1
|
Barnett D, Thijs C, Mommers M, Endika M, Klostermann C, Schols H, Smidt H, Nauta A, Arts I, Penders J. Why do babies cry? Exploring the role of the gut microbiota in infantile colic, constipation, and cramps in the KOALA birth cohort study. Gut Microbes 2025; 17:2485326. [PMID: 40159147 PMCID: PMC11959906 DOI: 10.1080/19490976.2025.2485326] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Gastrointestinal symptoms are common during infancy, including infantile colic. Colic can be loosely defined as prolonged and recurrent crying without obvious cause. The cause indeed remains unclear despite much research. Results on infant nutrition are inconclusive, but prior work has linked maternal mental health to infant crying. Recently, several small studies have described associations between gut microbiota and colic. We used a larger cohort to examine the role of the microbiota in infant gastrointestinal health, while also accounting for other biopsychosocial factors. Using fecal 16S rRNA gene amplicon sequencing data from 1,012 infants in the KOALA birth cohort, we examined associations between the 1-month gut microbiota and parent-reported functional gastrointestinal symptoms throughout infancy, including colic, constipation, and cramps. These analyses were adjusted for biopsychosocial factors that were associated with symptoms in a broader analysis involving 2,665 participants. In 257 infants, we also explored associations between breastmilk human milk oligosaccharides (HMOs) and gastrointestinal symptoms. Higher relative abundance of Staphylococcus at one month was associated with less constipation in the first three months of life. Conversely, Ruminococcus gnavus group abundance was associated with more colicky symptoms, particularly between four and seven months. Breastmilk concentrations of the HMOs lacto-N-hexaose (LNH) and lacto-N-neohexaose (LNnH) were associated with less constipation in the first three months. Our results support the conclusion that gut microbiota are relevant in infantile colic and constipation. However more work is needed to elucidate the underlying mechanisms, and explore their interplay with other relevant biopsychosocial factors such as maternal mental health.
Collapse
Affiliation(s)
- David Barnett
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Carel Thijs
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Monique Mommers
- CAPHRI Care and Public Health Research Institute, Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Martha Endika
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Cynthia Klostermann
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Henk Schols
- Department of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Arjen Nauta
- FrieslandCampina Ingredients, FrieslandCampina, Amersfoort, Netherlands
| | - Ilja Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - John Penders
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Medical Microbiology, Infectious Diseases and Infection Prevention, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
2
|
Maldarelli GA, Metz M, Oguntunmibi S, Tran N, Xiang G, Lukin D, Scherl EJ, Longman RS. IgG-seq identifies immune-reactive enteric bacteria in Crohn's disease with spondyloarthritis. Gut Microbes 2025; 17:2464221. [PMID: 39949039 PMCID: PMC11834481 DOI: 10.1080/19490976.2025.2464221] [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: 11/11/2024] [Revised: 01/08/2025] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
Joint inflammation is the most common extraintestinal manifestation of Crohn's disease (CD). Although alterations in the enteric microbiota are described in CD with spondyloarthritis (CD-SpA), it is not known whether distinct taxa serve as markers for clinical subtypes of axial (AxSpA) or peripheral SpA (pSpA) in CD. Moreover, it is not yet known whether these taxa generate a specific systemic IgG response. Here, we sequenced the fecal microbiome from 106 individuals (44 CD, 39 CD-SpA, 14 CD-AxSpA, and 9 healthy controls [HC]). This unique cohort revealed distinct taxonomic compositions of CD and CD-SpA compared to HC and demonstrates that the composition of the CD-AxSpA microbiome is distinct from that of CD-pSpA. Using autologous serum, we identified enteric bacteria recognized by serum IgG and demonstrate differences in the IgG coating index of specific bacterial genera associated with CD-SpA. The IgG coating index of Mediterraneibacter gnavus differentiated patients with CD-pSpA and is positively associated with joint disease activity. This work illustrates divergent microbiome compositions in CD-SpA subtypes, as well as the recognition of distinct enteric bacteria by serum IgG with the potential to serve as a marker of joint inflammation in CD.
Collapse
Affiliation(s)
- Grace A. Maldarelli
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Maeva Metz
- Department of Immunology and Microbial Pathogenesis, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Seun Oguntunmibi
- Department of Immunology and Microbial Pathogenesis, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Nancy Tran
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Grace Xiang
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Dana Lukin
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Ellen J. Scherl
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Randy S. Longman
- Department of Immunology and Microbial Pathogenesis, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Center for Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3
|
Geng P, Zhao N, Zhou Y, Harris RS, Ge Y. Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice. Gut Microbes 2025; 17:2455503. [PMID: 39841201 DOI: 10.1080/19490976.2025.2455503] [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: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The probiotic impact of microbes on host metabolism and health depends on both host genetics and bacterial genomic variation. Faecalibacterium prausnitzii is the predominant human gut commensal emerging as a next-generation probiotic. Although this bacterium exhibits substantial intraspecies diversity, it is unclear whether genetically distinct F. prausnitzii strains might lead to functional differences in the gut microbiome. Here, we isolated and characterized a novel F. prausnitzii strain (UT1) that belongs to the most prevalent but underappreciated phylogenetic clade in the global human population. Genome analysis showed that this butyrate-producing isolate carries multiple putative mobile genetic elements, a clade-specific defense system, and a range of carbohydrate catabolic enzymes. Multiomic approaches were used to profile the impact of UT1 on the gut microbiome and associated metabolic activity of C57BL/6 mice at homeostasis. Both 16S rRNA and metagenomic sequencing demonstrated that oral administration of UT1 resulted in profound microbial compositional changes including a significant enrichment of Lactobacillus, Bifidobacterium, and Turicibacter. Functional profiling of the fecal metagenomes revealed a markedly higher abundance of carbohydrate-active enzymes (CAZymes) in UT1-gavaged mice. Accordingly, UT1-conditioned microbiota possessed the elevated capability of utilizing starch in vitro and exhibited a lower availability of microbiota-accessible carbohydrates in the gut. Further analysis uncovered a functional network wherein UT1 reduced the abundance of mucin-degrading CAZymes and microbes, which correlated with a concomitant reduction of fecal mucin glycans. Collectively, our results reveal a crucial role of UT1 in facilitating the carbohydrate metabolism of the gut microbiome and expand our understanding of the genetic and phenotypic diversity of F. prausnitzii.
Collapse
Affiliation(s)
- Peiling Geng
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ni Zhao
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yong Ge
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
4
|
Zhang D, Wang Q, Li D, Chen S, Chen J, Zhu X, Bai F. Gut microbiome composition and metabolic activity in metabolic-associated fatty liver disease. Virulence 2025; 16:2482158. [PMID: 40122128 PMCID: PMC11959907 DOI: 10.1080/21505594.2025.2482158] [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] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/24/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Metabolic Associated Fatty Liver Disease (MAFLD) impacts approximately 25% of the global population. Between April 2023 and July 2023, 60 patients with MAFLD, along with 60 age, ethnicity, and sex-matched healthy controls (HCs), were enrolled from the Inner Mongolia Autonomous Region, China. Analysis of gut microbiota composition and plasma metabolic profiles was conducted using metagenome sequencing and LC-MS. LEfSe analysis identified five pivotal species: Eubacterium rectale, Dialister invisus, Pseudoruminococcus massiliensis, GGB3278 SGB4328, and Ruminococcaceae bacteria. In subgroup analysis, Eubacterium rectale tended to increase by more than 2 times and more than double in the non-obese MAFLD group, and MAFLD with moderate hepatic steatosis (HS), respectively. Plasma samples identified 172 metabolites mainly composed of fatty acid metabolites such as propionic acid and butyric acid analogues. Ruminococcaceae bacteria have a strong positive correlation with β-alanine, uric acid, and L-valine. Pseudoruminococcus massiliensis has a strong positive correlation with β-alanine. Combinations of phenomics and metabolomics yielded the highest accuracy (AUC = 0.97) in the MAFLD diagnosis. Combinations of phenomics and metagenomics yielded the highest accuracy (AUC = 0.94) in the prediction of the MAFLD HS progress. Increases in Eubacterium rectale and decreases in Dialister invisus seem to be indicative of MAFLD patients. Eubacterium rectale may predict HS degree of MAFLD and play an important role in the development of non-obese MAFLD. Eubacterium rectale can generate more propionic acid and butyric acid analogues to absorb energy and increase lipid synthesis and ultimately cause MAFLD.
Collapse
Affiliation(s)
- Daya Zhang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Qi Wang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Da Li
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Shiju Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Jinrun Chen
- Department of Gastroenterology, Otog Front Banner People 's Hospital, Otog Front Banner, China
| | - Xuli Zhu
- Department of Gastroenterology, Otog Front Banner People 's Hospital, Otog Front Banner, China
| | - Feihu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
5
|
Veseli I, Chen YT, Schechter MS, Vanni C, Fogarty EC, Watson AR, Jabri B, Blekhman R, Willis AD, Yu MK, Fernàndez-Guerra A, Füssel J, Eren AM. Microbes with higher metabolic independence are enriched in human gut microbiomes under stress. eLife 2025; 12:RP89862. [PMID: 40377187 PMCID: PMC12084026 DOI: 10.7554/elife.89862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
A wide variety of human diseases are associated with loss of microbial diversity in the human gut, inspiring a great interest in the diagnostic or therapeutic potential of the microbiota. However, the ecological forces that drive diversity reduction in disease states remain unclear, rendering it difficult to ascertain the role of the microbiota in disease emergence or severity. One hypothesis to explain this phenomenon is that microbial diversity is diminished as disease states select for microbial populations that are more fit to survive environmental stress caused by inflammation or other host factors. Here, we tested this hypothesis on a large scale, by developing a software framework to quantify the enrichment of microbial metabolisms in complex metagenomes as a function of microbial diversity. We applied this framework to over 400 gut metagenomes from individuals who are healthy or diagnosed with inflammatory bowel disease (IBD). We found that high metabolic independence (HMI) is a distinguishing characteristic of microbial communities associated with individuals diagnosed with IBD. A classifier we trained using the normalized copy numbers of 33 HMI-associated metabolic modules not only distinguished states of health vs IBD, but also tracked the recovery of the gut microbiome following antibiotic treatment, suggesting that HMI is a hallmark of microbial communities in stressed gut environments.
Collapse
Affiliation(s)
- Iva Veseli
- Biophysical Sciences Program, The University of ChicagoChicagoUnited States
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Yiqun T Chen
- Data Science Institute and Department of Biomedical Data Science, Stanford UniversityStanfordUnited States
| | - Matthew S Schechter
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Chiara Vanni
- MARUM Center for Marine Environmental Sciences, University of BremenBremenGermany
| | - Emily C Fogarty
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Andrea R Watson
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Bana Jabri
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Ran Blekhman
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Amy D Willis
- Department of Biostatistics, University of WashingtonSeattleUnited States
| | - Michael K Yu
- Toyota Technological Institute at ChicagoChicagoUnited States
| | - Antonio Fernàndez-Guerra
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of CopenhagenCopenhagenDenmark
| | - Jessika Füssel
- Department of Medicine, The University of ChicagoChicagoUnited States
- Institute for Chemistry and Biology of the Marine Environment, University of OldenburgOldenburgGermany
| | - A Murat Eren
- Department of Medicine, The University of ChicagoChicagoUnited States
- Institute for Chemistry and Biology of the Marine Environment, University of OldenburgOldenburgGermany
- Marine ‘Omics Bridging Group, Max Planck Institute for Marine MicrobiologyBremenGermany
- Helmholtz Institute for Functional Marine BiodiversityOldenburgGermany
- Alfred Wegener Institute for Polar and Marine ResearchBremerhavenGermany
| |
Collapse
|
6
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025; 32:653-677. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [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: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
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
|
8
|
Gong W, Jin G, Bao Y, Liu Q, Ni M, Wang J, Mao S, Zhang Y, Zheng Z. Characteristics and potential diagnostic value of gut microbiota in ovarian tumor patients. Sci Rep 2025; 15:16504. [PMID: 40360697 PMCID: PMC12075642 DOI: 10.1038/s41598-025-99912-x] [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: 12/04/2024] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The gut microbiota is closely related to the occurrence and development of cancer. However, the characteristics of gut microbiota associated with ovarian tumors remain elusive. In this study, fecal samples were collected from healthy control (HC) group and patients with ovarian tumor (OT) or with other benign tumor (OBT) for 16s rRNA sequencing to determine differential flora in gut microbiota. The composition of gut microbiota in the OT group, including bacterial abundance and diversity, was significantly different form HC and OBT groups. In the OT group, Escherichia_Shigella was markedly higher than in the HC group, while Coprococcus, Fusicatenibacter, Butyricicoccus and Oscillibacter were significantly lower than in HCs. The abundance of Fusicatenibacter, Butyricicoccus, Coprococcus Parasutterella, and Anaerotruncus in the OBT group was distinctly higher than that in the OT group, while the Lachnospiracae_ND3007_group was significantly lower. In addition, in OT patients, ovarian cancer (OC) and benign ovarian tumor (BOT) patients also showed a unique composition of gut microbiota. The random forest model was designed using different bacteria. Compared with HCs, area under curve (AUC) values for BOT and OC groups were 0.77 and 0.86, respectively. These findings suggest that some gut microbiota such as Escherichia_Shigella show a certain ability to distinguish between healthy individuals and patients with OT.
