1
|
Li C, Chen X, Zha W, Fang S, Shen J, Li L, Jiang H, Tian P. Impact of gut microbiota in chronic kidney disease: natural polyphenols as beneficial regulators. Ren Fail 2025; 47:2506810. [PMID: 40441674 PMCID: PMC12123969 DOI: 10.1080/0886022x.2025.2506810] [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: 12/14/2024] [Revised: 04/04/2025] [Accepted: 05/10/2025] [Indexed: 06/02/2025] Open
Abstract
Chronic kidney disease (CKD) poses a severe health risk with high morbidity and mortality, profoundly affecting patient quality of life and survival. Despite advancements in research, the pathophysiology of CKD remains incompletely understood. Growing evidence links CKD with shifts in gut microbiota function and composition. Natural compounds, particularly polyphenols, have shown promise in CKD treatment due to their antioxidant and anti-inflammatory properties and their ability to modulate gut microbiota. This review discusses recent progress in uncovering the connections between gut microbiota and CKD, including microbiota changes across different kidney diseases. We also examine metabolite alterations,such as trimethylamine-N-oxide, tryptophan derivatives, branched-chain amino acids, short-chain fatty acids, and bile acids,which contribute to CKD progression. Further, we outline the mechanisms through which polyphenols exert therapeutic effects on CKD, focusing on signaling pathways like nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), mammalian target of rapamycin (mTOR), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), phosphatidylin-ositol-3-kinase (PI3K)/protein kinase B (Akt), and toll like receptors (TLR), as well as their impact on gut microbiota. Lastly, we consider how dietary polyphenols could be harnessed as bioactive drugs to slow CKD progression. Future research should prioritize multi-omics approaches to identify patients who would benefit from polyphenolic interventions, enabling personalized treatment strategies to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Cheng Li
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Department of Nephrology, Jiujiang University affiliated Hospital, Jiu’jiang, Jiang’xi, China
| | - Xulong Chen
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Weiwei Zha
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Sitian Fang
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nan’chang, Jiangxi, China
| | - Jiangwen Shen
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Lin Li
- School of Clinical Medical, Jiujiang University, Jiu’jiang, Jiang’xi, China
| | - Hongli Jiang
- Department of Blood Purification, Kidney Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
| | - PuXun Tian
- Department of Kidney Transplantation, Nephropathy Hospital, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaan’xi, China
- Institute of Organ Transplantation, Xi’an Jiaotong University, Xi’an, Shaan’xi, China
| |
Collapse
|
2
|
Cantu-Jungles TM, Agamennone V, Van den Broek TJ, Schuren FHJ, Hamaker B. Systematically-designed mixtures outperform single fibers for gut microbiota support. Gut Microbes 2025; 17:2442521. [PMID: 39704614 DOI: 10.1080/19490976.2024.2442521] [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: 05/15/2024] [Revised: 10/03/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Dietary fiber interventions to modulate the gut microbiota have largely relied on isolated fibers or specific fiber sources. We hypothesized that fibers systematically blended could promote more health-related bacterial groups. Initially, pooled in vitro fecal fermentations were used to design dietary fiber mixtures to support complementary microbial groups related to health. Then, microbial responses were compared for the designed mixtures versus their single fiber components in vitro using fecal samples from a separate cohort of 10 healthy adults. The designed fiber mixtures outperformed individual fibers in supporting bacterial taxa across donors resulting in superior alpha diversity and unexpected higher SCFA production. Moreover, unique shifts in community structure and specific taxa were observed for fiber mixtures that were not observed for single fibers, suggesting a synergistic effect when certain fibers are put together. Fiber mixture responses were remarkably more consistent than individual fibers across donors in promoting several taxa, especially butyrate producers from the Clostridium cluster XIVa. This is the first demonstration of synergistic fiber interactions for superior support of a diverse group of important beneficial microbes consistent across people, and unexpectedly high SCFA production. Overall, harnessing the synergistic potential of designed fiber mixtures represents a promising and more efficacious avenue for future prebiotic development.
Collapse
Affiliation(s)
- T M Cantu-Jungles
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - V Agamennone
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - T J Van den Broek
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - F H J Schuren
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - B Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 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
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025; 17:1038-1063. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [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] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
5
|
Seo B, Jeon K, Kim WK, Jang YJ, Cha KH, Ko G. Strain-Specific Anti-Inflammatory Effects of Faecalibacterium prausnitzii Strain KBL1027 in Koreans. Probiotics Antimicrob Proteins 2025; 17:1711-1724. [PMID: 38411865 DOI: 10.1007/s12602-024-10213-7] [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] [Accepted: 01/04/2024] [Indexed: 02/28/2024]
Abstract
Faecalibacterium prausnitzii is one of the most dominant commensal bacteria in the human gut, and certain anti-inflammatory functions have been attributed to a single microbial anti-inflammatory molecule (MAM). Simultaneously, substantial diversity among F. prausnitzii strains is acknowledged, emphasizing the need for strain-level functional studies aimed at developing innovative probiotics. Here, two distinct F. prausnitzii strains, KBL1026 and KBL1027, were isolated from Korean donors, exhibiting notable differences in the relative abundance of F. prausnitzii. Both strains were identified as the core Faecalibacterium amplicon sequence variant (ASV) within the healthy Korean cohort, and their MAM sequences showed a high similarity of 98.6%. However, when a single strain was introduced to mice with dextran sulfate sodium (DSS)-induced colitis, KBL1027 showed the most significant ameliorative effects, including alleviation of colonic inflammation and restoration of gut microbial dysbiosis. Moreover, the supernatant from KBL1027 elevated the secretion of IL-10 cytokine more than that of KBL1026 in mouse bone marrow-derived macrophage (BMDM) cells, suggesting that the strain-specific, anti-inflammatory efficacy of KBL1027 might involve effector compounds other than MAM. Through analysis of the Faecalibacterium pan-genome and comparative genomics, strain-specific functions related to extracellular polysaccharide biosynthesis were identified in KBL1027, which could contribute to the observed morphological disparities. Collectively, our findings highlight the strain-specific, anti-inflammatory functions of F. prausnitzii, even within the same core ASV, emphasizing the influence of their human origin.
Collapse
Affiliation(s)
- Boram Seo
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Personalized Diet Research Group, Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Kyungchan Jeon
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Woon-Ki Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - You Jin Jang
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
- N-Bio, Seoul National University, Seoul, Republic of Korea.
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea.
- KoBioLabs Inc., Seoul, Republic of Korea.
| |
Collapse
|
6
|
Tiwari S, Paramanik V. Role of Probiotics in Depression: Connecting Dots of Gut-Brain-Axis Through Hypothalamic-Pituitary Adrenal Axis and Tryptophan/Kynurenic Pathway involving Indoleamine-2,3-dioxygenase. Mol Neurobiol 2025; 62:7230-7241. [PMID: 39875781 DOI: 10.1007/s12035-025-04708-9] [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: 06/20/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
Depression is one of the most disabling mental disorders worldwide and characterized by symptoms including worthlessness, anhedonia, sleep, and appetite disturbances. Recently, studies have suggested that tryptophan (Trp) metabolism plays a key role in depressed mood through serotonin and kynurenine pathway involving enzyme tryptophan 5-monooxygenase (TPH) and indoleamine-2,3-dioxygenase (IDO) respectively. Moreover, during neuroinflammation, IDO is activated by proinflammatory cytokines and affects neurogenesis, cognition, disturbed hypothalamic-pituitary-adrenal (HPA) axis, and gut homeostasis by altering the gut bacteria and its metabolites like Trp derivatives. Furthermore, over the decades, researchers have focused on understanding communication between the human microbiome, especially gut microbiota, and mental health, called gut-brain-axis (GBA), particularly through Trp metabolism. Supplementation of probiotics in depression has gained attention from researchers and clinicians. However, there is limited information about probiotics supplementation on depression involving enzyme IDO and kynurenine pathway metabolites. This review discussed the potential role of probiotics in depression through the tryptophan/kynurenine pathway.
Collapse
Affiliation(s)
- Sneha Tiwari
- Cellular and Molecular Neurobiology and Drug Targeting Laboratory, Department of Zoology, Indira Gandhi National Tribal University, Amarkantak-484 887, MP, India
| | - Vijay Paramanik
- Cellular and Molecular Neurobiology and Drug Targeting Laboratory, Department of Zoology, Indira Gandhi National Tribal University, Amarkantak-484 887, MP, India.
| |
Collapse
|
7
|
Guédon G, Charron-Bourgoin F, Lacroix T, Hamadouche T, Soler N, Douzi B, Chiapello H, Leblond-Bourget N. Massive acquisition of conjugative and mobilizable integrated elements fuels Faecalibacterium plasticity and hints at their adaptation to the gut. Sci Rep 2025; 15:17013. [PMID: 40379875 PMCID: PMC12084326 DOI: 10.1038/s41598-025-99981-y] [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: 11/15/2024] [Accepted: 04/24/2025] [Indexed: 05/19/2025] Open
Abstract
Faecalibacterium is one of the most abundant bacteria of the human gut microbiota of healthy adults and is recognized to have positive effects on health. Here, we precisely and comprehensively analyzed the conjugative mobilome of four complete Faecalibacterium genomes. Despite lacking any plasmid, these bacteria harbor a vast arsenal of 130 elements, including 17 integrative and conjugative elements (ICEs) and 83 integrative and mobilizable elements (IMEs), collectively comprising 14-23% of the genome. Genome comparison of two strains isolated from the same fecal sample (Faecalibacterium and Roseburia strains) revealed almost identical elements indicating that transfer of ICEs and IMEs shape gut microbiome. ICEs and IMEs from Faecalibacterium encode many and diverse predicted functions such as defense and stress response (phages, multidrug, antibiotics, oxidative stress, biliar salts, antimicrobial peptides), nutrient import and metabolisms (Fe3+, carbohydrates) and riboflavin synthesis. This hints at their important role in the survival and adaptation of Faecalibacterium strains to the gut ecosystem. A rapid survey of 29 additional Faecalibacterium genomes uncovered many putative ICEs and IMEs, reinforcing their role in the rapid and massive evolution of Faecalibacterium genomes.
Collapse
Affiliation(s)
- Gérard Guédon
- Université de Lorraine, INRAE, DynAMic, 54000, Nancy, France
| | | | - Thomas Lacroix
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France
| | | | - Nicolas Soler
- Université de Lorraine, INRAE, DynAMic, 54000, Nancy, France
| | | | - Hélène Chiapello
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France
| | - Nathalie Leblond-Bourget
- Université de Lorraine, INRAE, DynAMic, 54000, Nancy, France.
- Université de Lorraine, UMR1128 DynAMic UL-INRAE, 54000, Nancy, France.
| |
Collapse
|
8
|
Zhu Y, Liu Q, Alffenaar JW, Wang S, Cao J, Dong S, Zhou X, Li X, Li X, Xiong H, Zhu L, Hu Y, Wang W. Gut Microbiota in Patients with Tuberculosis Associated with Different Drug Exposures of Antituberculosis Drugs. Clin Pharmacol Ther 2025. [PMID: 40326511 DOI: 10.1002/cpt.3687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
Interindividual variability in drug exposure can significantly influence treatment outcomes and may lead to drug concentration-related side effects during tuberculosis (TB) treatment. Although the gut microbiota is known to affect drug metabolism, its impact on anti-TB drugs has not been thoroughly explored. This study sought to elucidate the relationship between pre-treatment gut microbiota and drug exposure levels among patients with pulmonary TB. Two cohorts were analyzed: a discovery cohort (N = 99) and a validation cohort (N = 32), both comprising patients undergoing anti-TB therapy with rifampicin, isoniazid, pyrazinamide, and ethambutol. The gut microbiota patterns of participants from the discovery cohort and the validation cohort were profiled by 16S rRNA gene sequencing and metagenomics, respectively. Analyses of both cohorts robustly established a positive association between pre-treatment microbial diversity and drug exposure, as well as significant differences in gut microbiota composition across various drug exposure groups. At the species level, Faecalibacterium prausnitzii was positively associated with drug exposure to rifampicin. Moreover, functional analysis revealed that starch and sucrose metabolism and secondary bile acid biosynthesis were more abundant in the high drug exposure group. To identify biomarkers capable of stratifying patients based on their drug exposure levels, 11 taxa, represented by Faecalibacterium, were selected in the discovery cohort (AUC = 0.992) and were confirmed in the validation cohort with high predictive accuracy (AUC = 0.894). This study demonstrated a correlation between microbial dysbiosis and reduced exposure to anti-TB medications. Optimizing treatment by regulating gut microbiota to improve drug exposure levels requires further validation through larger scale multicenter clinical trials.
Collapse
Affiliation(s)
- Yue Zhu
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Qiao Liu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jan-Willem Alffenaar
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
- Department of Clinical Pharmacology, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Institute for Infectious Diseases, University of Sydney, Sydney, New South Wales, Australia
| | - Shanshan Wang
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Jiayi Cao
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Shulan Dong
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xiangkang Zhou
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xiaoxue Li
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Xuliang Li
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Haiyan Xiong
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Limei Zhu
- Department of Chronic Communicable Disease, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Yi Hu
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Li X, He N, Wang H, Wu Z, Wang M, Liang H, Xiao L, Yang Z, Li C, Xu P, Dai T, Li S, Zou Y. Therapeutic effect of Faecalibacterium longum CM04-06 on DSS-induced ulcerative colitis in mice. J Appl Microbiol 2025; 136:lxaf119. [PMID: 40372371 DOI: 10.1093/jambio/lxaf119] [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: 12/12/2024] [Revised: 04/06/2025] [Accepted: 05/14/2025] [Indexed: 05/16/2025]
Abstract
AIMS This study explores the impact of Faecalibacterium longum CM04-06 on inflammatory bowel disease (IBD) by regulating gut microbiota in mice. METHODS AND RESULTS We reanalyzed the distribution of the CM04-06 genome in the metagenome of the IBD cohort and observed a significantly higher abundance of CM04-06 in healthy individuals compared to patients with UC or CD. The prophylactic administration of CM04-06 was evaluated for its effects on intestinal microbial diversity and community composition after a two-week trial in mice. The intestinal microbiota was characterized using metagenomic sequencing of fecal samples on the DNBSEQ platform. CM04-06 treatment resulted in a significant reduction in the Disease Activity Index (DAI) and histological scores, as well as a decrease in the levels of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in both the colon and serum of DSS-induced mice. Furthermore, supplementation with CM04-06 significantly reduced the levels of pro-inflammatory cytokines in both the colon and serum. Additionally, CM04-06 enhanced the integrity of the intestinal epithelial barrier by increasing the expression of tight junction proteins and mucin. Moreover, we observed greater abundances of Faecalibaculum rodentium, Alistipes onderdonkii, Alistipes shahii, and Bifidobacterium animalis after CM04-06 treatment. CONCLUSIONS CM04-06 prevents and alleviates intestinal inflammation by modulating the composition of the microbiota community, increasing the abundance of beneficial probiotics, and suppressing pro-inflammatory cytokine levels.
