potential is modulated by several factors, including the host's genetic background and the surrounding microbiota. Furthermore, diet has emerged as a critical factor influencing the gut microbiota, with some studies highlighting its role in modulating pathobionts. This review will delve into the role played by pathobionts in chronic intestinal inflammation, in both mouse models as well as in humans, with a focus on the interplay between dietary factors and pathobiont members of the intestinal microbiota. Understanding the complex relationships between diet, pathobionts, and chronic inflammation could pave the way for diet-based therapeutic strategies aimed at managing chronic inflammatory conditions.
Collapse
Affiliation(s)
- Héloïse Rytter
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
| | - Hannah Sturgeon
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France; CHRU Nancy, IHU Infiny, F54000 Nancy, France.
| |
Collapse
|
6
|
Luo Y, Pedersen CET, Eliasen AU, Brustad N, Chen L, Wang N, Jiang J, Trivedi U, Li X, Sørensen SJ, Chawes BL, Stokholm J, Thorsen J, Bønnelykke K. Maternal and child FUT2 secretor status affect gastroenteritis risk and gut microbiota composition in early life. Clin Microbiol Infect 2025:S1198-743X(25)00216-2. [PMID: 40339805 DOI: 10.1016/j.cmi.2025.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVE To investigate associations between maternal and child secretor status and early-life gastroenteritis risk, considering the roles of gut microbiota, breastfeeding, and daycare attendance. METHODS In the COPSAC2010 cohort (n=700), parents recorded gastroenteritis episodes during the first three years of life. Secretor status, rs601338 in the FUT2 gene, was genotyped in both parents and children. The association between secretor status and gastroenteritis was assessed using quasi-Poisson regression. Fecal samples were collected at 1 week, 1 month, 1 year after birth. The interaction between secretor status, breastfeeding and daycare attendance were analyzed through Cox regression. RESULTS Maternal secretor status increased first-year gastroenteritis risk (incidence rate ratio [IRR]=1.48, 95% confidence interval [CI]:1.05-2.16, p=0.033); child status increased second-year risk (IRR=1.56, 95%CI:1.11-2.27, p=0.015), especially after daycare attendance (interaction p=0.006). Maternal status associated with microbiota differences at 1 week (weighted UniFrac F=2.4, R2=0.47%, p=0.048) and 1 month (F=3.3, R2=0.62%, p=0.026); child status at 1 year (F=2.5, R2=0.45%, p=0.027). Secretor children showed lower Bacteroides vulgatus (Median [interquartile range (IQR)]:1.00% [0.04-12.92] vs. 5.00% [0.09-24.80], p=0.023) but higher Escherichia/Shigella (1.35% [IQR:0.28-7.42] vs. 0.56% [IQR:0.13-2.62], p=0.002). B. vulgatus mediated 14% of child status effects (average causal mediation effect [ACME] IRR=0.95, 95% CI: 0.89-0.99, p=0.014). CONCLUSION Maternal and child FUT2 status demonstrates age-specific impacts on gastroenteritis and microbiota in early life, providing new insights into gastrointestinal health genetics and host-microbiome dynamics.
Collapse
Affiliation(s)
- Yang Luo
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Casper-Emil T Pedersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Anders Ulrik Eliasen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Nicklas Brustad
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Liang Chen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Ni Wang
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Jie Jiang
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Urvish Trivedi
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Xuanji Li
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Søren Johannes Sørensen
- Department of Biology, Section of Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bo L Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, 4200 Slagelse, Denmark; Department of Food Science, University of Copenhagen, 1958 Frederiksberg, Copenhagen, Denmark
| | - Jonathan Thorsen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark.
| |
Collapse
|
7
|
Hitch TCA, Masson JM, Pauvert C, Bosch J, Nüchtern S, Treichel NS, Baloh M, Razavi S, Afrizal A, Kousetzi N, Aguirre AM, Wylensek D, Coates AC, Jennings SAV, Panyot A, Viehof A, Schmitz MA, Stuhrmann M, Deis EC, Bisdorf K, Chiotelli MD, Lissin A, Schober I, Witte J, Cramer T, Riedel T, Wende M, Winter KA, Amend L, Riva A, Trinh S, Mitchell L, Hartman J, Berry D, Seitz J, Bossert LC, Grognot M, Allers T, Strowig T, Pester M, Abt B, Reimer LC, Overmann J, Clavel T. HiBC: a publicly available collection of bacterial strains isolated from the human gut. Nat Commun 2025; 16:4203. [PMID: 40328737 PMCID: PMC12056005 DOI: 10.1038/s41467-025-59229-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Numerous bacteria in the human gut microbiome remain unknown and/or have yet to be cultured. While collections of human gut bacteria have been published, few strains are accessible to the scientific community. We have therefore created a publicly available collection of bacterial strains isolated from the human gut. The Human intestinal Bacteria Collection (HiBC) ( https://www.hibc.rwth-aachen.de ) contains 340 strains representing 198 species within 29 families and 7 phyla, of which 29 previously unknown species are taxonomically described and named. These included two butyrate-producing species of Faecalibacterium and new dominant species associated with health and inflammatory bowel disease, Ruminococcoides intestinale and Blautia intestinihominis, respectively. Plasmids were prolific within the HiBC isolates, with almost half (46%) of strains containing plasmids, with a maximum of six within a strain. This included a broadly occurring plasmid (pBAC) that exists in three diverse forms across Bacteroidales species. Megaplasmids were identified within two strains, the pMMCAT megaplasmid is globally present within multiple Bacteroidales species. This collection of easily searchable and publicly available gut bacterial isolates will facilitate functional studies of the gut microbiome.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Johannes M Masson
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Johanna Bosch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Selina Nüchtern
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Nicole S Treichel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Marko Baloh
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Soheila Razavi
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Afrizal Afrizal
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Ntana Kousetzi
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Andrea M Aguirre
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Amy C Coates
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Susan A V Jennings
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Atscharah Panyot
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Alina Viehof
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Matthias A Schmitz
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Maximilian Stuhrmann
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Evelyn C Deis
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Kevin Bisdorf
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Maria D Chiotelli
- Biophysics of Host-Microbe Interactions Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Artur Lissin
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Isabel Schober
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Julius Witte
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Thorsten Cramer
- Molecular Tumor Biology Research Group, Department of General, Visceral, Children and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | - Thomas Riedel
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katrin A Winter
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alessandra Riva
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Stefanie Trinh
- Institute of Neuroanatomy, University Hospital of RWTH Aachen, Aachen, Germany
| | - Laura Mitchell
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - David Berry
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | - Jochen Seitz
- Clinic for Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, LVR-University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Marianne Grognot
- Biophysics of Host-Microbe Interactions Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Thorsten Allers
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Till Strowig
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Michael Pester
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Technical University Braunschweig, Braunschweig, Germany
| | - Birte Abt
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Lorenz C Reimer
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Jörg Overmann
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
- Technical University Braunschweig, Braunschweig, Germany
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
8
|
Liu W, Chen X, Yang C, Lin Z, Huang X, Zhang Z, Liu J. Preventive effects of xanthohumol in APP/PS1 mice based on multi-omics atlas. Brain Res Bull 2025; 224:111316. [PMID: 40132750 DOI: 10.1016/j.brainresbull.2025.111316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Alzheimer's disease (AD) is a complex disease with unknown etiology and pathogenesis. We described a combined analysis of murine proteomics and microbiomics to find potential therapeutic targets of different doses of xanthohumol (Xn), with the goal of providing a biological basis for the treatment of early AD. Xn improved the spatial learning and memory ability of APP/PS1 mice; this was associated with an increased number of newborn neurons in the subgranular zone (SGZ) and dentate gyrus (DG) and a decreased inflammatory response. 108 proteins were significantly changed after 0.5 mg/kg Xn treatment while only 72 proteins changed by 5 mg/kg Xn. Eight significant microbiota were modulated by different doses of Xn at line discriminant analysis (LDA) score 3.0, but only three of which were regulated by 0.5 mg/kg Xn at LDA score 4.0. In addition, Xn treatment could significantly regulate the pathways of neurodegeneration- multiple diseases in the hippocampus and the penicillin and cephalosporin biosynthesis and atrazine degradation pathways in the gut. Interestingly, Nefl protein validated by correlation analysis was found in the common signaling pathway. 0.5 mg/kg Xn was able to reverse the correlation between hippocampal proteins and gut microbiota. Xn treatment significantly improved cognitive function in AD transgenic mice. Different doses of Xn caused significant differences in protein expression and flora composition and abundance, suggesting that the doses of Xn should be selected with caution, and low dose may be better in the prevention of AD.
Collapse
Affiliation(s)
- Wei Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.
| | - Xiao Chen
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Chen Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zequn Lin
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xinfeng Huang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zhen Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jianjun Liu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| |
Collapse
|
9
|
Turpin W, Lee SH, Croitoru K. Gut Microbiome Signature in Predisease Phase of Inflammatory Bowel Disease: Prediction to Pathogenesis to Prevention. Gastroenterology 2025; 168:902-913. [PMID: 39914464 DOI: 10.1053/j.gastro.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 03/23/2025]
Abstract
Advances in understanding the pathogenesis of inflammatory bowel disease (IBD) point toward a key role of the gut microbiome. We review the data describing the changes in the gut microbiome from IBD case-control studies and compare these findings with emerging data from studies of the preclinical phase of IBD. What is apparent is that assessing changes in the composition and function of the gut microbiome during the preclinical phase helps address confounding factors, such as disease activity and drug therapy, which can directly influence the gut microbiome. Understanding these changes in the predisease phase provides a means of predicting IBD in high-risk populations and offers insights into possible mechanisms involved in disease pathogenesis. Finally, we discuss strategies to use this information to design interventions aimed at modulating the microbiome as a means of preventing or delaying the onset of IBD.
Collapse
Affiliation(s)
- Williams Turpin
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Ahmadi A, Shokoohizadeh L, Sheikhesmaili F, Mirzaei MK, Mohammadi A, Nikkhoo B, Khodaei H, Alikhani MY, Yousefimashouf R. Gut microbiomes and treatment-resistant ulcerative colitis: a case-control study using qPCR. BMC Microbiol 2025; 25:254. [PMID: 40295906 PMCID: PMC12036124 DOI: 10.1186/s12866-025-03963-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The gut microbiome has been identified as a pivotal factor in ulcerative colitis (UC), given its role as the main reservoir of microbes in the body. This community of microorganisms, present in variable concentrations in the digestive tract, makes a wide range of beneficial roles for the host. However, the role of the gut microbiome in patients with refractory UC is still significant, so this study aimed to further investigate the role of these bacteria in patients with refractory UC. METHODS This case-control study was conducted on stool samples from four distinct groups: the first group comprised new patients diagnosed with ulcerative colitis (all of them had responded to treatment after follow-up) (N = 24); the second group consisted of patients with treatment-resistant ulcerative colitis (N = 23); the third group included first-degree relatives of group 1 patients (N = 24); and the fourth group consisted of first-degree relatives of group 2 patients (N = 23). The research tools employed in this study included a questionnaire, quantitative real-time PCR (qPCR) test, and culture on stool samples. RESULT The mean age of patients in groups 1 and 2 was 45.88 ± 18.51 and 41.30 ± 13.01 years, while the mean age of controls in groups 3 and 4 was 37.29 ± 9.62 and 40.96 ± 13.01 years, respectively. Stool culture results for pathogenic bacteria were negative in all four groups. The of history of consuming dairy products containing probiotics was highest in Group 1, with 22 (91.67%) subjects, while the lowest was observed in Group 3, with 16 (66.67%). The highest history of self-administered antibiotic use was observed in Group 2, with 13 cases (56.52%), while the lowest was noted in Group 3, with 4 cases (16.67%). The findings indicated a statistically significant relationship (P < 0.05) between Groups 2 and 4 with respect to the E. coli and Bifidobacterium ssp. microbial population. Additionally, a significant relationship was identified between the Lactobacillus ssp., Bifidobacterium ssp., and Bacteroides ssp. microbial community between groups 1 and 2 (P < 0.05). CONCLUSION The findings of this study demonstrated that several intestinal microbiomes have a substantial impact on the management of ulcerative colitis. The results of this study suggest that by comparing the gut microbiome of treatment-resistant and individuals newly diagnosed with ulcerative colitis, we can gain a better understanding of microbiome differences that may influence treatment outcomes. The results of this study may also lead to the identification of new therapeutic strategies that are based on regulating the gut microbiome. These strategies could include the use of fecal microbiome transplantation (FMT), probiotics, prebiotics, or specific bacteria-based therapies.