Collapse
Affiliation(s)
- Wangang Gong
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Gulei Jin
- Hangzhou Guhe Information and Technology Company, Hangzhou, Zhejiang, China
| | - Yejiang Bao
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Qi Liu
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Maowei Ni
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Junjian Wang
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Shuyu Mao
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yingli Zhang
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Zhiguo Zheng
- Zhejiang Cancer Hospital, Banshan Road, Hangzhou, 310022, Zhejiang, China.
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
9
|
Jacobsen GE, Gonzalez EE, Mendygral P, Faust KM, Hazime H, Fernandez I, Santander AM, Quintero MA, Jiang C, Damas OM, Deshpande AR, Kerman DH, Proksell S, Sendzischew Shane M, Sussman DA, Ghaddar B, Cickovsk T, Abreu MT. Deep Sequencing of Crohn's Disease Lamina Propria Phagocytes Identifies Pathobionts and Correlates With Pro-Inflammatory Gene Expression. Inflamm Bowel Dis 2025; 31:1203-1219. [PMID: 39951038 PMCID: PMC12069990 DOI: 10.1093/ibd/izae316] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Indexed: 05/14/2025]
Abstract
BACKGROUND Crohn's disease (CD) is characterized by an inflammatory response to gut microbiota. Macrophages and dendritic cells play an active role in CD inflammation. Specific microbiota have been implicated in the pathogenesis of ileal CD. We investigated the phagocyte-associated microbiome using an unbiased sequencing approach to identify potential pathobionts and elucidate the host response to these microbes. METHODS We collected ileal and colonic mucosal biopsies from CD patients and controls without inflammatory bowel disease (IBD), isolated lamina propria phagocytes (CD11b+ cells), and performed deep RNA sequencing (n = 37). Reads were mapped to the human genome for host gene expression analysis and a prokaryotic database for microbiome taxonomic and metatranscriptomic profiling. Results were confirmed in a second IBD cohort (n = 17). Lysed lamina propria cells were plated for bacterial culturing; isolated colonies underwent whole genome sequencing (n = 11). RESULTS Crohn's disease ileal phagocytes contained higher relative abundances of Escherichia coli, Ruminococcus gnavus, and Enterocloster spp. than those from controls. CD phagocyte-associated microbes had increased expression of lipopolysaccharide (LPS) biosynthesis pathways. Phagocytes with a higher pathobiont burden showed increased expression of pro-inflammatory and antimicrobial genes, including PI3 (antimicrobial peptide) and BPIFB1 (LPS-binding molecule). E. coli isolated from the CD lamina propria had more flagellar motility and antibiotic resistance genes than control-derived strains. CONCLUSIONS Lamina propria resident phagocytes harbor bacterial strains that may act as pathobionts in CD. Our findings shed light on the role of pathobionts and the immune response in CD pathogenesis and suggest new targets for therapies.
Collapse
Affiliation(s)
- Gillian E Jacobsen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eddy E Gonzalez
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Payton Mendygral
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Katerina M Faust
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hajar Hazime
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irina Fernandez
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ana M Santander
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria A Quintero
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Chunsu Jiang
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oriana M Damas
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amar R Deshpande
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David H Kerman
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Siobhan Proksell
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Morgan Sendzischew Shane
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel A Sussman
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bassel Ghaddar
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Trevor Cickovsk
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Maria T Abreu
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Crohn’s and Colitis Center, University of Miami Miller School of Medicine, Miami, FL, USA
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Lin D, Rezaei MJ. Plant polysaccharides and antioxidant benefits for exercise performance and gut health: from molecular pathways to clinic. Mol Cell Biochem 2025; 480:2827-2846. [PMID: 39692997 DOI: 10.1007/s11010-024-05178-8] [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: 09/24/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
In the last three decades, our understanding of how exercise induces oxidative stress has significantly advanced. Plant polysaccharides, such as dietary fibers and resistant starches, have been shown to enhance exercise performance by improving energy metabolism, reducing fatigue, increasing strength and stamina, mitigating oxidative stress post-exercise, facilitating muscle recovery, and aiding in detoxification. Moreover, antioxidants found in plant-based foods play a crucial role in protecting the body against oxidative stress induced by intense physical activity. By scavenging free radicals and reducing oxidative damage, antioxidants can improve exercise endurance, enhance recovery, and support immune function. Furthermore, the interaction between plant polysaccharides and antioxidants in the gut microbiota can lead to synergistic effects on overall health and performance. This review provides a comprehensive overview of the current research on plant polysaccharides and antioxidants in relation to exercise performance and gut health.
Collapse
Affiliation(s)
- Di Lin
- School of Sports, Zhengzhou Shengda University, Zhengzhou, 451191, Henan, China.
| | - Mohammad J Rezaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [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: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
12
|
Godny L, Elial-Fatal S, Arrouasse J, Sharar Fischler T, Reshef L, Kutukov Y, Cohen S, Pfeffer-Gik T, Barkan R, Shakhman S, Friedenberg A, Pauker MH, Rabinowitz KM, Shaham-Barda E, Goren I, Gophna U, Banai Eran H, Ollech JE, Snir Y, Broitman Y, Avni-Biron I, Yanai H, Dotan I. Mechanistic Implications of the Mediterranean Diet in Patients With Newly Diagnosed Crohn's Disease: Multiomic Results From a Prospective Cohort. Gastroenterology 2025; 168:952-964.e2. [PMID: 39814239 DOI: 10.1053/j.gastro.2024.12.031] [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: 06/13/2024] [Revised: 12/25/2024] [Accepted: 12/28/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND & AIMS To decipher the mechanisms underlying the protective role of the Mediterranean diet (MED) in Crohn's disease (CD), we explored the implications of adherence to MED on CD course, inflammatory markers, and microbial and metabolite composition. METHODS Patients with newly diagnosed CD were recruited and followed prospectively. MED adherence was assessed by repeated food frequency questionnaires (FFQs) using a predefined inflammatory bowel disease Mediterranean diet score (IBDMED score), alongside validated MED adherence screeners. Crohn's disease activity index (CDAI), C-reactive protein, fecal calprotectin, and microbial composition (16S-ribosomal RNA sequencing) were assessed each visit. Baseline serum and fecal samples were analyzed for targeted quantitative metabolomics. RESULTS Consecutive patients: 271 (52% men, average age 31 ± 12 years, B1 phenotype 75%). FFQs collected: 636 (range 1-5 FFQs per patient). Adherence to MED was associated with a noncomplicated CD course, and inversely correlated with CDAI, fecal calprotectin, C-reactive protein, and microbial dysbiosis index (all P < .05). Increasing adherence to MED over time correlated with reduced CDAI and inflammatory markers (P < .05). Adherence to MED correlated with a microbial cluster of commensals and short-chain fatty acid producers including Faecalibacterium, and with plant metabolites, vitamin derivatives, and amino acids. Conversely, adherence to MED inversely correlated with a cluster of oral genera, Escherichia coli and Ruminococcus gnavus, known CD-associated species, and with tryptophan metabolites, ceramides, and primary bile acids (false discovery rate < 0.2). CONCLUSION Adherence to MED is associated with beneficial clinical outcomes and decreased inflammatory markers. These may be driven by lower levels of primary bile acids and microbial dysbiosis and a beneficial microbial and metabolite composition. Randomized controlled trials are needed to evaluate the role of MED in CD management.
Collapse
Affiliation(s)
- Lihi Godny
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Nutrition Unit, Rabin Medical Center, Petah-Tikva, Israel.
| | | | - Jessica Arrouasse
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Faculty of Medical and Health Sciences, Tel Aviv, Israel
| | - Tali Sharar Fischler
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Leah Reshef
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Yelena Kutukov
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Shaked Cohen
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Tamar Pfeffer-Gik
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Nutrition Unit, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Revital Barkan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Shelly Shakhman
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Nutrition Unit, Rabin Medical Center, Petah-Tikva, Israel
| | - Adi Friedenberg
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Maor H Pauker
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Keren M Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Faculty of Medical and Health Sciences, Tel Aviv, Israel
| | - Efrat Shaham-Barda
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Felsenstein Medical Research Center, Faculty of Medical and Health Sciences, Tel Aviv, Israel
| | - Idan Goren
- Division of Gastroenterology, SUNY Upstate Medical University, Syracuse, New York
| | - Uri Gophna
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Hagar Banai Eran
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Jacob E Ollech
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yifat Snir
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yelena Broitman
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Irit Avni-Biron
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
13
|
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
|
14
|
Cui X, Li C, Zhong J, Liu Y, Xiao P, Liu C, Zhao M, Yang W. Gut microbiota - bidirectional modulator: role in inflammatory bowel disease and colorectal cancer. Front Immunol 2025; 16:1523584. [PMID: 40370465 PMCID: PMC12075242 DOI: 10.3389/fimmu.2025.1523584] [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] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
The gut microbiota is a diverse ecosystem that significantly impacts human health and disease. This article focuses on how the gut microbiota interacts with inflammatory bowel diseases and colorectal tumors, especially through immune regulation. The gut microbiota plays a role in immune system development and regulation, while the body's immune status can also affect the composition of the microbiota. These microorganisms exert pathogenic effects or correct disease states in gastrointestinal diseases through the actions of toxins and secretions, inhibition of immune responses, DNA damage, regulation of gene expression, and protein synthesis. The microbiota and its metabolites are essential in the development and progression of inflammatory bowel diseases and colorectal tumors. The complexity and bidirectionality of this connection with tumors and inflammation might render it a new therapeutic target. Hence, we explore therapeutic strategies for the gut microbiota, highlighting the potential of probiotics and fecal microbiota transplantation to restore or adjust the microbial community. Additionally, we address the challenges and future research directions in this area concerning inflammatory bowel diseases and colorectal tumors.