Collapse
Affiliation(s)
- Xiaofang Li
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
- BGI Research, Shenzhen 518083, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Haoyu Wang
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhinan Wu
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengmeng Wang
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Liang Xiao
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen 518083, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Cunyin Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Ping Xu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Yuanqiang Zou
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen 518083, China
| |
Collapse
|
10
|
Ncho CM, Gupta V, Goel A, Jeong CM, Jung JY, Ha SY, Eom JU, Yang HS, Yang JK, Choi YH. Impact of dietary polyphenols from shredded, steam-exploded pine on growth performance, organ indices, meat quality, and cecal microbiota of broiler chickens. Poult Sci 2025; 104:105088. [PMID: 40154182 PMCID: PMC11995072 DOI: 10.1016/j.psj.2025.105088] [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: 12/01/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025] Open
Abstract
The chicken's gastrointestinal tract is home to complex and diverse microbial communities that can be manipulated to enhance health and productivity. Although polyphenols have recently attracted the attention of researchers due to their potent antioxidant capabilities, their impact on the gut microbiota remains largely unexplored. Hence, in this study, we conducted a comprehensive analysis of the effects of dietary supplementation with polyphenol-rich extract from shredded, steam-exploded pine particles (PSPP) on growth, meat quality, and gut microbial dynamics in broiler chickens. Supplementation of PSPP was found to significantly improve birds' FCR until the third week of the trial but only marginally affected meat quality. Based on metataxonomic analyses of the cecal microbiotas of broilers fed increasing concentrations of PSPP, dietary PSPP modulated the composition of the cecal microbiota of the birds with a concomitant increase of Bacteroidetes and a decrease in the Firmicutes population. Similar trends were observed for the proportions of Alistipes and Faecalibacterium at the genus level. Additionally, 43 unique bacterial species were detected in the cecal microbiome of birds fed with PSPP. However, microbial diversity did not vary significantly among treatment groups. A particularly interesting finding was the specialization observed in the microbiome of birds receiving PSPP supplementation. Microbial co-occurrence network analyses revealed substantial modifications in their network structure when compared to control birds. Families like Rikenellaceae and Eubacteriaceae were notably absent, and the number of microbial interactions was drastically lower in the PSPP-fed group. Microbial taxa modeling revealed that the impact of increasing dietary PSPP levels primarily affected genus-level taxa, showing a decreasing trend. Overall, this offers compelling evidence that continuous PSPP supplementation may not only alter the composition of intestinal microbes but also have a profound effect on the interactions among different microbial species. Conversely, PSPP had minimal effects on broilers' performance and meat quality.
Collapse
Affiliation(s)
- Chris Major Ncho
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Vaishali Gupta
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Akshat Goel
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Chae-Mi Jeong
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ji-Young Jung
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Si-Young Ha
- Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jeong-Uk Eom
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Han-Sul Yang
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jae-Kyung Yang
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yang-Ho Choi
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea.
| |
Collapse
|
11
|
Chen B, Guan L, Wu C, Gong Y, Wu L, Zhang M, Cao Z, Chen Y, Yang C, Wang B, Li Y, Li B, Bi Y, Ning G, Wang J, Wang W, Liu R. Gut Microbiota-Butyrate-PPARγ Axis Modulates Adipose Regulatory T Cell Population. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411086. [PMID: 39998325 PMCID: PMC12120792 DOI: 10.1002/advs.202411086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/18/2025] [Indexed: 02/26/2025]
Abstract
Gut microbiota is essential for the function of peripherally-induced regulatory T (pTreg) cells. However, how commensal bacteria affect thymically derived fat-resident Treg cells that harbor a unique expression of peroxisome proliferator-activated receptor (PPAR)-γ and suppress inflammation in visceral adipose tissue (VAT), is not well defined. Here it is revealed that microbiota depletion causes a drastic decline in Treg cell population in VAT, particularly those expressing ST2 (ST2+ Treg), which are largely restored after gut microbiome reconstruction. Mechanistically, gut microbiota-derived butyrate increases VAT ST2+ Treg cells through binding PPARγ. Butyrate supplementation and high fiber diet increase VAT ST2+ Treg population in obese mice, and ameliorated glucose tolerance and visceral inflammation. Furthermore, human omental adipose Treg cells show positive correlation with fecal butyrate and certain butyrate-producing microbes. This study identifies the critical role of gut microbiota-butyrate-PPARγ axis in maintaining VAT Treg population, pinpointing a potential approach to augment VAT Treg population and ameliorate inflammation.
Collapse
Affiliation(s)
- Banru Chen
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Lizhi Guan
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chao Wu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yiwen Gong
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Lei Wu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Minchun Zhang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yufei Chen
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chengcan Yang
- Department of General SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011China
| | - Bing Wang
- Department of General SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011China
| | - Yunqi Li
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Bin Li
- Department of Immunology and MicrobiologyShanghai Institute of ImmunologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yufang Bi
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Guang Ning
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Weiqing Wang
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Ruixin Liu
- Department of Endocrine and Metabolic DiseasesShanghai Institute of Endocrine and Metabolic DiseasesRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Shanghai National Clinical Research Center for Metabolic DiseasesKey Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR ChinaShanghai Key Laboratory for Endocrine TumorState Key Laboratory of Medical GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
12
|
Kithama M, Hassan YI, Yin X, Tang J, Clairmont L, Sienkiewicz O, Ross K, Lau CHF, Lepp D, Zhao X, Kiarie EG, Diarra MS. Shift of Microbiota and Modulation of Resistome in the Ceca of Broiler Chicken Fed Berry Pomace Alone or in Combination of a Multienzyme Mixture. Microorganisms 2025; 13:1044. [PMID: 40431218 PMCID: PMC12114191 DOI: 10.3390/microorganisms13051044] [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: 03/13/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Alternative feed additives are being investigated due to the restriction of antibiotics use to decrease antimicrobial resistance (AMR) in food-producing animals. This study investigated the effects of dietary American cranberry (Vaccinium macrocarpon) and wild blueberry (V. angustifolium) pomaces on the cecal microbiota and resistome profiles as well as the short-chain fatty acid levels. Male broiler chickens Cobb500 were fed a basal diet with either 55 ppm bacitracin methylene disalicylate (BMD); 0.5% (CRP0.5) and 1% (CRP1) cranberry pomace; and 0.5% (LBP0.5) and 1% (LBP1) lowbush blueberry pomace with or without a multienzyme mixture (ENZ). The results showed that at 21 days of age, the total coliform counts decreased in the CRP0.5-fed birds compared to BMD (p < 0.05). The use of pomace significantly increased the abundance of Lactobacillus and Bacteroides regardless of ENZ, while CRP decreased the Proteobacteria phylum abundance. In-feed ENZ tended to increase the relative abundance of genes conferring aminoglycoside resistance. Treatment with CRP0.5 decreased the abundance of cepA genes encoding for macrolide (MACROLIDE) and lincomycin (InuD) resistance while increasing those for tetracycline (tetO and tetX) resistance. These results showed, for the first time, the potential of the studied enzymes in influencing berry pomace's effects on antimicrobial resistance gene profiles in broilers.
Collapse
Affiliation(s)
- Munene Kithama
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Yousef I. Hassan
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| | - Xianhua Yin
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| | - Joshua Tang
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| | - Lindsey Clairmont
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| | - Olimpia Sienkiewicz
- Department of Animal Science, McGill University, Montreal, QC H9X 3V9, Canada; (O.S.)
| | - Kelly Ross
- Summerland Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Summerland, BC V0H 1Z0, Canada
| | - Calvin Ho-Fung Lau
- Ottawa Laboratory (Carling), Canadian Food Inspection Agency, Ottawa, ON K1A 0C6, Canada
| | - Dion Lepp
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, QC H9X 3V9, Canada; (O.S.)
| | - Elijah G. Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Moussa S. Diarra
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON N1G 5C9, Canada; (M.K.); (J.T.); (L.C.); (D.L.)
| |
Collapse
|
13
|
Fu L, Baranova A, Cao H, Zhang F. Gut microbiome links obesity to type 2 diabetes: insights from Mendelian randomization. BMC Microbiol 2025; 25:253. [PMID: 40289103 PMCID: PMC12034155 DOI: 10.1186/s12866-025-03968-8] [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: 10/27/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Research has established links between the gut microbiome (GM) and both obesity and type 2 diabetes (T2D), which is much discussed, but underexplored. This study employed body mass index (BMI) as the measurement of obesity to delve deeper into the correlations from a genetic perspective. METHODS We performed the Mendelian randomization (MR) analysis to examine the causal effects of GM on T2D and BMI, and vice versa. Genome-wide association study (GWAS) summary datasets were utilized for the analysis, including T2D (N = 933,970), BMI (N = 806,834), and two GM datasets from the international consortium MiBioGen (211 taxa, N = 18,340) and the Dutch Microbiome Project (DMP) (207 taxa, N = 7,738). These datasets mainly cover European populations, with additional cohorts from Asia and other regions. To further explore the potential mediating role of GM in the connections between BMI and T2D, their interaction patterns were summarized into a network. RESULTS MR analysis identified 9 taxa that showed protective properties against T2D. Seven species were within the Firmicutes and Bacteroidales phyla in the DMP, and two were from the MiBioGen (Odds Ratio (OR): 0.94-0.95). Conversely, genetic components contributing to the abundance of 12 taxa were associated with increased risks of T2D (OR: 1.04-1.12). Furthermore, T2D may elevate the abundance of seven taxa (OR: 1.03-1.08) and reduce the abundance of six taxa (OR: 0.93-0.97). In the analysis of the influence of the genetic component of BMI on GM composition, BMI affected 52 bacterial taxa, with 28 decreasing (OR: 0.75-0.92) and 24 increasing (OR: 1.08-1.27). Besides, abundances of 25 taxa were negatively correlated with BMI (OR: 0.95-0.99), while positive correlations were detected for 14 taxa (OR: 1.01-1.05). Notably, we uncovered 11 taxa genetically associated with both BMI and T2D, which formed an interactive network. CONCLUSIONS Our findings provide evidence for the GM-mediated links between obesity and T2D. The identification of relevant GM taxa offers valuable insights into the potential role of the microbiome in these diseases.
Collapse
Affiliation(s)
- Li Fu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
- Research Centre for Medical Genetics, Moscow, 115478, Russia
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
14
|
Wu X, Mei J, Qiao S, Long W, Feng Z, Feng G. Causal relationships between gut microbiota and male reproductive inflammation and infertility: Insights from Mendelian randomization. Medicine (Baltimore) 2025; 104:e42323. [PMID: 40295237 PMCID: PMC12039986 DOI: 10.1097/md.0000000000042323] [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: 06/28/2024] [Revised: 09/08/2024] [Accepted: 10/30/2024] [Indexed: 04/30/2025] Open
Abstract
The study observed interactions between gut microbiota and male reproductive health, noting that the causal relationships were previously unclear. It aimed to explore the potential cause-and-effect relationship between gut bacteria and male reproductive problems such as inflammation, infertility, and sperm functionality, using a two-sample Mendelian randomization method to examine these connections. The analysis found that certain bacterial genera, such as Erysipelatoclostridium (0.71 [0.55-0.92]), Parasutterella (0.74 [0.57-0.96]), Ruminococcaceae UCG-009 (0.77 [0.60-0.98]), and Slackia (0.69 [0.49-0.96]), showed protective effects against prostatitis. In contrast, other genera like Faecalibacterium (1.59 [1.08-2.34]), Lachnospiraceae UCG004 (1.64 [1.15-2.34]), Odoribacter (1.68 [1.01-2.81]), Paraprevotella (1.28 [1.03-1.60]), and Sutterella (1.58 [1.13-2.19]) were detrimental. Additionally, causal relationships were identified between 2 genera and orchitis and epididymitis, 3 genera and male infertility, and 5 genera and abnormal spermatozoa. Further analysis of sperm-related proteins revealed causal associations between specific bacterial genera and proteins such as SPACA3, SPACA7, SPAG11A, SPAG11B, SPATA9, SPATA20, and ZPBP4. The results remained robust after sensitivity analysis and reverse Mendelian randomization analysis. The study concluded that specific bacterial genera have causal roles in reproductive inflammation, infertility, and sperm-associated proteins. This provides a novel strategy for the early diagnosis and identification of therapeutic targets in reproductive inflammation and infertility.
Collapse
Affiliation(s)
- Xiaohong Wu
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pediatric, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingwen Mei
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pediatric, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shicun Qiao
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pediatric, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wen Long
- Department of Radiology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhoushan Feng
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pediatric, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guo Feng
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Yokota H, Tanaka Y, Ohno H. Coculture of Bifidobacterium bifidum G9-1 With Butyrate-Producing Bacteria Promotes Butyrate Production. Microbiol Immunol 2025. [PMID: 40269463 DOI: 10.1111/1348-0421.13224] [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: 11/25/2024] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Supplementation with Bifidobacterium bifidum G9-1 (BBG9-1) has been established to enhance the production of butyrate, a short-chain fatty acid (SCFA) known for its beneficial effects in alleviating constipation. We hypothesized that BBG9-1 alters gut microbiota such that bacteria that produce butyric acid from lactate and acetate become more abundant. In this study, we sought to determine whether BBG9-1 promotes the growth of butyrate-producing bacteria and thereby enhances butyrate production. BBG9-1 was cocultured with different butyrate-producing bacteria to compare differences in the SCFA production of cocultures and monocultures. We indeed detected significant increases in the production of SCFAs in cocultures compared to monocultures. Moreover, lactate and butyrate production increased in a time-dependent manner in the BBG9-1 and Faecalibacterium prausnitzii ID 6052 coculture. In addition, acetate production in cocultures initially increased until 16 h, followed by a decline between 20 and 24 h, and a subsequent significant increase at 48 h. Comparatively, lactate and acetate production in the BBG9-1 and Anaerostipes caccae JCM 13470T coculture peaked at 16 h and declined thereafter, and butyrate production increased in a time-dependent manner. In contrast, lactate, acetate, and butyrate production in the BBG9-1 and Roseburia hominis JCM 17582T coculture increased in a time-dependent manner. These findings indicate that butyrate-producing bacteria increase butyrate production by utilizing BBG9-1-produced lactate and acetate. Thus, the butyrate-mediated physiological activity of BBG9-1 could be attributed to an indirect enhancement of butyrate production.