Collapse
Affiliation(s)
- Amjad Ahmadi
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leili Shokoohizadeh
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farshad Sheikhesmaili
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Munich, German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Prevention of Microbial Infectious Diseases, Central Institute of Disease Prevention, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Hakim Khodaei
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
11
|
Stentz R, Jones E, Gul L, Latousakis D, Parker A, Brion A, Goldson AJ, Gotts K, Carding SR. Proteomics of Bacterial and Mouse Extracellular Vesicles Released in the Gastrointestinal Tracts of Nutrient-Stressed Animals Reveals an Interplay Between Microbial Serine Proteases and Mammalian Serine Protease Inhibitors. Int J Mol Sci 2025; 26:4080. [PMID: 40362319 PMCID: PMC12071298 DOI: 10.3390/ijms26094080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Bacterial extracellular vesicles (BEVs) produced by members of the intestinal microbiota can not only contribute to digestion but also mediate microbe-host cell communication via the transfer of functional biomolecules to mammalian host cells. An unresolved question is which host factors and conditions influence BEV cargo and how they impact host cell function. To address this question, we analysed and compared the proteomes of BEVs released by the major human gastrointestinal tract (GIT) symbiont Bacteroides thetaiotaomicron (Bt) in vivo in fed versus fasted animals using nano-liquid chromatography with tandem mass spectrometry (LC-MSMS). Among the proteins whose abundance was negatively affected by fasting, nine of ten proteins of the serine protease family, including the regulatory protein dipeptidyl peptidase-4 (DPP-4), were significantly decreased in BEVs produced in the GITs of fasted animals. Strikingly, in extracellular vesicles produced by the intestinal epithelia of the same fasted mice, the proteins with the most increased abundance were serine protease inhibitors (serpins). Together, these findings suggest a dynamic interaction between GI bacteria and the host. Additionally, they indicate a regulatory role for the host in determining the balance between bacterial serine proteases and host serpins exported in bacterial and host extracellular vesicles.
Collapse
Affiliation(s)
- Régis Stentz
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
| | - Emily Jones
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
| | - Lejla Gul
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Dimitrios Latousakis
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
| | - Aimee Parker
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
| | - Arlaine Brion
- Core Science Resources, Quadram Institute Bioscience, Norwich NR4 7UQ, UK (A.J.G.); (K.G.)
| | - Andrew J. Goldson
- Core Science Resources, Quadram Institute Bioscience, Norwich NR4 7UQ, UK (A.J.G.); (K.G.)
| | - Kathryn Gotts
- Core Science Resources, Quadram Institute Bioscience, Norwich NR4 7UQ, UK (A.J.G.); (K.G.)
| | - Simon R. Carding
- Food, Microbiome and Health Research Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK; (E.J.); (L.G.); (D.L.); (A.P.); (S.R.C.)
- Norwich Medical School, University East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
12
|
Wu X, Li Y, Li P, Lu G, Wu J, Wang Z, Wen Q, Cui B, Wang J, Zhang F. Structural Variations in Ulcerative Colitis-associated Escherichia coli Reduce Fructose Utilization and Aggravate Inflammation Under High-Fructose Diet. Gastroenterology 2025:S0016-5085(25)00635-3. [PMID: 40250773 DOI: 10.1053/j.gastro.2025.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 02/16/2025] [Accepted: 03/09/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND AND AIMS Structural variations (SVs) have significant effects on microbial phenotypes. The underlying mechanism of functional changes caused by gut microbial SVs in the development of ulcerative colitis (UC) need further investigation. METHODS We performed long-read (Oxford Nanopore Technology-based) and short-read (Illumina-based) metagenomic sequencing on stool samples from 93 patients with UC and 100 healthy controls (HCs) and analyzed microbial SVs. A total of 648 Escherichia coli strains from fecal samples of patients with UC (UC-strains) and HCs (HC-strains) were isolated. SV-associated scrK gene deletion was verified via whole-genome sequencing or targeted polymerase chain reaction. Then, representative UC-strains, HC-strains, and scrK-knockout E coli were used for the in vitro and in vivo experiments to investigate the effects of specific SVs in E coli on fructose utilization ability and colitis. RESULTS E coli in UC with the highest fold change had SV-affected functional differences on fructose metabolism to that of HCs. The fructose utilization gene deletion was common in UC-strains, ostensibly reducing fructose utilization in vitro and leading to fructose-dependent aggravation of colitis in murine models. UC-strains and HC-strains induced comparable colitis under low fructose. However, high fructose exacerbated colitis severity exclusively in UC-strain-colonized mice, with elevated intestinal fructose residues, significant microbiome/metabolome changes, increased inflammation, and gut barrier disruption. These changes were mechanistically dependent on the deletion of the fructose utilization gene scrK. CONCLUSIONS SV-caused difference in fructose utilization and proinflammatory properties in E coli from patients with UC influence the development of UC, emphasizing the importance of fine-scale metagenomic studies in disease.
Collapse
Affiliation(s)
- Xia Wu
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuejuan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Pan Li
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gaochen Lu
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianyu Wu
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheyu Wang
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Quan Wen
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bota Cui
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Faming Zhang
- Department of Microbiota Medicine & Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Gilbert JA, Azad MB, Bäckhed F, Blaser MJ, Byndloss M, Chiu CY, Chu H, Dugas LR, Elinav E, Gibbons SM, Gilbert KE, Henn MR, Ishaq SL, Ley RE, Lynch SV, Segal E, Spector TD, Strandwitz P, Suez J, Tropini C, Whiteson K, Knight R. Clinical translation of microbiome research. Nat Med 2025; 31:1099-1113. [PMID: 40217076 DOI: 10.1038/s41591-025-03615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
The landscape of clinical microbiome research has dramatically evolved over the past decade. By leveraging in vivo and in vitro experimentation, multiomic approaches and computational biology, we have uncovered mechanisms of action and microbial metrics of association and identified effective ways to modify the microbiome in many diseases and treatment modalities. This Review explores recent advances in the clinical application of microbiome research over the past 5 years, while acknowledging existing barriers and highlighting opportunities. We focus on the translation of microbiome research into clinical practice, spearheaded by Food and Drug Administration (FDA)-approved microbiome therapies for recurrent Clostridioides difficile infections and the emerging fields of microbiome-based diagnostics and therapeutics. We highlight key examples of studies demonstrating how microbiome mechanisms, metrics and modifiers can advance clinical practice. We also discuss forward-looking perspectives on key challenges and opportunities toward integrating microbiome data into routine clinical practice, precision medicine and personalized healthcare and nutrition.
Collapse
Affiliation(s)
- Jack A Gilbert
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin J Blaser
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Mariana Byndloss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Fransisco, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Fransisco, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines, La Jolla, CA, USA
| | - Lara R Dugas
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome and Cancer Division, DKFZ, Heidelberg, Germany
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Katharine E Gilbert
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, ME, USA
- Microbes and Social Equity working group, Orono, ME, USA
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- ZOE Ltd, London, UK
| | | | - Jotham Suez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carolina Tropini
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
14
|
Wei X, Tang D. Effect of Bacteroides on Crohn's disease. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:393-402. [PMID: 39586813 DOI: 10.1055/a-2435-2659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Crohn's disease (CD), also known as cicatrizing enteritis, is an inflammatory bowel disease that occurs in the distal ileum and right colon of unknown cause and is also called inflammatory bowel disease (IBD) with ulcerative colitis (UC). In recent years, intestinal biota have been confirmed to play a significant role in various gastrointestinal diseases. Studies have found that intestinal microbiota disorders are closely associated with the onset and progression of Crohn's disease. Bacteroidetes, the second largest microbiota in the intestine, are crucial for equilibrium in the microbiota and intestinal environment. Certain Bacteroides can induce the development of Crohn's disease and aggravate intestinal inflammation directly or through their metabolites. Conversely, certain Bacteroides can reduce intestinal inflammation and symptoms of Crohn's disease. This article reviews the effect of several intestinal Bacteroides in the onset and progression of Crohn's disease and their impact on its treatment.
Collapse
Affiliation(s)
- Xuanyu Wei
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Nanjing University, Yangzhou, China
| |
Collapse
|
15
|
Jiang Y, Aton M, Zhu Q, Lu YY. Modeling microbiome-trait associations with taxonomy-adaptive neural networks. MICROBIOME 2025; 13:87. [PMID: 40158141 PMCID: PMC11954268 DOI: 10.1186/s40168-025-02080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
The human microbiome, a complex ecosystem of microorganisms inhabiting the body, plays a critical role in human health. Investigating its association with host traits is essential for understanding its impact on various diseases. Although shotgun metagenomic sequencing technologies have produced vast amounts of microbiome data, analyzing such data is highly challenging due to its sparsity, noisiness, and high feature dimensionality. Here, we develop MIOSTONE, an accurate and interpretable neural network model for microbiome-disease association that simulates a real taxonomy by encoding the relationships among microbial features. The taxonomy-encoding architecture provides a natural bridge from variations in microbial taxa abundance to variations in traits, encompassing increasingly coarse scales from species to domains. MIOSTONE has the ability to determine whether taxa within the corresponding taxonomic group provide a better explanation in a data-driven manner. MIOSTONE serves as an effective predictive model, as it not only accurately predicts microbiome-trait associations across extensive simulated and real datasets but also offers interpretability for scientific discovery. Both attributes are crucial for facilitating in silico investigations into the biological mechanisms underlying such associations among microbial taxa. Video Abstract.
Collapse
Affiliation(s)
- Yifan Jiang
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Matthew Aton
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| | - Yang Young Lu
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada.
| |
Collapse
|
16
|
Chen S, Yi M, Yi X, Zhou Y, Song H, Zeng M. Unveiling the fungal frontier: mycological insights into inflammatory bowel disease. Front Immunol 2025; 16:1551289. [PMID: 40207229 PMCID: PMC11979276 DOI: 10.3389/fimmu.2025.1551289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent gastrointestinal disease that seriously affects the quality of life of patients around the world. It is characterized by recurrent abdominal pain, diarrhea, and mucous bloody stools. There is an urgent need for more accurate diagnosis and effective treatment of IBD. Accumulated evidence suggests that gut microbiota plays an important role in the occurrence and development of gut inflammation. However, most studies on the role of gut microbiota in IBD have focused on bacteria, while fungal microorganisms have been neglected. Fungal dysbiosis can activate the host protective immune pathway related to the integrity of the epithelial barrier and release a variety of pro-inflammatory cytokines to trigger the inflammatory response. Dectin-1, CARD9, and IL-17 signaling pathways may be immune drivers of fungal dysbacteriosis in the development of IBD. In addition, fungal-bacterial interactions and fungal-derived metabolites also play an important role. Based on this information, we explored new strategies for IBD treatment targeting the intestinal fungal group and its metabolites, such as fungal probiotics, antifungal drugs, diet therapy, and fecal microbiota transplantation (FMT). This review aims to summarize the fungal dysbiosis and pathogenesis of IBD, and provide new insights and directions for further research in this emerging field.