Collapse
Affiliation(s)
- Xilun Cui
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jing Zhong
- Department of Medical Imaging, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengwei Zhao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
15
|
Ma X, Duan C, Wang X, Tao Y, Yang L, Teng Y, Pan Y, Zhang M, Xu J, Sheng J, Wang X, Jin P. Human gut microbiota adaptation to high-altitude exposure: longitudinal analysis over acute and prolonged periods. Microbiol Spectr 2025:e0291624. [PMID: 40257273 DOI: 10.1128/spectrum.02916-24] [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: 12/04/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
This study investigated the longitudinal effects of acute (7-day) and prolonged (3-month) high-altitude exposure on gut microbiota in healthy adult males, addressing the limited data available in human populations. A cohort of 406 healthy adult males was followed, and fecal samples were collected at three time points: baseline at 800 m (406 samples), 7 days after ascending to 4,500 m (406 samples), and 2 weeks post-return to 800 m following 3 months at high altitude (186 samples). High-throughput 16S ribosomal DNA sequencing was employed to analyze microbiota composition and diversity. Results revealed significant changes in alpha- and beta-diversity, with acute high-altitude exposure inducing more pronounced effects compared to prolonged exposure. Specifically, acute exposure increased opportunistic pathogens (Ruminococcus and Oscillibacter) but decreased beneficial short-chain fatty acid producers (Faecalibacterium and Bifidobacterium). Notably, these changes in microbiota persisted even after returning to low altitude, indicating long-term remodeling. Functional analyses revealed substantial changes in metabolic pathways, suggesting microbiota-driven adaptations to energy utilization under high-altitude hypoxic conditions. In summary, acute high-altitude exposure caused dramatic changes in gut microbiota, while prolonged exposure led to structural and functional reshaping. These findings enhance our understanding of how high-altitude environments reshape gut microbiota. IMPORTANCE This study is the first to investigate the impact of high-altitude exposure on gut microbiota adaptation in a large-scale longitudinal cohort. It seeks to enhance understanding of how high-altitude environments reshape gut microbiota. Acute exposure to high altitude significantly affected both α-diversity and β-diversity of gut microbiota, with acute exposure causing more pronounced changes than prolonged adaptation, indicating temporary disruptions in microbial communities. Notable shifts in microbial abundance were observed, including increased levels of genera linked to hypoxic stress (e.g., Gemmiger, Ruminococcus, and Parabacteroides) and decreased levels of beneficial bacteria (e.g., Faecalibacterium, Roseburia, and Bifidobacterium), suggesting possible adverse health effects. Functional analysis indicated changes in metabolism-related pathways post-exposure, supporting the idea that high-altitude adaptations involve metabolic adjustments for energy management. These findings enhance understanding of high-altitude physiology, illustrating the role of gut microbiota in hypoxic health.
Collapse
Affiliation(s)
- Xianzong Ma
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Xiaoying Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yurong Tao
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lang Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yongsheng Teng
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Mingjie Zhang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianqiu Sheng
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng Jin
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
16
|
Lotankar M, Houttu N, Benchraka C, Lahti L, Laitinen K. Links between gut microbiota with specific serum metabolite groups in pregnant women with overweight or obesity. Nutr Metab Cardiovasc Dis 2025:104095. [PMID: 40348632 DOI: 10.1016/j.numecd.2025.104095] [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: 06/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND AIM Gut microbiota may regulate metabolism but is incompletely characterized in pregnancy. Our objective was to investigate the relations using omics techniques. METHODS AND RESULTS In a cross-sectional setting, fecal and serum samples of 361 healthy pregnant women with overweight or obesity were analyzed with a combinatorial approach of metagenomics and targeted NMR-based metabolomics, with statistical and machine learning techniques to identify and analyze the extent to which the gut microbiota composition and predicted functions would be reflected in the serum metabolome. We identified five biclusters, each of which consisted of a set of gut microbial species and serum metabolites with correlated abundance profiles. Two of the biclusters included metabolites that have been linked to the cardiovascular health; one was linked with factors known to increase the risk i.e., various sizes of lipoprotein subclasses (VLDL and LDL), subclasses of relative lipoprotein lipid concentrations (VLDL, IDL, and LDL), apolipoprotein B, and an inflammation marker, glycoprotein acetylation. These metabolites were associated with abundances of species such as, Enterocloster bolteae and Ruminococcus gnavus. The second bicluster included metabolites linked with a reduced cardiovascular risk, such as different sizes of HDL (high-density lipoprotein), subclasses for relative lipoprotein lipid concentrations and mean diameter for HDL particles, and fatty acid ratios. These metabolites were associated with abundances of species, such as Bacteroides cellulosilyticus and Alistipes finegoldii. We did not observe any biclusters between predicted pathways and serum metabolites. CONCLUSION Overall, we identified five biclusters of co-abundant gut bacteria and serum metabolites , of which two were linked to pro-atherogenic and anti-atherogenic properties. TRIAL REGISTRATION www. CLINICALTRIALS Gov: NCT01922791.
Collapse
Affiliation(s)
- Mrunalini Lotankar
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, Finland; Nutrition and Food Research Center, Faculty of Medicine, University of Turku, Turku, Finland
| | - Noora Houttu
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, Finland; Nutrition and Food Research Center, Faculty of Medicine, University of Turku, Turku, Finland
| | - Chouaib Benchraka
- Department of Computing, Faculty of Technology, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, Faculty of Technology, University of Turku, Turku, Finland
| | - Kirsi Laitinen
- Integrative Physiology and Pharmacology Unit, Institute of Biomedicine, Faculty of Medicine, University of Turku, Finland; Nutrition and Food Research Center, Faculty of Medicine, University of Turku, Turku, Finland; Department of Obstetrics and Gynecology, Turku University Hospital, Wellbeing Services County of Southwest Finland, Turku, Finland.
| |
Collapse
|
17
|
Dingding H, Muhammad S, Manzoor I, Ghaffar SA, Alodaini HA, Moubayed NMS, Hatamleh AA, Songxiao X. Subtractive proteomics and reverse-vaccinology approaches for novel drug targets and designing a chimeric vaccine against Ruminococcus gnavus strain RJX1120. Front Immunol 2025; 16:1555741. [PMID: 40297578 PMCID: PMC12034673 DOI: 10.3389/fimmu.2025.1555741] [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: 01/05/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Mediterraneibacter gnavus, also known as Ruminococcus gnavus, is a Gram-positive anaerobic bacterium that resides in the human gut microbiota. Notably, this bacterium plays dual roles in health and disease. On one side it supports nutrient metabolism essential for bodily functions and on the other it contributes to the development of Inflammatory Bowel Disease (IBD) and other gastrointestinal disorders. R. gnavus strain RJX1120 is an encapsulated strain and has been linked to develop IBD. Despite the advances made on its role in gut homeostasis, limited information is available on strain-specific virulence factors, metabolic pathways, and regulatory mechanisms. The study of such aspects is crucial to make microbiota-targeted therapy and understand its implications in host health. A multi-epitope vaccine against R. gnavus strain RJX1120 was designed using reverse vaccinology-based subtractive proteomics approach. Among the 3,219 proteins identified in the R. gnavus strain RJX1120, two critical virulent and antigenic proteins, a Single-stranded DNA-binding protein SSB (A0A2N5PT08) and Cell division ATP-binding protein FtsE (A0A2N5NK05) were screened and identified as potential targets. The predicted B-cell and T-cell epitopes from these proteins were screened for essential immunological properties such as antigenicity, allergenicity, solubility, MHC binding affinity, and toxicity. Epitopes chosen were cross-linked using suitable spacers and an adjuvant to develop a multi-epitope vaccine. Structural refinement of the construct revealed that 95.7% of the amino acid residues were located in favored regions, indicating a high-quality structural model. Molecular docking analysis demonstrated a robust interaction between the vaccine construct and the human Toll-like receptor 4 (TLR4), with a binding energy of -1277.0 kcal/mol. The results of molecular dynamics simulations further confirmed the stability of the vaccine-receptor complex under physiological conditions. In silico cloning of the vaccine construct yielded a GC content of 48% and a Codon Adaptation Index (CAI) value of 1.0, indicating optimal expression in the host system. These results indicate the possibility of the designed vaccine construct as a candidate for the prevention of R. gnavus-associated diseases. However, experimental validation is required to confirm its immunogenicity and protective efficacy.
Collapse
Affiliation(s)
- Hou Dingding
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Sher Muhammad
- Faculty of Agriculture and Veterinary Sciences, Superior University Lahore, Lahore, Pakistan
| | - Irfan Manzoor
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | - Sana Abdul Ghaffar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad, Pakistan
| | | | - Nadine MS. Moubayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Xu Songxiao
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Grønbæk IMB, Halkjær SI, Mollerup S, Hansen EH, Paulsen SJ, Engel S, Theede K, Wilkens R, Boysen T, Petersen AM. The effects of probiotic treatment with Bifidobacterium breve, Bif195 for small intestinal Crohn's disease and the gut microbiome: results from a randomised, double-blind, placebo-controlled trial. Gut Pathog 2025; 17:19. [PMID: 40205497 PMCID: PMC11984114 DOI: 10.1186/s13099-025-00692-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The aetiology of Crohn's disease, a chronic inflammatory bowel disease, is multifactorial and not completely understood. However, the association with gut dysbiosis is well-established, and manipulation of the gut microbiota has gained interest as a treatment strategy. This study aimed to investigate the effects of the probiotic strain Bifidobacterium breve, Bif195™ (Bif195) on intestinal inflammation, symptoms, and the gut microbiome composition in patients with small intestinal Crohn's disease. METHODS This was a randomised, double-blind, placebo-controlled trial. Thirty-three patients with small intestinal Crohn's disease were assigned to eight weeks of treatment with Bif195 or placebo (1:1). The primary outcome was changes in bowel wall thickness measured by intestinal ultrasonography. Other outcomes were changes in symptom severity, quality of life, faecal calprotectin, fatigue, and specific inflammatory parameters on ultrasonography. Changes in the microbiome composition were also examined. RESULTS Bif195 did not affect the bowel wall thickness in the small intestine compared to placebo. Nor did we observe effects on secondary or clinical explorative outcomes. Analysis of the gut microbiome showed that the relative abundance of B. breve rose during the intervention in the Bif195 group, but the result was statistically non-significant. Surprisingly, we observed a clustering of baseline microbiome data into two groups that differed in several aspects including a statistically significant difference in the incidence of previous bowel resections among the participants. Furthermore, changes in symptom scores after eight weeks of intervention were significantly different across the two microbiome groups, with an interaction effect of p = 0.04. CONCLUSIONS Eight weeks of treatment with Bif195 did not affect clinical outcomes for Crohn's disease. However, variations in baseline microbiome data influenced the results. This underscores the importance of assessing baseline microbiome data in intervention studies in Crohn's disease. CLINICALTRIALS gov NCT04842149.
Collapse
Affiliation(s)
- Ida Marie Bruun Grønbæk
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark.
- Department of Clinical Microbiology, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark.
| | - Sofie Ingdam Halkjær
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Sarah Mollerup
- Department of Clinical Microbiology, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Esben Holm Hansen
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Sarah Juel Paulsen
- Department of Clinical Microbiology, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Sara Engel
- Chr. Hansen A/S, Part of Novonesis, Kogle Allé 6, 2970, Hørsholm, Denmark
| | - Klaus Theede
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Rune Wilkens
- Digestive Disease Center, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, 2400, Copenhagen NV, Denmark
| | - Trine Boysen
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
| | - Andreas Munk Petersen
- Gastrounit, Medical Section, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Amager and Hvidovre, Kettegård Alle 36, 2650, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| |
Collapse
|
19
|
Chen Q, Liu F, Zhang G, Qu Q, Chen Y, Li M, Huang Q, Fu H, Zhu X, He Y, Huang X, Zhang X. Progesterone regulates gut microbiota mediating bone marrow mesenchymal stem cell injury in immune thrombocytopenia patients during pregnancy. J Thromb Haemost 2025; 23:1428-1441. [PMID: 39756658 DOI: 10.1016/j.jtha.2024.12.027] [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: 08/25/2024] [Revised: 11/18/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Immune thrombocytopenia during pregnancy (PITP) is the most common cause of platelet reduction in early and mid-pregnancy. However, the pathogenesis of PITP is still unclear. OBJECTIVES To determine the characteristics of bone marrow mesenchymal stem cells (BM-MSCs) in PITP patients and to explore the associations between metabolites, the gut microbiota, and BM-MSCs in PITP. METHODS The characteristics of BM-MSCs were detected through in vitro and in vivo experiments. Nontargeted metabolomics was used to screen metabolites. The features of the gut microbiota were analyzed by 16S rDNA sequencing. PITP and fecal microbiota transplantation (FMT) mouse model were established to explore the associations between metabolites, gut microbiota, and BM-MSCs. RESULTS BM-MSCs from PITP patients had significant senescence and apoptosis, as well as impaired immunoregulatory function. Metabolomic analysis indicated that progesterone was the most significant specific metabolite in PITP patients. In vivo studies showed that progesterone mediated MSC injury. Further analysis of the gut microbiota and FMT experiments revealed that progesterone mediated BM-MSCs injury by regulating the composition of the gut microbiota in PITP. RNA sequencing analysis of BM-MSCs from FMT mice revealed abnormal expression of genes related to cell aging and the NOD-like receptor signaling pathway. CONCLUSION In conclusion, BM-MSCs in the PITP were significantly impaired, which was associated with increased progesterone and changes in the gut microbiota regulated by progesterone. Intervening with the gut microbiota may become a new treatment for PITP.
