1
|
Nepal S, Shi N, Hoyd R, Spakowicz DJ, Orwoll E, Shikany JM, Napoli N, Tabung FK. Role of insulinemic and inflammatory dietary patterns on gut microbial composition and circulating biomarkers of metabolic health among older American men. Gut Microbes 2025; 17:2497400. [PMID: 40296253 PMCID: PMC12045561 DOI: 10.1080/19490976.2025.2497400] [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: 01/28/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic low-grade inflammation and hyperinsulinemia are linked with metabolic dysfunction and dysbiosis. This study investigated the role of dietary inflammatory and insulinemic potential on gut microbiome and circulating health biomarkers in older men. Data from the Osteoporotic Fractures in Men (MrOS) study were analyzed. Reversed Empirical Dietary Inflammatory Pattern (rEDIP), Empirical Dietary Index for Hyperinsulinemia (rEDIH), and Healthy Eating Index (HEI)-2020 scores were computed from food frequency questionnaire data. Stool samples were profiled using 16S rRNA sequencing. Elastic net regression identified diet-associated microbial profiles and multivariable-adjusted linear regression assessed diet-biomarker associations. Higher rEDIP, rEDIH, and HEI-2020 scores were positively associated with gut microbiota alpha diversity. Specific genera, including Intestinibacter and Lachnospira, associated positively, while Dielma, Peptococcus, Feacalitalea, and Negativibaccilus associated inversely with healthier dietary patterns. When evaluating changes in dietary patterns between baseline and visit 4 ( ~ 14 years), these genera tended to define rEDIP, rEDIH more than HEI-2020. In addition, higher dietary quality was linked to better biomarker profiles, including lower creatinine, sodium, triglycerides, and insulin resistance. Beneficial effects of higher dietary quality on health may be mediated by the ability of diet to regulate gut microbial composition and metabolic biomarker profiles.
Collapse
Affiliation(s)
- Sushma Nepal
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ni Shi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel J. Spakowicz
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric Orwoll
- Department of Medicine, Oregon Health & Sciences University, Portland, OR, USA
| | - James M. Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
- Division of Bone and Mineral Diseases, Washington University, St Louis, MO, USA
| | - Fred K. Tabung
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
Sapra L, Saini C, Mishra PK, Garg B, Gupta S, Manhas V, Srivastava RK. Bacillus coagulans ameliorates inflammatory bone loss in post-menopausal osteoporosis via modulating the "Gut-Immune-Bone" axis. Gut Microbes 2025; 17:2492378. [PMID: 40275534 PMCID: PMC12036487 DOI: 10.1080/19490976.2025.2492378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/11/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Osteoporosis is a systemic skeletal disease that leads to lower bone mineral density and intensifies the risk of unexpected fractures. Recently, our group reported that numerical defect in the frequencies of Bregs along with their compromised tendency to produce IL-10 cytokine further aggravates inflammatory bone loss in post-menopausal osteoporosis (PMO). Dysbiosis induced mucosal injury and leaky gut are the predominant contributors involved in the progression of inflammatory diseases including PMO. Furthermore, several evidence suggest that gut microbial composition plays a crucial role in the development and differentiation of Bregs. Nevertheless, the potential role of dysbiotic gut microbiota (GM) and Bregs under estrogen deficient PMO conditions has never been deciphered. Here, we evaluated the role of GM in the onset and progression of PMO along with its role in modulating the anti-osteoporotic potential of Bregs. We found that enhancement in the endotoxin producing bacteria and concomitant reduction in the short chain fatty acids producing bacteria, both under pre-clinical and clinical osteoporotic condition augment inflammatory bone loss. This suggests that dysbiosis of GM potentially exacerbates bone deterioration under estrogen deficient PMO conditions. Remarkably, supplementation of probiotic Bacillus coagulans significantly improved the bone mineral density, bone strength, and bone microarchitecture by modulating the anti-osteoclastogenic, immunosuppressive and immunomodulatory potential of Bregs. The present study delves deeper into the role of immune homeostasis ("Breg-Treg-Th17" cell axis) and GM profile in the pathophysiology of PMO. Altogether, findings of the present study open novel therapeutic avenues, suggesting restoration of GM composition as one of the viable therapeutic options in mitigating inflammatory bone loss under PMO conditions via modulating the "Gut-Immune-Bone" axis.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), An ICMR-Collaborating Centre of Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), An ICMR-Collaborating Centre of Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pradyumna K. Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Bhavuk Garg
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sarika Gupta
- Molecular Science lab, National Institute of Immunology (NII), New Delhi, India
| | - Vikrant Manhas
- Department of Orthopaedics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rupesh K. Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), An ICMR-Collaborating Centre of Excellence in Bone Health, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
3
|
Qiao S, Wang T, Sun J, Han J, Dai H, Du M, Yang L, Guo CJ, Liu C, Liu SJ, Liu H. Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases. Gut Microbes 2025; 17:2489765. [PMID: 40190016 PMCID: PMC11980479 DOI: 10.1080/19490976.2025.2489765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/13/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Mengxuan Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Lan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
4
|
Herzog MKM, Peters A, Shayya N, Cazzaniga M, Kaka Bra K, Arora T, Barthel M, Gül E, Maurer L, Kiefer P, Christen P, Endhardt K, Vorholt JA, Frankel G, Heimesaat MM, Bereswill S, Gahan CGM, Claesson MJ, Domingo-Almenara X, Hardt WD. Comparing Campylobacter jejuni to three other enteric pathogens in OligoMM 12 mice reveals pathogen-specific host and microbiota responses. Gut Microbes 2025; 17:2447832. [PMID: 39835346 DOI: 10.1080/19490976.2024.2447832] [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: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
Campylobacter jejuni, non-typhoidal Salmonella spp., Listeria monocytogenes and enteropathogenic/enterohemorrhagic Escherichia coli (EPEC/EHEC) are leading causes of food-borne illness worldwide. Citrobacter rodentium has been used to model EPEC and EHEC infection in mice. The gut microbiome is well-known to affect gut colonization and host responses to many food-borne pathogens. Recent progress has established gnotobiotic mice as valuable models to study how microbiota affect the enteric infections by S. Typhimurium, C. rodentium and L. monocytogenes. However, for C. jejuni, we are still lacking a suitable gnotobiotic mouse model. Moreover, the limited comparability of data across laboratories is often negatively affected by variations between different research facilities or murine microbiotas. In this study, we applied the standardized gnotobiotic OligoMM12 microbiota mouse model and compared the infections in the same facility. We provide evidence of robust colonization and significant pathological changes in OligoMM12 mice following infection with these pathogens. Moreover, we offer insights into pathogen-specific host responses and metabolite signatures, highlighting the advantages of a standardized mouse model for direct comparisons of factors influencing the pathogenesis of major food-borne pathogens. Notably, we reveal for the first time that C. jejuni stably colonizes OligoMM12 mice, triggering inflammation. Additionally, our comparative approach successfully identifies pathogen-specific responses, including the detection of genes uniquely associated with C. jejuni infection in humans. These findings underscore the potential of the OligoMM12 model as a versatile tool for advancing our understanding of food-borne pathogen interactions.
Collapse
Affiliation(s)
- Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Audrey Peters
- Department of Life Sciences, MRC Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Nizar Shayya
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Monica Cazzaniga
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Kardokh Kaka Bra
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Trisha Arora
- Omic Sciences Unit, EURECAT - Technology Centre of Catalonia, Reus, Spain
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Luca Maurer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Katharina Endhardt
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Cormac G M Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Marcus J Claesson
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Lee JS, Kao DJ, Worledge CS, Villamaria ZF, Wang RX, Welch NM, Kostelecky RE, Colgan SP. E. coli genetically modified for purine nucleobase release promotes butyrate generation and colonic wound healing during DSS insult. Gut Microbes 2025; 17:2490211. [PMID: 40247632 PMCID: PMC12013446 DOI: 10.1080/19490976.2025.2490211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
The gut microbiota transforms energy stored as undigestible carbohydrates into a remarkable number of metabolites that fuel intestinal bacterial communities and the host tissue. Colonic epithelial cells at the microbiota-host interface depend upon such microbiota-derived metabolites (MDMs) to satisfy their energy requisite. Microbial dysbiosis eliciting MDM loss contributes to barrier dysfunction and mucosal disease. Recent work has identified a role for microbiota-sourced purines (MSPs), notably hypoxanthine, as an MDM salvaged by the colonic epithelium for nucleotide biogenesis and energy balance. Here, we investigated the role of MSPs in mice during disease-modeled colonic energetic stress using a strain of E. coli genetically modified for enhanced purine nucleobase release (E. coli Mutant). E. coli Mutant colonization protected against DSS-induced tissue damage and permeability while promoting proliferation for wound healing. Metabolite and metagenomic analyses suggested a colonic butyrate-purine nucleobase metabolic axis, wherein the E. coli Mutant provided purine substrate for Clostridia butyrate production and host purine salvage, altogether supplying the host substrate for efficient nucleotide biogenesis and energy balance.