Collapse
Affiliation(s)
- Silan Chen
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Meijing Yi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinying Yi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuxuan Zhou
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Houpan Song
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Meiyan Zeng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
17
|
Yang SY, Han SM, Lee JY, Kim KS, Lee JE, Lee DW. Advancing Gut Microbiome Research: The Shift from Metagenomics to Multi-Omics and Future Perspectives. J Microbiol Biotechnol 2025; 35:e2412001. [PMID: 40223273 PMCID: PMC12010094 DOI: 10.4014/jmb.2412.12001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
The gut microbiome, a dynamic and integral component of human health, has co-evolved with its host, playing essential roles in metabolism, immunity, and disease prevention. Traditional microbiome studies, primarily focused on microbial composition, have provided limited insights into the functional and mechanistic interactions between microbiota and their host. The advent of multi-omics technologies has transformed microbiome research by integrating genomics, transcriptomics, proteomics, and metabolomics, offering a comprehensive, systems-level understanding of microbial ecology and host-microbiome interactions. These advances have propelled innovations in personalized medicine, enabling more precise diagnostics and targeted therapeutic strategies. This review highlights recent breakthroughs in microbiome research, demonstrating how these approaches have elucidated microbial functions and their implications for health and disease. Additionally, it underscores the necessity of standardizing multi-omics methodologies, conducting large-scale cohort studies, and developing novel platforms for mechanistic studies, which are critical steps toward translating microbiome research into clinical applications and advancing precision medicine.
Collapse
Affiliation(s)
- So-Yeon Yang
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Min Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji-Young Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Eun Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Liu Y, Li X, Chen Y, Yao Q, Zhou J, Wang X, Meng Q, Ji J, Yu Z, Chen X. Fecal microbiota transplantation: application scenarios, efficacy prediction, and factors impacting donor-recipient interplay. Front Microbiol 2025; 16:1556827. [PMID: 40201444 PMCID: PMC11975908 DOI: 10.3389/fmicb.2025.1556827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Fecal microbiota transplantation (FMT) represents a therapeutic approach that directly regulates the gut microbiota of recipients, normalizes its composition and reaping therapeutic rewards. Currently, in addition to its general application in treating Clostridium difficile (C. difficile) infection (CDI), FMT treatment has also been extended to the fields of other gastrointestinal diseases, infections, gut-liver or gut-brain axis disorders, metabolic diseases and cancer, etc. Prior to FMT, rigorous donor screening is essential to reduce the occurrence of adverse events. In addition, it is imperative to evaluate whether the recipient can safely and effectively undergo FMT treatment. However, the efficacy of FMT is influenced by the complex interactions between the gut microbiota of donor and recipient, the degree of donor microbiota engraftment is not necessarily positively related with the success rate of FMT. Furthermore, an increasing number of novel factors affecting FMT outcomes are being identified in recent clinical trials and animal experiments, broadening our understanding of FMT treatment. This article provides a comprehensive review of the application scenarios of FMT, the factors influencing the safety and efficacy of FMT from the aspects of both the donors and the recipients, and summarizes how these emerging novel regulatory factors can be combined to predict the clinical outcomes of patients undergoing FMT.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinru Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuchao Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinjie Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxuan Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Jiang Z, Li P, Qiu K, Liao Y, Chen X, Xuan J, Wang F, Ma H, Wang Y, Zhu M. Proteus mirabilis exacerbates ulcerative colitis by inhibiting mucin production. Front Microbiol 2025; 16:1556953. [PMID: 40201443 PMCID: PMC11975560 DOI: 10.3389/fmicb.2025.1556953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration in colonic mucosa, accompanied by a defective epithelial barrier. Proteus mirabilis (P. mirabilis) bacterium is a putative intestinal pathogen with invasive ability, yet its role in UC inflammation and gut barrier disruption is unclear. This study aims to investigate its epidemiological presence, pathogenic roles and preventive strategy during UC inflammation. Method P. mirabilis culture and PCR amplification of the P. mirabilis-specific ureR gene were used to detect fecal P. mirabilis and determine its prevalence in UC and control stool specimens. P. mirabilis isolated from UC stool specimens was gavaged into dextran sulfate sodium (DSS)-treated mice. Inflammation and the mucus layer of colons were assessed through histological examination and cytokine quantification. Bacteriophages were screened and used to eliminate P. mirabilis in colitis animals. Results and discussion The fecal P. mirabilis bacteria were detected by PCR amplification of P. mirabilis-specific ureR gene. Of 41 UC patients, 65.9% patients were P. mirabilis positive, which was significantly higher than the controls. Administration of P. mirabilis aggravated DSS-induced colitis symptom and mucosal inflammation in mice. Interestingly, the colonic mucus layer, an essential component of the epithelial barrier, of the animals was dramatically disrupted, which was consistent with the alteration of human UC colon. The disrupted mucus layer was mediated by the down-regulation of IL-18 in intestinal epithelium. Importantly, a bacteriophage cocktail targeting P. mirabilis could restore the mucus barrier and alleviate the enteric inflammation. Thus, our results suggest that P. mirabilis is a UC pathobiont bacterium, which exacerbates the severity of UC inflammation owing to down-regulation of mucin production and IL-18 expression. Bacteriophage-mediated elimination of P. mirabilis may be effective in limiting UC inflammation.
Collapse
Affiliation(s)
- Zhihui Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Pengpeng Li
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Kehui Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Yang Liao
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Ji Xuan
- Department of Gastroenterology, Jinling Hospital, The Medical School of Nanjing University, Nanjing, China
| | - Fangyu Wang
- Department of Gastroenterology, Jinling Hospital, The Medical School of Nanjing University, Nanjing, China
| | - Hongfeng Ma
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
- Department of Rehabilitation Medicine, Huzhou Rehabilitation Hospital, Huzhou, China
| | - Ye Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| | - Minsheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Suqian Scientific Research Institute of Nanjing University Medical School, Gulou Hospital of the Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
20
|
Wang L, Zhao H, Wu F, Chen J, Xu H, Gong W, Wen S, Yang M, Xia J, Chen Y, Chen D. Bile-Liver phenotype: Exploring the microbiota landscape in bile and intratumor of cholangiocarcinoma. Comput Struct Biotechnol J 2025; 27:1173-1186. [PMID: 40206347 PMCID: PMC11981758 DOI: 10.1016/j.csbj.2025.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Cholangiocarcinoma (CCA) arises within the peritumoral bile microenvironment, yet microbial translocation from bile to intracholangiocarcinoma (IntraCCA) tissues remains poorly understood. Previous studies on bile microbiota alterations from biliary benign disease (BBD) to CCA have yielded inconsistent results, highlighting the need for cross-study analysis. We presented a comprehensive analysis of five cohorts (N = 266), including our newly established 16S rRNA gene profiling (n = 42), to elucidate these microbiota transitions. The concordance of bacteria between CCA bile and intraCCA tissue, represented by Enterococcus and Staphylococcus, suggested microbiota migration from bile to intratumoral tissues. A computational random forest machine learning model effectively distinguished intraCCA tissue from CCA bile, identifying Rhodococcus and Ralstonia as diagnostically significant. The model also excelled in differentiating CCA bile from BBD bile, achieving an AUC value of 0.931 in external validation. Using unsupervised hierarchical clustering, we established Biletypes based on microbial signatures in our cohort. A combination of 17 genera effectively stratified patients into Biletype A and Biletype B. Biletype B robustly discerned CCA from BBD, with Sub-Biletype B1 correlating with advanced TNM stage and poorer prognosis. Among the 17 genera, bacterial Cluster 1, composed of Sphingomonas, Staphylococcus, Massilia, Paenibacillus, Porphyrobacter, Lawsonella, and Aerococcus, was enriched in Biletype B1 and predicted CCA with an AUC of 0.96. Staphylococcus emerged as a promising single-genus predictor for CCA diagnosis and staging. In conclusion, this study delineates a potential microbiota transition pathway from the gut through CCA bile to intra-CCA tissue, proposing Biletypes and Staphylococcus as biomarkers for CCA prognosis.
Collapse
Affiliation(s)
- Lei Wang
- Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of Hepatopancreatobiliary Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Hui Zhao
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Fan Wu
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Jiale Chen
- Department of Hospital Infection Management,Wujin Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Changzhou 213161, China
| | - Hanjie Xu
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| | - Wanwan Gong
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Sijia Wen
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Mengmeng Yang
- Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China
| | - Jiazeng Xia
- Department of General Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China
- Department of General Surgery, Jiangnan University Medical Center, Wuxi 214002, China
| | - Yu Chen
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| | - Daozhen Chen
- Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
- Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214002, China
| |
Collapse
|
21
|
Adachi A, Zhang F, Kanaya S, Ono N. Quantifying uncertainty in microbiome-based prediction using Gaussian processes with microbial community dissimilarities. BIOINFORMATICS ADVANCES 2025; 5:vbaf045. [PMID: 40110560 PMCID: PMC11919817 DOI: 10.1093/bioadv/vbaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/24/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
Summary The human microbiome is closely associated with the health and disease of the human host. Machine learning models have recently utilized the human microbiome to predict health conditions and disease status. Quantifying predictive uncertainty is essential for the reliable application of these microbiome-based prediction models in clinical settings. However, uncertainty quantification in such prediction models remains unexplored. In this study, we have developed a probabilistic prediction model using a Gaussian process (GP) with a kernel function that incorporates microbial community dissimilarities. We evaluated the performance of probabilistic prediction across three regression tasks: chronological age, body mass index, and disease severity, using publicly available human gut microbiome datasets. The results demonstrated that our model outperformed existing methods in terms of probabilistic prediction accuracy. Furthermore, we found that the confidence levels closely matched the empirical coverage and that data points predicted with lower uncertainty corresponded to lower prediction errors. These findings suggest that GP regression models incorporating community dissimilarities effectively capture the characteristics of phylogenetic, high-dimensional, and sparse microbial abundance data. Our study provides a more reliable framework for microbiome-based prediction, potentially advancing the application of microbiome data in health monitoring and disease diagnosis in clinical settings. Availability and implementation The code is available at https://github.com/asahiadachi/gp4microbiome.
Collapse
Affiliation(s)
- Asahi Adachi
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Fan Zhang
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Shigehiko Kanaya
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Data Science Center, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Naoaki Ono
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Data Science Center, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| |
Collapse
|
22
|
Valdés-Mas R, Leshem A, Zheng D, Cohen Y, Kern L, Zmora N, He Y, Katina C, Eliyahu-Miller S, Yosef-Hevroni T, Richman L, Raykhel B, Allswang S, Better R, Shmueli M, Saftien A, Cullin N, Slamovitz F, Ciocan D, Ouyang KS, Mor U, Dori-Bachash M, Molina S, Levin Y, Atarashi K, Jona G, Puschhof J, Harmelin A, Stettner N, Chen M, Suez J, Honda K, Lieb W, Bang C, Kori M, Maharshak N, Merbl Y, Shibolet O, Halpern Z, Shouval DS, Shamir R, Franke A, Abdeen SK, Shapiro H, Savidor A, Elinav E. Metagenome-informed metaproteomics of the human gut microbiome, host, and dietary exposome uncovers signatures of health and inflammatory bowel disease. Cell 2025; 188:1062-1083.e36. [PMID: 39837331 DOI: 10.1016/j.cell.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Host-microbiome-dietary interactions play crucial roles in regulating human health, yet their direct functional assessment remains challenging. We adopted metagenome-informed metaproteomics (MIM), in mice and humans, to non-invasively explore species-level microbiome-host interactions during commensal and pathogen colonization, nutritional modification, and antibiotic-induced perturbation. Simultaneously, fecal MIM accurately characterized the nutritional exposure landscape in multiple clinical and dietary contexts. Implementation of MIM in murine auto-inflammation and in human inflammatory bowel disease (IBD) characterized a "compositional dysbiosis" and a concomitant species-specific "functional dysbiosis" driven by suppressed commensal responses to inflammatory host signals. Microbiome transfers unraveled early-onset kinetics of these host-commensal cross-responsive patterns, while predictive analyses identified candidate fecal host-microbiome IBD biomarker protein pairs outperforming S100A8/S100A9 (calprotectin). Importantly, a simultaneous fecal nutritional MIM assessment enabled the determination of IBD-related consumption patterns, dietary treatment compliance, and small intestinal digestive aberrations. Collectively, a parallelized dietary-bacterial-host MIM assessment functionally uncovers trans-kingdom interactomes shaping gastrointestinal ecology while offering personalized diagnostic and therapeutic insights into microbiome-associated disease.