Collapse
Affiliation(s)
- Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Fengqi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Gaochao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qingyuan Qu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yuxiu Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Menglin Li
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qiusha Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Haixia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiaolu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yun He
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Clinical Research Center for Hematologic Disease, Beijing, China.
| |
Collapse
|
20
|
Cavon J, Basso M, Kadosh KC, Gibbons SM. The human gut microbiome and sleep across adulthood: associations and therapeutic potential. Lett Appl Microbiol 2025; 78:ovaf043. [PMID: 40113228 PMCID: PMC11959190 DOI: 10.1093/lambio/ovaf043] [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: 10/18/2024] [Revised: 02/25/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025]
Abstract
Sleep is an essential homeostatic process that undergoes dynamic changes throughout the lifespan, with distinct life stages predisposed to specific sleep pathologies. Similarly, the gut microbiome also varies with age, with different signatures associated with health and disease in the latest decades of life. Emerging research has shown significant cross-talk between the gut microbiota and the brain through several pathways, suggesting the microbiota may influence sleep, though the specific mechanisms remain to be elucidated. Here, we critically examine the existing literature on the potential impacts of the gut microbiome on sleep and how this relationship varies across adulthood. We suggest that age-related shifts in gut microbiome composition and immune function may, in part, drive age-related changes in sleep. We conclude with an outlook on the therapeutic potential of microbiome-targeted interventions aimed at improving sleep across adulthood, particularly for individuals experiencing high stress or with sleep complaints.
Collapse
Affiliation(s)
- Jacob Cavon
- Institute for Systems Biology, Seattle, WA 98109, United States
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA 98195, United States
| | - Melissa Basso
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7HX, United Kingdom
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom
| | - Kathrin Cohen Kadosh
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7HX, United Kingdom
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA 98109, United States
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA 98195, United States
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, United States
- eScience Institute, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
21
|
Lu J, Huang Y, Yin Y, Tang B. Exploring blood immune cells in the protective effects of gut microbiota on rheumatic heart disease based on Mendelian randomization analysis. Sci Rep 2025; 15:10745. [PMID: 40155605 PMCID: PMC11953348 DOI: 10.1038/s41598-025-92356-3] [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: 10/16/2024] [Accepted: 02/27/2025] [Indexed: 04/01/2025] Open
Abstract
Rheumatic Heart Disease (RHD) remains a significant health burden, particularly in regions with scarce healthcare resources, research on its immunological aspects remains insufficient. This study employed a two-sample Mendelian Randomization approach, utilizing GWAS data from the largest available datasets for gut microbiota and immune cells as exposures, with outcome data for Rheumatic Valve Diseases (RVD) and Rheumatic Heart Disease affecting other parts of the heart (RHD-other) obtained from the FinnGen study. The primary analytical method was the Inverse Variance Weighted (IVW) approach, complemented by heterogeneity analyses and MR-Egger regression to assess horizontal pleiotropy. Furthermore, a two-step mediation analysis was conducted to investigate the potential mediating role of immune cells in the association between gut microbiota and RHD. This study revealed significant inverse associations between gut microbiota abundance and Rheumatic Heart Disease (RHD) risk. Specifically, the gut abundance of genus Blautia was negatively correlated with RHD-other risk (P_IVW: 0.00932, OR [95%CI]: 0.000734[3.22e-06, 0.16937]), and genus Ruminococcaceae UCG005 showed a similar negative association (P_IVW: 0.038, OR [95%CI]: 0.165[0.02994, 0.90811]). Additionally, the proportions of CD4-CD8- T cell %leukocyte and CD4-CD8- T cell %T cell were inversely related to RHD-other risk (P_IVW: 0.02222, OR [95%CI]: 5.08027 [1.26133, 20.46191] and P: 0.01601, OR[95%CI]: 6.55576 [1.4196, 30.27582], respectively). Moreover, IgD on IgD + CD24 + B cells was found to be negatively correlated with RHD-other risk (P_IVW: 0.01867, OR [95%CI]: 2.17171 [1.1380, 4.14443]). The study also highlighted the protective effects of gut microbiota through mediation analyses: Blautia's impact via IgD on IgD + CD24 + B cells showed a mediation proportion of 8.62514%; Ruminococcaceae UCG005's influence via CD4-CD8- T cell %T cell and CD4-CD8- T cell %leukocyte resulted in mediation proportions of 35.25817% and 30.86827%, respectively. Significant inverse associations were observed between gut microbiota abundance and risk of Rheumatic Heart Disease (RHD), with specific findings for Rheumatic Valve Disease (RVD) and RHD affecting other parts of the heart (RHD-other). For RHD-other, higher abundance of Blautia (OR: 0.0007, 95% CI: 3.22e-06 to 0.169, p = 0.009) and Ruminococcaceae UCG005 (OR: 0.165, 95% CI: 0.030 to 0.908, p = 0.038) were associated with lower risk. Additionally, lower proportions of CD4-CD8- T cells (%leukocyte and %T cell) and IgD on IgD + CD24 + B cells were inversely related to RHD-other risk (ORs: 5.08 and 6.56, p = 0.022 and p = 0.016, respectively). For RVD, higher abundance of Candidatus Soleaferrea was protective (OR: 0.670, 95% CI: 0.460 to 0.976, p = 0.037), while higher levels of CD11c on granulocytes were associated with increased risk (OR: 1.310, 95% CI: 1.023 to 1.679, p = 0.032). Mediation analyses indicated that gut microbiota influence RHD risk through distinct immune pathways, with Blautia affecting RHD-other via IgD on B cells (8.62% mediation), Ruminococcaceae UCG005 via CD4-CD8- T cells (%T cell: 35.26%, %leukocyte: 30.87%). Genus Candidatus Soleaferrea affecting RVD through CD11c on granulocyte (15.01% mediation). The study concludes that higher gut abundance of Candidatus Soleaferrea protects against RVD through the mechanism involving CD11c on granulocytes. Additionally, Blautia exerts a protective effect against RHD-other through its influence on IgD on IgD + CD24 + B cells. Similarly, the abundance of genus Ruminococcaceae UCG005 provides protection against RHD-other by influencing CD4-CD8- T cell %T cell and CD4-CD8- T cell %leukocyte.
Collapse
Affiliation(s)
- Juexiu Lu
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yujie Huang
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yangguang Yin
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Biqiong Tang
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| |
Collapse
|
22
|
Neylan CJ, Levin MG, Hartmann K, Beigel K, Khodursky S, DePaolo JS, Abramowitz S, Furth EE, Heuckeroth RO, Damrauer SM, Maguire LH. Genome-wide association meta-analysis identifies 126 novel loci for diverticular disease and implicates connective tissue and colonic motility. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.27.25324777. [PMID: 40196262 PMCID: PMC11974943 DOI: 10.1101/2025.03.27.25324777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Diverticular disease is a common and morbid complex phenotype influenced by both innate and environmental risk factors. We performed the largest genome-wide association study meta-analysis for diverticular disease, identifying 126 novel loci. Employing multiple downstream analytic strategies, including tissue and pathway enrichment, statistical fine-mapping, allele-specific expression, protein quantitative trait loci and drug-target investigations, and linkage disequilibrium score regression, we prioritized causal genes and produced several lines of evidence linking diverticular disease to connective tissue biology and colonic motility. We substantiated these findings by integrating single-cell RNA sequencing data, showing that prioritized diverticular disease-associated genes are enriched for expression in colonic smooth muscle, fibroblasts, and interstitial cells of Cajal. In quantitative analysis of surgical specimens, we found a substantial reduction in the density of elastin present in the sigmoid colon in severe diverticulitis.
Collapse
Affiliation(s)
- Christopher J. Neylan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Michael G. Levin
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Katherine Hartmann
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Katherine Beigel
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Sam Khodursky
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John S. DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sarah Abramowitz
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Robert O. Heuckeroth
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104
- The Children’s Hospital of Philadelphia Research Institute and Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lillias H. Maguire
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
- Division of Colon and Rectal Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
23
|
Lewis N, Villani A, Lagopoulos J. Gut dysbiosis as a driver of neuroinflammation in attention-deficit/hyperactivity disorder: A review of current evidence. Neuroscience 2025; 569:298-321. [PMID: 39848564 DOI: 10.1016/j.neuroscience.2025.01.031] [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: 07/11/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
There is mounting evidence for the involvement of the immune system, neuroinflammation and disturbed gut microbiota, or dysbiosis, in attention-deficit/hyperactivity disorder (ADHD). Gut dysbiosis is strongly implicated in many physical, autoimmune, neurological, and neuropsychiatric conditions, however knowledge of its particular pathogenic role in ADHD is sparse. As such, this narrative review examines and synthesizes the available evidence related to inflammation, dysbiosis, and neural processes in ADHD. Minimal differences in microbiota diversity measures between cases and controls were found, however many relative abundance differences were observed at all classification levels (phylum to strain). Compositional differences of taxa important to key gut-brain axis pathways, in particular Bacteroides species and Faecalibacterium, may contribute to inflammation, brain functioning differences, and symptoms, in ADHD. We have identified one possible model of ADHD etiopathogenesis involving systemic inflammation, an impaired blood-brain barrier, and neural disturbances as downstream consequences of gut dysbiosis. Nevertheless, studies conducted to date have varied degrees of methodological rigour and involve diverse participant characteristics and analytical techniques, highlighting a need for additional research.
Collapse
Affiliation(s)
- Naomi Lewis
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia; Thompson Institute, University of the Sunshine Coast, 12 Innovation Pkwy, Birtinya, QLD 4575, Australia.
| | - Anthony Villani
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia.
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Eccles Blvd, Birtinya, QLD 4575, Australia.
| |
Collapse
|
24
|
Ye Y, Abulizi A, Zhang Y, Lu F, An Y, Ren C, Zhang H, Wang Y, Lin D, Lu D, Li M, Yang B. Ganoderic Acid Ameliorates Ulcerative Colitis by Improving Intestinal Barrier Function via Gut Microbiota Modulation. Int J Mol Sci 2025; 26:2466. [PMID: 40141109 PMCID: PMC11942431 DOI: 10.3390/ijms26062466] [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: 01/06/2025] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic and recurrent gastrointestinal disease that affects millions of humans worldwide and imposes a huge social and economic burden. It is necessary to find safe and efficient drugs for preventing and treating UC. The aim of this study was to determine whether ganoderic acid (GA), the main bioactive components of Ganoderma lucidum, has preventive and therapeutic effect on UC in a dextran sulfate sodium (DSS)-induced UC mouse model. Our experimental results showed that GA significantly ameliorated the body weight loss and disease activity index (DAI) of UC mice. GA significantly restored 11% of the colon length and 69% of the spleen index compared to UC mice. GA significantly decreased the intestinal inflammatory response and improved the barrier function of the intestine by upregulating the tight junction proteins Zonula occludens-1 (ZO-1), occludin and claudin-1. A co-housing experiment showed that gut microbiota accounted for the therapeutic activity of GA on UC, which was confirmed by fecal microbiota transplantation from GA-treated mice to the UC mice. Furthermore, 16S rDNA high-throughput sequencing of fecal bacteria showed that GA significantly enriched the abundance of Lactobacillus, Oscillospira, Odoribacter and Ruminococcus, which were positively correlated with colon length. Furthermore, this study found the functional metabolites, including Indole-3-acetaldehyde (IAAld), Glutamine (Gln) and Glutathione (GSH), reduced barrier damage in the Caco-2 cell model. In conclusion, this study suggests that GA could ameliorate UC by improving intestinal barrier function via modulating gut microbiota and associated metabolites.