Collapse
Affiliation(s)
- J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Daniel J. Kao
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Corey S. Worledge
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Zachary F. Villamaria
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Nichole M. Welch
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Rachael E. Kostelecky
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| |
Collapse
|
6
|
Moore M, Whittington HD, Knickmeyer R, Azcarate-Peril MA, Bruno-Bárcena JM. Non-stochastic reassembly of a metabolically cohesive gut consortium shaped by N-acetyl-lactosamine-enriched fibers. Gut Microbes 2025; 17:2440120. [PMID: 39695352 DOI: 10.1080/19490976.2024.2440120] [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: 04/30/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Diet is one of the main factors shaping the human microbiome, yet our understanding of how specific dietary components influence microbial consortia assembly and subsequent stability in response to press disturbances - such as increasing resource availability (feeding rate) - is still incomplete. This study explores the reproducible re-assembly, metabolic interplay, and compositional stability within microbial consortia derived from pooled stool samples of three healthy infants. Using a single-step packed-bed reactor (PBR) system, we assessed the reassembly and metabolic output of consortia exposed to lactose, glucose, galacto-oligosaccharides (GOS), and humanized GOS (hGOS). Our findings reveal that complex carbohydrates, especially those containing low inclusion (~1.25 gL-1) components present in human milk, such as N-acetyl-lactosamine (LacNAc), promote taxonomic, and metabolic stability under varying feeding rates, as shown by diversity metrics and network analysis. Targeted metabolomics highlighted distinct metabolic responses to different carbohydrates: GOS was linked to increased lactate, lactose to propionate, sucrose to butyrate, and CO2, and the introduction of bile salts with GOS or hGOS resulted in butyrate reduction and increased hydrogen production. This study validates the use of single-step PBRs for reliably studying microbial consortium stability and functionality in response to nutritional press disturbances, offering insights into the dietary modulation of microbial consortia and their ecological dynamics.
Collapse
Affiliation(s)
- Madison Moore
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Hunter D Whittington
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Rebecca Knickmeyer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose M Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
7
|
Zhang Y, Wang A, Zhao W, Qin J, Zhang Y, Liu B, Yao C, Long J, Yuan M, Yan D. Microbial succinate promotes the response to metformin by upregulating secretory immunoglobulin a in intestinal immunity. Gut Microbes 2025; 17:2450871. [PMID: 39812329 PMCID: PMC11740685 DOI: 10.1080/19490976.2025.2450871] [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: 01/07/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Metformin is the first-line pharmacotherapy for type 2 diabetes mellitus; however, many patients respond poorly to this drug in clinical practice. The potential involvement of microbiota-mediated intestinal immunity and related signals in metformin responsiveness has not been previously investigated. In this study, we successfully constructed a humanized mouse model by fecal transplantation of the gut microbiota from clinical metformin-treated - responders and non-responders, and reproduced the difference in clinical phenotypes of responsiveness to metformin. The abundance of Bacteroides thetaiotaomicron, considered a representative differential bacterium of metformin responsiveness, and the level of secretory immunoglobulin A (SIgA) in intestinal immunity increased significantly in responder recipient mice following metformin treatment. In contrast, no significant alterations in B. thetaiotaomicron and SIgA were observed in non-responder recipient mice. The study of IgA-/- mice confirmed that downregulated expression or deficiency of SIgA resulted in non-response to metformin, meaning that metformin was unable to improve dysfunctional glucose metabolism and reduce intestinal and adipose tissue inflammation, ultimately leading to systemic insulin resistance. Furthermore, supplementation with succinate, a microbial product of B. thetaiotaomicron, potentially reversed the non-response to metformin by inducing the production of SIgA. In conclusion, we demonstrated that upregulated SIgA, which could be regulated by succinate, was functionally involved in metformin response through its influence on immune cell-mediated inflammation and insulin resistance. Conversely, an inability to regulate SIgA may result in a lack of response to metformin.
Collapse
Affiliation(s)
- Ying Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiting Wang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zhao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia’an Qin
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bing Liu
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengcheng Yao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingxia Yuan
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Mukhopadhya I, Martin JC, Shaw S, Gutierrez-Torrejon M, Boteva N, McKinley AJ, Gratz SW, Scott KP. Novel insights into carbohydrate utilisation, antimicrobial resistance, and sporulation potential in Roseburia intestinalis isolates across diverse geographical locations. Gut Microbes 2025; 17:2473516. [PMID: 40089923 PMCID: PMC11913394 DOI: 10.1080/19490976.2025.2473516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Roseburia intestinalis is one of the most abundant and important butyrate-producing human gut anaerobic bacteria that plays an important role in maintaining health and is a potential next-generation probiotic. We investigated the pangenome of 16 distinct strains, isolated over several decades, identifying local and time-specific adaptations. More than 50% of the genes in each individual strain were assigned to the core genome, and 77% of the cloud genes were unique to individual strains, revealing the high level of genome conservation. Co-carriage of the same enzymes involved in carbohydrate binding and degradation in all strains highlighted major pathways in carbohydrate utilization and reveal the importance of xylan, starch and mannose as key growth substrates. A single strain had adapted to use rhamnose as a sole growth substrate, the first time this has been reported. The ubiquitous presence of motility and sporulation gene clusters demonstrates the importance of these phenotypes for gut survival and acquisition of this bacterium. More than half the strains contained functional, potentially transferable, tetracycline resistance genes. This study advances our understanding of the importance of R. intestinalis within the gut ecosystem by elucidating conserved metabolic characteristics among different strains, isolated from different locations. This information will help to devise dietary strategies to increase the abundance of this species providing health benefits.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
- Microbiology and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jennifer C. Martin
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, UK
- All Wales Medical Genomics Service, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, UK
| | | | - Nikoleta Boteva
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Aileen J. McKinley
- Department of Surgery, Aberdeen Royal Infirmary Foresterhill, Aberdeen, UK
| | - Silvia W. Gratz
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Karen P. Scott
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
9
|
Charlesson B, Jones J, Abbiss C, Peeling P, Watts S, Christophersen C. Training load influences gut microbiome of highly trained rowing athletes. J Int Soc Sports Nutr 2025; 22:2507952. [PMID: 40400144 PMCID: PMC12100958 DOI: 10.1080/15502783.2025.2507952] [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: 02/07/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Despite the importance of the gut microbiome on physical performance and health, little is known on the impact of training on an athlete's gut health. OBJECTIVE This study investigates the effect of training load on markers of gut health. METHODS Whole stool (24 h) samples were collected from 23 highly trained rowers (mean ± SD; age 19.2 ± 1.1 y; weight 80.1 ± 11.4 kg; height 1.83 ± 0.09 m) following periods of high (HT) and low training load (LT). The microbiome and short-chain fatty acid concentrations were characterized from the whole stool samples. Three-day weighted food records were used to determine diet quality (ADIcore), macronutrient, and fiber intakes during HT and LT. RESULTS By design, training duration (147%) and intensity (130%) were greater during (HT), compared with (LT) (p < 0.001). Carbohydrate, fat, protein, and fiber intake remained stable, but ADIcore was higher in HT (55 ± 10) compared with LT (49 ± 9; t(15) = 2.78, p = 0.014; CI: 1.34 to 10.155). Stool frequency (1.11 ± 0.47 vs 0.67 ± 0.76; p = 0.007) was lower in HT compared with LT, and a greater number of participants were unable to produce a stool sample during LT (8% vs 47%). Short chain fatty acid (SCFA), propionic (120.64 ± 30.06 mm vs 91.35 ± 34.91 mm; p = 0.007), and butyric acid (104.76 ± 50.02 vs 64.23 ± 22.05 mm, p = 0.003) concentrations were lower in HT compared with LT. Alpha diversity, Shannon-Wiener diversity index (3.43 ± 0.37 vs 3.67 ± 0.34, p = 0.09) was lower in HT than LT. The abundance of the dominant Bacteroidia was greater at HT compared to LT and ratio of firmicutes to Bacteroidota (n = 16, 1.31 ± 1.19 vs 4.29 ± 3.88, t(15) = -3.44, p = 0.04, CI = -4.82 to -1.13) was lower in HT compared to LT. CONCLUSION Results of this study indicate that gut microbiome, SCFA concentrations, stool frequency, and diet quality vary between periods of high and low training load in athletes. The relationship between these factors and impact of such changes in gut health is currently unclear and warrants further investigation.