Collapse
|
16
|
Sun X, Li W, Chen G, Hu G, Jia J. Faecalibacterium duncaniae Mitigates Intestinal Barrier Damage in Mice Induced by High-Altitude Exposure by Increasing Levels of 2-Ketoglutaric Acid. Nutrients 2025; 17:1380. [PMID: 40284246 PMCID: PMC12030221 DOI: 10.3390/nu17081380] [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: 03/16/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Exposure to high altitudes often results in gastrointestinal disorders. This study aimed to identify probiotic strains that can alleviate such disorders. Methods: We conducted a microbiome analysis to investigate the differences in gut microbiota among volunteers during the acute response and acclimatization phases at high altitudes. Subsequently, we established a mouse model of intestinal barrier damage induced by high-altitude exposure to further investigate the roles of probiotic strains and 2-ketoglutaric acid. Additionally, we performed untargeted metabolomics and transcriptomic analyses to elucidate the underlying mechanisms. Results: The microbiome analysis revealed a significant increase in the abundance of Faecalibacterium prausnitzii during the acclimatization phase. Faecalibacterium duncaniae (F. duncaniae) significantly mitigated damage to the intestinal barrier and the reduction of 2-ketoglutaric acid levels in the cecal contents induced by high-altitude exposure in mice. Immunohistochemistry and TUNEL staining demonstrated that high-altitude exposure significantly decreased the expression of ZO-1 and occludin while increasing apoptosis in ileal tissues. In contrast, treatment with F. duncaniae alleviated the loss of ZO-1 and occludin, as well as the apoptosis induced by high-altitude exposure. Furthermore, 2-ketoglutaric acid also mitigated this damage, reducing the loss of occludin and apoptosis in mice. Transcriptomic analysis indicated that high-altitude exposure significantly affects the calcium signaling pathway; conversely, the administration of F. duncaniae significantly influenced the PPAR signaling pathway, mineral absorption, and the regulation of lipolysis in adipocytes. Additionally, the expression of the FBJ osteosarcoma oncogene (Fos) was markedly reduced following the administration of F. duncaniae. Conclusions:F. duncaniae mitigates hypoxia-induced intestinal barrier damage by increasing levels of 2-ketoglutaric acid and shows promise as a probiotic, ultimately aiding travelers in adapting to high-altitude environments.
Collapse
Affiliation(s)
| | | | | | - Gaosheng Hu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; (X.S.); (W.L.); (G.C.)
| | - Jingming Jia
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; (X.S.); (W.L.); (G.C.)
| |
Collapse
|
17
|
Sabit H, Abouelnour S, Hassen BM, Magdy S, Yasser A, Wadan AHS, Abdel-Ghany S, Radwan F, Alqosaibi AI, Hafiz H, Awlya OFA, Arneth B. Anticancer Potential of Prebiotics: Targeting Estrogen Receptors and PI3K/AKT/mTOR in Breast Cancer. Biomedicines 2025; 13:990. [PMID: 40299687 PMCID: PMC12025111 DOI: 10.3390/biomedicines13040990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Estrogen receptors (ERs) play a critical role in breast cancer (BC) development and progression, with ERα being oncogenic and ERβ exhibiting tumor-suppressive properties. The interaction between ER signaling and other molecular pathways, such as PI3K/AKT/mTOR, influences tumor growth and endocrine resistance. Emerging research highlights the role of prebiotics in modulating gut microbiota, which may influence estrogen metabolism, immune function, and therapeutic responses in BC. This review explores the impact of prebiotics on estrogen receptor modulation, gut microbiota composition, immune regulation, and metabolic pathways in breast cancer. The potential of prebiotics as adjunctive therapies to enhance treatment efficacy and mitigate chemotherapy-related side effects is discussed. A comprehensive analysis of recent preclinical and clinical studies was conducted, examining the role of prebiotics in gut microbiota modulation, immune regulation, and metabolic reprogramming in breast cancer. The impact of short-chain fatty acids (SCFAs) derived from prebiotic fermentation on epigenetic regulation and endocrine resistance was also evaluated. Prebiotics were found to modulate the gut microbiota-estrogen axis, reduce inflammation, and influence immune responses. SCFAs demonstrated selective estrogen receptor downregulation and metabolic reprogramming, suppressing tumor growth. Synbiotic interventions mitigate chemotherapy-related side effects, improving the quality of life in breast cancer patients. Prebiotics offer a promising avenue for breast cancer prevention and therapy by modulating estrogen metabolism, immune function, and metabolic pathways. Future clinical trials are needed to validate their efficacy as adjunctive treatments in breast cancer management.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Sama Abouelnour
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Bassel M. Hassen
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Salma Magdy
- Department of Agri-Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Ahmed Yasser
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, Suez Governorate 15888, Egypt;
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Faisal Radwan
- Center for Coastal Environmental Health and Biomolecular Research, NCCOS/NOS/NOAA, Charleston, SC 29412, USA
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Hala Hafiz
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ohaad F. A. Awlya
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldingerstr. 1, 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr. 12, 35392 Giessen, Germany
| |
Collapse
|
18
|
Wang XM, Huang HJ, Sun XW, Wei RQ, Chen HY, Liu C, Liu SJ. Identification and Characterization of Two Novel Members of the Family Eubacteriaceae, Anaerofustis butyriciformans sp. nov. and Pseudoramibacter faecis sp. nov., Isolated from Human Feces. Microorganisms 2025; 13:916. [PMID: 40284751 PMCID: PMC12029686 DOI: 10.3390/microorganisms13040916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Members of Eubacteriaceae are involved in host health and diseases. Two Gram-stain-positive, strictly anaerobic, non-motile, non-spore-forming, and rod-shaped bacterial strains, HA2171T and HA2172T, were isolated from the feces of Chinese healthy donors. Based on 16S rRNA gene sequences, HA2171T and HA2172T belonged to the family Eubacteriaceae. Physiological and biochemical characterizations indicated that HA2171T and HA2172T were neutrophilic, mesophilic, and tolerant to low-concentration NaCl. The major cellular fatty acids (>10.0%) of HA2171T were C16:0, C14:0, C18:1ω7c, and C17:0 2-OH, and those of HA2172T were C14:0 and C16:0. MK-6 was the respiratory quinone in both strains. Phylogenetic and phylogenomic analyses showed that HA2171T was closest to Anaerofustis stercorihominis ATCC BAA-858T and that HA2172T as closest to Pseudoramibacter alactolyticus ATCC 23263T. Genome annotation revealed that the HA2171T and HA2172T were able to metabolize carbohydrates and produce acetate and butyrate. HA2172T contains genes associated with hydrogen sulfide production, which is a potential risk for diseases. Based on the phylogenetic, phenotypic, and chemotaxonomic characteristics, we propose that HA2171T and HA2172T represent two novel species, and the names Anaerofustis butyriciformans sp. nov. and Pseudoramibacter faecis sp. nov. are proposed.
Collapse
Affiliation(s)
- Xiao-Meng Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Hao-Jie Huang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Xin-Wei Sun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Rui-Qi Wei
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Hao-Yu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (X.-M.W.); (H.-J.H.); (X.-W.S.); (R.-Q.W.); (H.-Y.C.)
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
19
|
Kalkan AE, BinMowyna MN, Raposo A, Ahmad MF, Ahmed F, Otayf AY, Carrascosa C, Saraiva A, Karav S. Beyond the Gut: Unveiling Butyrate's Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025; 17:1305. [PMID: 40284169 PMCID: PMC12029953 DOI: 10.3390/nu17081305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate's role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate's growing recognition as a modulator of neurological health via its interaction with the gut-brain axis. Additionally, butyrate's neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer's, Parkinson's, autism spectrum disorder, and Huntington's disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected.
Collapse
Affiliation(s)
- Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia;
| | - Abdullah Y. Otayf
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain;
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
20
|
Gofron KK, Wasilewski A, Małgorzewicz S. Effects of GLP-1 Analogues and Agonists on the Gut Microbiota: A Systematic Review. Nutrients 2025; 17:1303. [PMID: 40284168 PMCID: PMC12029897 DOI: 10.3390/nu17081303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND GLP-1 analogues are a relatively new class of medications that form the cornerstone of diabetes treatment. They possess invaluable glucose-lowering properties without hypoglycemic effects as well as strong cardioprotective effects. The gut microbiome has become the focus of numerous studies, demonstrating its influence not only on the gut but also on the overall well-being of the entire body. However, the effects of GLP-1 analogs on gut microbiota remain uncertain. SCOPE OF REVIEW Our systematic review (based on PRISMA guidelines) aimed to gather knowledge on the effects of GLP-1 analogue medications on the composition, richness, and abundance of gut microbiota in both animal and human models. CONCLUSIONS Thirty-eight studies were included in this systematic review. GLP-1 analogues have demonstrated a notable impact on the composition, richness, and diversity of gut microbiota. We can conclude, following the obtained research results of our study, that liraglutide promotes the growth of beneficial genera relevant for beneficial metabolic functions. Exenatide and exendin-4 administration showed various effects on the microbiome composition in animal and human studies. In animal models, it increased genera associated with improved metabolism; however, in human models, genera linked to better metabolic functions and escalated inflammation increased. Following dulaglutide administration, increases in Bacteroides, Akkermansia, and Ruminococcus, genera connected to an improved metabolic model, were significant. Finally, varied results were obtained after semaglutide treatment, in which A. muciniphila, known for its positive metabolic functions, increased; however, microbial diversity decreased. Semaglutide treatment provided various results indicating many confounding factors in semaglutide's impact on the gut microbiota. Results varied due to dissimilarities in the studied populations and the duration of the studies. Further research is essential to confirm these findings and to better recognize their implications for the clinical outcomes of patients.
Collapse
Affiliation(s)
- Krzysztof Ksawery Gofron
- Student Scientific Circle at Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland
| | - Andrzej Wasilewski
- Student Scientific Association of Medical Chemistry and Immunochemistry, Wroclaw Medical University, Marii Skłodowskiej-Curie 48/50, 59-369 Wroclaw, Poland;
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland;
- Department of Nephrology, Transplantology, and Internal Medicine, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland
| |
Collapse
|
21
|
Eslami M, Adampour Z, Fadaee Dowlat B, Yaghmayee S, Motallebi Tabaei F, Oksenych V, Naderian R. A Novel Frontier in Gut-Brain Axis Research: The Transplantation of Fecal Microbiota in Neurodegenerative Disorders. Biomedicines 2025; 13:915. [PMID: 40299512 PMCID: PMC12025253 DOI: 10.3390/biomedicines13040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
The gut-brain axis (GBA) represents a sophisticated bidirectional communication system connecting the central nervous system (CNS) and the gastrointestinal (GI) tract. This interplay occurs primarily through neuronal, immune, and metabolic pathways. Dysbiosis in gut microbiota has been associated with multiple neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). In recent years, fecal microbiota transplantation (FMT) has gained attention as an innovative therapeutic approach, aiming to restore microbial balance in the gut while influencing neuroinflammatory and neurodegenerative pathways. This review explores the mechanisms by which FMT impacts the gut-brain axis. Key areas of focus include its ability to reduce neuroinflammation, strengthen gut barrier integrity, regulate neurotransmitter production, and reinstate microbial diversity. Both preclinical and clinical studies indicate that FMT can alleviate motor and cognitive deficits in PD and AD, lower neuroinflammatory markers in MS, and enhance respiratory and neuromuscular functions in ALS. Despite these findings, several challenges remain, including donor selection complexities, uncertainties about long-term safety, and inconsistencies in clinical outcomes. Innovations such as synthetic microbial communities, engineered probiotics, and AI-driven analysis of the microbiome hold the potential to improve the precision and effectiveness of FMT in managing neurodegenerative conditions. Although FMT presents considerable promise as a therapeutic development, its widespread application for neurodegenerative diseases requires thorough validation through well-designed, large-scale clinical trials. It is essential to establish standardized protocols, refine donor selection processes, and deepen our understanding of the molecular mechanisms behind its efficacy.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Zarifeh Adampour
- Institute of Science, Biotechnology and Biosafety Department, Eskishehir Osmangazi University, Eskishehir 26040, Türkiye;
| | - Bahram Fadaee Dowlat
- School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Shayan Yaghmayee
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Faezeh Motallebi Tabaei
- Department of Medical Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
22
|
Rytter H, Naimi S, Wu G, Lewis J, Duquesnoy M, Vigué L, Tenaillon O, Belda E, Vazquez-Gomez M, Touly N, Arnone D, Hao F, Ley RE, Clément K, Peyrin-Biroulet L, Patterson AD, Gewirtz AT, Chassaing B. In vitro microbiota model recapitulates and predicts individualised sensitivity to dietary emulsifier. Gut 2025; 74:761-774. [PMID: 39870396 PMCID: PMC12013555 DOI: 10.1136/gutjnl-2024-333925] [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: 09/19/2024] [Accepted: 12/26/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND Non-absorbed dietary emulsifiers, including carboxymethylcellulose (CMC), directly disturb intestinal microbiota, thereby promoting chronic intestinal inflammation in mice. A randomised controlled-feeding study (Functional Research on Emulsifiers in Humans, FRESH) found that CMC also detrimentally impacts intestinal microbiota in some, but not all, healthy individuals. OBJECTIVES This study aimed to establish an approach for predicting an individual's sensitivity to dietary emulsifiers via their baseline microbiota. DESIGN We evaluated the ability of an in vitro microbiota model (MiniBioReactor Arrray, MBRA) to reproduce and predict an individual donor's sensitivity to emulsifiers. Metagenomes were analysed to identify signatures of emulsifier sensitivity. RESULTS Exposure of human microbiotas, maintained in the MBRA, to CMC recapitulated the differential CMC sensitivity previously observed in FRESH subjects. Furthermore, select FRESH control subjects (ie, not fed CMC) had microbiotas that were highly perturbed by CMC exposure in the MBRA model. CMC-induced microbiota perturbability was associated with a baseline metagenomic signature, suggesting the possibility of using one's metagenome to predict sensitivity to dietary emulsifiers. Transplant of human microbiotas that the MBRA model deemed CMC-sensitive, but not those deemed insensitive, into IL-10-/- germfree mice resulted in overt colitis following CMC feeding. CONCLUSION These results suggest that an individual's sensitivity to emulsifier is a consequence of, and can thus be predicted by, examining their baseline microbiota, paving the way to microbiota-based personalised nutrition.