Collapse
Affiliation(s)
- Yuwei Ye
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Abudumijiti Abulizi
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yukun Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Feng Lu
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yongpan An
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Chaoqun Ren
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Hang Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Yiming Wang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Dongmei Lin
- JUNCAO Technology Research Institute, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
| | - Dan Lu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing 100191, China;
| | - Min Li
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| | - Baoxue Yang
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Y.); (A.A.); (Y.Z.); (F.L.); (Y.A.); (C.R.); (H.Z.); (Y.W.)
| |
Collapse
|
25
|
Iliev ID, Ananthakrishnan AN, Guo CJ. Microbiota in inflammatory bowel disease: mechanisms of disease and therapeutic opportunities. Nat Rev Microbiol 2025:10.1038/s41579-025-01163-0. [PMID: 40065181 DOI: 10.1038/s41579-025-01163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/26/2025]
Abstract
Perturbations in the intestinal microbiome are strongly linked to the pathogenesis of inflammatory bowel disease (IBD). Bacteria, fungi and viruses all make up part of a complex multi-kingdom community colonizing the gastrointestinal tract, often referred to as the gut microbiome. They can exert various effects on the host that can contribute to an inflammatory state. Advances in screening, multiomics and experimental approaches have revealed insights into host-microbiota interactions in IBD and have identified numerous mechanisms through which the microbiota and its metabolites can exert a major influence on the gastrointestinal tract. Looking into the future, the microbiome and microbiota-associated processes will be likely to provide unparalleled opportunities for novel diagnostic, therapeutic and diet-inspired solutions for the management of IBD through harnessing rationally designed microbial communities, powerful bacterial and fungal metabolites, individually or in combination, to foster intestinal health. In this Review, we examine the current understanding of the cross-kingdom gut microbiome in IBD, focusing on bacterial and fungal components and metabolites. We examine therapeutic and diagnostic opportunities, the microbial metabolism, immunity, neuroimmunology and microbiome-inspired interventions to link mechanisms of disease and identify novel research and therapeutic opportunities for IBD.
Collapse
Affiliation(s)
- Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology and Hepatology Division, Weill Cornell Medicine, New York, NY, USA.
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Chun-Jun Guo
- Joan and Sanford I. Weill Department of Medicine, Gastroenterology and Hepatology Division, Weill Cornell Medicine, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
26
|
Li S, Wang H, Li B, Lu H, Zhao J, Gao A, An Y, Yang J, Ma T. Multi-Omics Analysis Reveals the Negative Effects of High-Concentrate Diets on the Colonic Epithelium of Dumont Lambs. Animals (Basel) 2025; 15:749. [PMID: 40076032 PMCID: PMC11898968 DOI: 10.3390/ani15050749] [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: 02/13/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Feeding HC diets has been found to induce metabolic dysregulation in the colon. However, the mechanisms by which changes in colonic flora and metabolites damage the colonic epithelium are poorly studied. Therefore, the present experiment used a multi-omics technique to investigate the mechanism of colonic injury induced by high-concentrate diets in lambs. Twelve male Dumont lambs were randomly split into two groups: a low-concentrate diet (LC = concentrate/forage = 30:70) group and a high-concentrate diet (HC = concentrate/forage = 70:30) group. The results showed that the HC group presented significantly increased lipopolysaccharide (LPS) concentrations in the colonic epithelium and significantly decreased serum total cholesterol (TC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) levels (p < 0.05), which led to cavities and inflammatory cell infiltration in the colonic epithelium. The HC group had significantly lower pH and less VFAs in colon contents, as well as a significantly increased abundance of bacteria of the genera [Eubacterium]_coprostanoligenes_group, Rikenellaceae_RC9_gut_group, Treponema, Clostridia_UCG-014, Alistipes, Ruminococcus, Christensenellaceae_R-7_group, UCG-002, Bacteroidales_RF16_group and Lachnospiraceae_AC2044_group compared to the LC diet group. These microorganisms significantly increased the level of metabolites of cholic acid, chenodeoxycholic acid, LysoPA (P-16:0/0:0), methapyrilene, and fusaric acid. A transcriptome analysis showed that cytokine-cytokine receptor interaction, glutathione metabolism, and the peroxisome signaling pathway were downregulated in the colon epithelium of the lambs fed the HC diet. Therefore, the HC diet caused epithelial inflammation and oxidative damage by affecting the interaction between the microbial flora of the colon and metabolites and the host epithelium, which eventually disrupted colon homeostasis and had a negative impact on sheep health.
Collapse
Affiliation(s)
- Shufang Li
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Hairong Wang
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Boyang Li
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Henan Lu
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Jianxin Zhao
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Aiwu Gao
- Food Science, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Yawen An
- Veterinary Research Institute, Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot 010018, China;
| | - Jinli Yang
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| | - Tian Ma
- Animal Nutrition and Feed Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (S.L.); (B.L.); (H.L.); (J.Z.); (J.Y.); (T.M.)
| |
Collapse
|
27
|
Li W, Tu J, Zheng J, Das A, Yan Q, Jiang X, Ding W, Bai X, Lai K, Yang S, Yang C, Zou J, Diwan AD, Zheng Z. Gut Microbiome and Metabolome Changes in Chronic Low Back Pain Patients With Vertebral Bone Marrow Lesions. JOR Spine 2025; 8:e70042. [PMID: 39877797 PMCID: PMC11772216 DOI: 10.1002/jsp2.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Background Chronic low back pain (LBP) is a significant global health concern, often linked to vertebral bone marrow lesions (BML), particularly fatty replacement (FR). This study aims to explore the relationship between the gut microbiome, serum metabolome, and FR in chronic LBP patients. Methods Serum metabolomic profiling and gut microbiome analysis were conducted in chronic LBP patients with and without FR (LBP + FR, n = 40; LBP, n = 40) and Healthy Controls (HC, n = 31). The study investigates alterations in branched-chain amino acids (BCAAs) levels and identifies key microbial species associated with BCAA metabolism. In vitro experiments elucidate the role of BCAAs in adipogenesis of bone marrow mesenchymal stem cells (BM-MSCs) via the SIRT4 pathway. Results Chronic LBP patients with FR exhibit depleted BCAA levels in their serum metabolome, along with alterations in the gut microbiome. Specific microbial species, including Ruminococcus gnavus, Roseburia hominis, and Lachnospiraceae bacterium 8 1 57FAA, are identified as influential in BCAA metabolism and BM-MSCs metabolism. In vitro experiments demonstrate the ability of BCAAs to induce BM-MSCs adipogenesis through SIRT4 pathway activation. Conclusion This study sheds light on the intricate relationship between the disturbed gut ecosystem, serum metabolites, and FR in chronic LBP. Dysbiosis in the gut microbiome may contribute to altered BCAA degradation, subsequently promoting BM-MSCs adipogenesis and FR. Understanding these interactions provides insights for targeted therapeutic strategies to mitigate chronic LBP associated with FR by restoring gut microbial balance and modulating serum metabolite profiles.
Collapse
Affiliation(s)
- Wentian Li
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Gulbali Institute, School of Agricultural, Environmental and Veterinary SciencesCharles Sturt UniversityWagga WaggaAustralia
| | - Ji Tu
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Nepean HospitalNepean Blue Mountains Local Health DistrictPenrithAustralia
| | - Jinjian Zheng
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Abhirup Das
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
| | - Qi Yan
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaotao Jiang
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Medicine and HealthThe University of new South WalesSydneyAustralia
| | - Wenyuan Ding
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Hebei Joint International Research Centre for Spinal DiseasesCenter for Innovation & Translational Medicine, the First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Xupeng Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Charles Perkins Centre and School of Medical SciencesUniversity of SydneySydneyAustralia
| | - Kaitao Lai
- Northcott Neuroscience LaboratoryANZAC Research Institute, Concord HospitalSydneyAustralia
- Tissue Engineering and Microfluidics Laboratory (TE&M)Australian Institute for Bioengineering and Nanotechnology (AIBN), the University of QueenslandSt LuciaAustralia
| | - Sidong Yang
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cao Yang
- Spine ServiceDepartment of Orthopaedic Surgery, St. George HospitalKogarahAustralia
| | - Jun Zou
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ashish D. Diwan
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhaomin Zheng
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
28
|
Barra NG, Fang H, Bhatwa A, Schmidt AM, Syed SA, Steinberg GR, Morrison KM, Surette MG, Wade MG, Holloway AC, Schertzer JD. Food supply toxicants and additives alter the gut microbiota and risk of metabolic disease. Am J Physiol Endocrinol Metab 2025; 328:E337-E353. [PMID: 39871724 DOI: 10.1152/ajpendo.00364.2024] [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: 09/17/2024] [Revised: 10/09/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025]
Abstract
Metabolic disease is rising along with both global industrialization and the use of new commercial, agricultural, and industrial chemicals and food additives. Exposure to these compounds may contribute to aspects of metabolic diseases such as obesity, diabetes, and fatty liver disease. Ingesting compounds in the food supply is a key route of human exposure, resulting in the interaction between toxicants or additives and the intestinal microbiota. Toxicants can influence the composition and function of the gut microbiota, and these microbes can metabolize and transform toxicants and food additives. Microbe-toxicant interactions in the intestine can alter host mucosal barrier function, immunity, and metabolism, which may contribute to the risk or severity of metabolic disease development. Targeting the connection between toxicants, food, and immunity in the gut using strategies such as fermentable fiber (i.e., inulin) may mitigate some of the effects of these compounds on host metabolism. Understanding causative factors in the microbe-host relationship that promote toxicant-induced dysmetabolism is an important goal. This review highlights the role of common toxicants (i.e., persistent organic pollutants, pesticides, and fungicides) and food additives (emulsifiers and artificial sweeteners) found in our food supply that alter the gut microbiota and promote metabolic disease development.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Arshpreet Bhatwa
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Angela M Schmidt
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Saad A Syed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Alison C Holloway
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
29
|
Tashkent Y, Choo JM, Richard A, Wang Z, Calzadilla‐Bertot L, Vasil E, Miller S, Taylor SL, Ivey KL, Woodman R, Adler B, Ayonrinde OT, Olynyk JK, Beilin LJ, Mori TA, Wigg AJ, Muller KR, Adams LA, Rogers GB. Steatotic Liver Disease in Younger Adults is Associated With Altered Gut Microbiology. Liver Int 2025; 45:e70032. [PMID: 39999013 PMCID: PMC11855901 DOI: 10.1111/liv.70032] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD) is a leading cause of chronic liver disease worldwide. As SLD pathogenesis has been linked to gut microbiome alterations, we aimed to identify SLD-associated gut microbiome features early in SLD development by utilising a highly characterised cohort of community-dwelling younger adults. METHODS AND RESULTS At age 27 years, 588 participants of the Raine Study Generation 2 underwent cross-sectional assessment. Hepatic steatosis was quantified using a validated magnetic resonance imaging (MRI) volumetric liver fat fraction (VLFF) equation (HepaFat). Of the 588 participants, 488 (83%) were classified as having 'no SLD' (VLFF ≤ 3.55%), 76 (12.9%) with 'mild-moderate' SLD (VLFF: 3.56%-13.4%) and 24 (4.10%) with 'severe' SLD (VLFF > 13.4%). Stool microbiome profiling identified an association between severe SLD and lower microbiota alpha diversity (observed features [p = 0.015], Pielou evenness [p = 0.001] and Shannon diversity [p = 0.002]) compared to no SLD. Faecal microbiota composition differed significantly between no SLD and both mild-moderate (p = 0.004) and severe SLD groups (p = 0.001). There was no significant difference in microbiota dispersion between SLD groups. Reduced relative abundance of short-chain fatty acid producing bacteria, and higher levels of proinflammatory bacterial taxa, were both significantly associated with severe SLD (q < 0.05). CONCLUSIONS SLD in younger adults is associated with reduced intestinal microbial diversity and a pattern of bacterial taxa depletion that is consistent with other chronic inflammatory conditions. Our characterisation of gut microbiome characteristics in early SLD development provides a potential basis for risk identification and reduction. TRIAL REGISTRATION The Raine Study is registered in the Australian New Zealand Clinical Trials Registry (ACTRN12617001599369).