Collapse
Affiliation(s)
- B. Charlesson
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
- Western Australian Institute of Sport, Perth, Australia
| | - J. Jones
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| | - C. Abbiss
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| | - P. Peeling
- Western Australian Institute of Sport, Perth, Australia
- University of Western Australia, School of Human Sciences, Perth, Australia
| | - S. Watts
- Western Australian Institute of Sport, Perth, Australia
- University of Western Australia, School of Human Sciences, Perth, Australia
| | - C.T. Christophersen
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| |
Collapse
|
10
|
Kim CW, Kim HJ, Kang I, Ku KB, Kim Y, Park JH, Lim J, Kang BH, Park WH, La J, Chang S, Hwang I, Kim M, Ahn S, Lee HK. Gut dysbiosis from high-salt diet promotes glioma via propionate-mediated TGF-β activation. J Exp Med 2025; 222:e20241135. [PMID: 40402148 PMCID: PMC12097148 DOI: 10.1084/jem.20241135] [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: 07/02/2024] [Revised: 03/08/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025] Open
Abstract
The purpose of this study is to investigate the impact of a high-salt diet (HSD), which is commonly found in Western countries, on the progression of glioma. Our research shows that the alterations in gut microbiota caused by an HSD facilitated the development of glioma. Mice fed an HSD have elevated levels of intestinal propionate, which accelerated the growth of glioma cells. We also find that propionate supplementation enhanced the response of glioma cells to low oxygen levels. Moreover, we identify a link between TGF-β signaling, response to low oxygen levels, and invasion-related pathways. Propionate treatment increases the expression of HIF-1α, leading to an increase in TGF-β1 production. Additionally, propionate treatment promotes glioma cell invasion through TGF-β signaling. Our findings suggest that an HSD-induced increase in propionate plays a crucial role in glioma progression by facilitating invasion through the hypoxic response and TGF-β signaling pathways, thereby establishing a significant connection between gut microbiota and the progression of glioma.
Collapse
Affiliation(s)
- Chae Won Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - Hyun-Jin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - In Kang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Keun Bon Ku
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Yumin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jang Hyun Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - Juhee Lim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Byeong Hoon Kang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Won Hyung Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jeongwoo La
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Sungwoo Chang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Inju Hwang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Minji Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
11
|
Liu S, Zheng Y, Cui B, Yang J, Yuan B, Cao Y, Zhao Z, Sun Z, Wang Q, Yang X, Pan W, He C. Gut microbiota-derived butyrate alleviates the impairment of mice intestinal integrity caused by Toxoplasma gondii infection. Life Sci 2025; 374:123709. [PMID: 40368048 DOI: 10.1016/j.lfs.2025.123709] [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: 01/24/2025] [Revised: 05/04/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Chronic infection with Toxoplasma gondii (T. gondii) results in severe damages to the integrity of intestinal barrier, however, both the underlying mechanism and feasible intervention strategies are still little known. Here, we found that both the chronic infection of T. gondii and transplanting gut microbiota from T. gondii-infected mice severely impaired the mice intestinal integrity, which was characterized by significantly decreased thickness of inner mucus layer and down-regulated expression of three tight junction proteins Occludin, ZO-1, and Claudin (p < 0.05). Moreover, T. gondii infection also led to mice intestinal microbiota dysbiosis, especially butyrate-producing bacteria, and significantly changed the expression of several senescence-associated markers, including 6- and 7- fold upregulation for P16, P21, and 6-fold downregulation for Lamin B1 at mRNA levels, and 2-fold downregulation for β-galactosidase at protein levels (p < 0.05). Interestingly, subsequent administration with dietary butyrate could alleviate T. gondii-induced intestinal integrity impairment and cell senescence, revealing a significant increase of the inner mucus layer thickness (p < 0.001), and a remarkable decrease in P16, P21, β-galactosidase expression levels while an upregulation of Lamin B1 expression (p < 0.05). Taken together, our study revealed that T. gondii-induced dysbiosis of gut microbiota, especially butyrate-producing bacteria, contributes to the intestinal impairment, potentially via promoting cell senescence. In addition, administration with the metabolite, butyrate, could be a promising therapeutic measure against T. gondii infection.
Collapse
Affiliation(s)
- Shuni Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yutao Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Bingqian Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiayi Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bohui Yuan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China
| | - Yuhan Cao
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zimu Zhao
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhuo Sun
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qingling Wang
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, China.
| | - Cheng He
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, China; Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, China.
| |
Collapse
|
12
|
Chen J, Li H, Long X, Tong H, Xin X, Zhang H, Li Y, Liu P, He X, Chen Z. Causal association between gut microbiota and endometrial cancer in European and East Asian populations: a two-sample Mendelian randomization study. BMC Womens Health 2025; 25:283. [PMID: 40483441 PMCID: PMC12145594 DOI: 10.1186/s12905-025-03789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a significant global health concern. While observational epidemiological studies suggest a potential link between gut microbiota dysbiosis and the development of EC, the direction and causality of this association remain uncertain. METHODS We performed Mendelian randomization (MR) analysis to investigate the causal relationship between gut microbiota and EC. Exposure data were obtained from the MiBioGen study consortium (N = 18,340), and outcome data were sourced from the IEU OpenGWAS database, specifically datasets "ebi-a-GCST006464" (N = 121,885) and "bbj-a-113" (N = 90,730). The inverse variance-weighted(IVW) method was applied to evaluate the association between gut microbiota composition and EC risk. Sensitivity analyses were conducted to ensure the robustness of the findings. RESULTS Our study identified several microbial taxa linked to EC risk. In Europeans, genera such as Marvinbryantia, RuminococcaceaeUCG014, and Dorea exhibited protective effects, while family Erysipelotrichaceae (OR:1.224) and FamilyXI (OR:1.090) were significantly correlated with high EC risk. In East Asians, genera Lachnospira (OR:3.561) and family Bifidobacteriaceae (OR:1.715) were found associated with EC risk. Genera Lachnoclostridium and ErysipelotrichaceaeUCG003, family Coriobacteriaceae positively served as protective factors. Sensitivity analyses confirmed the reliability of our results, and there was no evidence of pleiotropy or heterogeneity. Our analysis identified several microbial taxa associated with EC risk. In Europeans, genera such as Marvinbryantia, Ruminococcaceae UCG014, and Dorea demonstrated protective effects, while the families Erysipelotrichaceae (OR: 1.224) and FamilyXI (OR: 1.090) were significantly associated with increased EC risk. In East Asians, the genus Lachnospira (OR: 3.561) and the family Bifidobacteriaceae (OR: 1.715) were linked to higher EC risk, whereas the genera Lachnoclostridium and Erysipelotrichaceae UCG003 and the family Coriobacteriaceae were identified as protective factors. Sensitivity analyses confirmed the reliability of these results, with no evidence of pleiotropy or heterogeneity. CONCLUSION This study highlights a relationship between gut microbiota and EC, emphasizing the potential of gut microbiota as therapeutic targets and biomarkers for assessing EC prognosis and treatment efficacy. These findings provide novel insights into the role of gut microbiota in the development and progression of EC.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
- Department of Radiology, Union Hospital, Tongji Medical College,, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan, 430022, China
| | - Haiqing Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xinrui Long
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Tong
- Department of Spine Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Xin Xin
- Department of Orthopedics of Yan'an Hospital, Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Han Zhang
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yeyao Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Ping Liu
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaolin He
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhuoyuan Chen
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Yan'an Hospital Affiliated to, Kunming Medical University, Kunming, Yunnan Province, China.