Collapse
Affiliation(s)
- Rafael Valdés-Mas
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Avner Leshem
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Danping Zheng
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yotam Cohen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Corine Katina
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | | | - Tal Yosef-Hevroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Richman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Barbara Raykhel
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Allswang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Better
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Shmueli
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nyssa Cullin
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Fernando Slamovitz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dragos Ciocan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Uria Mor
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Mally Dori-Bachash
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shahar Molina
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ghil Jona
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Stettner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jotham Suez
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank Popgen, University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Michal Kori
- Pediatric Gastroenterology Unit, Kaplan Medical Center, Rehovot, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitsan Maharshak
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yifat Merbl
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Zamir Halpern
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Dror S Shouval
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Raanan Shamir
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Shapiro
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
23
|
Xu L, Wang C, Liu Y, Zhang Y, Li Z, Pang L. MASP1 modulation as a novel therapeutic target in severe pediatric pertussis: insights from a multi-omics approach. Infect Immun 2025; 93:e0027124. [PMID: 39841046 PMCID: PMC11834402 DOI: 10.1128/iai.00271-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025] Open
Abstract
Pertussis, a severe infectious disease in children, has become increasingly prominent in recent years. This study aims to investigate the role of the MASP1 protein in severe pertussis in children through multi-omics analysis, providing a theoretical basis for the development of novel therapeutic strategies. The study retrieved macro-genome and 16S rRNA data of pediatric pertussis from public databases to analyze microbial diversity and specific flora abundance, conducting pathway functional enrichment analysis. Differential expression analysis of transcriptome data and Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis, combined with machine learning, identified the key gene MASP1. A Bordetella pertussis infection model was established using human bronchial epithelial cell line HBE135-E6E7 to validate MASP1 expression changes and investigate its relationship with airway epithelial cell damage by constructing cell lines overexpressing and knocking down MASP1. Finally, the impact of inhibiting MASP1 expression on infection symptoms was evaluated using a mouse pertussis infection model. The results revealed significant differences in microbial diversity and specific flora abundance between healthy children and those with pertussis, with MASP1 significantly upregulated in severe pertussis and its inhibition alleviating infection symptoms. The study highlights the critical role of MASP1 in pertussis, providing a crucial foundation for developing therapeutic strategies targeting MASP1.
Collapse
Affiliation(s)
- Lin Xu
- Department of Pediatrics, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Caiying Wang
- Department of Pediatrics, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yuhuan Liu
- Department of Pediatrics, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yanlan Zhang
- Department of Pediatrics, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Chaoyang District Center for Disease Control and Prevention, Beijing, China
| | - Lin Pang
- Department of Pediatrics, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Gao Z, Tang J, Wu C, Ding W, Wang X, Long Y, Wang Y, Liu H. In Vitro Assessment of Bacillus thuringiensis Exopolysaccharides and Their Effects on Gut Microbiota from Ulcerative Colitis In Vitro. Int J Mol Sci 2025; 26:1692. [PMID: 40004156 PMCID: PMC11855630 DOI: 10.3390/ijms26041692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Bacillus thuringiensis exopolysaccharide BPS-2 inhibits malondialdehyde secretion, enhances antioxidant enzyme activities, and significantly improves the antioxidant status of inflammatory cells. In the present study, the apparent morphology and spatial conformation of BPS-2 were analyzed further, and several functional properties were investigated. The results demonstrated that BPS-2 was a polymeric straight-chain polysaccharide with good thermal stability, exhibiting non-Newtonian properties and good antioxidant and anticancer activities. Notably, this study systematically investigated the impact of BPS-2 on the intestinal microbiota composition in patients diagnosed with ulcerative colitis. Through in vitro fermentation of fecal bacteria collected from six volunteers, it was found that BPS-2 exerted a positive influence on the intestinal flora of ulcerative colitis patients, augmenting the secretion of short-chain fatty acids and facilitating an increase in the relative abundance of Bifidobacterium spp. These results suggest that BPS-2 has the potential to be a food additive for suppressing ulcerative colitis and for other medically related applications.
Collapse
Affiliation(s)
- Zexin Gao
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China;
| | - Jie Tang
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
| | - Chuanchao Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China;
| | - Wenping Ding
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
| | - Xianyi Wang
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
| | - Yaohang Long
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
| | - Yaping Wang
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
| | - Hongmei Liu
- Engineering Research Center of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, China; (Z.G.); (J.T.); (W.D.); (X.W.); (Y.L.); (Y.W.)
- School of Basic Medicine Science, Guizhou Medical University, Guiyang 550031, China
| |
Collapse
|
25
|
Elmassry MM, Sugihara K, Chankhamjon P, Kim Y, Camacho FR, Wang S, Sugimoto Y, Chatterjee S, Chen LA, Kamada N, Donia MS. A meta-analysis of the gut microbiome in inflammatory bowel disease patients identifies disease-associated small molecules. Cell Host Microbe 2025; 33:218-234.e12. [PMID: 39947133 DOI: 10.1016/j.chom.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 02/19/2025]
Abstract
Gut microbiome changes have been associated with several human diseases, but the molecular and functional details underlying these associations remain largely unknown. Here, we performed a meta-analysis of small molecule biosynthetic gene clusters (BGCs) in metagenomic samples of the gut microbiome from inflammatory bowel disease (IBD) patients and matched healthy subjects and identified two Clostridia-derived BGCs that are significantly associated with Crohn's disease (CD), a main IBD type. Using synthetic biology, we discovered and solved the structures of six fatty acid amides as the products of the CD-enriched BGCs, which we subsequently detected in fecal samples from IBD patients. Finally, we show that the discovered molecules disrupt gut permeability and exacerbate disease in chemically or genetically susceptible mouse models of colitis. These findings suggest that microbiome-derived small molecules may play a role in the etiology of IBD and represent a generalizable approach for discovering molecular mediators of disease-relevant microbiome-host interactions.
Collapse
Affiliation(s)
- Moamen M Elmassry
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Kohei Sugihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Yeji Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francine R Camacho
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Shuo Wang
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Yuki Sugimoto
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Seema Chatterjee
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lea Ann Chen
- Department of Medicine, Division of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
26
|
Agudelo C, Kateete DP, Nasinghe E, Kamulegeya R, Lubega C, Mbabazi M, Baker N, Lin KY, Liu CC, Kasambula AS, Kigozi E, Komakech K, Mukisa J, Mulumba K, Mwachan P, Nakalanda BS, Nalubega GP, Nsubuga J, Sitenda D, Ssenfuka H, Cirolia GT, Gustafson JT, Wang R, Nsubuga ML, Yiga F, Stanley SA, Bagaya BS, Elliott A, Joloba M, Wolf AR. Enterococcus and Eggerthella species are enriched in the gut microbiomes of COVID-19 cases in Uganda. Gut Pathog 2025; 17:9. [PMID: 39905557 DOI: 10.1186/s13099-025-00678-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Infection with the COVID-19-causing pathogen SARS-CoV-2 is associated with disruption in the human gut microbiome. The gut microbiome enables protection against diverse pathogens and exhibits dysbiosis during infectious and autoimmune disease. Studies based in the United States and China have found that severe COVID-19 cases have altered gut microbiome composition when compared to mild COVID-19 cases. We present the first study to investigate the gut microbiome composition of COVID-19 cases in a population from Sub-Saharan Africa. Given the impact of geography and cultural traditions on microbiome composition, it is important to investigate the microbiome globally and not draw broad conclusions from homogenous populations. RESULTS We used stool samples in a Ugandan biobank collected from COVID-19 cases during 2020-2022. We profiled the gut microbiomes of 83 symptomatic individuals who tested positive for SARS-CoV-2 along with 43 household contacts who did not present any symptoms of COVID-19. The inclusion of healthy controls enables us to generate hypotheses about bacterial strains potentially related to susceptibility to COVID-19 disease, which is highly heterogeneous. Comparison of the COVID-19 patients and their household contacts revealed decreased alpha diversity and blooms of Enterococcus and Eggerthella in COVID-19 cases. CONCLUSIONS Our study finds that the microbiome of COVID-19 individuals is more likely to be disrupted, as indicated by decreased diversity and increased pathobiont levels. This is either a consequence of the disease or may indicate that certain microbiome states increase susceptibility to COVID-19 disease. Our findings enable comparison with cohorts previously published in the Global North, as well as support new hypotheses about the interaction between the gut microbiome and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carolina Agudelo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Emmanuel Nasinghe
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Rogers Kamulegeya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Christopher Lubega
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Monica Mbabazi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Noah Baker
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Kathryn Y Lin
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Chang C Liu
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Arthur Shem Kasambula
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Edgar Kigozi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Kevin Komakech
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - John Mukisa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Kassim Mulumba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Patricia Mwachan
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Brenda Sharon Nakalanda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Gloria Patricia Nalubega
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Julius Nsubuga
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Diana Sitenda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Henry Ssenfuka
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Giana T Cirolia
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Biophysics PhD Program, University of California, Berkeley, Berkeley, CA, USA
| | - Jeshua T Gustafson
- College of Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Ruohong Wang
- College of Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Moses Luutu Nsubuga
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Fahim Yiga
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Sarah A Stanley
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda
| | - Alison Elliott
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Moses Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda.
- Integrated Biorepository of H3Africa Uganda (IBRH3AU), Kampala, Uganda.
| | - Ashley R Wolf
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
27
|
Jiang Y, Liao D, Zhu Q, Lu YY. PhyloMix: enhancing microbiome-trait association prediction through phylogeny-mixing augmentation. Bioinformatics 2025; 41:btaf014. [PMID: 39799515 PMCID: PMC11849959 DOI: 10.1093/bioinformatics/btaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
MOTIVATION Understanding the associations between traits and microbial composition is a fundamental objective in microbiome research. Recently, researchers have turned to machine learning (ML) models to achieve this goal with promising results. However, the effectiveness of advanced ML models is often limited by the unique characteristics of microbiome data, which are typically high-dimensional, compositional, and imbalanced. These characteristics can hinder the models' ability to fully explore the relationships among taxa in predictive analyses. To address this challenge, data augmentation has become crucial. It involves generating synthetic samples with artificial labels based on existing data and incorporating these samples into the training set to improve ML model performance. RESULTS Here, we propose PhyloMix, a novel data augmentation method specifically designed for microbiome data to enhance predictive analyses. PhyloMix leverages the phylogenetic relationships among microbiome taxa as an informative prior to guide the generation of synthetic microbial samples. Leveraging phylogeny, PhyloMix creates new samples by removing a subtree from one sample and combining it with the corresponding subtree from another sample. Notably, PhyloMix is designed to address the compositional nature of microbiome data, effectively handling both raw counts and relative abundances. This approach introduces sufficient diversity into the augmented samples, leading to improved predictive performance. We empirically evaluated PhyloMix on six real microbiome datasets across five commonly used ML models. PhyloMix significantly outperforms distinct baseline methods including sample-mixing-based data augmentation techniques like vanilla mixup and compositional cutmix, as well as the phylogeny-based method TADA. We also demonstrated the wide applicability of PhyloMix in both supervised learning and contrastive representation learning. AVAILABILITY AND IMPLEMENTATION The Apache-licensed source code is available at (https://github.com/batmen-lab/phylomix).