Collapse
Affiliation(s)
- Héloïse Rytter
- Microbiome-Host Interactions, INSERM U1306, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France
- INSERM U1016, CNRS UMR8104, Mucosal Microbiota in Chronic Inflammatory Diseases, Université de Paris, Paris, France
| | - Sabrine Naimi
- INSERM U1016, CNRS UMR8104, Mucosal Microbiota in Chronic Inflammatory Diseases, Université de Paris, Paris, France
| | - Gary Wu
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jim Lewis
- Center for Clinical Epidemiology and Biostatistics, Division of Gastroenterology and Hepatology,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maeva Duquesnoy
- Microbiome-Host Interactions, INSERM U1306, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France
- INSERM U1016, CNRS UMR8104, Mucosal Microbiota in Chronic Inflammatory Diseases, Université de Paris, Paris, France
| | - Lucile Vigué
- Robustness and Evolvability of Life, CNRS UMR10 8104, INSERM U1016, Université Paris Cité, Paris, France
| | - Olivier Tenaillon
- Robustness and Evolvability of Life, CNRS UMR10 8104, INSERM U1016, Université Paris Cité, Paris, France
| | - Eugeni Belda
- Inserm, Nutrition and Obesities: Systemic Approaches Research Unit, NutriOmics, Sorbonne University, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, IRD, Sorbonne Université, Bondy, France
| | - Marta Vazquez-Gomez
- Inserm, Nutrition and Obesities: Systemic Approaches Research Unit, NutriOmics, Sorbonne University, Paris, France
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, IRD, Sorbonne Université, Bondy, France
| | - Nina Touly
- INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France
- INSERM, NGERE, University of Lorraine, Nancy, France
- FHU-CURE, Nancy University Hospital, Vandoeuvre les Nancy, France
- CHRU Nancy, IHU Infiny, Vandoeuvre-les-Nancy, France
| | - Djésia Arnone
- INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France
- INSERM, NGERE, University of Lorraine, Nancy, France
- FHU-CURE, Nancy University Hospital, Vandoeuvre les Nancy, France
- CHRU Nancy, IHU Infiny, Vandoeuvre-les-Nancy, France
| | - Fuhua Hao
- Center for Molecular Toxicology and Carcinogenesis, Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ruth E Ley
- Microbiome Science, Max-Planck-Institute for Biology, Tübingen, Germany
| | - Karine Clément
- Inserm, Nutrition and Obesities: Systemic Approaches Research Unit, NutriOmics, Sorbonne University, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Laurent Peyrin-Biroulet
- INFINY Institute, Nancy University Hospital, Vandœuvre-lès-Nancy, France
- INSERM, NGERE, University of Lorraine, Nancy, France
- FHU-CURE, Nancy University Hospital, Vandoeuvre les Nancy, France
- CHRU Nancy, IHU Infiny, Vandoeuvre-les-Nancy, France
- Department of Gastroenterology, Nancy University Hospital, Vandoeuvre-lès-Nancy, France
| | - Andrew D Patterson
- Center for Molecular Toxicology and Carcinogenesis, Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Benoit Chassaing
- Microbiome-Host Interactions, INSERM U1306, CNRS UMR6047, Institut Pasteur, Université Paris Cité, Paris, France
- INSERM U1016, CNRS UMR8104, Mucosal Microbiota in Chronic Inflammatory Diseases, Université de Paris, Paris, France
- CHRU Nancy, IHU Infiny, Vandoeuvre-les-Nancy, France
| |
Collapse
|
23
|
Vitetta L, Bambling M, Strodl E. Persister Intestinal Bacteria, Epigenetics and Major Depression. FRONT BIOSCI-LANDMRK 2025; 30:26837. [PMID: 40302324 DOI: 10.31083/fbl26837] [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: 10/04/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 05/02/2025]
Abstract
The microbiota-gut-brain axis has been proposed as a potential modulator of mood disorders such as major depression. Complex bidirectional biochemical activities in this axis have been posited to participate in adverse mood disorders. Environmental and genetic factors have dominated recent discussions on depression. The prescription of antibiotics, antidepressants, adverse negative DNA methylation reactions and a dysbiotic gut microbiome have been cited as causal for the development and progression of depression. While research continues to investigate the microbiome-gut-brain axis, this review will explore the state of persistence of gut bacteria that underpins bacterial dormancy, possibly due to adverse environmental conditions and/or pharmaceutical prescriptions. Bacterial dormancy persistence in the intestinal microbial cohort could affect the role of bacterial epigenomes and DNA methylations. DNA methylations are highly motif driven exerting significant control on bacterial phenotypes that can disrupt bacterial metabolism and neurotransmitter formation in the gut, outcomes that can support adverse mood dispositions.
Collapse
Affiliation(s)
- Luis Vitetta
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2005, Australia
| | - Matthew Bambling
- Faculty of Medicine and Health, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Esben Strodl
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4058, Australia
| |
Collapse
|
24
|
Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int J Mol Sci 2025; 26:3059. [PMID: 40243712 PMCID: PMC11988433 DOI: 10.3390/ijms26073059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn's disease. The gut microbiota's role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies-including prebiotics, probiotics, postbiotics, and symbiotics-have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients' quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Pierluigi Puca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Federica di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Alfredo Papa
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
| |
Collapse
|
25
|
Pimenta M, Alexa M, Mabandza DB, Dulaurent S, Huynh BT, Gaschet M, Opatowski L, Breurec S, Ploy MC, Dagot C. Wastewater-based AMR surveillance associated with tourism on a Caribbean island (Guadeloupe). J Glob Antimicrob Resist 2025; 43:27-34. [PMID: 40154781 DOI: 10.1016/j.jgar.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/23/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVES Antimicrobial resistance (AMR) is a major public health concern worldwide. International travel is a risk factor for acquiring antibiotic-resistant bacteria (ARB) and antibiotic-resistance genes (ARGs). Therefore, understanding the transmission of ARB and ARGs is instrumental in tackling AMR. This longitudinal study aimed to assess the benefit of wastewater monitoring in Guadeloupe to evaluate the role of tourism in the spread of AMR. METHODS A wastewater-based surveillance (WBS) study was conducted to monitor AMR in Guadeloupe in 2022 during dry and wet seasons. We characterized the resistome, microbiome and exposome of water samples collected in wastewater treatment facilities of two cities with different levels of tourism activities, in the content of aircraft toilets, and the pumping station receiving effluents from hotels. RESULTS The results show that the WBS approach facilitates the differentiation of various untreated effluents concerning exposome, microbiome, and resistome, offering insights into AMR dissemination. Additionally, the findings reveal that microbiome and exposome are comparable across sites and seasons, while resistome characterisation at specific locations may be pertinent for health surveillance. The microbiome of aircraft was predominantly composed of anaerobic bacteria from human intestinal microbiota, whereas the other locations exhibited a blend of human and environmental bacteria. Notably, individuals arriving by air have not introduced clinically significant resistance genes. Exposome compounds have been shown to influence the resistome's variance. CONCLUSIONS Clear differences were seen between the aircraft and the local sampling sites, indicating that the contribution of tourism to the observed resistance in Guadeloupe is not significant.
Collapse
Affiliation(s)
- Mélanie Pimenta
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Maria Alexa
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Degrâce Batantou Mabandza
- Transmission, Reservoir and Diversity of Pathogens Unit, Pasteur Institute of Guadeloupe, Pointe-à-Pitre, France
| | - Sylvain Dulaurent
- Department of Pharmacology, Toxicology and Pharmacovigilance, Limoges University Hospital, Limoges, France
| | - Bich-Tram Huynh
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Margaux Gaschet
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Lulla Opatowski
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Sébastien Breurec
- Transmission, Reservoir and Diversity of Pathogens Unit, Pasteur Institute of Guadeloupe, Pointe-à-Pitre, France; Faculty of Medicine Hyacinthe Bastaraud, University of Antilles, Pointe-à-Pitre, France; Centre for Clinical Investigation 1424, INSERM, Pointe-à-Pitre/Les Abymes, France; Laboratory of clinical microbiology, University hospital Center of Guadeloupe, Pointe-à-Pitre/Les Abymes, France
| | - Marie-Cécile Ploy
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Christophe Dagot
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France.
| |
Collapse
|
26
|
Shi Z, Li M, Zhang C, Li H, Zhang Y, Zhang L, Li X, Li L, Wang X, Fu X, Sun Z, Zhang X, Tian L, Zhang M, Chen WH, Li Z. Butyrate-producing Faecalibacterium prausnitzii suppresses natural killer/T-cell lymphoma by dampening the JAK-STAT pathway. Gut 2025; 74:557-570. [PMID: 39653411 PMCID: PMC12013593 DOI: 10.1136/gutjnl-2024-333530] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Natural killer/T-cell lymphoma (NKTCL) is a highly aggressive malignancy with a dismal prognosis, and gaps remain in understanding the determinants influencing disease outcomes. OBJECTIVE To characterise the gut microbiota feature and identify potential probiotics that could ameliorate the development of NKTCL. DESIGN This cross-sectional study employed shotgun metagenomic sequencing to profile the gut microbiota in two Chinese NKTCL cohorts, with validation conducted in an independent Korean cohort. Univariable and multivariable Cox proportional hazards analyses were applied to assess associations between identified marker species and patient outcomes. Tumour-suppressing effects were investigated using comprehensive in vivo and in vitro models. In addition, metabolomics, RNA sequencing, chromatin immunoprecipitation sequencing, Western blot analysis, immunohistochemistry and lentiviral-mediated gene knockdown system were used to elucidate the underlying mechanisms. RESULTS We first unveiled significant gut microbiota dysbiosis in NKTCL patients, prominently marked by a notable reduction in Faecalibacterium prausnitzii which correlated strongly with shorter survival among patients. Subsequently, we substantiated the antitumour properties of F. prausnitzii in NKTCL mouse models. Furthermore, F. prausnitzii culture supernatant demonstrated significant efficacy in inhibiting NKTCL cell growth. Metabolomics analysis revealed butyrate as a critical metabolite underlying these tumour-suppressing effects, validated in three human NKTCL cell lines and multiple tumour-bearing mouse models. Mechanistically, butyrate suppressed the activation of Janus kinase-signal transducer and activator of transcription pathway through enhancing histone acetylation, promoting the expression of suppressor of cytokine signalling 1. CONCLUSION These findings uncover a distinctive gut microbiota profile in NKTCL and provide a novel perspective on leveraging the therapeutic potential of F. prausnitzii to ameliorate this malignancy.
Collapse
Affiliation(s)
- Zhuangzhuang Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Min Li
- Department of Bioinformatics and Systems Biology, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhang
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
- Chinese PLA General Hospital and Medical School, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongwen Li
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yue Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Lei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Ling Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xiaorui Fu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Zhenchang Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Li Tian
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| | - Wei-Hua Chen
- Department of Bioinformatics and Systems Biology, Huazhong University of Science and Technology, Wuhan, China
- School of Biological Science, Jining Medical University, Rizhao, Shandong, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Quan R, Decraecker L, Appeltans I, Cuende-Estévez M, Van Remoortel S, Aguilera-Lizarraga J, Wang Z, Hicks G, Wykosky J, McLean P, Denadai-Souza A, Hussein H, Boeckxstaens GE. Fecal Proteolytic Bacteria and Staphylococcal Superantigens Are Associated With Abdominal Pain Severity in Irritable Bowel Syndrome. Am J Gastroenterol 2025; 120:603-613. [PMID: 39166748 PMCID: PMC11864055 DOI: 10.14309/ajg.0000000000003042] [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: 06/04/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Changes in the composition of the gut microbiota have been associated with the development of irritable bowel syndrome (IBS). However, to what extent specific bacterial species relate to clinical symptoms remains poorly characterized. We investigated the clinical relevance of bacterial species linked with increased proteolytic activity, histamine production, and superantigen (SAg) production in patients with IBS. METHODS Fecal (n = 309) and nasal (n = 214) samples were collected from patients with IBS and healthy volunteers (HV). Clinical symptoms and gut transit time were evaluated. Bacterial abundance in feces and nasal swabs as well as fecal trypsin-like activity were assessed. RESULTS The percentage of fecal samples containing Staphylococcus aureus was significantly higher in IBS compared with HV. Forty-nine percent of S. aureus -positive fecal samples from patients with IBS were also positive for SAgs, compared with 12% of HV. Patients with IBS and positive fecal SAg-producing S. aureus reported higher pain scores than those without S. aureus . Moreover, increased fecal proteolytic activity was associated with abdominal pain. Fecal abundance of Paraprevotella clara and Alistipes putredinis was significantly decreased in IBS, particularly in samples with higher proteolytic activity. Patients with lower Alistipes putredinis or Faecalibacterium prausnitzii abundance reported more severe abdominal pain. DISCUSSION In keeping with our preclinical findings, we show that increased presence of SAg-producing S. aureus in fecal samples of patients with IBS is associated with increased levels of abdominal pain. We also show that increased fecal proteolytic activity is associated with increased abdominal pain in patients with IBS.