- Department of Orthopedics of Yan'an Hospital, Affiliated to Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
13
|
Neacsu M, Sayegh M, Vaughan NJ, Duncan GJ, Cantlay L, Anderson S, Henderson D, Fyfe C, Farquharson F, Saibu S, Horgan G, Louis P, Johnstone AM, Russell WR. Fava bean and buckwheat are sustainable food sources which support satiety and beneficially modulate several biomarkers, bacteria and metabolites associated with human health. Eur J Nutr 2025; 64:211. [PMID: 40481954 PMCID: PMC12145301 DOI: 10.1007/s00394-025-03726-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 05/23/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND The world's population requires adequate food supply, satisfying specific nutrient requirements to meet dietary recommendations, promote nutrition security and sustain health, while stimulating agriculture biodiversity. This study assessed the potential of buckwheat and fava bean to diversify the source of dietary nutrients. METHODS Twenty healthy volunteers (n = 6 men, n = 14 women; 42.08 ± 12.12 years; body mass index 24.72 ± 4.69 kg/m2) were recruited in a randomised controlled crossover study consisting of two seven-day intervention periods, buckwheat- and fava bean-based diets were provided to meet individual volunteers resting metabolic rate requirements. The study assessed subjective hunger and the impact of the diets on the gut microbiota composition and the plasma profiles of lipids, glucose, insulin, urea and homocysteine. Plasma, urine and faecal metabolites were also measured before and after consumption of each diet using targeted metabolomics (LC- and GC-MS). RESULTS Both intervention diets were as satiating as the volunteers' habitual diets (p = 0.234). The fava bean diet significantly reduced fasted plasma glucose and insulin and increased plasma homocysteine (p < 0.05). Buckwheat diet decreased plasma homocysteine (p < 0.01) and increased plasma, urine and faecal concentrations of salicylic acid and 2,3-dihydroxybenzoic acid. Both diets significantly increased plasma non-esterified fatty acids values, reduced plasma urea and faecal deoxycholic acid concentrations (p < 0.05). The fava bean diet provided significantly higher amounts of dietary fibre (both in comparison with habitual and buckwheat diet) significantly increasing the urine indole-3-propionic acid concentration (p < 0.01) (Day 0 vs. Day 7) and the faecal, plasma and urine indole-3-propionic acid concentrations (p < 0.01) (on Day 7 buckwheat vs. Day 7 fava bean diet). Furthermore, the fava bean diet promoted the growth of the gut bacterium Coprococcus eutactus (p < 0.05). CONCLUSION Buckwheat and fava bean contribute in a sustainable way to meet dietary recommendations and to promote dietary diversification. Diets rich in buckwheat and fava bean were found to be satiating and to beneficially modulate several biomarkers, bacteria and metabolites which are correlated with prevention of metabolic disorders such as cardiovascular disease and type 2 diabetes.
Collapse
Affiliation(s)
- Madalina Neacsu
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| | - Marietta Sayegh
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | | | - Gary J Duncan
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Louise Cantlay
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Susan Anderson
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Donna Henderson
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Claire Fyfe
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Freda Farquharson
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Salifu Saibu
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Graham Horgan
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
- Biomathematics and Statistics Scotland, AB25 2ZD, Aberdeen, UK
| | - Petra Louis
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | | | - Wendy R Russell
- The Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
14
|
Hadiono MA, Kazen AB, Aboulalazm FA, Burnett CML, Reho JJ, Kindel TL, Grobe JL, Kirby JR. Reutericyclin mitigates risperidone-induced suppression of anaerobic energy expenditure. Am J Physiol Regul Integr Comp Physiol 2025; 328:R741-R757. [PMID: 40235074 DOI: 10.1152/ajpregu.00190.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
Recent studies from our laboratory demonstrated that the gut microbial community represents a thermogenic biomass, as cecectomy causes an ∼8% decrease in total energy expenditure (EE) via suppression of anaerobic EE. The composition of the microbial community also dictates the EE of the microbial biomass as treatment with the antipsychotic, risperidone, suppresses anaerobic EE in a microbiome-dependent manner. Finally, we have determined that a specialized metabolite produced by Limosilactobacillus reuteri, reutericyclin (RTC), opposes the weight-gain effects of risperidone. In the present study, we performed comprehensive evaluations of energy balance in female C57BL/6J mice treated with risperidone, RTC, or both, to identify mechanisms by which RTC affects energy balance to mitigate risperidone-induced weight gain. We observed that risperidone suppressed anaerobic EE, and that RTC coadministration ameliorated the anaerobic EE suppression and weight gain induced by risperidone. Because anaerobic EE is dependent on the gut microbiota, we performed 16S and whole genome shotgun sequencing on stool and cecal samples following whole animal calorimetry. Risperidone and RTC treatments reciprocally modified the relative abundance of taxa known to participate in fermentation, especially for the production of short-chain fatty acids, which have been correlated with health and leanness in both humans and mice. Together, our data demonstrate that treatment with RTC positively modulates anaerobic EE, possibly by enhancing fermentation of the gut microbial community, and may represent a novel therapeutic in the treatment of obesity.NEW & NOTEWORTHY The gut microbial community represents a thermogenic biomass. The composition of the microbial community dictates energy expenditure of the microbial biomass and is altered by xenobiotics and bacterial metabolites. This study demonstrates that treatment with reutericyclin positively modulates anaerobic energy expenditure and may represent a novel therapeutic in the treatment of obesity.
Collapse
Affiliation(s)
- Matthew A Hadiono
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Alexis B Kazen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Fatima A Aboulalazm
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Colin M L Burnett
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Tammy L Kindel
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John R Kirby
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
15
|
Corona-Cervantes K, Sánchez-Salguero E, Zárate-Segura PB, Krishnakumar A, Piña-Escobedo A, Rangel-Calvillo MN, Ramírez-Lozada T, Acosta-Altamirano G, Lázaro-Pérez NDS, Sierra-Martínez M, Santos-Argumedo L, García-Mena J. Maternal immunoglobulins differentially bind a diverse bacterial community in human colostrum and the stool of breastfed neonates. Immunol Lett 2025; 273:106978. [PMID: 39924004 DOI: 10.1016/j.imlet.2025.106978] [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: 07/24/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
In the early days, maternal immunoglobulins are essential for sustaining a balanced gut environment by influencing the interaction between the host and the microbiome. The successional establishment of the pioneer strains is an interesting topic of research where maternal immunoglobulins appear to be important. This proof-of-concept study explored the binding pattern of IgA1, IgA2, IgM, and IgG classes to a commensal bacterial in human colostrum and the stool of breastfed neonates. We used flow cytometry coupled with 16S rRNA gene sequencing in human colostrum and neonatal feces samples to characterize this Ig-microbiota association. We observed that in human colostrum samples, IgA2 and IgM bind alfa and beta Proteobacteria, which can potentially stimulate neonatal immune system development in the gut. Other immunoglobulins like IgG predominantly bind facultative anaerobes belonging to the Firmicutes phylum, reported as part of human milk microbiota and pioneer colonizers of the neonatal gut. Maternal immunoglobulins also bind a wide diversity of bacteria in the neonatal stool. For instance, IgA2 and IgM bound more members of the phylum Bacteroidetes in comparison to IgG, these Bacteroidetes and some firmicutes have been reported as late colonizers of the neonatal gut, and their presence is important due to their ability to produce important short chain fatty acids like propionate and butyrate. Our results support the current view that microbial and immunoglobulin transference is crucial for developing the neonate's immune system and individual gut microbiota.
Collapse
Affiliation(s)
- Karina Corona-Cervantes
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), México City, Mexico
| | - Erick Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Mexico City, Mexico
| | - Paola Berenice Zárate-Segura
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, 11340, Mexico
| | - Aparna Krishnakumar
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), México City, Mexico
| | - Alberto Piña-Escobedo
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), México City, Mexico
| | | | - Tito Ramírez-Lozada
- Unidad de Ginecología y Obstetricia, Hospital Regional de Alta Especialidad de Ixtapaluca, Carretera Federal México-Puebla Km. 34.5, Col. Zoquiapan, Ixtapaluca, 56530, Mexico
| | - Gustavo Acosta-Altamirano
- Dirección de Investigación, Hospital General de México, Dr. Balmis 148 Col. Doctores, Cuauhtémoc, 06720, Ciudad de México, Mexico
| | - Noemí Del Socorro Lázaro-Pérez
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), México City, Mexico
| | - Mónica Sierra-Martínez
- Unidad de Investigación en Salud, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS Bienestar. Carretera Federal México-Puebla Km. 34.5, Col. Zoquiapan, Ixtapaluca, 56530, Mexico.