Collapse
Affiliation(s)
- Yifan Jiang
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Disen Liao
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Qiyun Zhu
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, United States
| | - Yang Young Lu
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
28
|
Gao X, Liang H, Hu T, Zou Y, Xiao L. Cultivated genome references for protein database construction and high-resolution taxonomic annotation in metaproteomics. Microbiol Spectr 2025; 13:e0175524. [PMID: 39665565 PMCID: PMC11792528 DOI: 10.1128/spectrum.01755-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 12/13/2024] Open
Abstract
Metaproteomics offers a profound understanding of the functional dynamics of the gut microbiome, which is crucial for personalized healthcare strategies. The selection of an appropriate database is a critical step for the identification of peptides and proteins, as well as for the provision of accurate taxonomic and functional annotations. The matched metagenomic-derived database is considered to be the best, but its limitations include the identification of low-abundance organisms and taxonomic resolution. Herein, we constructed a protein database (DBCGR2) based on Cultivated Genome Reference 2 (CGR2) and developed a complete peptide-centric analysis workflow for database searching and for the annotation of taxonomy and function. This workflow was subsequently appraised in comparison with metagenomics-derived databases for the analysis of metaproteomic data. Our findings suggested that the performance of DBCGR2 in identification was comparable with metagenomics-derived databases with improvement in identification rates of peptides from low-abundance species. The database searching results could be fully annotated using the pepTaxa taxonomic annotation approach developed in this study, and the taxonomic resolution was enhanced to strain level. Additionally, the results demonstrated that the sensitivity of functional annotation could be enhanced by employing DBCGR2. Overall, the DBCGR2 combined with pepTaxa can be considered an alternative for metaproteomic data analysis with superior analysis performances.IMPORTANCEMass spectrometry-based metaproteomics offers a profound understanding of the gut microbial taxonomy and functionality. The databases utilized in the analysis of metaproteomic data are crucial, as they determine the identification of proteins that can be recognized and linked to overall human health, in addition to the quality of taxonomic and functional annotation. Among the most effective approaches for constructing protein databases is the utilization of metagenomic sequencing to create matched databases. However, the database, derived from isolated genomes, has yet to undergo rigorous testing for their efficacy and accuracy in protein identification and taxonomic and functional annotation. Here, we constructed a protein database DBCGR2 derived from Cultivated Genome Reference 2 (CGR2) and a complete workflow for data analysis. We compared the performances of DBCGR2 and metagenomics-derived databases. Our results indicated that DBCGR2 can be regarded as an alternative to metagenomics-derived databases, which contribute to metaproteomic data analysis.
Collapse
|
29
|
Li Y, Zhao J, Ma Q, Xu J, Yuan Y, Zhang Q, Yan W, Xu W, Yang Z, Li G, Gao L, Wang B. Comparison of clinical characteristics, risk factors, and outcomes of patients infected with Bacteroides fragilis group at a tertiary care hospital in central China between 2017 and March 2024. Anaerobe 2025; 91:102922. [PMID: 39581456 DOI: 10.1016/j.anaerobe.2024.102922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES This study aimed to explore the epidemiology, clinical presentation, and risk factors for treatment failure in patients infected with various Bacteroides fragilis group (BFG) species. METHODS We analyzed clinical and laboratory data from 178 patients infected with BFG who were admitted to our hospital between 2017 and 2024. We analyzed categorical data using Chi-square and Fisher exact tests, continuous variables using Student's t-tests or Mann-Whitney U-tests, and risk factors using multivariate logistic regression and Pearson's chi-squared tests. RESULTS Bacteroides fragilis (69.7 %) and Bacteroides thetaiotaomicron (16.9 %) were the most prevalent species among 178 BFG isolates. Most patients had polymicrobial infections, with the following pathogens isolated from concurrent samples: Escherichia coli, Klebsiella spp., and Enterococcus spp. Intra-abdominal sites were most frequently infected with BFG. Patients aged >50 years and those who had intravascular catheters were infected with more other Bacteroides species than B. fragilis. Admission to an ICU, respiratory, renal and cardiovascular diseases, and chest drainage were associated more often with B. thetaiotaomicron than with B. fragilis. Gastrointestinal diseases, tracheal intubation, and ICU admission were associated more frequently with treatment failure in patients infected with B. fragilis whereas solid cancers, renal disease, multiple organ dysfunction syndrome, and tracheal intubation were more likely to be associated with treatment failure in patients infected with other Bacteroides spp. CONCLUSIONS The most prevalent BFG species in the patients were B. fragilis and B. thetaiotaomicron. The demographic characteristics of the patients, underlying diseases, and risk factors for poor clinical outcomes clearly differed among species.
Collapse
Affiliation(s)
- Yongmei Li
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Jing Zhao
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Qiong Ma
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Junhong Xu
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Youhua Yuan
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Qi Zhang
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Wenjuan Yan
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Wenbo Xu
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Zhiyu Yang
- Department of Digestion, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Gang Li
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Lan Gao
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Baoya Wang
- Department of Clinical Laboratory, People's Hospital of Henan University, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China.
| |
Collapse
|
30
|
Misselwitz B, Haller D. [The intestinal microbiota in inflammatory bowel diseases]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:146-155. [PMID: 39870907 DOI: 10.1007/s00108-024-01845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND The intestinal microbiota comprises all living microorganisms in the gastrointestinal tract and is crucial for its function. Clinical observations and laboratory findings confirm a central role of the microbiota in chronic inflammatory bowel diseases (IBD). However, many mechanistic details remain unclear. OBJECTIVES Changes in the microbiota and the causal relationship with the pathogenesis of IBD are described and current and future diagnostic and therapeutic options are discussed. MATERIALS AND METHODS Narrative review. RESULTS The intestinal microbiota is altered in composition, diversity, and function in IBD patients, but specific (universal) IBD-defining bacteria have not been identified. The healthy microbiota has numerous anti-inflammatory functions such as the production of short-chain fatty acids or competition with pathogens. In contrast, the IBD microbiota promotes inflammation through the destruction of the intestinal barrier and direct interaction with the immune system. The balance between pro- and anti-inflammatory effects of the microbiota appears to be crucial for the development of intestinal inflammation. Microbiota-based IBD diagnostics show promise but are not yet ready for clinical use. Probiotics and fecal microbiota transplantation have clinical effects, especially in ulcerative colitis, but the potential of microbiota-based therapies is far from being fully realized. CONCLUSION IBD dysbiosis remains undefined so far. It is unclear how the many parallel pro- and anti-inflammatory mechanisms contribute to IBD pathogenesis. An inadequate mechanistic understanding hinders the development of microbiota-based diagnostics and therapies.
Collapse
Affiliation(s)
- Benjamin Misselwitz
- Medizinische Klinik und Poliklinik II, Ludwig-Maximilians-Universität München, Marchioninistraße 15, 83477, München, Deutschland.
| | - Dirk Haller
- Lehrstuhl für Ernährung und Immunologie, School of Life Sciences, Technische Universität München, Gregor-Mendel-Straße 2, 85354, Freising, Deutschland.
| |
Collapse
|
31
|
Li J, Ji W, Chen G, Yu K, Zeng J, Zhang Q, Xiong G, Du C, Peng Y, Zeng X, Chen C. Peonidin-3-O-(3,6-O-dimalonyl-β-D-glucoside), a polyacylated anthocyanin isolated from the black corncobs, alleviates colitis by modulating gut microbiota in DSS-induced mice. Food Res Int 2025; 202:115688. [PMID: 39967148 DOI: 10.1016/j.foodres.2025.115688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025]
Abstract
Polyacylated anthocyanins are known for their enhanced stability and immunosuppressive properties. Although peonidin-3-O-(3,6-O-dimalonyl-β-D-glucoside) (P3GdM) from black corncobs has demonstrated notable antibacterial and stress-resistance effects in plants, its regulatory role in inflammatory bowel disease (IBD) remains unexplored. In this study, P3GdM was isolated from black corncobs, and its potential as a treatment for dextran sulfate sodium (DSS)-induced colitis in mice was evaluated. The findings revealed that P3GdM significantly mitigated clinical symptoms, reduced the disease activity index (DAI), suppressed the production of pro-inflammatory cytokines and endotoxins, and repaired the intestinal barrier. Furthermore, P3GdM markedly improved DSS-induced gut microbiota dysbiosis, significantly increasing microbial diversity and enhancing the relative abundance of critical bacterial species such as Akkermansia muciniphila and Lactobacillus reuteri, while also stimulating the production of short-chain fatty acids (SCFAs) and lactic acid. Correlation analyses further revealed strong associations between key microbial taxa, pro-inflammatory factors, clinical symptoms, tight junction proteins, and SCFAs. These findings provide support for the potential of P3GdM as an adjunct therapy for intestinal disorders, particularly colitis.
Collapse
Affiliation(s)
- Junjie Li
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Wenting Ji
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences & Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Kun Yu
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Jianhua Zeng
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Qi Zhang
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Guoyuan Xiong
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Chuanlai Du
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China
| | - Yujia Peng
- Key Laboratory for Waste Plastics Biocatalytic Degradation and Recycling, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Chunxu Chen
- College of Food Engineering, Anhui Science and Technology University, Chuzhou 233100, Anhui, China; College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Anhui Provincial Key Laboratory of Functional Agriculture and Functional Foods, Chuzhou 233100, China.
| |
Collapse
|
32
|
Wasney M, Briscoe L, Wolff R, Ghezzi H, Tropini C, Garud N. Uniform bacterial genetic diversity along the guts of mice inoculated with human stool. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635365. [PMID: 39974986 PMCID: PMC11838389 DOI: 10.1101/2025.01.28.635365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Environmental gradients exist throughout the digestive tract, driving spatial variation in the membership and abundance of bacterial species along the gut. However, less is known about the distribution of genetic diversity within bacterial species along the gut. Understanding this distribution is important because bacterial genetic variants confer traits important for the functioning of the microbiome and are also known to impart phenotypes to the hosts, including local inflammation along the gut and the ability to digest food. Thus, to be able to understand how the microbiome functions at a mechanistic level, it is essential to understand how genetic diversity is organized along the gut and the ecological and evolutionary processes that give rise to this organization. In this study, we analyzed bacterial genetic diversity of approximately 30 common gut commensals in five regions along the gut lumen in germ-free mice colonized with the same healthy human stool sample. While species membership and abundances varied considerably along the gut, genetic diversity within species was substantially more uniform. Driving this uniformity were similar strain frequencies along the gut, implying that multiple, genetically divergent strains of the same species can coexist within a host without spatially segregating. Additionally, the approximately 60 unique evolutionary adaptations arising within mice tended to sweep throughout the gut, showing little specificity for particular gut regions. Together, our findings show that genetic diversity may be more uniform along the gut than species diversity, which implies that species presence-absence may play a larger role than genetic variation in responding to varied environments along the gut.
Collapse
Affiliation(s)
- Michael Wasney
- University of California, Los Angeles, Human Genetics, Los Angeles, CA
| | - Leah Briscoe
- University of California, Los Angeles, Interdepartmental Program in Bioinformatics, Los Angeles, CA
| | - Richard Wolff
- University of California, Los Angeles, Ecology and Evolutionary Biology, Los Angeles, CA
| | - Hans Ghezzi
- University of British Columbia, Department of Bioinformatics, Vancouver, Canada
| | - Carolina Tropini
- University of British Columbia, Department of Microbiology and Immunology, Vancouver, Canada
- University of British Columbia, School of Biomedical Engineering, Vancouver, Canada
- Canadian Institute for Advanced Research, Humans and the Microbiome Program, Toronto, Canada
| | - Nandita Garud
- University of California, Los Angeles, Human Genetics, Los Angeles, CA
- University of California, Los Angeles, Interdepartmental Program in Bioinformatics, Los Angeles, CA
- University of California, Los Angeles, Ecology and Evolutionary Biology, Los Angeles, CA
| |
Collapse
|
33
|
Pangga GM, Star-Shirko B, Psifidi A, Xia D, Corcionivoschi N, Kelly C, Hughes C, Lavery U, Richmond A, Ijaz UZ, Gundogdu O. Impact of commercial gut health interventions on caecal metagenome and broiler performance. MICROBIOME 2025; 13:30. [PMID: 39881387 PMCID: PMC11776324 DOI: 10.1186/s40168-024-02012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Maintaining gut health is a persistent and unresolved challenge in the poultry industry. Given the critical role of gut health in chicken performance and welfare, there is a pressing need to identify effective gut health intervention (GHI) strategies to ensure optimal outcomes in poultry farming. In this study, across three broiler production cycles, we compared the metagenomes and performance of broilers provided with ionophores (as the control group) against birds subjected to five different GHI combinations involving vaccination, probiotics, prebiotics, essential oils, and reduction of ionophore use. RESULTS Using a binning strategy, 84 (≥ 75% completeness, ≤ 5% contamination) metagenome-assembled genomes (MAGs) from 118 caecal samples were recovered and annotated for their metabolic potential. The majority of these (n = 52, 61%) had a differential response across all cohorts and are associated with the performance parameter - European poultry efficiency factor (EPEF). The control group exhibited the highest EPEF, followed closely by the cohort where probiotics are used in conjunction with vaccination. The use of probiotics B, a commercial Bacillus strain-based formulation, was determined to contribute to the superior performance of birds. GHI supplementation generally affected the abundance of microbial enzymes relating to carbohydrate and protein digestion and metabolic pathways relating to energy, nucleotide synthesis, short-chain fatty acid synthesis, and drug-transport systems. These shifts are hypothesised to differentiate performance among groups and cycles, highlighting the beneficial role of several bacteria, including Rikenella microfusus and UBA7160 species. CONCLUSIONS All GHIs are shown to be effective methods for gut microbial modulation, with varying influences on MAG diversity, composition, and microbial functions. These metagenomic insights greatly enhance our understanding of microbiota-related metabolic pathways, enabling us to devise strategies against enteric pathogens related to poultry products and presenting new opportunities to improve overall poultry performance and health. Video Abstract.