Collapse
Affiliation(s)
- Yasmina Tashkent
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Jocelyn M. Choo
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Alyson Richard
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Zhengyi Wang
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Luis Calzadilla‐Bertot
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of HepatologySir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Egi Vasil
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Sophie Miller
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Steven L. Taylor
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Kerry L. Ivey
- Division of Aging, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Richard Woodman
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Brendan Adler
- Envision Medical ImagingWembleyWestern AustraliaAustralia
| | - Oyekoya T. Ayonrinde
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of Gastroenterology and HepatologyFiona Stanley HospitalMurdochWestern AustraliaAustralia
- Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - John K. Olynyk
- Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - Lawrence J. Beilin
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Trevor A. Mori
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Alan J. Wigg
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Kate R. Muller
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Leon A. Adams
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of HepatologySir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Geraint B. Rogers
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| |
Collapse
|
30
|
Kume M, Din J, Zegarra-Ruiz DF. Dysregulated Intestinal Host-Microbe Interactions in Systemic Lupus Erythematosus: Insights from Patients and Mouse Models. Microorganisms 2025; 13:556. [PMID: 40142449 PMCID: PMC11944652 DOI: 10.3390/microorganisms13030556] [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: 02/03/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by chronic inflammation that affects multiple organs, with its prevalence varying by ethnicity. Intestinal dysbiosis has been observed in both SLE patients and murine models. Additionally, intestinal barrier impairment is thought to contribute to the ability of pathobionts to evade and breach immune defenses, resulting in antigen cross-reactivity, microbial translocation, subsequent immune activation, and, ultimately, multiple organ failure. Since the detailed mechanisms underlying these processes are difficult to examine using human samples, murine models are crucial. Various SLE murine models, including genetically modified spontaneous and inducible murine models, offer insights into pathobionts and how they dysregulate systemic immune systems. Furthermore, since microbial metabolites modulate systemic immune responses, bacteria and their metabolites can be targeted for treatment. Based on human and mouse research insights, this review examines how lupus pathobionts trigger intestinal and systemic immune dysregulation. Therapeutic approaches, such as fecal microbiota transplantation and dietary adjustments, show potential as cost-effective and safe methods for preventing and treating SLE. Understanding the complex interactions between the microbiota, host factors, and immune dysregulation is essential for developing novel, personalized therapies to tackle this multifaceted disease.
Collapse
Affiliation(s)
| | | | - Daniel F. Zegarra-Ruiz
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; (M.K.); (J.D.)
| |
Collapse
|
31
|
Deng D, Zhao L, Song H, Wang H, Cao H, Cui H, Zhou Y, Cui R. Microbiome analysis of gut microbiota in patients with colorectal polyps and healthy individuals. Sci Rep 2025; 15:7126. [PMID: 40021742 PMCID: PMC11871317 DOI: 10.1038/s41598-025-91626-4] [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: 10/15/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
Colorectal polyps serve as the primary precursors for colorectal cancer. A close relationship has been observed between colorectal polyps and gut microbiota. However, the composition and role of the microbiome associated with tubular adenoma are not well understood. In this study, we prospectively evaluated alterations in gut microbiota among patients with colorectal polyps. A total of 60 subjects were enrolled in this study, including 30 patients with colorectal polyps (CP group) and 30 healthy controls (control group). The 16S rRNA sequencing was employed to characterize the gut microbiome in fecal samples. The results revealed that the beta diversity of the gut microbiota in the CP group significantly differs from that of the control group (p = 0.001). At the phylum level, the relative abundance of Bacteroides, Fusobacteria, and Proteobacteria was higher in the CP group compared to the control group (p < 0.05), whereas the relative abundance of Actinobacteria was higher in the control group in comparison to the CP group (p < 0.05). At the genus level, the abundance of Bacteroides increased in the CP group (p < 0.05), while Bifidobacterium declined in the CP group (p < 0.05). At the species level, the abundance of Clostridium perfringens, unidentified_Bacteroides, unidentified_Dorea, Escherichia coli, Clostridium ramosum, and Ruminococcus gnavus was higher (p < 0.05), whereas the abundance of Bifidobacterium adolescentis, unclassified_Bifidobacterium, Bifidobacterium longum, Faecalibacterium prausnitzii, and unidentified_Bifidobacterium is lower in CP group compared to the control group (p < 0.05). There was a structural imbalance in the composition of intestinal colonization flora for CP patients, characterized by a decrease in beneficial bacteria and an increase in harmful bacteria. Escherichia, Shigella, and Bacteroides may serve as promising biomarkers for early detection of colorectal polyps.
Collapse
Affiliation(s)
- Dayi Deng
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Lin Zhao
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Hui Song
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Houming Wang
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Hengjie Cao
- Department of Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Huimin Cui
- Department of Surgery, Jinan Licheng District Hospital of Chinese Medicine, Jinan, 250000, China
| | - Yong Zhou
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China.
| | - Rong Cui
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China.
| |
Collapse
|
32
|
Liu M, Li XW, Sun H, Yan YQ, Xia ZY, Refaie A, Zhang NY, Wang S, Tan C, Sun LH. T-2 toxin-induced splenic injury by disrupting the gut microbiota-spleen axis via promoting IL-6/JAK/STAT1 signaling-mediated inflammation and apoptosis and its mitigation by elemental nano-selenium. Arch Toxicol 2025:10.1007/s00204-025-04005-3. [PMID: 40014112 DOI: 10.1007/s00204-025-04005-3] [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: 01/15/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
T-2 toxin is one of the most toxic A trichothecene mycotoxins prevalent in the environment and food chain, which brings severe health hazards to both animals and humans and it can significantly damage immune function. In this study, we comprehensively explained the impact of T-2 toxin on the spleen through gut microbiota-spleen axis by integrating the transcriptome and microbiome. Our results revealed that dietary T-2 toxin ≥ 1.0 mg/kg exposure significantly inhibited the growth performance and caused spleen injury in broilers chicks, accompanied by oxidative stress and histopathological damage. Cecal microbiome analysis suggested that T-2 toxin exposure caused gut microbial dysbiosis, especially leading to the decrease of some beneficial bacteria genera that enhanced gut barrier and reduced inflammation, including Blautia, Coprococcus, Lachnospira and Anaerostipes belonging to Lachnospiraceae family. Transcriptome analysis suggested that T-2 toxin exposure directly caused splenic inflammation and immune-related signaling, such as cytokine-cytokine receptor interaction, Toll-like receptor signaling pathway, NOD-like receptor signaling pathway and JAK-STAT signaling pathway. Furthermore, by integrating the transcriptome and microbiome analysis, we found that spleen damage induced by T-2 toxin was associated with the abnormal activation of IL-6/JAK/STAT1 signaling pathway-mediated inflammation and apoptosis, which was further verified by western bolt analysis. Notably, dietary selenium supplementation could protect chicks from T-2 toxin-induced adverse effects on growth performance and spleen injury by inhibiting the expression of the IL-6/JAK/STAT1 signaling-related genes. In summary, our findings provided new insights into the immunotoxicity mechanisms of T-2 toxin in the chickens' spleen and highlighted the potential of selenium to alleviate T-2 toxin-induced immunotoxicity.
Collapse
Affiliation(s)
- Meng Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xue-Wu Li
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Newhope Liuhe Co. Ltd., Beijing, China
| | - Hua Sun
- Inner Mongolia Academy of Agriculture and Animal Husbandry Science, Hohhot, 010031, Inner Mongolia, China
| | - Yi-Qin Yan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhi-Yuan Xia
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Alainaa Refaie
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ni-Ya Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shuai Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, Key Laboratory of Smart Farming Technology for Agricultural Animals of Ministry of Agriculture and Rural Affairs, College of Animal Science & Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
33
|
Santangelo BE, Bada M, Hunter LE, Lozupone C. Hypothesizing mechanistic links between microbes and disease using knowledge graphs. Sci Rep 2025; 15:6905. [PMID: 40011529 PMCID: PMC11865272 DOI: 10.1038/s41598-025-91230-6] [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: 10/08/2024] [Accepted: 02/19/2025] [Indexed: 02/28/2025] Open
Abstract
Knowledge graphs have been a useful tool for many biomedical applications because of their effective representation of biological concepts. Plentiful evidence exists linking the gut microbiome to disease in a correlative context, but uncovering the mechanistic explanation for those associations remains a challenge. Here we demonstrate the potential of knowledge graphs to hypothesize plausible mechanistic accounts of host-microbe interactions in disease. We have constructed a knowledge graph of linked microbes, genes and metabolites called MGMLink, and, using a shortest path or template-based search through the graph and a novel path-prioritization methodology based on the structure of the knowledge graph, we show that this knowledge supports inference of mechanistic hypotheses that explain observed relationships between microbes and disease phenotypes. We discuss specific applications of this methodology in inflammatory bowel disease and Parkinson's disease. This approach enables mechanistic hypotheses surrounding the complex interactions between gut microbes and disease to be generated in a scalable and comprehensive manner.
Collapse
Affiliation(s)
- Brook E Santangelo
- Department of Biomedical Informatics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.
| | - Michael Bada
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | | | - Catherine Lozupone
- Department of Biomedical Informatics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
34
|
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
|
35
|
Wu D, Yin M, Cao D, Zhang X, Zhu Y, Wei Y, Li Y, Wen C, Zhou J. Disruption of Gut Microbiota and Associated Fecal Metabolites in Collagen-Induced Arthritis Mice During the Early Stage. J Inflamm Res 2025; 18:1703-1717. [PMID: 39925933 PMCID: PMC11806705 DOI: 10.2147/jir.s502980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease and increasing evidence suggests that disturbances in the composition and function of gut microbiota are potentially implicated in the progression of RA. Further revealing the microbiota and related metabolic disorders in the preclinical stage of RA (pre-RA) is of great significance for exploration of disease mechanisms. Methods DBA/1 mice were injected with type II collagen on days 0 and 21 to establish collagen-induced arthritis (CIA) mouse model. Footpad thickness, serum autoantibodies, and joint histopathology were used to assess the progression of RA. A combination of 16S rRNA sequencing, untargeted metabolomics and targeted short-chain fatty acids (SCFAs) analysis were employed to comprehensively investigate the alterations of gut microbiota and fecal metabolites in CIA during the pre-RA stage. Results 20 days after the initial collagen immunization, CIA mice showed immune responses without joint symptoms, alongside gut microbiota disruption. Alterations were observed in 20 microbial taxa, including Oscillospira, Bifidobacterium, Ruminococcus, Allobaculum, Alistipes, Lactobacillus, and Candidatus_Arthromitus, etc. Untargeted and targeted metabolomics identified 33 altered fecal metabolites, mainly including sugars and their derivatives, amino acids, long-chain fatty acids and SCFAs, etc. Correlation analysis showed significant correlations between specific gut microbial abundances and fecal metabolite levels. Especially, SCFAs were strongly associated with Bifidobacterium, Alistipes, Ruminococcus, Anaerotruncus, and Allobaculum. Conclusion These findings suggest that collagen immunization leads to disruption of gut microbiome and induces changes of fecal metabolites in mice, which may play a key role in early development of RA in CIA mice.
Collapse
Affiliation(s)
- Dehong Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, People’s Republic of China
| | - Mengdi Yin
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Dandan Cao
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xiafeng Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yichun Zhu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Ying Wei
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yiling Li
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jia Zhou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
36
|
Kamegai K, Hayakawa K, Saito S, Mezaki K, Sakurai A, Ohmagari N. Unveiling Ruminococcus gnavus bacteremia: Clinical characteristics and implications. J Infect Chemother 2025; 31:102558. [PMID: 39547409 DOI: 10.1016/j.jiac.2024.11.008] [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: 05/25/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Ruminococcus gnavus is a microbiota-forming, gram-positive coccus reportedly associated with several diseases, such as Crohn's disease. The number of in vitro studies on it is increasing. However, its clinical information is lacking in the literature, with only a few case reports published to date. To elucidate the significance of this organism, we describe its clinical characteristics in this study. METHODS During the study period (2013-2022), we identified 11 patients with R. gnavus bacteremia and conducted a retrospective chart review. Cases identified to be contaminated were excluded. We found 11 reports of R. gnavus bacteremia on PubMed and reviewed their clinical characteristics. RESULTS Eleven R. gnavus bloodstream infection cases were identified in our facility. The median age of the patients was 83 years (interquartile range: 73.75-87.25). Seven cases had at least one documented intestinal lesion including three with malignancy cases, and two cases had uncompensated cirrhosis. In most cases, bacterial translocation was suspected as the entry mechanism. Among the 11 R. gnavus bloodstream infections, 7 (63.6 %) were associated with intestinal lesions, and 2 (18.2 %) had a history of suspected bacterial translocation without documented intestinal lesions. CONCLUSION To the best of our knowledge, this is the largest cohort study on R. gnavus bloodstream infections. Intestinal entry was suspected in more than 80 % of cases in both our cohort and the literature review cohort. For cases of bacteremia with an unknown etiology due to R. gnavus, a thorough examination of gastrointestinal lesions should be performed.