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), Mexico City, Mexico.
| | - Jaime García-Mena
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav), México City, Mexico.
| |
Collapse
|
16
|
Fontinha F, Martins N, Magalhães R, Peres H, Oliva-Teles A. Dietary short-chain fatty acid supplementation does not affect growth performance, metabolism, and oxidative status of European seabass (Dicentrarchus labrax) juveniles. Comp Biochem Physiol B Biochem Mol Biol 2025; 278:111096. [PMID: 40157714 DOI: 10.1016/j.cbpb.2025.111096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
This study investigated the effects of dietary short-chain fatty acid (SCFA) supplementation, on growth performance, metabolism, and antioxidant status of European seabass juveniles. Six isoproteic (43 % crude protein) and isolipidic (18 % crude lipid) diets were formulated to include 0.25 and 0.50 % Sodium acetate (SA), Sodium propionate (SP), or Sodium butyrate (SB). A diet without SCFA supplementation was used as a control. The diets were fed to triplicate groups of European seabass juveniles (initial body weight of 15 g) for 56 days. The supplementation of SCFA in the diet had no impact on the growth, feed utilization, or body composition of seabass. In the intestine, gene expression of pyruvate kinase (pk) and glucokinase (gk), phosphoenolpyruvate carboxykinase (pepck), glucose facilitative carrier type 2 (glut2), and citrate synthase (cs) was lower in fish fed the SP0.50 diet than in the other groups. Moreover, fatty acid synthase (fas) gene expression was lower in fish fed the SA0.25, SA0.50, and SB0.25 diets than in the other groups. Further, catalase (CAT) and glutathione reductase (GR) activity and lipid peroxidation (LPO) levels showed no differences between groups. In contrast, glutathione peroxidase (GPX) activity was higher in fish fed the SP0.50 diet. In the liver, GR activity and LPO levels showed no differences between groups. In contrast, CAT activity was lower in all dietary treatments than in control, and GPX and G6PDH activity was lower in fish fed with the SB (0.25 and 0.50 %) diet than in the other diets. Overall, SCFA supplementation did not affect growth performance and feed utilization and only had minor effects on metabolism and antioxidant defense mechanisms.
Collapse
Affiliation(s)
- F Fontinha
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av, General Norton de Matos s/n., 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, University of Porto, Rua do Campo Alegre s/n, Ed. FC4, 4169-007 Porto, Portugal
| | - N Martins
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av, General Norton de Matos s/n., 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, University of Porto, Rua do Campo Alegre s/n, Ed. FC4, 4169-007 Porto, Portugal.
| | - R Magalhães
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av, General Norton de Matos s/n., 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, University of Porto, Rua do Campo Alegre s/n, Ed. FC4, 4169-007 Porto, Portugal
| | - H Peres
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av, General Norton de Matos s/n., 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, University of Porto, Rua do Campo Alegre s/n, Ed. FC4, 4169-007 Porto, Portugal
| | - A Oliva-Teles
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av, General Norton de Matos s/n., 4450-208 Matosinhos, Portugal; Departamento de Biologia, Faculdade de Ciências, University of Porto, Rua do Campo Alegre s/n, Ed. FC4, 4169-007 Porto, Portugal
| |
Collapse
|
17
|
Salsinha AS, Araújo-Rodrigues H, Dias C, Cima A, Rodríguez-Alcalá LM, Relvas JB, Pintado M. Omega-3 and conjugated fatty acids impact on human microbiota modulation using an in vitro fecal fermentation model. Clin Nutr 2025; 49:102-117. [PMID: 40262394 DOI: 10.1016/j.clnu.2025.04.007] [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: 03/14/2024] [Revised: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND & AIMS Gut microbiota has been gaining increasing attention and its important role in the maintenance of a general good health condition is already established. The potential of gut microbiota modulation through diet is an important research focus to be considered. Lipids, as omega-3 fatty acids, are well known for their beneficial role on organs and corresponding diseases. However, their impact on gut microbiota is still poorly defined, and studies on the role of other polyunsaturated fatty acids, such as conjugated linoleic and linolenic acids, are even scarcer. METHODS By using an in vitro human fermentation model, we assessed the effect of omega-3, CLA isomers, and punicic acid on microbiota modulation. RESULTS Fish oil, Omega-3, and CLA samples positively impact Akkermansia spp. and Bifidobacterium spp. growth. Moreover, all the samples supported Roseburia spp. growth after 24 h of fermentation and, importantly, they were able to maintain the Firmicutes: Bacteroidetes ratio near 1. All the bioactive fatty acid samples, except Pomegranate oil, were able to significantly increase butyrate levels compared to those found in the positive control (FOS) sample. Moreover, Fish oil and Omega-3 samples were able to increase the concentration of GABA, alanine, tyrosine, phenylalanine, isoleucine, and leucine between 12 and 24 h of fermentation. CONCLUSIONS The impact of the assessed polyunsaturated fatty acids in gut microbiota has been observed in its impact on key bacteria (Akkermansia, Bifidobacterium, and Roseburia) as well as their metabolic byproducts, including butyrate and amino acids, which could potentially play a role in modulating the gut-brain axis.
Collapse
Affiliation(s)
- Ana Sofia Salsinha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal; Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Helena Araújo-Rodrigues
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal; Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Cindy Dias
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - André Cima
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - Luís Miguel Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Manuela Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
18
|
Rong X, Zhu L, Shu Q. Synergistic gut microbiome-mediated degradation of Astragalus membranaceus polysaccharides and Codonopsis pilosula polysaccharides into butyric acid: a metatranscriptomic analysis. Microbiol Spectr 2025:e0303924. [PMID: 40422281 DOI: 10.1128/spectrum.03039-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/16/2025] [Indexed: 05/28/2025] Open
Abstract
Astragalus membranaceus and Codonopsis pilosula are traditional Chinese medicines known for their tonifying effects, which are linked to the metabolism of their polysaccharide components in the gut. However, the role of gut microbiota in the degradation of these polysaccharides to butyric acid remains unclear. This study aims to investigate the in vitro degradation of polysaccharides from Astragalus membranaceus and Codonopsis pilosula by healthy mice fecal microbiota, focusing on butyric acid production and the associated microbial gene expression. We conducted an in vitro analysis of the degradation of homogeneous polysaccharides. from Astragalus membranaceus and Codonopsis pilosula using fecal microbiota cultures derived from healthy mice. The fecal microbiota was cultured with the polysaccharides for 48 hours, after which the degradation liquid was collected for butyric acid quantification and metatranscriptome analysis of the microbiota. The degradation of Astragalus membranaceus polysaccharide resulted in a significant increase in butyric acid levels compared to those produced from Codonopsis pilosula polysaccharide or fructooligosaccharide (control). Differential gene expression analysis indicated an upregulation of carbohydrate-active enzymes and genes associated with butyrate production during the degradation of Astragalus membranaceus polysaccharides. Additionally, the findings suggested that synergistic interactions between polysaccharide-degrading and butyrate-producing bacteria play a crucial role in the microbiota's response to specific polysaccharides. This study highlights the potential of Astragalus polysaccharides to enhance butyric acid production through specific gut microbiota interactions, suggesting their beneficial effects on gut health and metabolism. Further research may provide insights into the therapeutic applications of these traditional medicines in modulating gut microbiota and improving health outcomes.IMPORTANCEThis study significantly advances our understanding of the role of gut microbiota in the metabolism of traditional Chinese medicinal polysaccharides, specifically those from Astragalus membranaceus and Codonopsis pilosula. By demonstrating that Astragalus membranaceus polysaccharide enhances butyric acid production more effectively than Codonopsis pilosula polysaccharide or fructooligosaccharides, the research highlights the potential of these natural compounds in modulating gut health. The identification of upregulated carbohydrate-active enzymes and butyrate production genes provides valuable insights into the microbial mechanisms underlying polysaccharide degradation. This work not only contributes to the field of microbiome research but also supports the development of functional foods and therapeutics aimed at enhancing gut health through targeted polysaccharide consumption.