Collapse
Affiliation(s)
- Gladys Maria Pangga
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Banaz Star-Shirko
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | | | - Dong Xia
- Royal Veterinary College, London, UK
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Agri-Food and Biosciences Institute, Veterinary Sciences Division, Belfast, UK
- Faculty of Bioengineering of Animal Resources, University of Life Sciences King Mihai Timișoara, Timișoara, Romania
| | - Carmel Kelly
- Bacteriology Branch, Agri-Food and Biosciences Institute, Veterinary Sciences Division, Belfast, UK
| | | | | | | | - Umer Zeeshan Ijaz
- James Watt School of Engineering, University of Glasgow, Glasgow, UK.
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
34
|
Zhang Y, Hao R, Chen J, Huang K, Li S, Cao H, Guan X. Gut-Derived Ursodeoxycholic Acid from Saponins of Quinoa Regulated Colitis via Inhibiting the TLR4/NF-κB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2415-2429. [PMID: 39827465 DOI: 10.1021/acs.jafc.4c09151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Alteration of the gut microbiota and its metabolites plays a key role in the development of inflammatory bowel disease (IBD). Here, we investigated the mechanism of saponins, a byproduct from quinoa (SQ) processing, in regulating IBD. SQ ameliorated gut microbiota dysbiosis revealed by 16S rRNA sequencing and improved colonic antioxidant activities and barrier integrity in dextran sulfate sodium (DSS)-treated mice. Broad-spectrum antibiotics further proved that the gut-protective effects of SQ were mediated by gut microbiota. Next, fecal microbiota transplantation (FMT) of SQ-induced gut microbiota/metabolites to inoculate DSS-treated mice alleviated colitis significantly. Untargeted metabolomics and lipidomics revealed that ursodeoxycholic acid (UDCA) was enriched as a microbial metabolite after SQ supplementation. UDCA was then found to attenuate DSS-induced colitis in vivo by targeting the TLR4/NF-κB pathway, which was also verified in a Caco-2 cell model treated with a TLR4 agonist/antagonist. Overall, our findings established that gut microbiota-UDCA-TLR4/NF-κB signaling plays a key role in mediating the protective effects of SQ.
Collapse
Affiliation(s)
- Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Ruojie Hao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junda Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Hongwei Cao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| |
Collapse
|
35
|
Chen S, Zhang D, Li D, Zeng F, Chen C, Bai F. Microbiome characterization of patients with Crohn disease and the use of fecal microbiota transplantation: A review. Medicine (Baltimore) 2025; 104:e41262. [PMID: 39854760 PMCID: PMC11771716 DOI: 10.1097/md.0000000000041262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/19/2024] [Accepted: 11/15/2024] [Indexed: 01/26/2025] Open
Abstract
Inflammatory bowel disease is a chronic inflammatory condition predominantly affecting the intestines, encompassing both ulcerative colitis and Crohn disease (CD). As one of the most common gastrointestinal disorders, CD's pathogenesis is closely linked with the intestinal microbiota. Recently, fecal microbiota transplantation (FMT) has gained attention as a potential treatment for CD, with the effective reestablishment of intestinal microecology considered a crucial mechanism of FMT therapy. This article synthesizes the findings of population-based cohort studies to enhance our understanding of gut microbial characteristics in patients with CD. It delves into the roles of "beneficial" and "pathogenic" bacteria in CD's development. This article systematically reviews and compares data on clinical response rates, remission rates, adverse events, and shifts in bacterial microbiota. Among these studies, gut microbiome analysis was conducted in only 7, and a single study examined the metabolome. Overall, FMT has demonstrated a partial restoration of typical CD-associated microbiological alterations, leading to increased α-diversity in responders and a moderate shift in patient microbiota toward the donor profile. Several factors, including donor selection, delivery route, microbial state (fresh or frozen), and recipient condition, are identified as pivotal in influencing FMT's effectiveness. Future prospective clinical studies with larger patient cohorts and improved methodologies are imperative. In addition, standardization of FMT procedures, coupled with advanced genomic techniques such as macroproteomics and culture genomics, is necessary. These advancements will further clarify the bacterial microbiota alterations that significantly contribute to FMT's therapeutic effects in CD treatment, as well as elucidate the underlying mechanisms of action.
Collapse
Affiliation(s)
- Shiju Chen
- Graduate School, Hainan Medical University, Haikou, China
| | - Daya Zhang
- Graduate School, Hainan Medical University, Haikou, China
| | - Da Li
- Graduate School, Hainan Medical University, Haikou, China
| | - Fan Zeng
- Graduate School, Hainan Medical University, Haikou, China
| | - Chen Chen
- Graduate School, Hainan Medical University, Haikou, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
36
|
Lakemeyer M, Latorre R, Blazkova K, Jensen D, Wood HM, Shakil N, Thomas SC, Saxena D, Mulpuri Y, Poolman D, de Haro PD, Keller LJ, Reed DE, Schmidt BL, Lomax AE, Bunnett NW, Bogyo M. Identification of a secreted protease from Bacteroides fragilis that induces intestinal pain and inflammation by cleavage of PAR 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633241. [PMID: 39868234 PMCID: PMC11761754 DOI: 10.1101/2025.01.15.633241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Protease-activated receptor 2 (PAR2) is a central regulator of intestinal barrier function, inflammation and pain. Upregulated intestinal proteolysis and PAR2-signaling are implicated in inflammatory bowel diseases (IBDs) and irritable bowel syndrome (IBS). To identify potential bacterial regulators of PAR2 activity, we developed a functional assay for PAR2 processing and used it to screen conditioned media from a library of diverse gut commensal microbes. We found that multiple bacteria secrete proteases that cleave host PAR2. Using chemoproteomic profiling with a covalent irreversible inhibitor, we identified a previously uncharacterized Bacteroides fragilis serine protease Bfp1, and showed that it cleaves and activates PAR2 in multicellular and murine models. PAR2 cleavage by Bfp1 disrupts the intestinal barrier, sensitizes nociceptors, and triggers colonic inflammation and abdominal pain. Collectively, our findings uncover Bfp1-mediated PAR2-processing as a new axis of host-commensal-interaction in the gut that has the potential to be targeted for therapeutic intervention in IBD or IBS.
Collapse
Affiliation(s)
- Markus Lakemeyer
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena; Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena; Jena, Germany
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - Rocco Latorre
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Kristyna Blazkova
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - Dane Jensen
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Hannah M Wood
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Nayab Shakil
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena; Jena, Germany
| | - Scott C Thomas
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - Deepak Saxena
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - Yatendra Mulpuri
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - David Poolman
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Paz Duran de Haro
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Laura J Keller
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
37
|
Zheng ZL, Zheng QF, Wang LQ, Liu Y. Bowel preparation before colonoscopy: Consequences, mechanisms, and treatment of intestinal dysbiosis. World J Gastroenterol 2025; 31:100589. [PMID: 39811511 PMCID: PMC11684204 DOI: 10.3748/wjg.v31.i2.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
The term "gut microbiota" primarily refers to the ecological community of various microorganisms in the gut, which constitutes the largest microbial community in the human body. Although adequate bowel preparation can improve the results of colonoscopy, it may interfere with the gut microbiota. Bowel preparation for colonoscopy can lead to transient changes in the gut microbiota, potentially affecting an individual's health, especially in vulnerable populations, such as patients with inflammatory bowel disease. However, measures such as oral probiotics may ameliorate these adverse effects. We focused on the bowel preparation-induced changes in the gut microbiota and host health status, hypothesized the factors influencing these changes, and attempted to identify measures that may reduce dysbiosis, thereby providing more information for individualized bowel preparation for colonoscopy in the future.
Collapse
Affiliation(s)
- Ze-Long Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Qing-Fan Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Li-Qiang Wang
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yi Liu
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
38
|
Dmitrijeva M, Ruscheweyh HJ, Feer L, Li K, Miravet-Verde S, Sintsova A, Mende DR, Zeller G, Sunagawa S. The mOTUs online database provides web-accessible genomic context to taxonomic profiling of microbial communities. Nucleic Acids Res 2025; 53:D797-D805. [PMID: 39526369 PMCID: PMC11701688 DOI: 10.1093/nar/gkae1004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/03/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Determining the taxonomic composition (taxonomic profiling) is a fundamental task in studying environmental and host-associated microbial communities. However, genome-resolved microbial diversity on Earth remains undersampled, and accessing the genomic context of taxa detected during taxonomic profiling remains a challenging task. Here, we present the mOTUs online database (mOTUs-db), which is consistent with and interfaces with the mOTUs taxonomic profiling tool. It comprises 2.83 million metagenome-assembled genomes (MAGs) and 919 090 single-cell and isolate genomes from 124 295 species-level taxonomic units. In addition to being one of the largest prokaryotic genome resources to date, all MAGs in the mOTUs-db were reconstructed de novo in 117 902 individual samples by abundance correlation of scaffolds across multiple samples for improved quality metrics. The database complements the Genome Taxonomy Database, with over 50% of its species-level taxonomic groups being unique. It also offers interactive querying, enabling users to explore and download genomes at various taxonomic levels. The mOTUs-db is accessible at https://motus-db.org.
Collapse
Affiliation(s)
- Marija Dmitrijeva
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Hans-Joachim Ruscheweyh
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Lilith Feer
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Kang Li
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Samuel Miravet-Verde
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Anna Sintsova
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| | - Daniel R Mende
- Medical Microbiology and Infection Prevention (MMI), Amsterdam University Medical Center, 1105AZ Amsterdam, The Netherlands
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, 2333ZA Leiden, Netherlands
| | - Shinichi Sunagawa
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
39
|
Zhou H, Balint D, Shi Q, Vartanian T, Kriegel MA, Brito I. Lupus and inflammatory bowel disease share a common set of microbiome features distinct from other autoimmune disorders. Ann Rheum Dis 2025; 84:93-105. [PMID: 39874239 PMCID: PMC11868722 DOI: 10.1136/ard-2024-225829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES This study aims to elucidate the microbial signatures associated with autoimmune diseases, particularly systemic lupus erythematosus (SLE) and inflammatory bowel disease (IBD), compared with colorectal cancer (CRC), to identify unique biomarkers and shared microbial mechanisms that could inform specific treatment protocols. METHODS We analysed metagenomic datasets from patient cohorts with six autoimmune conditions-SLE, IBD, multiple sclerosis, myasthenia gravis, Graves' disease and ankylosing spondylitis-contrasting these with CRC metagenomes to delineate disease-specific microbial profiles. The study focused on identifying predictive biomarkers from species profiles and functional genes, integrating protein-protein interaction analyses to explore effector-like proteins and their targets in key signalling pathways. RESULTS Distinct microbial signatures were identified across autoimmune disorders, with notable overlaps between SLE and IBD, suggesting shared microbial underpinnings. Significant predictive biomarkers highlighted the diverse microbial influences across these conditions. Protein-protein interaction analyses revealed interactions targeting glucocorticoid signalling, antigen presentation and interleukin-12 signalling pathways, offering insights into possible common disease mechanisms. Experimental validation confirmed interactions between the host protein glucocorticoid receptor (NR3C1) and specific gut bacteria-derived proteins, which may have therapeutic implications for inflammatory disorders like SLE and IBD. CONCLUSIONS Our findings underscore the gut microbiome's critical role in autoimmune diseases, offering insights into shared and distinct microbial signatures. The study highlights the potential importance of microbial biomarkers in understanding disease mechanisms and guiding treatment strategies, paving the way for novel therapeutic approaches based on microbial profiles. TRIAL REGISTRATION NUMBER NCT02394964.