Collapse
Affiliation(s)
- Runze Quan
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Lisse Decraecker
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - María Cuende-Estévez
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Samuel Van Remoortel
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Javier Aguilera-Lizarraga
- Laboratory of Sensory Neurophysiology and Pain, Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Zheng Wang
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | | | | | | | - Alexandre Denadai-Souza
- Laboratory of Mucosal Biology, Hepatology Research Unit, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Guy E. Boeckxstaens
- Center for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Chen B, Li Y, Li Z, Hu X, Zhen H, Chen H, Nie C, Hou Y, Zhu S, Xiao L, Li T. Vitamin E ameliorates blood cholesterol level and alters gut microbiota composition: A randomized controlled trial. Nutr Metab Cardiovasc Dis 2025:103964. [PMID: 40087044 DOI: 10.1016/j.numecd.2025.103964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND AIMS Antioxidants, including vitamin E (VE) and grape seed extract (GSE), as anti-aging supplementation have been widely used to improve human health. The gut microbiota plays a crucial role in health and affects the treatment effect of various interventions. However, the role of gut microbiota in VE remains unclear. This study aimed to assess the longitudinal impact of VE treatment on body health and the gut microbiota. METHODS AND RESULTS A randomized controlled trial was conducted with 90 healthy individuals. The participants were randomly assigned to three groups: a treatment group receiving VE, another antioxidant treatment group receiving GSE, and a control group receiving a placebo. We found that VE ameliorated blood cholesterol levels by reducing the levels of low-density lipoprotein cholesterol (LDL-C) in healthy volunteers. After the intervention, there was an increase in the relative abundance of short-chain fatty acid (SCFA)-producing bacteria and bile acid metabolizers. Specifically, the abundances of Lachnospira sp. and Faecalibacterium spp. increased in the VE. Interestingly, the gut microbiota of poor responders harbored a greater proportion of disease-associated bacterial species. CONCLUSIONS VE could promote health by lowering LDL-C, partly and indirectly by affecting gut bacteria with the ability to produce SCFAs or metabolize bile acids. REGISTRATION NUMBER FOR CLINICAL TRIALS The clinical trial was registered on August 28, 2021. Registration number was ChiCTR2100050567 (https://www.chictr.org.cn).
Collapse
Affiliation(s)
- Bangwei Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China; BGI Genomics, Shenzhen, China
| | - Yaxin Li
- BGI Genomics, Shenzhen, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Xiaojie Hu
- Department of Gastrointestinal Surgery, Hebei General Hospital, Shijiazhuang, China
| | | | | | | | | | | | | | - Tao Li
- BGI Genomics, Shenzhen, China; BGI Research, Shenzhen, China.
| |
Collapse
|
29
|
Ali AY, Zahran SA, Eissa M, Kashef MT, Ali AE. Gut microbiota dysbiosis and associated immune response in systemic lupus erythematosus: impact of disease and treatment. Gut Pathog 2025; 17:10. [PMID: 39966979 PMCID: PMC11834511 DOI: 10.1186/s13099-025-00683-7] [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: 11/10/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Gut microbial dysbiosis and leaky gut play a role in systemic lupus erythematosus (SLE). Geographical location and dietary habits affect the microbiome composition in diverse populations. This study explored the gut microbiome dysbiosis, leaky gut, and systemic immune response to gut bacterial consortium in patients with SLE exhibiting mild/moderate and severe disease activity. METHODS Fecal and blood samples were collected from patients with SLE and healthy volunteers. Genomic DNA was extracted from the stool samples and subjected to 16S rRNA amplicon sequencing and microbiome profiling. Additionally, enzyme-linked immunosorbent assays were employed to determine the serum lipopolysaccharide level, as an assessment of gut permeability, and the systemic immune response against gut bacteria. RESULTS Patients with SLE showed significantly lower gut bacterial richness and diversity, indicated by observed OTUs (56.6 vs. 74.44; p = 0.0289), Shannon (3.05 vs. 3.45; p = 0.017) and Simpson indices (0.91 vs. 0.94; p = 0.033). A lower Firmicutes-to-Bacteroidetes ratio (1.07 vs. 1.69; p = 0.01) was observed, with reduced genera such as Ruminococcus 2 (0.003 vs. 0.026; p = 0.0009) and Agathobacter (0.003 vs. 0.012; p < 0.0001) and elevated Escherichia-Shigella (0.04 vs. 0.006; p < 0.0001) and Bacteroides (0.206 vs. 0.094; p = 0.033). Disease severity was associated with a higher relative abundance of Prevotella (0.001 vs. 0.0001; p = 0.04). Medication effects included lower Romboutsia (0.0009 vs. 0.011; p = 0.005) with azathioprine and higher Prevotella (0.003 vs. 0.0002; p = 0.038) with cyclophosphamide. Furthermore, categorization by prednisolone dosage revealed significantly higher relative abundances of Slackia (0.0007 vs. 0.00002; p = 0.0088), Romboutsia (0.009 vs. 0.002; p = 0.0366), and Comamonas (0.002 vs. 0.00007; p = 0.0249) in patients receiving high-dose prednisolone (> 10 mg/day). No differences in serum lipopolysaccharide levels were found, but SLE patients exhibited elevated serum gut bacterial antibody levels, suggesting a systemic immune response. CONCLUSION This study confirms the gut microbiome dysbiosis in patients with SLE, influenced by disease severity and specific medication usage.
Collapse
Affiliation(s)
- Aya Y Ali
- Microbiology & Immunology Department, Faculty of Pharmacy, Future University in Egypt, Cairo, 12311, Egypt
| | - Sara A Zahran
- Microbiology & Immunology Department, Faculty of Pharmacy, Future University in Egypt, Cairo, 12311, Egypt.
| | - Mervat Eissa
- Rheumatology & Rehabilitation Department, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| | - Mona T Kashef
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Amal Emad Ali
- Microbiology & Immunology Department, Faculty of Pharmacy, Future University in Egypt, Cairo, 12311, Egypt
| |
Collapse
|
30
|
Yassin LK, Nakhal MM, Alderei A, Almehairbi A, Mydeen AB, Akour A, Hamad MIK. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat 2025; 19:1504065. [PMID: 40012737 PMCID: PMC11860919 DOI: 10.3389/fnana.2025.1504065] [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: 09/30/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a significant role in the maintenance of brain structure and function. The MGBA serves as a conduit between the CNS and the ENS, facilitating communication between the emotional and cognitive centers of the brain via diverse pathways. In the initial stages of this review, we will examine the way how MGBA affects neurogenesis, neuronal dendritic morphology, axonal myelination, microglia structure, brain blood barrier (BBB) structure and permeability, and synaptic structure. Furthermore, we will review the potential mechanistic pathways of neuroplasticity through MGBA influence. The short-chain fatty acids (SCFAs) play a pivotal role in the MGBA, where they can modify the BBB. We will therefore discuss how SCFAs can influence microglia, neuronal, and astrocyte function, as well as their role in brain disorders such as Alzheimer's disease (AD), and Parkinson's disease (PD). Subsequently, we will examine the technical strategies employed to study MGBA interactions, including using germ-free (GF) animals, probiotics, fecal microbiota transplantation (FMT), and antibiotics-induced dysbiosis. Finally, we will examine how particular bacterial strains can affect brain structure and function. By gaining a deeper understanding of the MGBA, it may be possible to facilitate research into microbial-based pharmacological interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
31
|
Cai X, Ren F, Yao Y. Gut microbiota and their metabolites in the immune response of rheumatoid arthritis: Therapeutic potential and future directions. Int Immunopharmacol 2025; 147:114034. [PMID: 39805176 DOI: 10.1016/j.intimp.2025.114034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent joint inflammation, damage, and loss of function. In recent years, the role of gut microbiota and its metabolites in immune regulation has attracted increasing attention. The gut microbiota influences the host immune system's homeostasis through various mechanisms, regulating the differentiation, function, and immune tolerance of immune cells. Dysbiosis of the gut microbiota in RA patients is closely associated with abnormal activation of immune cells and excessive secretion of inflammatory cytokines. Metabolites produced by the gut microbiota, such as short-chain fatty acids (SCFAs), tryptophan metabolites, bile acids, and amino acid metabolites, play a critical role in immune responses, regulating the functions of immune cells like T cells, B cells, and macrophages, and inhibiting the release of pro-inflammatory cytokines. Restoring the balance of the gut microbiota and optimizing the production of metabolic products may become a new strategy for RA treatment. This review discusses the role of gut microbiota and its metabolites in the immune response of RA, exploring how they influence the immunopathological process of RA through the regulation of immune cells and key immune factors. It also provides a theoretical basis for future therapeutic strategies based on gut microbiota modulation.
Collapse
Affiliation(s)
- Xiaoyu Cai
- Department of Pharmacy Hangzhou First People's Hospital Hangzhou China.
| | - Fujia Ren
- Department of Pharmacy Hangzhou Women's Hospital Hangzhou China
| | - Yao Yao
- Department of Pharmacy Women's Hospital School of Medicine Zhejiang University Hangzhou China
| |
Collapse
|
32
|
Gulyaeva A, Liu L, Garmaeva S, Kruk M, Weersma RK, Harmsen HJM, Zhernakova A. Identification and characterization of Faecalibacterium prophages rich in diversity-generating retroelements. Microbiol Spectr 2025; 13:e0106624. [PMID: 39745426 PMCID: PMC11792537 DOI: 10.1128/spectrum.01066-24] [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: 04/28/2024] [Accepted: 11/25/2024] [Indexed: 02/05/2025] Open
Abstract
Metagenomics has revealed the incredible diversity of phages within the human gut. However, very few of these phages have been subjected to in-depth experimental characterization. One promising method of obtaining novel phages for experimental characterization is through induction of the prophages integrated into the genomes of cultured gut bacteria. Here, we developed a bioinformatic approach to prophage identification that builds on prophage genomic properties, existing prophage-detecting software, and publicly available virome sequencing data. We applied our approach to 22 strains of bacteria belonging to the genus Faecalibacterium, resulting in identification of 15 candidate prophages, and validated the approach by demonstrating the activity of five prophages from four of the strains. The genomes of three active phages were identical or similar to those of known phages, while the other two active phages were not represented in the Viral RefSeq database. Four of the active phages possessed a diversity-generating retroelement (DGR), and one retroelement had two variable regions. DGRs of two phages were active at the time of the induction experiments, as evidenced by nucleotide variation in sequencing reads. We also predicted that the host range of two active phages may include multiple bacterial species. Finally, we noted that four phages were less prevalent in the metagenomes of inflammatory bowel disease patients compared to a general population cohort, a difference mainly explained by differences in the abundance of the host bacteria. Our study highlights the utility of prophage identification and induction for unraveling phage molecular mechanisms and ecological interactions.IMPORTANCEWhile hundreds of thousands of phage genomes have been discovered in metagenomics studies, only a few of these phages have been characterized experimentally. Here, we explore phage characterization through bioinformatic identification of prophages in genomes of cultured bacteria, followed by prophage induction. Using this approach, we detect the activity of five prophages in four strains of commensal gut bacteria Faecalibacterium. We further note that four of the prophages possess diversity-generating retroelements implicated in rapid mutation of phage genome loci associated with phage-host and phage-environment interactions and analyze the intricate patterns of retroelement activity. Our study highlights the potential of prophage characterization for elucidating complex molecular mechanisms employed by the phages.
Collapse
Affiliation(s)
- Anastasia Gulyaeva
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Sanzhima Garmaeva
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Marloes Kruk
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Rinse K. Weersma
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, the Netherlands
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
33
|
Qu Q, Dou Q, Xiang Z, Yu B, Chen L, Fan Z, Zhao X, Yang S, Zeng P. Population-level gut microbiome and its associations with environmental factors and metabolic disorders in Southwest China. NPJ Biofilms Microbiomes 2025; 11:24. [PMID: 39905038 PMCID: PMC11794850 DOI: 10.1038/s41522-025-00661-6] [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: 08/31/2023] [Accepted: 01/26/2025] [Indexed: 02/06/2025] Open
Abstract
Gut microbiota affects host health and disease. Large-scale cohorts have explored the interactions between the microbiota, host, and environment to reveal the disease-associated microbiota variation. A population-level gut metagenomic cohort is still rare in China. Here, we performed metagenomic sequencing on fecal samples from the CMEC Microbiome Project in Southwest China. In this study, we identified host socioeconomics, diet, lifestyle, and medical measurements that were significantly associated with microbiome function and composition. We revealed extensive novel associations between the host microbiome and common metabolic disorders. Our results provide new insight into associations of gut microbiota with metabolic disorders so as to support the translation of gut microbiome findings into potential clinical practice.
Collapse
Affiliation(s)
- Qianyu Qu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qingyu Dou
- National Clinical Research Center of Geriatrics, Geriatric Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhejun Xiang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Bin Yu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, China
| | - Lili Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zhenxin Fan
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Shujuan Yang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Peibin Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
34
|
Chen J, Guo S, Shi S. Effects of water acidifiers on the growth performance, intestinal function and gut microflora in broilers. Br Poult Sci 2025:1-8. [PMID: 39898934 DOI: 10.1080/00071668.2025.2454958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025]
Abstract
1. This study evaluated the effect of acidified drinking water on the gastrointestinal function and intestinal health of broilers.2. A total of 630 one-day-old male broilers (Arbor Acre) were randomly assigned to one of three treatment groups: drinking water treatment (CON), drinking water + 0.5 ml Selko pH®/L (Selko pH), or + 0.85 ml Forticoat®/L (Forticoat) treated groups. Performance data, gut and digesta samples were collected from the broilers at the age of 21 and 42 d.3. The results showed that acidifying drinking water had no significant effect on body weight or average daily gain (ADG). However, addition of Forticoat significantly increased (p < 0.05) feed conversion ratio (FCR) throughout the experimental period and significantly increased (p < 0.05) pepsin activity on d 21. The Selko pH supplemented drinking water significantly increased (p < 0.05) the relative length of the duodenum and jejunum on d 21. The relative length of the jejunum and caecum on d 42 compared to birds receiving CON. The addition of the Forticoat to drinking water significantly increased (p < 0.05) the relative length of the jejunum and caecum on d 42 than for samples from birds in the CON group. In the caecal chyme, abundance of Blautia, Bifidobasterium, Faecalibacterium, Limosilactobacillus and Akkermania spp. on d 21 were significantly higher (p < 0.05) in the caecum of birds receiving Selko pH than those in CON group and the number of Escherichia Shigella in Selko pH and Forticoat group were significantly lower (p < 0.05).4. Overall, adding Seiko pH and Forticoat to drinking water improved pepsin activity, reduced the number of caecal pathogens, increased the number of beneficial bacteria and improved intestinal health in broilers.