Collapse
Affiliation(s)
- XinQian Rong
- College of traditional Chinese medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - LingFeng Zhu
- College of traditional Chinese medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - QingLong Shu
- College of traditional Chinese medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Jan A, Bayle P, Mohellibi N, Lemoine C, Pepke F, Béguet-Crespel F, Jouanin I, Tremblay-Franco M, Laroche B, Serror P, Rigottier-Gois L. A consortium of seven commensal bacteria promotes gut microbiota recovery and strengthens ecological barrier against vancomycin-resistant enterococci. MICROBIOME 2025; 13:129. [PMID: 40414934 DOI: 10.1186/s40168-025-02127-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 04/29/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Vancomycin-resistant enterococci (VRE) often originate from the gastrointestinal tract, where their proliferation precedes dissemination into the bloodstream, and can lead to systemic infection. Uncovering the actors and mechanisms reducing the intestinal colonisation by VRE is essential to control infection. We aimed to identify commensal bacteria that interfere with VRE gut colonisation or act as an ecological barrier. RESULTS We performed a 3-week longitudinal analysis of the gut microbiota composition and VRE carriage levels during microbiota recovery in mice colonised with VRE after antibiotic-induced dysbiosis. By combining biological data and mathematical modelling, we identified 15 molecular species (OTUs) that negatively correlated with VRE overgrowth. Six strains representative of these OTUs were collected, cultivated and used in mixture with a seventh strain (Mix7) in two different mouse lines challenged with VRE. Of the seven strains, three belonged to Lachnospiraceae, one to Muribaculaceae, one to Ruminococcaceae and two to Lactobacillaceae. We found that Mix7 led to a better recovery of the gut microbiota composition and reduced VRE carriage. Differences in the effect of Mix7 were observed between responder and non-responder mice. These differences were associated with variations in the composition of the initial microbiota and during recovery and represent potential biomarkers for predicting response to Mix7. In a mouse model of alternative stable state of dysbiosis, response to Mix7 was associated with higher concentrations of short-chain fatty acids (acetate, propionate, butyrate) and a range of metabolites including bile acids, reflecting the recovery of the microbiota back to initial state. Furthermore, Muribaculum intestinale strain was required to obtain the Mix7 effect on VRE reduction in vivo, but the presence of at least one of the other six strains was needed. None of the supernatant of the seven strains, alone or in combination, inhibited VRE growth in vitro. Interestingly, five strains belong to species shared among humans and mice, and the other two have human functional equivalents. CONCLUSIONS An innovative approach based on mathematical modelling of the microbiota composition permitted to identify a mixture of commensal bacterial strains, which improves the ecological barrier effect against VRE. The mechanisms are dependent on the recovery and initial composition of the microbiota. Ultimately, this work will enable a move towards a personalised medicine by targeting predisposed patients presenting a risk of infection, such as neutropenic or bone-marrow transplant patients, and likely to respond to supplementation with commensal strains, providing new live biotherapeutic products and biomarkers to predict response to supplementation. Video Abstract.
Collapse
Affiliation(s)
- Alan Jan
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Perrine Bayle
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Nacer Mohellibi
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Clara Lemoine
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Frédéric Pepke
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Fabienne Béguet-Crespel
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Isabelle Jouanin
- Toxalim - Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, Toulouse University, UT3, Toulouse, F-31300, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | - Marie Tremblay-Franco
- Toxalim - Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, Toulouse University, UT3, Toulouse, F-31300, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | - Béatrice Laroche
- MaIAGE, INRAE, Université Paris-Saclay, Jouy-en-Josas, 78350, France
- MUSCA, INRIA, Université Paris-Saclay, Palaiseau, 91120, France
| | - Pascale Serror
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France
| | - Lionel Rigottier-Gois
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, F-78350, France.
| |
Collapse
|
20
|
Zhang Y, Wu H, Jin M, Feng G, Wang S. The gut-heart axis: unveiling the roles of gut microbiota in cardiovascular diseases. Front Cardiovasc Med 2025; 12:1572948. [PMID: 40491716 PMCID: PMC12146390 DOI: 10.3389/fcvm.2025.1572948] [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: 02/13/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
The gut microbiome refers to the collective genomes of the approximately 1,000-1,150 microbial species found in the human gut, called the gut microbiota. Changing the gut microbiota composition has been shown to affect cardiovascular health significantly. Numerous studies have demonstrated the part that gut microbiota and its metabolites play in the development and course of several illnesses, including colon cancer, heart failure, stroke, hypertension, and inflammatory bowel disease. With cardiovascular diseases responsible for more than 31% of all fatalities globally, conditions like hypertension, atherosclerosis, and heart failure are serious global health issues. Developing preventive measures to fight cardiovascular diseases requires understanding how the gut microbiota interacts with the cardiovascular system. Understanding the distinctive gut microbiota linked to cardiovascular diseases has been made possible by microbial sequencing analysis. The gut microbiota and cardiovascular diseases are closely related, and more profound knowledge of this association may result in treatment strategies and broad guidelines for enhancing cardiovascular health through gut microbiome modification. This review summarizes the role of gut microbiota in cardiovascular diseases, highlighting their influence on disease progression and potential therapeutic implications.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, International Mongolian Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China
| | - Huimin Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mu Jin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guirong Feng
- Department of Anesthesiology, International Mongolian Hospital of Inner Mongolia, Hohhot, Inner Mongolia, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Linzhi People’s Hospital, Linzhi, Tibet, China
| |
Collapse
|
21
|
Tang H, Chai X, Qin X, Lian YE, Gao Y, Wang H, Guo J, Wang B, Wang Y. The impact of mechanical unloading on the gut microbiota and the mitigating role of butyrate in bone loss. Int Immunopharmacol 2025; 159:114909. [PMID: 40424649 DOI: 10.1016/j.intimp.2025.114909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/07/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025]
Abstract
Bone loss induced by mechanical unloading is a critical concern in aerospace medicine and for patients subjected to prolonged bed rest. In this study, we investigated the effects of mechanical unloading on the gut microbiota and short-chain fatty acid (SCFA) metabolism, as well as the potential role of butyrate in mitigating bone loss. Through a combination of a hindlimb unloading model, microbiological assessments, and metabolic profiling, we demonstrated that mechanical unloading significantly reduced gut microbiota diversity, altered the Firmicutes-to-Bacteroidetes ratio, and decreased total SCFA levels. Furthermore, butyrate supplementation mitigated the adverse effects of mechanical unloading on the bone microstructure by increasing ELK1 protein expression through HDAC2 inhibition, thus promoting osteoblast differentiation and bone formation. These findings highlight the intricate relationships among gut microbiota alterations, SCFA metabolism, and bone health during mechanical unloading, suggesting that butyrate may be a potential therapeutic agent to counteract bone loss in microgravity environments.
Collapse
Affiliation(s)
- Hanqin Tang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China; Department of Basic Medical Laboratory, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Xin Chai
- Department of Emergency, An Ning Attached Medical Area, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Xuezhi Qin
- Graduate School of Gansu University of Traditional Chinese Medicine, Lanzhou 730050, China
| | - Yu-E Lian
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China; Graduate School of Gansu University of Traditional Chinese Medicine, Lanzhou 730050, China
| | - Yuhai Gao
- Department of Basic Medical Laboratory, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Hongli Wang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Jing Guo
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Biaomeng Wang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China
| | - Yixuan Wang
- Department of Gastroenterology, the 940th Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou 730050, China.
| |
Collapse
|
22
|
Yin D, Cao X, Li S, Zhang Y, Gao C, Zhu M, Li D. Antiallergic activities of polysaccharides derived from Coptis chinensis. Int J Biol Macromol 2025; 315:144681. [PMID: 40425117 DOI: 10.1016/j.ijbiomac.2025.144681] [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: 08/12/2024] [Revised: 05/20/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
To assess the effects and underlying mechanisms of Coptis chinensis, a traditional Chinese herb known for its antiallergic properties, crude polysaccharides (CP1) were extracted via water extraction, and homopolysaccharides were purified by column chromatography. This study analyzed the sugar and protein content, molecular weight distribution, monosaccharide composition, and functional groups of C. chinensis polysaccharides and evaluated their antiallergic effects in mice sensitized to ovalbumin (OVA). The results showed that the purified homopolysaccharide (CP2-3-1, 138.78 kDa, primarily composed of glucose) was more effective than CP1 in alleviating allergic symptoms. CP2-3-1 treatment led to improvements in anaphylactic scores, diarrhea rates, rectal temperatures, jejunum structure, and levels of immunoglobulin E (IgE), mouse mast cell protease-1 (mMcep1), and interleukin-4 (IL-4) antibodies in mouse serum, which were similar to those in the phosphate-buffered saline (PBS) control group. In addition, CP2-3-1 treatment significantly increased the richness and diversity of the gut microbiota in OVA-sensitized mice, restoring core microbiota to near-PBS levels, including key phyla (Firmicutes and Bacteroidetes) and important families (Lachnospiraceae, Lactobacillaceae, Rikenellaceae, and Muribaculaceae). This research confirms the antiallergic effects of polysaccharides from C. chinensis and highlights the potential for developing new food allergy therapies based on C. chinensis homopolysaccharides.