Collapse
Affiliation(s)
- Hao Zhou
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Diana Balint
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Qiaojuan Shi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | | | - Martin A Kriegel
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, Münster, Germany; Section of Rheumatology and Clinical Immunology, University Hospital Münster, Münster, Germany; Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ilana Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
40
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
41
|
Peng Z, Li D, Wu N, Wang XY, Sun GX, Gao HB, Li HX. Safety and efficacy of curcumin in the treatment of ulcerative colitis: An updated systematic review and meta-analysis of randomized controlled trials. Explore (NY) 2025; 21:103083. [PMID: 39612780 DOI: 10.1016/j.explore.2024.103083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVE This study aims to systematically evaluate the safety and efficacy of curcumin as an adjunctive treatment for patients with ulcerative colitis (UC) and to assess the methodological quality of the published studies. METHODS A comprehensive search was conducted in PubMed, Embase, and CENTRAL databases for randomized controlled trials published up to August 18, 2023. Two independent reviewers screened studies based on predefined criteria. Meta-analysis was performed using a random-effects model with RevMan 5.4. Heterogeneity was assessed using Cochran's Q test and I² statistic. RESULTS Eight randomized controlled trials involving 482 patients were included. Seven studies reported clinical remission, and three reported endoscopic remission. Compared to the placebo group, adjunctive curcumin therapy significantly improved clinical remission (RR=2.33, 95 % CI: 1.25 to 4.34; P = 0.008; I²=80 %). Although endoscopic remission showed an increasing trend, it was not statistically significant (RR=4.17, 95 % CI: 0.63 to 27.71; P = 0.14; I²=80 %). Significant improvements were also observed in clinical improvement (RR=1.93, 95 % CI: 1.10 to 3.36; P = 0.02; I²=56 %) and endoscopic improvement (RR=1.76, 95 % CI: 1.12 to 2.77; P = 0.01; I²=62 %) in the curcumin group. No serious adverse events were reported. Subgroup analysis indicated a positive correlation between treatment efficacy and dosage, with no significant impact of administration method or follow-up duration on the pooled results or heterogeneity. CONCLUSION Curcumin as an adjunctive treatment shows promise in improving clinical and endoscopic outcomes in UC patients without significant adverse effects. However, due to the limited number of studies and substantial heterogeneity, further large-scale randomized controlled trials are necessary to confirm these findings.
Collapse
Affiliation(s)
- Zhao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China.
| | - Duo Li
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Na Wu
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Xiao-Yuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Gai-Xia Sun
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Hui-Bin Gao
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Hai-Xia Li
- Department of Gastroenterology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, China
| |
Collapse
|
42
|
Marzano V, Levi Mortera S, Putignani L. Insights on Wet and Dry Workflows for Human Gut Metaproteomics. Proteomics 2024:e202400242. [PMID: 39740098 DOI: 10.1002/pmic.202400242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
The human gut microbiota (GM) is a community of microorganisms that resides in the gastrointestinal (GI) tract. Recognized as a critical element of human health, the functions of the GM extend beyond GI well-being to influence overall systemic health and susceptibility to disease. Among the other omic sciences, metaproteomics highlights additional facets that make it a highly valuable discipline in the study of GM. Indeed, it allows the protein inventory of complex microbial communities. Proteins with associated taxonomic membership and function are identified and quantified from their constituent peptides by liquid chromatography coupled to mass spectrometry analyses and by querying specific databases (DBs). The aim of this review was to compile comprehensive information on metaproteomic studies of the human GM, with a focus on the bacterial component, to assist newcomers in understanding the methods and types of research conducted in this field. The review outlines key steps in a metaproteomic-based study, such as protein extraction, DB selection, and bioinformatic workflow. The importance of standardization is emphasized. In addition, a list of previously published studies is provided as hints for researchers interested in investigating the role of GM in health and disease states.
Collapse
Affiliation(s)
- Valeria Marzano
- Research Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefano Levi Mortera
- Research Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Research Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
43
|
Monshizadeh M, Hong Y, Ye Y. Multitask knowledge-primed neural network for predicting missing metadata and host phenotype based on human microbiome. BIOINFORMATICS ADVANCES 2024; 5:vbae203. [PMID: 39735577 PMCID: PMC11676323 DOI: 10.1093/bioadv/vbae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/27/2024] [Accepted: 12/11/2024] [Indexed: 12/31/2024]
Abstract
Motivation Microbial signatures in the human microbiome are closely associated with various human diseases, driving the development of machine learning models for microbiome-based disease prediction. Despite progress, challenges remain in enhancing prediction accuracy, generalizability, and interpretability. Confounding factors, such as host's gender, age, and body mass index, significantly influence the human microbiome, complicating microbiome-based predictions. Results To address these challenges, we developed MicroKPNN-MT, a unified model for predicting human phenotype based on microbiome data, as well as additional metadata like age and gender. This model builds on our earlier MicroKPNN framework, which incorporates prior knowledge of microbial species into neural networks to enhance prediction accuracy and interpretability. In MicroKPNN-MT, metadata, when available, serves as additional input features for prediction. Otherwise, the model predicts metadata from microbiome data using additional decoders. We applied MicroKPNN-MT to microbiome data collected in mBodyMap, covering healthy individuals and 25 different diseases, and demonstrated its potential as a predictive tool for multiple diseases, which at the same time provided predictions for the missing metadata. Our results showed that incorporating real or predicted metadata helped improve the accuracy of disease predictions, and more importantly, helped improve the generalizability of the predictive models. Availability and implementation https://github.com/mgtools/MicroKPNN-MT.
Collapse
Affiliation(s)
- Mahsa Monshizadeh
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| | - Yuhui Hong
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| | - Yuzhen Ye
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| |
Collapse
|
44
|
Kreimeyer H, Gonzalez CG, Fondevila MF, Hsu CL, Hartmann P, Zhang X, Stärkel P, Bosques-Padilla F, Verna EC, Abraldes JG, Brown RS, Vargas V, Altamirano J, Caballería J, Shawcross DL, Louvet A, Lucey MR, Mathurin P, Garcia-Tsao G, Bataller R, Investigators A, Gonzalez DJ, Schnabl B. Faecal proteomics links neutrophil degranulation with mortality in patients with alcohol-associated hepatitis. Gut 2024; 74:103-115. [PMID: 39033024 PMCID: PMC11631684 DOI: 10.1136/gutjnl-2024-332730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE Patients with alcohol-associated hepatitis (AH) have a high mortality. Alcohol exacerbates liver damage by inducing gut dysbiosis, bacterial translocation and inflammation, which is characterised by increased numbers of circulating and hepatic neutrophils. DESIGN In this study, we performed tandem mass tag (TMT) proteomics to analyse proteins in the faeces of controls (n=19), patients with alcohol-use disorder (AUD; n=20) and AH (n=80) from a multicentre cohort (InTeam). To identify protein groups that are disproportionately represented, we conducted over-representation analysis using Reactome pathway analysis and Gene Ontology to determine the proteins with the most significant impact. A faecal biomarker and its prognostic effect were validated by ELISA in faecal samples from patients with AH (n=70), who were recruited in a second and independent multicentre cohort (AlcHepNet). RESULT Faecal proteomic profiles were overall significantly different between controls, patients with AUD and AH (principal component analysis p=0.001, dissimilarity index calculated by the method of Bray-Curtis). Proteins that showed notable differences across all three groups and displayed a progressive increase in accordance with the severity of alcohol-associated liver disease were predominantly those located in neutrophil granules. Over-representation and Reactome analyses confirmed that differentially regulated proteins are part of granules in neutrophils and the neutrophil degranulation pathway. Myeloperoxidase (MPO), the marker protein of neutrophil granules, correlates with disease severity and predicts 60-day mortality. Using an independent validation cohort, we confirmed that faecal MPO levels can predict short-term survival at 60 days. CONCLUSIONS We found an increased abundance of faecal proteins linked to neutrophil degranulation in patients with AH, which is predictive of short-term survival and could serve as a prognostic non-invasive marker.
Collapse
Affiliation(s)
- Henriette Kreimeyer
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Carlos G Gonzalez
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Marcos F Fondevila
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Cynthia L Hsu
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Divison of Gastroenterology, Hepatology and Nutrition, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Xinlian Zhang
- Division of Biostatistics and Bioinformatics, Herbert Wertehim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, USA
| | - Peter Stärkel
- Department of Hepatology and Gastroenterology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Francisco Bosques-Padilla
- Hospital Universitario, Departamento de Gastroenterología, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Elizabeth C Verna
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Juan G Abraldes
- Division of Gastroenterology (Liver Unit), University of Alberta, Edmonton, Alberta, Canada
| | - Robert S Brown
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA
| | - Victor Vargas
- Liver Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Jose Altamirano
- Liver Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Caballería
- Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Liver Unit, Hospital Clinic, Barcelona, Catalunya, Spain
| | - Debbie L Shawcross
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Alexandre Louvet
- Service des Maladies de L'appareil Digestif et Unité INFINITE 1286, Hôpital Huriez, Lille, France
| | - Michael R Lucey
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Philippe Mathurin
- Service des Maladies de L'appareil Digestif et Unité INFINITE 1286, Hôpital Huriez, Lille, France
| | - Guadalupe Garcia-Tsao
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- Section of Digestive Diseases, VA-CT Healthcare System, West Haven, CT, USA
| | - Ramón Bataller
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - David J Gonzalez
- Department of Pharmacology, University of California San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
45
|
Cui D, Li M, Liu M, Shen M, Liu Y, Wang K, Tang H, Lu X, Li S, Yao J, Zhang G. Synergistic effect and mechanism of monoacylglycerol lipase inhibitor and Icaritin in the treatment of ulcerative colitis. Int Immunopharmacol 2024; 142:113155. [PMID: 39276456 DOI: 10.1016/j.intimp.2024.113155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Ulcerative colitis (UC) is a chronic, relapsing nonspecific intestinal inflammatory disease. It is difficult for a single drug to treat UC effectively and maintain long-term efficacy. There is an urgent need to find new drugs and treatment strategies. MAGL11 is a new kind of single acylglycerol lipase (MAGL) inhibitor. Icaritin (Y003) is the major metabolite of icariin in vivo. Several studies have confirmed the role of MAGL inhibitors and icariin in anti-inflammatory and regulation of intestinal stability. Therefore, this study adopted a new strategy of combining MAGL inhibitor with Icaritin to further explore the role and mechanism of drugs in the treatment of UC. Enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin staining (HE), immunohistochemical (IHC) and Western blot were used to detect the synergistic protective effects of MAGL11 and Y003 on intestinal pathological injury, intestinal mucosal permeability and inflammation in UC mice. 16S rDNA sequencing was used to detect the synergistic effect of MAGL11 and Y003 on gut microbiota. The effects of MAGL11 and Y003 combined therapy on serum and fecal metabolism of UC mice were analyzed by untargeted metabolomics. Proteomics method was applied to investigate the molecular mechanisms underlying MAGL11 and Y003 synergy in the treatment of UC. The results showed that MAGL11 and Y003 could synergistically improve the clinical symptoms, reduce intestinal inflammation and pathological damage, and improve intestinal mucosal permeability in UC mice. The mechanism study found that MAGL11 and Y003 could synergistically inhibit Toll-like receptors 4 (TLR4) / Myeloid differentiation primary response gene (Myd88)/Nuclear factor kappa-B (NF-κB) pathway and further regulate gut microbiota imbalance and metabolic disorders to treat UC.