Collapse
Affiliation(s)
- Kohei Kamegai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Japan.
| | - Kayoko Hayakawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Japan
| | - Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Japan
| | - Kazuhisa Mezaki
- Laboratory of Bacteriology, National Center for Global Health and Medicine, Japan
| | - Ayana Sakurai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Japan
| |
Collapse
|
37
|
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
|
38
|
Navid F, Chen L, Bowness P, Colbert RA. HLA-B27 and spondyloarthritis: at the crossroads of innate and adaptive immunity. Nat Rev Rheumatol 2025; 21:77-87. [PMID: 39623156 DOI: 10.1038/s41584-024-01189-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 01/29/2025]
Abstract
HLA-B*27 confers a strong risk of developing spondyloarthritis (SpA), which includes axial SpA with or without peripheral arthritis, enthesitis, acute anterior uveitis and gastrointestinal inflammation. Although no definitive mechanism has been established to explain the role of this HLA class I protein in the pathogenesis of SpA, three main hypotheses have emerged. First is the idea that self-peptides displayed by HLA-B27 resemble microorganism-derived peptides, leading to the expansion of autoreactive CD8+ T cells that trigger disease. The second and third hypotheses focus on aberrant properties of HLA-B27, including its tendency to form cell-surface dimers that can activate innate killer immunoglobulin-like receptors on CD4+ T helper 17 cells, triggering the production of pathogenic cytokines. HLA-B27 also misfolds in the endoplasmic reticulum, which can activate the unfolded protein response, increasing IL-23 expression and thereby promoting the production of type 17 cytokines. HLA-B27 misfolding in mesenchymal stem cells has also been linked to enhanced bone formation by mesenchymal stem cell-derived osteoblasts, which could contribute to structural damage in axial SpA. In this Review we summarize prevailing ideas about the role of HLA-B27 in SpA, discuss the latest developments as well as the gaps in current knowledge, and provide recommendations for future research to address these unmet needs.
Collapse
Affiliation(s)
- Fatemeh Navid
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liye Chen
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Paul Bowness
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Robert A Colbert
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Cyphert EL, Clare S, Dash A, Nixon JC, Raphael J, Harrison J, Heilbronner A, Kim HJ, Cunningham M, Lebl D, Schwab F, Hernandez CJ, Stein EM. A Pilot Study of the Gut Microbiota in Spine Fusion Surgery Patients. HSS J 2025; 21:65-72. [PMID: 39846059 PMCID: PMC11748416 DOI: 10.1177/15563316231201410] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 01/24/2025]
Abstract
Background The microbiome has been identified as a contributor to bone quality. As skeletal health is critical to success of orthopedic surgery, the gut microbiome may be a modifiable factor associated with postoperative outcomes. For spine fusion surgery in particular, de novo bone formation and sufficient bone mineral density are essential for successful outcomes. Given the prevalence and complexity of these procedures, the identification of novel factors that may be related to operative success is important. Questions/purposes We sought to investigate how the composition of the microbiota related to bone health in a focused spinal fusion surgery cohort. Methods We investigated the composition of the microbiome in a cohort of 31 patients prior to spinal fusion surgery, as well as changes in the microbiome over 6 weeks postoperatively. Preoperative areal bone mineral density (BMD) was measured by dual-energy X-ray absorptiometry. Results Composition of gut microbiota differed among spinal fusion patients with low bone mass (T-score ≤ -1.0) and those with normal BMD (P = .03). There was no significant change in composition of the gut microbiota between preoperative evaluation and 6 weeks postoperatively. Conclusions Our findings in this small sample suggest there may be a relationship between BMD and composition of the gut microbiome in patients who undergo spinal fusion surgery. Further work is needed to investigate these relationships as well as potential interventions to foster a favorable microbial composition in spinal fusion surgery patients.
Collapse
Affiliation(s)
- Erika L. Cyphert
- Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | | | - Jacob C. Nixon
- Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | | | | | | - Han Jo Kim
- Hospital for Special Surgery, New York, NY, USA
| | | | - Darren Lebl
- Hospital for Special Surgery, New York, NY, USA
| | | | - Christopher J. Hernandez
- Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Hospital for Special Surgery, New York, NY, USA
| | | |
Collapse
|
40
|
Chen X, Chang H, Gao C, Zhu X. Genetic insights into the gut microbiota-duodenal diseases interplay: A Mendelian randomization and Bayesian weighting study. Microb Pathog 2025; 199:107181. [PMID: 39615706 DOI: 10.1016/j.micpath.2024.107181] [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: 04/14/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND Many observational studies have shown a close association between gut microbiota and the risk of various duodenal diseases. Therefore, we urgently explore the potential causal relationship between gut microbiota and some duodenal diseases, mainly including duodenal ulcers and duodenitis. METHODS We conducted Mendelian randomization (MR) studies using genetic instrumental variables for gut microbiota from GWAS and duodenal disease datasets. Causal relationships were examined using multiple MR methods with Bonferroni correction. Bayesian Weighted Mendelian Randomization (BWMR) assessed causal relationships, employing ELBO and weighted data. Reverse MR analysis was conducted on microbiota showing significant causal relationships with duodenal diseases. RESULTS Through MR analysis, we identified three gut microbiota that promote the occurrence of duodenal ulcers (family. Coriobacteriaceae: OR = 1.003; 95 % CI = 1.0005-1.0056; p = 0.016, genus.RuminococcaceaeUCG003: OR = 1.006; 95 % CI = 1.002-1.007; p = 0.002, order. Coriobacteriales: OR = 1.003; 95 % CI = 1.0005-1.0056; p = 0.016), one microbiota that inhibits the occurrence of duodenitis (family. Acidaminococcaceae: OR = 0.994; 95 % CI = 0.988-0.999; p = 0.046), and one microbiota that promotes the occurrence of duodenitis (genus.Eubacteriumcoprostanoligenesgroup: OR = 1.006; 95 % CI = 1.0005-1.013; p = 0.033). Further confirmation of the occurrence of duodenal ulcers and the production of family.Coriobacteriaceae and order.Coriobacteriales microbiota was obtained through reverse MR analysis, indicating that the occurrence of duodenal ulcers also promotes the growth of these microbiota. CONCLUSION Our study employs Mendelian randomization techniques to demonstrate a causal relationship between specific gut microbiota and duodenal ulcers and duodenitis. Additionally, our analysis suggests that duodenal ulcer occurrence promotes the growth of certain microbiota, emphasizing the intricate interplay between gut microbiota composition and these diseases.
Collapse
Affiliation(s)
- Xuehui Chen
- The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Hong Chang
- Department of Pathology, Qingdao Third People's Hospital, Qingdao, China
| | - Chuanmei Gao
- Department of Endoscopy Center, Zibo Central Hospital, Zibo, China
| | - Xiao Zhu
- The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
41
|
Nooij S, Plomp N, Sanders IMJG, Schout L, van der Meulen AE, Terveer EM, Norman JM, Karcher N, Larralde MF, Vossen RHAM, Kloet SL, Faber KN, Harmsen HJM, Zeller GF, Kuijper EJ, Smits WK, Ducarmon QR. Metagenomic global survey and in-depth genomic analyses of Ruminococcus gnavus reveal differences across host lifestyle and health status. Nat Commun 2025; 16:1182. [PMID: 39885121 PMCID: PMC11782615 DOI: 10.1038/s41467-025-56449-x] [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: 07/12/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
Ruminococcus gnavus is a gut bacterium found in > 90% of healthy individuals, but its increased abundance is also associated with chronic inflammatory diseases, particularly Crohn's disease. Nevertheless, its global distribution and intraspecies genomic variation remain understudied. By surveying 12,791 gut metagenomes, we recapitulated known associations with metabolic diseases and inflammatory bowel disease. We uncovered a higher prevalence and abundance of R. gnavus in Westernized populations and observed bacterial relative abundances up to 83% in newborns. Next, we built a resource of R. gnavus isolates (N = 45) from healthy individuals and Crohn's disease patients and generated complete R. gnavus genomes using PacBio circular consensus sequencing. Analysis of these genomes and publicly available high-quality draft genomes (N = 333 genomes) revealed multiple clades which separated Crohn's-derived isolates from healthy-derived isolates. Presumed R. gnavus virulence factors could not explain this separation. Bacterial genome-wide association study revealed that Crohn's-derived isolates were enriched in genes related to mobile elements and mucin foraging. Together, we present a large R. gnavus resource that will be available to the scientific community and provide novel biological insights into the global distribution and genomic variation of R. gnavus.
Collapse
Affiliation(s)
- S Nooij
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Donor Feces Bank (NDFB), Leiden University Medical Center, Leiden, the Netherlands
| | - N Plomp
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - I M J G Sanders
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - L Schout
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - A E van der Meulen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - E M Terveer
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Donor Feces Bank (NDFB), Leiden University Medical Center, Leiden, the Netherlands
| | - J M Norman
- Vedanta Biosciences, Inc., Cambridge, Massachusetts, USA
| | - N Karcher
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - M F Larralde
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
| | - R H A M Vossen
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - S L Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - K N Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H J M Harmsen
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G F Zeller
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - E J Kuijper
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - W K Smits
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Q R Ducarmon
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands.
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands.
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
42
|
Bang S, Shin YH, Park SM, Deng L, Williamson RT, Graham DB, Xavier RJ, Clardy J. Unusual Phospholipids from Morganella morganii Linked to Depression. J Am Chem Soc 2025; 147:2998-3002. [PMID: 39818770 PMCID: PMC11783507 DOI: 10.1021/jacs.4c15158] [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: 10/28/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
A multifactorial association study detected a probable causal connection between the prevalence of Morganella morganii in the gut microbiome and the incidence of major depressive disorder (MDD) in the human host. A bioassay-guided fractionation approach identified bacterially produced metabolites that induced pro-inflammatory immune responses. The metabolites are unusual phospholipids that resemble conventional cardiolipins, in which diethanolamine (DEA) replaces the central glycerol. These molecular chimeras of endogenous metabolites from phospholipid biosynthetic pathways and the industrially produced micropollutant DEA activate TLR2/TLR1 receptors and induce the production of pro-inflammatory cytokines, especially IL-6. Their activity in conventional immunomodulatory assays largely parallels that of immunogenic cardiolipins with conventional structures. The molecular mechanism connecting these chimeric cardiolipins to MDD is supported by other studies and has implications for conditions other than MDD.
Collapse
Affiliation(s)
- Sunghee Bang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Yern-Hyerk Shin
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sung-Moo Park
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Lei Deng
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - R. Thomas Williamson
- Department
of Chemistry and Biochemistry, University
of North Carolina Wilmington, Wilmington, North Carolina 28409, United States
| | - Daniel B. Graham
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ramnik J. Xavier
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
43
|
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
|
44
|
Bhusri B, Sutheeworapong S, Kittichotirat W, Kusonmano K, Thammarongtham C, Lertampaiporn S, Prommeenate P, Praphanphoj V, Kittitharaphan W, Dulsawat S, Paenkaew P, Cheevadhanarak S. Characterization of gut microbiota on gender and age groups bias in Thai patients with autism spectrum disorder. Sci Rep 2025; 15:2587. [PMID: 39833480 PMCID: PMC11747245 DOI: 10.1038/s41598-025-86740-2] [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: 08/03/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication and interaction problems. The prevalence of ASD is increasing globally, with a higher ratio of males to females. Gastrointestinal symptoms are common in individuals with ASD, and gut microbiota has been implicated in the disorder's development. This study aimed to investigate the gut microbiota alteration in Thai individuals with ASD compared to healthy controls using 16S rRNA gene sequencing. The influence of gender and age on gut microbiota composition and function was also examined. A total of 65 ASD individuals and 30 neurotypical (NT) individuals were included in the analysis. The results revealed notable differences in gut microbiota composition between the ASD and NT groups, with variations observed in microbial richness and the presence of enriched microbial taxa. These differences were influenced by both gender and age. Fusobacteriota, Fusobacteriaceae, and Fusobacterium were found to be enriched in individuals with ASD. Furthermore, the study identified gender-related taxa, such as Bacteroides plebeius, enriched in ASD females. Age-related taxa, including Veillonella, known to be associated with poor oral hygiene, were also observed in ASD children. The analysis of differentially abundant pathways highlighted the enrichment of various metabolic pathways in individuals with ASD, including those related to endocrine-disrupting chemicals. These findings underscore the importance of considering gender and age when studying gut microbiota in ASD. They provide valuable insights into the potential role of gut microbiota dysbiosis in ASD pathogenesis and highlight the influence of environmental factors.