Collapse
Affiliation(s)
- Dangyue Yin
- College of Forestry, Northwest A&F University, Yangling 712100, Shaanxi, China; School of Astronautics, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China
| | - Xinyuan Cao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham, Ningbo 315048, Zhejiang, China
| | - Shanshan Li
- College of Forestry, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ying Zhang
- College of Forestry, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Changyuan Gao
- College of Forestry, Shanxi Agricultural University, Taigu 030801, China
| | - Mingqiang Zhu
- College of Mechanical and Electronic Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Dongbing Li
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham, Ningbo 315048, Zhejiang, China.
| |
Collapse
|
23
|
Wang W, Wu H, Li J, Chen Z, Liu J, Xu E, Hassanin AA, Rahman SU, Chen L, Zheng R. The effect of anthocyanin extract from Lycium ruthenicum Murray on intestinal barrier function in Bamei ternary pigs. PROTOPLASMA 2025:10.1007/s00709-025-02075-9. [PMID: 40410595 DOI: 10.1007/s00709-025-02075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 05/14/2025] [Indexed: 05/25/2025]
Abstract
The intestinal barrier is a critical defense against external pathogens and plays a central role in immune regulation and nutrient absorption. Oxidative stress and chronic inflammation in high-altitude environments can exacerbate the damage to the intestinal barrier in Baimei ternary pigs. Anthocyanin extract of Lycium ruthenicum Murray (AEL), has garnered widespread attention due to its rich anthocyanin flavonoid content, which exhibits antioxidant and anti-inflammatory properties. These properties help alleviate inflammation and oxidative stress, thereby enhancing gut function in animals. Based on this, the study employed Bamei ternary pigs and supplemented their basic diet with varying concentrations of AEL to investigate its impact on gut barrier function. The results demonstrated that AEL inhibited key factors of the intestinal Toll-like receptor pathway, including Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), tumor necrosis factor receptor-associated protein 6 (TRAF6), and nuclear factor kappa B (NF-κB), affecting gene transcription and protein expression levels. This led to a reduction in pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), an increase in anti-inflammatory IL-10 production, and improved antioxidant capacity by enhancing total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activity, while decreasing malondialdehyde (MDA) production. Additionally, AEL improved intestinal morphology and facilitated the transcription and expression of tight junction proteins, including zonula occludens-1 (ZO-1), claudin-1 (CLDN-1), and occludin (OCLN). AEL also elevated the transcription levels of mucin 1 (MUC1) and mucin 2 (MUC2), as well as the secretion levels of polymeric immunoglobulin receptor (pIgR) and secretory immunoglobulin A (SIgA), while increased the number of intestinal goblet cells. Furthermore, dietary supplementation with AEL altered the structure of the intestinal microbiota, enhancing the abundance of beneficial bacterial genera such as Verrucomicrobiaceae, Rikenellaceae, Butyricicoccaceae, UCG-005、Rikenellaceae_RC9_gut_group、norank_f_Ruminococcaceae、Eubacterium_oxidoreducens_group, thereby promoting the production of intestinal short-chain fatty acids (SCFAs). In conclusion, AEL inhibits the Toll-like receptor pathway, reduces the production of inflammatory factors, enhances antioxidant levels, improves intestinal morphology and microbiota structure,, thereby reinforcing intestinal barrier function.
Collapse
Affiliation(s)
- Wensheng Wang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Hua Wu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China.
| | - Jinming Li
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Zixin Chen
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Jiayi Liu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Enron Xu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Abdallah A Hassanin
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Siddiq Ur Rahman
- Department of Computer Science and Bioinformatics, Khushal Khan Khattak University, Karak, Pakistan
| | - Lin Chen
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Runtao Zheng
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| |
Collapse
|
24
|
Iqbal M, Yu Q, Tang J, Xiang J. Unraveling the gut microbiota's role in obesity: key metabolites, microbial species, and therapeutic insights. J Bacteriol 2025; 207:e0047924. [PMID: 40183584 PMCID: PMC12096833 DOI: 10.1128/jb.00479-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Obesity, characterized by excessive fat accumulation, stems from an imbalance between energy intake and expenditure, with the gut microbiota playing a crucial role. This review highlights how gut microbiota influences metabolic pathways, inflammation, and adipose tissue regulation in obesity. Specific bacteria and metabolites, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), modulate gut permeability, inflammation, and energy harvest, impacting obesity development. Certain gut bacteria, including Clostridium XIVb, Dorea spp., Enterobacter cloacae, and Collinsella aerofaciens, promote obesity by increasing energy harvest, gut permeability, and inflammatory response through LPS translocation into the bloodstream. Conversely, beneficial bacteria like Akkermansia muciniphila, Lactobacillus spp., and Bifidobacterium spp. enhance gut barrier integrity, regulate SCFA production, and modulate fasting-induced adipose factor, which collectively support metabolic health by reducing fat storage and inflammation. Metabolites such as SCFAs (acetate, propionate, and butyrate) interact with G-protein coupled receptors to regulate lipid metabolism and promote the browning of white adipose tissue (WAT), thus enhancing thermogenesis and energy expenditure. However, LPS contributes to insulin resistance and fat accumulation, highlighting the dual roles of these microbial metabolites in both supporting and disrupting metabolic function. Therapeutic interventions targeting gut microbiota, such as promoting WAT browning and activating brown adipose tissue (BAT), hold promise for obesity management. However, personalized approaches are necessary due to individual microbiome variability. Further research is essential to translate these insights into microbiota-based clinical therapies.
Collapse
Affiliation(s)
- Majid Iqbal
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Yu
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
de Moraes Arnoso BJ, de Araújo CA, Ramundo GD, de Bem GF, Ognibene DT, Fontes-Dantas FL, Martins BC, Daleprane JB, de Souza MO, Resende AC, da Costa CA. Açaí seed extract mitigates intestinal and hypothalamic alterations in obese mice. Mol Cell Endocrinol 2025; 606:112574. [PMID: 40409530 DOI: 10.1016/j.mce.2025.112574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/16/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Obesity is a significant health concern, significantly contributing to increased morbidity and mortality by disrupting multiple physiological systems. It is strongly associated with metabolic dysfunctions, including impaired glycemic homeostasis, compromised intestinal barrier integrity, and gut microbiota imbalances, all exacerbating the risk of chronic diseases. The hydroalcoholic extract of açaí seeds (ASE), rich in phenolic compounds, has demonstrated beneficial effects on obesity and hyperglycemia; however, its impacts on gut health and gut-hypothalamus communication remain unclear. This study aimed to investigate the therapeutic effect of ASE in intestinal and hypothalamic alterations associated with obesity and compare it with Metformin. Male C57BL/6 mice were fed a high-fat or standard diet for 14 weeks. The ASE (300 mg/kg/day) and Metformin (300 mg/kg/day) treatments started in the tenth week until the fourteenth week, totaling four weeks of treatment. Our data show that the treatment with ASE and Metformin reduced body weight, ameliorated lipid profile, hyperglycemia, and plasma hyperleptinemia, and decreased the oxidative damage in the gut by reducing immunostaining of 8-isoprostane and NOX-4 expression, and improved the intestinal parameters and hypothalamic gene expression. Obesity-induced dysbiosis in the HF group was marked by reduced Proteobacteria and elevated LPS plasma levels, which were improved by treatments with ASE and Metformin. These findings suggest that ASE and Metformin are promising strategies to counteract the adverse effects of obesity on intestinal health and gut-hypothalamus communication, though they act through distinct mechanisms. Therefore, we can suggest that ASE is a promising natural product for treating the intestinal alterations associated with obesity.