Collapse
Affiliation(s)
- Deyu Cui
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Meng Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Mingfei Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 276005, China
| | - Yifan Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Kun Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Hongguang Tang
- School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiaoyan Lu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 276005, China
| | - Shirong Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 276005, China.
| | - Guimin Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 276005, China.
| |
Collapse
|
46
|
Zhang Y, Schluter J, Zhang L, Cao X, Jenq RR, Feng H, Haines J, Zhang L. Review and revamp of compositional data transformation: A new framework combining proportion conversion and contrast transformation. Comput Struct Biotechnol J 2024; 23:4088-4107. [PMID: 39624165 PMCID: PMC11609487 DOI: 10.1016/j.csbj.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 01/03/2025] Open
Abstract
Due to the development of next-generation sequencing technology and an increased appreciation of their role in modulating host immunity and their potential as therapeutic agents, the human microbiome has emerged as a key area of interest in various biological investigations of human health and disease. However, microbiome data present a number of statistical challenges not addressed by existing methods, such as the varying sequencing depth, the compositionality, and zero inflation. Solutions like scaling and transformation methods help to mitigate heterogeneity and release constraints, but often introduce biases and yield inconsistent results on the same data. To address these issues, we conduct a systematic review of compositional data transformation, with a particular focus on the connection and distinction of existing techniques. Additionally, we create a new framework that enables the development of new transformations by combining proportion conversion with contrast transformations. This framework includes well-known methods such as Additive Log Ratio (ALR) and Centered Log Ratio (CLR) as special cases. Using this framework, we develop two novel transformations-Centered Arcsine Contrast (CAC) and Additive Arcsine Contrast (AAC)-which show enhanced performance in scenarios with high zero-inflation. Moreover, our findings suggest that ALR and CLR transformations are more effective when zero values are less prevalent. This comprehensive review and the innovative framework provide microbiome researchers with a significant direction to enhance data transformation procedures and improve analytical outcomes.
Collapse
Affiliation(s)
- Yiqian Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, 2109 Adelbert Rd, Cleveland, 44106, OH, USA
- Department of Statistics, University of Illinois Urbana-Champaign, 605 E. Springfield Ave., Champaign, 61820, IL, USA
| | - Jonas Schluter
- Institute for Systems Genetics, Department of Microbiology, New York University Grossman School of Medicine, 435 East 30th Street, New York, 10016, NY, USA
| | - Lijun Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, 2109 Adelbert Rd, Cleveland, 44106, OH, USA
| | - Xuan Cao
- Division of Statistics and Data Science, Department of Mathematical Sciences, University of Cincinnati, 2815 Commons Way, Cincinnati, 45219, OH, USA
| | - Robert R. Jenq
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, 1500 East Duarte Road, Duarte, 91010, CA, USA
| | - Hao Feng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, 2109 Adelbert Rd, Cleveland, 44106, OH, USA
| | - Jonathan Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, 2109 Adelbert Rd, Cleveland, 44106, OH, USA
| | - Liangliang Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, 2109 Adelbert Rd, Cleveland, 44106, OH, USA
- Case Comprehensive Cancer Center, 2103 Cornell Road, Cleveland, 44106, OH, USA
| |
Collapse
|
47
|
Hertz S, Anderson JM, Nielsen HL, Schachtschneider C, McCauley KE, Özçam M, Larsen L, Lynch SV, Nielsen H. Fecal microbiota is associated with extraintestinal manifestations in inflammatory bowel disease. Ann Med 2024; 56:2338244. [PMID: 38648495 PMCID: PMC11036898 DOI: 10.1080/07853890.2024.2338244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/15/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION A large proportion of patients with inflammatory bowel disease (IBD) experience IBD-related inflammatory conditions outside of the gastrointestinal tract, termed extraintestinal manifestations (EIMs) which further decreases quality of life and, in extreme cases, can be life threatening. The pathogenesis of EIMs remains unknown, and although gut microbiota alterations are a well-known characteristic of patients with IBD, its relationship with EIMs remains sparsely investigated. This study aimed to compare the gut microbiota of patients with IBD with and without EIMs. METHODS A total of 131 Danish patients with IBD were included in the study, of whom 86 had a history of EIMs (IBD-EIM) and 45 did not (IBD-C). Stool samples underwent 16S rRNA sequencing. Amplicon sequence variants (ASVs) were mapped to the Silva database. Diversity indices and distance matrices were compared between IBD-EIM and IBD-C. Differentially abundant ASVs were identified using a custom multiple model statistical analysis approach, and modules of co-associated bacteria were identified using sparse correlations for compositional data (SparCC) and related to patient EIM status. RESULTS Patients with IBD and EIMs exhibited increased disease activity, body mass index, increased fecal calprotectin levels and circulating monocytes and neutrophils. Microbiologically, IBD-EIM exhibited lower fecal microbial diversity than IBD-C (Mann-Whitney's test, p = .01) and distinct fecal microbiota composition (permutational multivariate analysis of variance; weighted UniFrac, R2 = 0.018, p = .01). A total of 26 ASVs exhibited differential relative abundances between IBD-EIM and IBD-C, including decreased Agathobacter and Blautia and increased Eggerthella lenta in the IBD-EIM group. SparCC analysis identified 27 bacterial co-association modules, three of which were negatively related to EIM (logistic regression, p < .05) and included important health-associated bacteria, such as Agathobacter and Faecalibacterium. CONCLUSIONS The fecal microbiota in IBD patients with EIMs is distinct from that in IBD patients without EIM and could be important for EIM pathogenesis.
Collapse
Affiliation(s)
- Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline Moltzau Anderson
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Hans Linde Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Claire Schachtschneider
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Kathryn E. McCauley
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Mustafa Özçam
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Lone Larsen
- Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Center for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Aalborg University, Aalborg, Denmark
| | - Susan V. Lynch
- Department of Medicine, Division of Gastroenterology, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
48
|
Wang H, Cai Y, Wu W, Zhang M, Dai Y, Wang Q. Exploring the role of gut microbiome in autoimmune diseases: A comprehensive review. Autoimmun Rev 2024; 23:103654. [PMID: 39384149 DOI: 10.1016/j.autrev.2024.103654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/04/2024] [Accepted: 09/28/2024] [Indexed: 10/11/2024]
Abstract
As the industrialized society advances, there has been a gradual increase in the prevalence of autoimmune disorders. A probe into the fundamental causes has disclosed several factors in modern society that have an influence on the gut microbiome. These dramatic shifts in the gut microbiome are likely to be one of the reasons for the disarray in the immune system, and the relationship between the immune system and the gut microbiome emerging as a perennial hot topic of research. This review enumerates the findings from sequencing studies of gut microbiota on seven autoimmune diseases (ADs): Rheumatoid Arthritis (RA), Systemic Lupus Erythematosus (SLE), Ankylosing Spondylitis (AS), Systemic Sclerosis (SSc), Sjögren's Syndrome (SjS), Juvenile Idiopathic Arthritis (JIA), and Behçet's Disease (BD). It aims to identify commonalities in changes in the gut microbiome within the autoimmune disease cohort and characteristics specific to each disease. The dysregulation of the gut microbiome involves a disruption of the internal balance and the balance between the external environment and the host. This dysregulation impacts the host's immune system, potentially playing a role in the development of ADs. Damage to the gut epithelial barrier allows potential pathogens to translocate to the mucosal layer, contacting epithelial cells, disrupting tight junctions, and being recognized by antigen-presenting cells, which triggers an immune response. Primed T-cells assist B-cells in producing antibodies against pathogens; if antigen mimicry occurs, an immune response is generated in extraintestinal organs during immune cell circulation, clinically manifesting as ADs. However, current research is limited; advancements in sequencing technology, large-scale cohort studies, and fecal microbiota transplantation (FMT) research are expected to propel this field to new peaks.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yueshu Cai
- Department of Urology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Wenqi Wu
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Miaomiao Zhang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Yong Dai
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China; The Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China.
| |
Collapse
|
49
|
Xu L, Wang Y, Yan D, Li M, Qiao L, Chen Z, Wu M, Zhong G. Albumin binding domain fusion improved the therapeutic efficacy of Inhibitor of Differentiation-2 protein in colitis mice. Life Sci 2024; 359:123237. [PMID: 39532259 DOI: 10.1016/j.lfs.2024.123237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
AIMS The human Inhibitor of Differentiation-2 (hID2) protein is a promising candidate for the treatment of colitis. However, its relatively low molecular weight limits its clinical application. To extend the therapeutic half-life, an albumin-binding domain (ABD), known for its high affinity for human serum albumin (HSA), was fused to hID2, resulting in a recombinant ABD-hID2. The anti-colitis bioactivity of ABD-hID2 than that of hID2 was evaluated in this study. MAIN METHODS Western blotting, size-exclusion high-performance chromatography, HSA binding assay, and pharmacokinetic studies were used to characterise ABD-hID2, which was induced by dextran sulfate sodium salt (DSS), Citrobacter rodentium (CR), and ABD-hID2 and hID2. The Disease Activity Index, histological pathologies, inflammatory response, Alcian blue or tuft cell staining, and tight junction proteins were determined. Alterations in the intestinal microbiota after ABD-hID2 treatment were analysed via 16S rRNA gene sequencing. KEY FINDINGS Compared with hID2, ABD-hID2 exhibited a decreased dimer complex, bound to HSA with high affinity, and demonstrated an extended blood retention time in vivo. Consequently, ABD-hID2 exhibited increased therapeutic efficacy in both DSS- and CR-induced colitis mouse models, as evidenced by the alleviation of colitis symptoms, preservation of goblet and tuft cell functions, restoration of the intestinal mucus barrier, and suppression of abnormal immune-inflammatory responses. Additionally, the modulation of the gut microbiota may play a role in the protective effects of ABD-hID2 in mice with CR-induced ulcerative colitis. SIGNIFICANCE ABD-hID2 enhances the bioactivity of hID2 and has the potential for further development as a treatment for colitis.
Collapse
Affiliation(s)
- Lingyun Xu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, Hunan, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yuxin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Dong Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Min Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Lin Qiao
- Department of Medical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Zhiguo Chen
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, Hunan, China
| | - Minna Wu
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, Hunan, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Genshen Zhong
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, Hunan, China.
| |
Collapse
|
50
|
Probul N, Huang Z, Saak CC, Baumbach J, List M. AI in microbiome-related healthcare. Microb Biotechnol 2024; 17:e70027. [PMID: 39487766 PMCID: PMC11530995 DOI: 10.1111/1751-7915.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 11/04/2024] Open
Abstract
Artificial intelligence (AI) has the potential to transform clinical practice and healthcare. Following impressive advancements in fields such as computer vision and medical imaging, AI is poised to drive changes in microbiome-based healthcare while facing challenges specific to the field. This review describes the state-of-the-art use of AI in microbiome-related healthcare. It points out limitations across topics such as data handling, AI modelling and safeguarding patient privacy. Furthermore, we indicate how these current shortcomings could be overcome in the future and discuss the influence and opportunities of increasingly complex data on microbiome-based healthcare.
Collapse
Affiliation(s)
- Niklas Probul
- Institute for Computational Systems BiologyUniversity of HamburgHamburgGermany
| | - Zihua Huang
- Data Science in Systems Biology, TUM School of Life SciencesTechnical University of MunichFreisingGermany
| | | | - Jan Baumbach
- Institute for Computational Systems BiologyUniversity of HamburgHamburgGermany
- Computational Biomedicine Lab, Department of Mathematics and Computer ScienceUniversity of Southern DenmarkOdenseDenmark
| | - Markus List
- Data Science in Systems Biology, TUM School of Life SciencesTechnical University of MunichFreisingGermany
- Munich Data Science InstituteTechnical University of MunichGarchingGermany
| |
Collapse
|