Collapse
Affiliation(s)
- Benjaporn Bhusri
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Sawannee Sutheeworapong
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Kanthida Kusonmano
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Chinae Thammarongtham
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Supatcha Lertampaiporn
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Peerada Prommeenate
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | | | - Wiranpat Kittitharaphan
- Yuwaprasart Waithayopathum Child and Adolescent Psychiatric Hospital, Ministry of Public Health, Samut Prakan, 10270, Thailand
| | - Sudarat Dulsawat
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Prasobsook Paenkaew
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Supapon Cheevadhanarak
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
- School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
| |
Collapse
|
45
|
Brown EM, Nguyen PNU, Xavier RJ. Emerging biochemical, microbial and immunological evidence in the search for why HLA-B ∗27 confers risk for spondyloarthritis. Cell Chem Biol 2025; 32:12-24. [PMID: 39168118 PMCID: PMC11741937 DOI: 10.1016/j.chembiol.2024.07.012] [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: 03/08/2024] [Revised: 05/25/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
The strong association of the human leukocyte antigen B∗27 alleles (HLA-B∗27) with spondyloarthritis and related rheumatic conditions has long fascinated researchers, yet the precise mechanisms underlying its pathogenicity remain elusive. Here, we review how interplay between the microbiome, the immune system, and the enigmatic HLA-B∗27 could trigger spondyloarthritis, with a focus on whether HLA-B∗27 presents an arthritogenic peptide. We propose mechanisms by which the unique biochemical characteristics of the HLA-B∗27 protein structure, particularly its peptide binding groove, could dictate its propensity to induce pathological T cell responses. We further provide new insights into how TRBV9+ CD8+ T cells are implicated in the disease process, as well as how the immunometabolism of T cells modulates tissue-specific inflammatory responses in spondyloarthritis. Finally, we present testable models and suggest approaches to this problem in future studies given recent advances in computational biology, chemical biology, structural biology, and small-molecule therapeutics.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
46
|
Qiao T, Wen XH. Exploring gut microbiota as a novel therapeutic target in Crohn's disease: Insights and emerging strategies. World J Gastroenterol 2025; 31:100827. [PMID: 39811502 PMCID: PMC11684203 DOI: 10.3748/wjg.v31.i2.100827] [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: 08/28/2024] [Revised: 09/30/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Extensive research has investigated the etiology of Crohn's disease (CD), encompassing genetic predisposition, lifestyle factors, and environmental triggers. Recently, the gut microbiome, recognized as the human body's second-largest gene pool, has garnered significant attention for its crucial role in the pathogenesis of CD. This paper investigates the mechanisms underlying CD, focusing on the role of 'creeping fat' in disease progression and exploring emerging therapeutic strategies, including fecal microbiota transplantation, enteral nutrition, and therapeutic diets. Creeping fat has been identified as a unique pathological feature of CD and has recently been found to be associated with dysbiosis of the gut microbiome. We characterize this dysbiotic state by identifying key microbiome-bacteria, fungi, viruses, and archaea, and their contributions to CD pathogenesis. Additionally, this paper reviews contemporary therapies, emphasizing the potential of biological therapies like fecal microbiota transplantation and dietary interventions. By elucidating the complex interactions between host-microbiome dynamics and CD pathology, this article aims to advance our understanding of the disease and guide the development of more effective therapeutic strategies for managing CD.
Collapse
Affiliation(s)
- Tong Qiao
- Department of Clinical Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xian-Hui Wen
- College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
47
|
Jangi S, Zhao N, Hsia K, Park YS, Michaud DS, Yoon H. Specific Bacterial Co-abundance Groups Are Associated With Inflammatory Status in Patients With Ulcerative Colitis. J Crohns Colitis 2025; 19:jjae125. [PMID: 39126385 PMCID: PMC11725523 DOI: 10.1093/ecco-jcc/jjae125] [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: 05/20/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND AIMS While there is increasing interest in microbiome-directed therapies for patients with ulcerative colitis (UC), the identification of microbial targets remains elusive, underlining the need for novel approaches. METHODS Utilizing metagenomic data from the Study of a Prospective Adult Research Cohort with Inflammatory Bowel Disease (SPARC IBD), available via the IBD Plexus Program of the Crohn's & Colitis Foundation, we used a tree-based dichotomous approach to assemble distinct clusters of species-level bacterial co-abundance groups (CAGs). We evaluated the abundance of bacterial CAGs and fungal taxa during remission (n = 166) and activity (n = 46). We examined if the bacterial CAGs identified in our cohorts were conserved in 2 healthy cohorts and a Korean UC cohort. RESULTS CAG3 and CAG8, dominated by bacteria from the family Lachnospiraceae, were associated with remission. Low abundance of CAG8 and elevated abundance of Candida genus were predictive of active UC. Constituents from CAG8 were influential hub species of the remission-associated microbial UC network, including Ruminococcus gnavus, Erysipelatoclostridium ramosum, Blautia, and Dorea species. These hub species interactions were preserved in 2 healthy cohorts and were partially recapitulated in a Korean UC cohort. CAG8 abundance correlated with the secondary bile acid production pathway. Bacterial CAGs did not correlate with Candida; however, Bifidobacterium adolescentis and Alistipes putredinis were negatively associated with Candida. CONCLUSIONS Lachnospiraceae-dominated bacterial CAGs were associated with remission in UC, with key bacterial interactions within the CAG also observed in 2 healthy cohorts and a Korean UC cohort. Bacterial CAG-based analyses may aid in designing candidate consortia for microbiome-based therapeutics.
Collapse
Affiliation(s)
- Sushrut Jangi
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Naisi Zhao
- Tufts University School of Medicine, Public Health and Community Medicine, Boston, MA, USA
| | - Katie Hsia
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dominique S Michaud
- Tufts University School of Medicine, Public Health and Community Medicine, Boston, MA, USA
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
48
|
Nishijima S, Stankevic E, Aasmets O, Schmidt TSB, Nagata N, Keller MI, Ferretti P, Juel HB, Fullam A, Robbani SM, Schudoma C, Hansen JK, Holm LA, Israelsen M, Schierwagen R, Torp N, Telzerow A, Hercog R, Kandels S, Hazenbrink DHM, Arumugam M, Bendtsen F, Brøns C, Fonvig CE, Holm JC, Nielsen T, Pedersen JS, Thiele MS, Trebicka J, Org E, Krag A, Hansen T, Kuhn M, Bork P. Fecal microbial load is a major determinant of gut microbiome variation and a confounder for disease associations. Cell 2025; 188:222-236.e15. [PMID: 39541968 DOI: 10.1016/j.cell.2024.10.022] [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: 03/08/2024] [Revised: 07/12/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
The microbiota in individual habitats differ in both relative composition and absolute abundance. While sequencing approaches determine the relative abundances of taxa and genes, they do not provide information on their absolute abundances. Here, we developed a machine-learning approach to predict fecal microbial loads (microbial cells per gram) solely from relative abundance data. Applying our prediction model to a large-scale metagenomic dataset (n = 34,539), we demonstrated that microbial load is the major determinant of gut microbiome variation and is associated with numerous host factors, including age, diet, and medication. We further found that for several diseases, changes in microbial load, rather than the disease condition itself, more strongly explained alterations in patients' gut microbiome. Adjusting for this effect substantially reduced the statistical significance of the majority of disease-associated species. Our analysis reveals that the fecal microbial load is a major confounder in microbiome studies, highlighting its importance for understanding microbiome variation in health and disease.
Collapse
Affiliation(s)
- Suguru Nishijima
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Evelina Stankevic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Aasmets
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Thomas S B Schmidt
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Marisa Isabell Keller
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pamela Ferretti
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Anthony Fullam
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Christian Schudoma
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johanne Kragh Hansen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Mads Israelsen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Robert Schierwagen
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Nikolaj Torp
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Anja Telzerow
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rajna Hercog
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stefanie Kandels
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Diënty H M Hazenbrink
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Charlotte Brøns
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Medical department, University Hospital Zeeland, Køge, Denmark
| | - Julie Steen Pedersen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Maja Sofie Thiele
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Münster, Germany; European Foundation for the Study of Chronic Liver Failure, EFCLIF, Barcelona, Spain
| | - Elin Org
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Aleksander Krag
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kuhn
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Peer Bork
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Max Delbrück Centre for Molecular Medicine, Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
49
|
Fliegerová KO, Mahayri TM, Sechovcová H, Mekadim C, Mrázek J, Jarošíková R, Dubský M, Fejfarová V. Diabetes and gut microbiome. Front Microbiol 2025; 15:1451054. [PMID: 39839113 PMCID: PMC11747157 DOI: 10.3389/fmicb.2024.1451054] [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: 06/18/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Diabetes mellitus represents a significant global health problem. The number of people suffering from this metabolic disease is constantly rising and although the incidence is heterogeneous depending on region, country, economic situation, lifestyle, diet and level of medical care, it is increasing worldwide, especially among youths and children, mainly due to lifestyle and environmental changes. The pathogenesis of the two most common subtypes of diabetes mellitus, type 1 (T1DM) and type 2 (T2DM), is substantially different, so each form is characterized by a different causation, etiology, pathophysiology, presentation, and treatment. Research in recent decades increasingly indicates the potential role of the gut microbiome in the initiation, development, and progression of this disease. Intestinal microbes and their fermentation products have an important impact on host metabolism, immune system, nutrient digestion and absorption, gut barrier integrity and protection against pathogens. This review summarizes the current evidence on the changes in gut microbial populations in both types of diabetes mellitus. Attention is focused on changes in the abundance of specific bacterial groups at different taxonomic levels in humans, and microbiome shift is also assessed in relation to geographic location, age, diet and antidiabetic drug. The causal relationship between gut bacteria and diabetes is still unclear, and future studies applying new methodological approaches to a broader range of microorganisms inhabiting the digestive tract are urgently needed. This would not only provide a better understanding of the role of the gut microbiome in this metabolic disease, but also the use of beneficial bacterial species in the form of probiotics for the treatment of diabetes.
Collapse
Affiliation(s)
- Kateřina Olša Fliegerová
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Prague, Czechia
| | - Tiziana Maria Mahayri
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Prague, Czechia
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Hana Sechovcová
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Prague, Czechia
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, Prague, Czechia
| | - Chahrazed Mekadim
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Prague, Czechia
| | - Jakub Mrázek
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Prague, Czechia
| | - Radka Jarošíková
- Institute for Clinical and Experimental Medicine, Diabetes Centre, Prague, Czechia
- Department of Internal Medicine, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Michal Dubský
- Institute for Clinical and Experimental Medicine, Diabetes Centre, Prague, Czechia
| | - Vladimíra Fejfarová
- Institute for Clinical and Experimental Medicine, Diabetes Centre, Prague, Czechia
- Department of Internal Medicine, Second Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
50
|
Yan Z, Chen Q, Ren Y, Shi J, Xu Z, Xue Y, Geng Y. Maltodextrin alleviates constipation induced by loperamide hydrochloride in mice. FOOD BIOSCI 2025; 63:105675. [DOI: 10.1016/j.fbio.2024.105675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|