Collapse
Affiliation(s)
- J Chen
- Department of Feed and Nutrition, Jiangsu Institute of Poultry Science, Yangzhou, China
| | - S Guo
- Department of Feed and Nutrition, Jiangsu Institute of Poultry Science, Yangzhou, China
| | - S Shi
- Department of Feed and Nutrition, Jiangsu Institute of Poultry Science, Yangzhou, China
| |
Collapse
|
35
|
Chen E, Ajami NJ, White DL, Liu Y, Gurwara S, Hoffman K, Graham DY, El-Serag HB, Petrosino JF, Jiao L. Dairy Consumption and the Colonic Mucosa-Associated Gut Microbiota in Humans-A Preliminary Investigation. Nutrients 2025; 17:567. [PMID: 39940425 PMCID: PMC11820694 DOI: 10.3390/nu17030567] [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: 01/01/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Dairy consumption has been associated with various health outcomes that may be mediated by changes in gut microbiota. METHODS This cross-sectional study investigated the association between the colonic mucosa-associated gut microbiota and the self-reported intake of total dairy, milk, cheese, and yogurt. A total of 97 colonic mucosal biopsies collected from 34 polyp-free individuals were analyzed. Dairy consumption in the past year was assessed using a food frequency questionnaire. The 16S rRNA gene V4 region was amplified and sequenced. Operational taxonomic unit (OTU) classification was performed using the UPARSE and SILVA databases. OTU diversity and relative abundance were compared between lower vs. higher dairy consumption groups. Multivariable negative binomial regression models for panel data were used to estimate the incidence rate ratio and 95% confidence interval for bacterial counts and dairy consumption. False discovery rate-adjusted p values (q value) < 0.05 indicated statistical significance. RESULTS Higher total dairy and milk consumption and lower cheese consumption were associated with higher alpha microbial diversity (adjusted p values < 0.05). Higher total dairy and milk consumption was also associated with higher relative abundance of Faecalibacterium. Higher milk consumption was associated with higher relative abundance of Akkermansia. Higher total dairy and cheese consumption was associated with lower relative abundance of Bacteroides. CONCLUSIONS Dairy consumption may influence host health by modulating the structure and composition of the colonic adherent gut microbiota.
Collapse
Affiliation(s)
- Ellie Chen
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
| | - Nadim J. Ajami
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Donna L. White
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
- Houston VA HSR&D Center for Innovations in Quality, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Yanhong Liu
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Shawn Gurwara
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
| | - Kristi Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - David Y. Graham
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Section of Gastroenterology, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, USA
| | - Hashem B. El-Serag
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
- Houston VA HSR&D Center for Innovations in Quality, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine (BCM), Houston, TX 77030, USA
- Section of Gastroenterology, Effectiveness and Safety, Michael E DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, USA
| | - Joseph F. Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine (BCM), Houston, TX 77030, USA
- Texas Medical Center Digestive Disease Center, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Li Jiao
- Department of Medicine, Baylor College of Medicine (BCM), Houston, TX 77030, USA (D.Y.G.); (H.B.E.-S.)
| |
Collapse
|
36
|
Richardson M, Zhao S, Lin L, Sheth RU, Qu Y, Lee J, Moody T, Ricaurte D, Huang Y, Velez-Cortes F, Urtecho G, Wang HH. SAMPL-seq reveals micron-scale spatial hubs in the human gut microbiome. Nat Microbiol 2025; 10:527-540. [PMID: 39901058 PMCID: PMC12120799 DOI: 10.1038/s41564-024-01914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/12/2024] [Indexed: 02/05/2025]
Abstract
The local arrangement of microbes can profoundly impact community assembly, function and stability. However, our understanding of the spatial organization of the human gut microbiome at the micron scale is limited. Here we describe a high-throughput and streamlined method called Split-And-pool Metagenomic Plot-sampling sequencing (SAMPL-seq) to capture spatial co-localization in a complex microbial consortium. The method obtains microbial composition of micron-scale subcommunities through split-and-pool barcoding. SAMPL-seq analysis of the healthy human gut microbiome identified bacterial taxa pairs that consistently co-occurred both over time and across multiple individuals. These co-localized microbes organize into spatially distinct groups or 'spatial hubs' dominated by Bacteroidaceae, Ruminococcaceae and Lachnospiraceae families. Using inulin as a dietary perturbation, we observed reversible spatial rearrangement of the gut microbiome where specific taxa form new local partnerships. Spatial metagenomics using SAMPL-seq can unlock insights into microbiomes at the micron scale.
Collapse
Affiliation(s)
- Miles Richardson
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Shijie Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Liyuan Lin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Ravi U Sheth
- Department of Systems Biology, Columbia University, New York, NY, USA
- Kingdom Supercultures, Brooklyn, NY, USA
| | - Yiming Qu
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Jeongchan Lee
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Thomas Moody
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Deirdre Ricaurte
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Yiming Huang
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Florencia Velez-Cortes
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Guillaume Urtecho
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Harris H Wang
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
37
|
Trezzi S, Scaccabarozzi G, Nossa R, Piazza C, Bianchi AR, Rosi E, Tizzoni F, Mauri M, Camillo L, Baragetti A, Molteni M, Campanella V, Mauro L, Cremonesi P, Severgnini M, Monroy MM, Castiglioni B, Sparvoli F, Pisano S, Pozzi M, Crippa A, Nobile M. Behavioural, cognitive, and neurophysiological effects of a synbiotic supplementation enriched with pigmented corn extract or cornstarch in drug-naïve children with attention-deficit hyperactivity disorder: A randomised, double-blind, comparison-controlled clinical trial. Clin Nutr ESPEN 2025; 65:408-417. [PMID: 39710171 DOI: 10.1016/j.clnesp.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/21/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND & AIMS Considerable interest has been recently given to the potential role of the gut-brain axis (GBA) -a two-way communication network between the gut microbiota and the central nervous system- in the pathogenesis of attention-deficit hyperactivity disorder (ADHD), suggesting the potential usefulness of probiotic and synbiotic supplementations. In light of the limited available evidence, synbiotic efficacy in ADHD children not taking medications should be clarified. This study aimed to investigate the efficacy of a synbiotic dietary supplementation on fatty acids levels as well as on microbiota composition, behaviour, cognition, and brain function in children with ADHD. METHODS A total of 41 drug-naïve school-aged children diagnosed with ADHD were enrolled in a 3-month randomised, double-blind, comparison-controlled clinical trial, receiving either a synbiotic mix (COMP group) or the same synbiotic mix enriched with an additional extract from pigmented corn (EXP group). Changes in levels of some specific short-chain and branched-chain fatty acids were considered as primary outcomes. Secondary outcome measures included gut microbiota profiling, Child Behaviour Checklist, Conners Parent Rating Scale-revised, computerised cognitive tasks, and haemodynamic response to a Go-NoGo task measured by fNIRS. RESULTS No superiority of the EXP synbiotic mix was observed. Analysis of fatty acids did not reveal any significant difference between groups. Children in the COMP group reported a slightly greater improvement than those in the EXP group in focused attention and in the haemodynamic response to a cognitive task. CONCLUSIONS This study shows that pigmented corn extract does not enhance the effects of the synbiotic supplementation in ADHD children in terms of fatty acid production, microbiota composition, clinical, cognitive and neurophysiological measures. However, a synbiotic mix of probiotics plus prebiotic acacia fibre and cornstarch could have some promising effects on ADHD symptoms, which warrants further research. Future studies should also continue to explore the potential of fNIRS for monitoring the effects of interventions that target the GBA. TRIAL REGISTRATION ClinicalTrials.gov: NCT06005506.
Collapse
Affiliation(s)
- Sara Trezzi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Gaia Scaccabarozzi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Roberta Nossa
- Bioengineering Lab, Scientific Institute, IRCSS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Caterina Piazza
- Bioengineering Lab, Scientific Institute, IRCSS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Alessandro Rodolfo Bianchi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Eleonora Rosi
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Federica Tizzoni
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Maddalena Mauri
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Laura Camillo
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Massimo Molteni
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| | | | - Luca Mauro
- NEMO Clinical Centre, Serena Onlus Foundation, Milan, MI, Italy
| | - Paola Cremonesi
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, LO, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, MI, Italy
| | - Mariela Mejia Monroy
- Institute of Agricultural Biology and Biotechnology, National Research Council, Pisa, PI, Italy
| | - Bianca Castiglioni
- Institute of Agricultural Biology and Biotechnology, National Research Council, Lodi, LO, Italy
| | - Francesca Sparvoli
- Institute of Agricultural Biology and Biotechnology, National Research Council, Milan, MI, Italy
| | - Simone Pisano
- Department of Neuroscience, AORN Santobono-Pausilipon, Via Mario Fiore 6, Naples, Italy; Department of Translational Medical Sciences, Federico II University, Via Pansini 5, Naples, Italy
| | - Marco Pozzi
- Child and Adolescent Psychiatry Unit, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, LC, Italy
| | - Alessandro Crippa
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy.
| | - Maria Nobile
- Child Psychopathology Unit, Scientific Institute, IRCSS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, LC, Italy
| |
Collapse
|
38
|
Hindle VK, Veasley NM, Holscher HD. Microbiota-Focused Dietary Approaches to Support Health: A Systematic Review. J Nutr 2025; 155:381-401. [PMID: 39486521 PMCID: PMC11867136 DOI: 10.1016/j.tjnut.2024.10.043] [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: 07/10/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Diet affects the intestinal microbiota. Increasingly, research is linking the intestinal microbiota to various human health outcomes. Consumption of traditional prebiotics (inulin, fructo-oligosaccharides, and galacto-oligosaccharides) confers health benefits through substrate utilization by select intestinal microorganisms, namely Bifidobacterium and Lactobacilli spp. A similar but distinct concept focused on microorganisms to support human health is through direct consumption of certain live microorganisms recognized as probiotics, which classically include Lactobacilli or Bifidobacterium strains. With advances in sequencing technologies and culturing techniques, other novel functional intestinal microorganisms are being increasingly identified and studied to determine how they may underpin human health benefits. These novel microorganisms are targeted for enrichment within the autochthonous intestinal microbiota through dietary approaches and are also gaining interest as next-generation probiotics because of their purported beneficial properties. Thus, characterizing dietary approaches that nourish select microorganisms in situ is necessary to propel biotic-focused research forward. As such, we reviewed the literature to summarize findings on dietary approaches that nourish the human intestinal microbiota and benefit health to help fill the gap in knowledge on the connections between certain microorganisms, the metabolome, and host physiology. The overall objective of this systematic review was to summarize the impact of dietary interventions with the propensity to nourish certain intestinal bacteria, affect microbial metabolite concentrations, and support gastrointestinal, metabolic, and cognitive health in healthy adults. Findings from the 17 randomized controlled studies identified in this systematic review indicated that dietary interventions providing dietary fibers, phytonutrients, or unsaturated fatty acids differentially enriched Akkermansia, Bacteroides, Clostridium, Eubacterium, Faecalibacterium, Roseburia, and Ruminococcus species, with variable effects on microbial metabolites and subsequent associations with physiologic markers of gastrointestinal and metabolic health. These findings have implications for biotic-focused research on candidate prebiotic substrates as well as next-generation probiotics.
Collapse
Affiliation(s)
- Veronica K Hindle
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Nadine M Veasley
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States; Personalized Nutrition Initiative, University of Illinois Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
39
|
Guan JL, Xu TT, Lin Y, Mo YS, He BY, Han YY, Li JY, Xia SH, Zhou YN, Liao JZ, Li PY. High-dose dual therapy for Helicobacter pylori eradication inducing less impact on the gut microbiota. Gut Pathog 2025; 17:7. [PMID: 39885529 PMCID: PMC11783801 DOI: 10.1186/s13099-025-00682-8] [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: 08/08/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) eradication regimens may have different effects on the gut microbiota. Few studies have analyzed the safety of high-dose dual therapy (HDDT) from a micro-ecological perspective. This study aimed to compare the impact of H. pylori eradication with HDDT and bismuth quadruple therapy (BQT) on gut microbiota. PATIENTS AND METHODS H. Pylori-infected treatment-naive patients were recruited and screened from September 2023 to April 2024 and randomly assigned to the HDDT group (esomeprazole 20 mg, amoxicillin 750 mg, qid, 14 days) or BQT group (esomeprazole 20 mg, amoxicillin 1000 mg, clarithromycin 500 mg, and bismuth potassium citrate 600 mg, bid, 14 days). Fresh stool specimens were collected and stored before treatment and at week 2 and week 8 after treatment. The diversity and composition of the gut microbiota were compared and analyzed in both groups using 16 S rRNA gene sequencing. RESULTS Forty-nine H. pylori positive patients were enrolled and randomly assigned to either the HDDT (n = 24) or the BQT group (n = 25) group. Compared with baseline, alpha and beta diversities significantly changed at week 2 after receiving BQT and did not recover fully at week 8. However, in the HDDT group, the diversities at week 2 changed mildly without statistical significance, compared to baseline. Additionally, a greater number of species had alterations in their abundances in the BQT group compared to the HDDT group at week 2. However, the abundances of these species were restored to their previous levels at week 8 in both the HDDT and BQT groups. CONCLUSIONS Compared to BQT, HDDT exerted less impact on the diversity and composition of the gut microbiota. CLINICAL TRIAL REGISTRATION ChiCTR2100053268.
Collapse
Affiliation(s)
- Jia-Lun Guan
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ting-Ting Xu
- Department of Gastroenterology, Wenchang People's Hospital, Wenchang, China
| | - Ya Lin
- Department of Gastroenterology, Wenchang People's Hospital, Wenchang, China
| | - Yan-Shuai Mo
- Department of Anesthesiology, Wenchang People's Hospital, Wenchang, China
| | - Bi-Yu He
- Department of Gastroenterology, Wenchang People's Hospital, Wenchang, China
| | - Ying-Ying Han
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ji-Yan Li
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Su-Hong Xia
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ya-Ni Zhou
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Jia-Zhi Liao
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Pei-Yuan Li
- Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
- Department of Gastroenterology, Wenchang People's Hospital, Wenchang, China.
| |
Collapse
|
40
|
Yang Y, Zhou HY, Zhou GM, Chen J, Ming R, Zhang D, Jiang HW. The impact of different gastrointestinal reconstruction techniques on gut microbiota after gastric cancer surgery. Front Microbiol 2025; 15:1494049. [PMID: 39925886 PMCID: PMC11804259 DOI: 10.3389/fmicb.2024.1494049] [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: 09/10/2024] [Accepted: 12/24/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction Gastric cancer is one of the common malignant tumors in the digestive tract, characterized by high incidence and mortality rates. This is particularly significant in China, where a large proportion of global new cases of gastric cancer and related deaths occur. In recent years, with the continuous development of molecular biology technology, people have gained a deeper understanding of the gastrointestinal microbiome, and studies have shown that it is closely related to the occurrence, development, and therapeutic response of gastric cancer. Although surgical intervention is crucial in significantly extending the survival of gastric cancer patients, the disruption of the balance of the intestinal microbiota caused by surgery itself should not be overlooked, as it may affect postoperative recovery. Methods This study was approved by the Biomedical Ethics Committee of Sichuan Mianyang 404 Hospital. A random sampling method was used to select patients who underwent gastric cancer surgery at the hospital from January 2023 to December 2023. All patients signed written informed consent forms. Standardized perioperative management was conducted for the patients in the study, including preoperative preparation, intraoperative handling, and postoperative treatment. Fecal samples were collected from patients before surgery (before bowel preparation) and around one week after surgery for 16S rRNA sequencing analysis, through which differential biomarkers and related functional genes were sought. Results The study results indicated that there was no significant difference in the diversity of the gut microbiota between the two groups. Compared with the R-Y group, the DTR surgical method significantly altered the structure of the gut microbiota, affecting the types, quantities, and proportions of intestinal bacteria. Furthermore, the DTR group exhibited poorer postoperative nutritional absorption capacity compared to the R-Y group, as indicated by a lower F/B ratio. The R-Y group showed a richer abundance of Bacteroidetes and a lower abundance of Proteobacteria, as well as a higher F/B ratio after surgery. These findings provide new insights into the changes in the gut microbiota following gastric cancer surgery, which may be of significant importance for postoperative recovery and long-term health management. Discussion This study reveals the impact of different gastrointestinal reconstruction techniques on the postoperative gut microbiota of gastric cancer patients, providing new insights into the physiological changes during the postoperative recovery period. Although there was no significant difference in microbial diversity between the DTR group and the R-Y group, the DTR group showed more pronounced changes in microbial structure postoperatively, which may be associated with an increased risk of postoperative infection. These findings emphasize the importance of considering the impact on the gut microbiota when selecting gastric cancer surgery methods. However, the study had a limited sample size and did not delve into changes in metabolites. Future studies should expand the sample size and conduct metabolomic analyses to further validate these preliminary findings.
Collapse
Affiliation(s)
- Yu Yang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Hang-Yu Zhou
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Guo-Min Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Jin Chen
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Rui Ming
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Dong Zhang
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Huai-Wu Jiang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Gastrointestinal Surgery, Mianyang 404 Hospital, Mianyang, Sichuan, China
| |
Collapse
|
41
|
Florêncio GP, Xavier AR, Natal ACDC, Sadoyama LP, Röder DVDDB, Menezes RDP, Sadoyama Leal G, Patrizzi LJ, Pena GDG. Synergistic Effects of Probiotics and Lifestyle Interventions on Intestinal Microbiota Composition and Clinical Outcomes in Obese Adults. Metabolites 2025; 15:70. [PMID: 39997695 PMCID: PMC11857521 DOI: 10.3390/metabo15020070] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/25/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Obesity is a growing global epidemic. The composition of the intestinal microbiota can be influenced by several factors. Studies highlight the role of intestinal bacteria in the pathophysiology of obesity. So, the objective of this study was to investigate whether the use of probiotics, together with healthy lifestyle habits, contributes to weight reduction in obese individuals by analyzing the intestinal microbiota profile. METHODS A prospective study was carried out with 45 adults with obesity. Participants underwent guidance on healthy lifestyle habits, received a probiotic component containing different microbiological strains and were followed for 60 days. Clinical parameters, body composition, biochemical analysis, and intestinal microbiota assessment were performed before and after treatment. After 60 days, it was observed that the bacterial strains present in the probiotic were present in the patients' intestinal microbiota. Participants also showed improvements in physical activity, sleep quality, and anxiety management, as well as changes in some eating habits, such as a reduction in the consumption of processed foods and a significant increase in water intake. RESULTS A reduction in BMI, fasting glucose, insulin, HOMA-IR, LDL cholesterol, and triglycerides was observed, in addition to an increase in HDL cholesterol, improvement in bowel movement frequency, and stool consistency. Analysis of the intestinal microbiota revealed an increase in microbial diversity and a better balance between the bacterial phyla Firmicutes and Bacteroidetes. CONCLUSIONS The changes related to improving the composition of the intestinal microbiota, dietary habits, increased physical activity, reduced anxiety, and better sleep quality have significantly contributed to weight loss and improvements in physiological parameters in obese individuals.
Collapse
Affiliation(s)
- Glauber Pimentel Florêncio
- School of Medicine, Federal University of Uberlândia, Uberlândia 38405-320, MG, Brazil; (G.P.F.); (A.R.X.); (A.C.d.C.N.); (L.P.S.)
| | - Analicy Rodrigues Xavier
- School of Medicine, Federal University of Uberlândia, Uberlândia 38405-320, MG, Brazil; (G.P.F.); (A.R.X.); (A.C.d.C.N.); (L.P.S.)
| | - Ana Catarina de Castro Natal
- School of Medicine, Federal University of Uberlândia, Uberlândia 38405-320, MG, Brazil; (G.P.F.); (A.R.X.); (A.C.d.C.N.); (L.P.S.)
| | - Lorena Prado Sadoyama
- School of Medicine, Federal University of Uberlândia, Uberlândia 38405-320, MG, Brazil; (G.P.F.); (A.R.X.); (A.C.d.C.N.); (L.P.S.)
| | | | - Ralciane de Paula Menezes
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38405-318, MG, Brazil;
| | - Geraldo Sadoyama Leal
- Institute of Biotechnology, Federal University of Catalão, Catalão 75704-020, GO, Brazil;
| | - Lislei Jorge Patrizzi
- Department of Physiotherapy, Federal University of Triângulo Mineiro, Uberaba 38025-350, MG, Brazil;
| | - Geórgia das Graças Pena
- School of Medicine, Federal University of Uberlândia, Uberlândia 38405-320, MG, Brazil; (G.P.F.); (A.R.X.); (A.C.d.C.N.); (L.P.S.)
| |
Collapse
|
42
|
Neumann CJ, Mohammadzadeh R, Woh PY, Kobal T, Pausan MR, Shinde T, Haid V, Mertelj P, Weiss EC, Kolovetsiou-Kreiner V, Mahnert A, Kumpitsch C, Jantscher-Krenn E, Moissl-Eichinger C. First-year dynamics of the anaerobic microbiome and archaeome in infants' oral and gastrointestinal systems. mSystems 2025; 10:e0107124. [PMID: 39714161 PMCID: PMC11756582 DOI: 10.1128/msystems.01071-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Recent research provides new insights into the early establishment of the infant gut microbiome, emphasizing the influence of breastfeeding on the development of gastrointestinal microbiomes. In our study, we longitudinally examined the taxonomic and functional dynamics of the oral and gastrointestinal tract (GIT) microbiomes of healthy infants (n = 30) in their first year, focusing on the often-over-looked aspects, the development of archaeal and anaerobic microbiomes. Breastfed (BF) infants exhibit a more defined transitional phase in their oral microbiome compared to non-breastfed (NBF) infants, marked by a decrease in Streptococcus and the emergence of anaerobic genera such as Granulicatella. This phase, characterized by increased alpha-diversity and significant changes in beta-diversity, occurs earlier in NBF infants (months 1-3) than in BF infants (months 4-6), suggesting that breastfeeding supports later, more defined microbiome maturation. We demonstrated the presence of archaea in the infant oral cavity and GIT microbiome from early infancy, with Methanobrevibacter being the predominant genus. Still, transient patterns show that no stable archaeome is formed. The GIT microbiome exhibited gradual development, with BF infants showing increased diversity and complexity between the third and eighth months, marked by anaerobic microbial networks. NBF infants showed complex microbial co-occurrence patterns from the start. These strong differences between BF and NBF infants' GIT microbiomes are less pronounced on functional levels than on taxonomic levels. Overall, the infant microbiome differentiates and stabilizes over the first year, with breastfeeding playing a crucial role in shaping anaerobic microbial networks and overall microbiome maturation. IMPORTANCE The first year of life is a crucial period for establishing a healthy human microbiome. Our study analyses the role of archaea and obligate anaerobes in the development of the human oral and gut microbiome, with a specific focus on the impact of breastfeeding in this process. Our findings demonstrated that the oral and gut microbiomes of breastfed infants undergo distinct phases of increased dynamics within the first year of life. In contrast, the microbiomes of non-breastfed infants are more mature from the first month, leading to a steadier development without distinct transitional phases in the first year. Additionally, we found that archaeal signatures are present in infants under 1 year of age, but they do not form a stable archaeome. In contrast to this, we could track specific bacterial strains transitioning from oral to gut or persisting in the gut over time.
Collapse
Affiliation(s)
- Charlotte J. Neumann
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Rokhsareh Mohammadzadeh
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Pei Yee Woh
- Department of Food
Science and Nutrition, The Hong Kong Polytechnic
University, Hong Kong,
Hong Kong
- Research Institute for
Future Food (RiFood), The Hong Kong Polytechnic
University, Hong Kong SAR,
China
| | - Tanja Kobal
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Manuela-Raluca Pausan
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
- BBMRI-ERIC, Graz,
Styria, Austria
| | - Tejus Shinde
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Victoria Haid
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Polona Mertelj
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Eva-Christine Weiss
- Department of
Obstetrics and Gynecology, Medical University of
Graz, Graz,
Styria, Austria
| | | | - Alexander Mahnert
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Christina Kumpitsch
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
| | - Evelyn Jantscher-Krenn
- Department of
Obstetrics and Gynecology, Medical University of
Graz, Graz,
Styria, Austria
- Research Unit Early
Life Determinants (ELiD), Medical University of
Graz, Graz,
Styria, Austria
- BioTechMed,
Graz, Styria, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and
Research Institute of Hygiene, Microbiology and Environmental Medicine,
Medical University of Graz,
Graz, Styria, Austria
- BioTechMed,
Graz, Styria, Austria
| |
Collapse
|
43
|
Węsierska E, Micek P, Adamski MG, Gondek K, Lis M, Trela M, Wojtysiak D, Kowal J, Wyrobisz-Papiewska A, Kunstman G, Mosiołek S, Smoroń K. Changes in the intestinal microbiota of broiler chicken induced by dietary supplementation of the diatomite-bentonite mixture. BMC Vet Res 2025; 21:13. [PMID: 39799366 PMCID: PMC11724591 DOI: 10.1186/s12917-024-04439-4] [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: 02/26/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Diatomite is a source of biologically available silicon but in feed industry its insecticide and anti-caking properties have been also widely recognized. The aim of the study was to evaluate the effect of dietary diatomite-bentonite mixture (DBM) supplementation on the quantitative and qualitative composition of the bacterial microbiome of the broiler chicken gut. The trial was carried out on 960 Ross 308 broiler chickens divided into 2 experimental groups throughout the entire rearing period lasting 6 weeks. The birds were fed complete granulated diets without (group C) or with DBM (group E) in an amount of 1% from the 11 day of life. Two nutritionally balanced diets were used, tailored to the age of the broilers: a grower diet (from day 11 to 34) and a finisher diet (from day 35 to 42 of life). RESULTS Diatomite used in a mixture with bentonite significantly altered the microbiome. Restricting the description to species that comprise a minimum of 1% of all analyzed sequences, 36 species in group E (with diatomite) and 30 species in group C (without diatomite) were selected. Several bacteria species were identified in intestinal contents of chickens for the first time. Thirteen species occurred only in group E: Agathobaculum butyriciproducens, Anaerobutyricum hallii, Anaerobutyricum soehngenii, Blautia producta ATCC 27,340 = DSM 2950, Gordonibacter pamelaeae 7-10-1-b, Helicobacter pullorum NCTC 12,824, Lactobacillus crispatus, L. helveticus DSM 20,075 = CGMCC 1.1877, Mucispirillum schaedleri, Phascolarctobacterium faecium, Phocaeicola coprocola DSM 17,136, P. massiliensis, and Ruthenibacterium lactatiformans. CONCLUSIONS The findings highlight the intricate and potentially consequential relationship between diet, specifically diatomite-bentonite mixture supplementation, and gut microbiota composition.
Collapse
Affiliation(s)
- E Węsierska
- Department of Infectious Diseases and Public Health, University of Agriculture in Krakow, Al. Mickiewicza 21, Krakow, 31-120, Poland.
| | - P Micek
- Department of Nutrition, Animal Biotechnology, and Fisheries, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - M G Adamski
- SPARK-TECH, Sp. z o.o., Rynek Główny 28, Krakow, 31-010, Poland
| | - K Gondek
- Department of Agricultural and Environmental Chemistry, University of Agriculture in Krakow, Al. Mickiewicza 21, Krakow, 31-120, Poland
| | - M Lis
- Department of Zoology and Animal Welfare, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - M Trela
- Department of Zoology and Animal Welfare, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - D Wojtysiak
- Department of Genetics, Animal Breeding and Ethology, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - J Kowal
- Department of Zoology and Animal Welfare, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - A Wyrobisz-Papiewska
- Department of Zoology and Animal Welfare, University of Agriculture in Krakow, Al. Mickiewicza 24/28, Krakow, 30-059, Poland
| | - G Kunstman
- SPARK-TECH, Sp. z o.o., Rynek Główny 28, Krakow, 31-010, Poland
| | - S Mosiołek
- SPARK-TECH, Sp. z o.o., Rynek Główny 28, Krakow, 31-010, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow, 30-348, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, Krakow, 30-348, Poland
| | - K Smoroń
- Specialized Mining Company "Górtech" Sp. z o.o, ul. Wielicka 50, Krakow, 30-552, Poland
| |
Collapse
|
44
|
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
|
45
|
Li R, Chen X, Shi C, Zhu Y. Study on the Effect of Radish Sprouts on Short-Chain Fatty Acids and Gut Microbial Diversity in Healthy Individuals. Foods 2025; 14:170. [PMID: 39856836 PMCID: PMC11765271 DOI: 10.3390/foods14020170] [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: 12/02/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
This study aimed to assess the impact of radish sprouts on the gut microbiota of healthy individuals. Radish sprout additives, subjected to short-term storage and steam treatment, were used to intervene in an in vitro culture of human gut microbiota. The influence of radish sprouts on the gut microbiota was evaluated by monitoring short-chain fatty acid (SCFA) content and proportion in the fermentation broth, and microbial diversity was assessed using 16S rDNA amplicon sequencing. The results indicated that the gut microbiota produced a substantial amount of SCFA within 48 h of fermentation, with a right-skewed distribution across all groups. The addition of both digestates enhanced Firmicutes diversity, while Bacteroidetes and Proteobacteria diversity remained stable between the control and fresh sprout groups. The 30 s steam treatment group showed an increase in Bacteroidetes and a decrease in Proteobacteria diversity. The abundance of Bacilli, Bacillaceae, and Bacillus was significantly higher in both the fresh and steam-treated groups compared to the control. Both fresh and steam-treated radish sprout digestates enriched gut microbiota diversity, with steam treatment showing superior effects. These findings suggest that radish sprout consumption may positively influence gut microbiota, with steam treatment potentially enhancing these benefits.
Collapse
Affiliation(s)
- Ru Li
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xuehong Chen
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
| | - Cong Shi
- College of Food and Biological Engineering, Xuzhou Institute of Technology, Xuzhou 221018, China; (R.L.); (X.C.); (C.S.)
| | - Yi Zhu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
46
|
Correa Lopes B, Turck J, Tolbert MK, Giaretta PR, Suchodolski JS, Pilla R. Prolonged storage reduces viability of Peptacetobacter (Clostridium) hiranonis and core intestinal bacteria in fecal microbiota transplantation preparations for dogs. Front Microbiol 2025; 15:1502452. [PMID: 39839105 PMCID: PMC11747423 DOI: 10.3389/fmicb.2024.1502452] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Fecal microbiota transplantation (FMT) has been described useful as an adjunct treatment for chronic enteropathy in dogs. Different protocols can be used to prepare and store FMT preparations, however, the effect of these methods on microbial viability is unknown. We aimed (1) to assess the viability of several core intestinal bacterial species by qPCR and (2) to assess Peptacetobacter (Clostridium) hiranonis viability through culture to further characterize bacterial viability in different protocols for FMT preparations. Methods Bacterial abundances were assessed in feces from six healthy dogs by qPCR after propidium monoazide (PMA-qPCR) treatment for selective quantitation of viable bacteria. Conservation methods tested included lyophilization (stored at 4°C and at -20°C) and freezing with glycerol-saline solution (12.5%) and without any cryoprotectant (stored at -20°C). Additionally, the abundance of P. hiranonis was quantified using bacterial culture. Results Using PMA-qPCR, the viability of Faecalibacterium, Escherichia coli, Streptococcus, Blautia, Fusobacterium, and P. hiranonis was reduced in lyophilized fecal samples kept at 4°C and -20°C up to 6 months (p < 0.05). In frozen feces without cryoprotectant, only Streptococcus and E. coli were not significantly reduced for up to 3 months (p > 0.05). Lastly, no differences were observed in the viability of those species in glycerol-preserved samples up to 6 months (p > 0.05). When using culture to evaluate the viability of P. hiranonis, we observed that P. hiranonis abundance was lower in lyophilized samples kept at 4°C than -20°C; and P. hiranonis abundance was higher in glycerol-preserved samples for up to 6 months than in samples preserved without glycerol for up to 3 months. Moreover, the highest abundance of P. hiranonis was observed in glycerol-preserved feces. After 3 months, P. hiranonis was undetectable by culture in 83% (5/6) of the frozen samples without glycerol. Discussion While the lyophilization procedure initially reduced P. hiranonis abundance, P. hiranonis viability was stable thereafter for up to 6 months at -20°C. The higher bacterial viability detected in fecal samples preserved with glycerol confirms the use of this cryoprotectant as a reliable method to keep bacteria alive in the presence of fecal matrix for FMT purposes.
Collapse
Affiliation(s)
- Bruna Correa Lopes
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jonathan Turck
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - M. Katherine Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Paula R. Giaretta
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
- Department of Veterinary Pathology, Hygiene and Public Health, University of Milan, Milan, Italy
| |
Collapse
|
47
|
Sun X, Yun L, Xie K, Liu R, Ren X, Zeng B, Cao X, Li Z, Zhou G, Liu B, Peng L, Yuan L. Probiotic Bacillus pumilus LV149 enhances gut repair, modulates microbiota, and alters transcriptome in DSS-induced colitis mice. Front Microbiol 2025; 15:1507979. [PMID: 39845056 PMCID: PMC11753000 DOI: 10.3389/fmicb.2024.1507979] [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: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose Gut microbiota dysbiosis significantly impacts ulcerative colitis (UC) progression and exacerbation. Probiotics show promise in UC management. This study evaluated the effects of different doses of Bacillus pumilus LV149, an aquatic-derived probiotic, on gut injury repair in male C57BL/6 mice with dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) and investigated the underlying mechanisms. Methods UC was induced by allowing mice free access to a 3% DSS solution for 7 days, with concurrent daily oral gavage of either a low (LV149-L, 1 × 108 CFU/day/mouse) or high (LV149-H, 1 × 109 CFU/day/mouse) dose of LV149. The effects were assessed through physiological parameters, intestinal barrier integrity, inflammation, gut microbiota composition, and transcriptomic changes. Results LV149 significantly improved pathological symptoms, including weight loss and disease activity index (DAI), and reduced colon shortening in a dose-dependent manner and inflammatory damage. The intervention also restored gut barrier function by upregulating mucins, goblet cell counts, and tight junction proteins (ZO-1, occludin, and claudin-1) in colonic tissue, along with reducing serum lipopolysaccharide (LPS) levels. Notably, only the LV149-H significantly decreased the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, while both doses increased the expression of the anti-inflammatory cytokine IL-10 in a dose-dependent in colonic tissue. LV149 further modulated the gut microbiota, increasing beneficial bacteria and reducing pathogenic populations. Transcriptomic analysis indicated that LV149-L may exert gut repair effects via the IL-17 signaling pathway, whereas LV149-H appears to act through the JAK-STAT signaling pathway. Conclusion This study demonstrated that LV149, particularly at a higher dose, effectively mitigated DSS-induced colonic injury by modulating gut microbiota, enhancing gut barrier integrity, and reducing inflammation. The dose-dependent effects underscored LV149-H's potential as a therapeutic agent for UC due to its stronger anti-inflammatory properties and gut-protective effects.
Collapse
Affiliation(s)
- Xinyu Sun
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Long Yun
- Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Keming Xie
- Medical College of Jiaying University, Jiaying University, Meizhou, China
| | - Renhui Liu
- School of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Xinyue Ren
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bokun Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xudong Cao
- Department of Chemical and Biological Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Zhi Li
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Guihao Zhou
- Division of Medicine, University College London, London, United Kingdom
| | - Bang Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Luo Peng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Lihong Yuan
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
48
|
Khan MF, Khodve G, Yadav S, Mallick K, Banerjee S. Probiotic treatment improves post-traumatic stress disorder outcomes in mice. Behav Brain Res 2025; 476:115246. [PMID: 39255901 DOI: 10.1016/j.bbr.2024.115246] [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: 05/23/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a mental disorder resulting from traumatic events which are characterized primarily by anxiety and depressive disorder. In this study, we determine the role of gut bacteria in PTSD. PTSD-like symptoms were produced by single prolonged stress (SPS). SPS animals showed increased levels of anxiety as measured by the elevated plus maze test, while depressive behaviour was confirmed using sucrose preference, force swim, and tail suspension tests. Gut dysbiosis was confirmed in PTSD animals by next-generation sequencing of 16 s RNA of faecal samples, while these animals also showed increased intestinal permeability and altered intestinal ultrastructure. Probiotic treatment increases beneficial microbiota, improves intestinal health and reduces PTSD-associated anxiety and depression. We also found a decrease in cortical BDNF levels in PTSD animals, which was reversed after probiotic administration. Here, we establish the link between gut dysbiosis and PTSD and show that probiotic treatment may improve the outcome of PTSD like symptoms in mice.
Collapse
Affiliation(s)
- Mohd Faiz Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Gopal Khodve
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Sanjay Yadav
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Keya Mallick
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India.
| |
Collapse
|
49
|
Negishi H, Ichikawa A, Takahashi S, Kano H, Makino S. Targeted prebiotic application of gluconic acid-containing oligosaccharides promotes Faecalibacterium growth through microbial cross-feeding networks. THE ISME JOURNAL 2025; 19:wraf027. [PMID: 39936592 PMCID: PMC11922316 DOI: 10.1093/ismejo/wraf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/19/2024] [Accepted: 02/10/2025] [Indexed: 02/13/2025]
Abstract
The gut microbiome plays a crucial role in human health, and certain bacterial species, such as Faecalibacterium prausnitzii, are particularly beneficial. This study conducted a comprehensive investigation of prebiotic compounds that showed potential for specifically promoting beneficial gut bacteria. Using in vitro fecal cultures and a human intervention study, we identified maltobionic acid and lactobionic acid as compounds that specifically promoted Faecalibacterium growth both in vitro and in vivo without significantly affecting Bifidobacterium, which is typically increased by traditional prebiotics. In a human intervention study (n = 27), a significant increase was observed in Faecalibacterium abundance following maltobionic acid supplementation, with effectiveness correlating with the initial Parabacteroides abundance. Mechanistic investigations revealed a cross-feeding pathway between gut bacteria. In this pathway, Parabacteroides species converted the gluconic acid moiety of maltobionic and lactobionic acids to glucuronic acid, which was then preferentially utilized by Faecalibacterium. These findings suggest that gluconic acid-containing oligosaccharides are promising prebiotics for the targeted enhancement of beneficial Faecalibacterium and underscore the importance of microbial interactions in prebiotic research, offering new avenues for personalized microbiome modulation strategies.
Collapse
Affiliation(s)
- Hiroki Negishi
- Wellness Science Labs, Meiji Holdings Co., Ltd., Tokyo 192-0919, Japan
| | - Ayumi Ichikawa
- Wellness Science Labs, Meiji Holdings Co., Ltd., Tokyo 192-0919, Japan
| | - Saori Takahashi
- Wellness Science Labs, Meiji Holdings Co., Ltd., Tokyo 192-0919, Japan
| | - Hiroshi Kano
- Wellness Science Labs, Meiji Holdings Co., Ltd., Tokyo 192-0919, Japan
| | - Seiya Makino
- Wellness Science Labs, Meiji Holdings Co., Ltd., Tokyo 192-0919, Japan
| |
Collapse
|
50
|
Bhutta NK, Xu X, Jian C, Wang Y, Liu Y, Sun J, Han B, Wu S, Javeed A. Gut microbiota mediated T cells regulation and autoimmune diseases. Front Microbiol 2024; 15:1477187. [PMID: 39749132 PMCID: PMC11694513 DOI: 10.3389/fmicb.2024.1477187] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Gut microbiota regulates the immune system, the development and progression of autoimmune diseases (AIDs) and overall health. Recent studies have played a crucial part in understanding the specific role of different gut bacterial strains and their metabolites in different AIDs. Microbial signatures in AIDs are revealed by advanced sequencing and metabolomics studies. Microbes such as Faecalibacterium prausnitzii, Akkermansia muciniphila, Anaerostipes caccae, Bacteroides sp., Roseburia sp., Blautia sp., Blautia faecis, Clostridium lavalense, Christensenellaceae sp., Coprococcus sp., Firmicutes sp., Ruminococcaceae sp., Lachnospiraceae sp., Megamonas sp., Monoglobus sp., Streptococcus pneumoniae and Bifidobacterium sp. help maintain immune homeostasis; whereas, Prevotella copri, Ruminococcus gnavus, Lactobacillus salivarius, Enterococcus gallinarum, Elizabeth menigoseptica, Collinsella sp., Escherichia sp., Fusobacterium sp., Enterobacter ludwigii, Enterobacteriaceae sp., Proteobacteria, Porphyromonas gingivalis, Porphyromonas nigrescens, Dorea sp., and Clostridium sp. cause immuno-pathogenesis. A complex web of interactions is revealed by understanding the influence of gut microbiota on immune cells and various T cell subsets such as CD4+ T cells, CD8+ T cells, natural killer T cells, γδ T cells, etc. Certain AIDs, including rheumatoid arthritis, diabetes mellitus, atopic asthma, inflammatory bowel disease and non-alcoholic fatty liver disease exhibit a state of dysbiosis, characterized by alterations in microbial diversity and relative abundance of specific taxa. This review summarizes recent developments in understanding the role of certain microbiota composition in specific AIDs, and the factors affecting specific regulatory T cells through certain microbial metabolites and also focuses the potential application and therapeutic significance of gut microbiota-based interventions as novel adjunctive therapies for AIDs. Further research to determine the precise association of each gut bacterial strain in specific diseases is required.
Collapse
Affiliation(s)
- Nabeel Khalid Bhutta
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiujin Xu
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Cuiqin Jian
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yifan Wang
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yi Liu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Jinlyu Sun
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, Department of Allergy, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bingnan Han
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Shandong Wu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Ansar Javeed
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|