Collapse
Affiliation(s)
| | - Caroline Alves de Araújo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Giovana Dias Ramundo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Fabricia Lima Fontes-Dantas
- Neurogenetics Laboratory, Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Bruna Cadete Martins
- Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
26
|
Cao Q, Shen M, Li R, Liu Y, Zeng Z, Zhou J, Niu D, Zhang Q, Wang R, Yao J, Zhang G. Elucidating the specific mechanisms of the gut-brain axis: the short-chain fatty acids-microglia pathway. J Neuroinflammation 2025; 22:133. [PMID: 40400035 PMCID: PMC12093714 DOI: 10.1186/s12974-025-03454-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/22/2025] [Indexed: 05/23/2025] Open
Abstract
In recent years, the gut microbiota has been increasingly recognized for its influence on various central nervous system diseases mediated by microglia, yet the underlying mechanisms remain unclear. As key metabolites of the gut microbiota, short-chain fatty acids (SCFAs) have emerged as a focal point in understanding microglia-related interactions. In this review, we further refine the connection between the gut microbiota and microglia by introducing the concept of the "SCFAs-microglia" pathway. We summarize current knowledge on this pathway, recent discoveries regarding its role in neurological diseases, and potential pharmacological strategies targeting it. Finally, we outlined the current challenges and limitations in this field of research. We hope this review provides new insights into the role of the gut microbiota in neuroimmune regulation.
Collapse
Affiliation(s)
- Qingyu Cao
- College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Ruoqiu Li
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yan Liu
- School of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Quancai Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China
| | - Rongrong Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China.
| | - Guimin Zhang
- College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd, Linyi, 276005, China.
| |
Collapse
|
27
|
Throat S, Bhattacharya S. The Role of RS Type 2 (High-Amylose Maize Starch) in the Inhibition of Colon Cancer: A Comprehensive Review of Short-Chain Fatty Acid (SCFA) Production and Anticancer Mechanisms. Mol Nutr Food Res 2025:e70107. [PMID: 40392033 DOI: 10.1002/mnfr.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/30/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Dietary fiber, especially resistant starch (RS) Type 2 (RS2) found in high-amylose maize starch (HAMS), is vital for gut health and helps prevent colon cancer. In contrast to most nutrients, dietary fiber is not degraded by the intestinal enzymes; it reaches the distal parts of the gut, where it is fermented by the gut microbiota into short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate. SCFAs energize colonocytes, reduce inflammation, and enhance gut immunity. HAMS is absorbed in the colon, where it ferments to create SCFAs that feed good gut flora and have antiinflammatory and antiproliferative effects. RS2 in HAMS modulates gene signaling, activates tumor-suppressor genes like tumor suppressor protein (p53), exhibits antidiabetic, cholesterol-lowering, and antiinflammatory effects. Incorporation of RS2-rich sources enhances gut barriers, decreases colorectal cancer biomarkers, and counteracts the negative impacts of low-fiber Western diets, making HAMS a promising functional food for chronic disease prevention and health promotion.
Collapse
Affiliation(s)
- Siddhi Throat
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| |
Collapse
|
28
|
Ma T, Li Y, Yang N, Wang H, Shi X, Liu Y, Jin H, Kwok LY, Sun Z, Zhang H. Efficacy of a postbiotic and its components in promoting colonic transit and alleviating chronic constipation in humans and mice. Cell Rep Med 2025; 6:102093. [PMID: 40286792 DOI: 10.1016/j.xcrm.2025.102093] [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: 06/26/2024] [Revised: 12/06/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
This study evaluates the efficacy of the postbiotic Probio-Eco in alleviating constipation in humans and mice. A randomized, double-blind, placebo-controlled crossover trial involving 110 adults with chronic constipation (Rome IV criteria) demonstrates that a 3-week Probio-Eco intervention significantly improves constipation symptoms, stool straining, and worry scores. Gut microbiota and metabolomic analyses reveal modulations in specific gut microbiota, succinate, tryptophan derivatives, deoxycholate, propionate, butyrate, and cortisol, correlating with symptom relief. A loperamide-induced mouse model confirms that Probio-Eco and its bioactive components (succinate, 3-indoleacrylic acid, and 5-hydroxytryptophan) alleviate constipation by stimulating mucin-2 secretion, regulating intestinal transport hormones, and promoting anti-inflammatory responses. Multi-omics integration identifies key pathways, including succinate-short-chain fatty acid, tryptophan-5-hydroxytryptophan-serotonin, and tryptophan-3-indoleacrylic acid, driving intestinal homeostasis and motility. These findings highlight the comprehensive efficacy of Probio-Eco and provide robust evidence for its clinical application in constipation management. This study was registered at Chinese Clinical Trial Registry (ChiCTR2100054376).
Collapse
Affiliation(s)
- Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Ni Yang
- State Key Laboratory of Research and Development of Classical Prescription and Modern Chinese Medicine, 1899 Meiling Road, Nanchang 330103, China
| | - Huan Wang
- Inner Mongolia People's Hospital, Hohhot, China
| | - Xuan Shi
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanfang Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
29
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
30
|
Maldonado-Gomez MX, Ng KM, Drexler RA, Conner AMS, Vierra CG, Krishnakumar N, Gerber HM, Taylor ZR, Treon JL, Ellis M, Garcia JKA, Cerney JP, Chapin DG, Kerns RT, Marcobal AM, Watkins SM, Amicucci MJ. A diverse set of solubilized natural fibers drives structure-dependent metabolism and modulation of the human gut microbiota. mBio 2025; 16:e0047025. [PMID: 40214223 PMCID: PMC12077125 DOI: 10.1128/mbio.00470-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
Growing evidence suggests that inadequate dietary fiber intake, termed the "fiber gap," is linked to disease states through disruption of the gut microbiota. Despite this, our understanding of how various fiber structures influence the microbiota and health is limited by the lack of diverse commercially available fibers. Studies have primarily focused on a limited range of fibers, rather than the diverse array of fibers representative of those commonly found in our diets. In this study, we aimed to investigate how naturally derived fibers impact the human microbiota and their metabolic products. We performed a comprehensive structural characterization and functional evaluation of a unique and highly diverse set of new, highly soluble fibers with varied monosaccharide compositions, glycosidic linkages, and polymer lengths. Using an ex vivo high-throughput human microbiota platform coupled with metabolomic profiling, we demonstrate that these diverse fibers drive distinct and consistent microbial and metabolic profiles across cohorts of donors in a structure-dependent manner. These metabolic effects were accompanied by both general and donor-specific changes in microbial taxa. Finally, we demonstrate that integrating detailed glycomic characterization with microbial and metabolomic data allowed for prediction of functional outcomes driven by a novel material, pineapple pulp fiber. This work highlights the potential for targeted dietary fiber interventions to modulate the microbiota and improve health outcomes, paving the way for the development of new fiber-rich products with specific health benefits.IMPORTANCEFiber deficiency is associated with numerous disease states, many of which are linked to disruption of the gut microbiota. This study encompasses the first systematic and comprehensive characterization of a diverse collection of naturally derived solubilized fibers and their impacts on the microbiota. The results expand our understanding of the beneficial effects of specific carbohydrate structures naturally found in the human diet, highlighting the potential for designing fiber-based health interventions. The high solubility of these fibers increases both the range of products they can be incorporated in as well as their assayability in experiments, enabling a widespread increase in fiber consumption and positive health impacts.
Collapse
|
31
|
Bao L, Cui X, Zeng T, Liu G, Lai W, Zhao H, Gao F, Wu J, Leong KW, Chen C. Incorporation of polylactic acid microplastics into the carbon cycle as a carbon source to remodel the endogenous metabolism of the gut. Proc Natl Acad Sci U S A 2025; 122:e2417104122. [PMID: 40324088 DOI: 10.1073/pnas.2417104122] [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: 08/22/2024] [Accepted: 04/02/2025] [Indexed: 05/07/2025] Open
Abstract
Biodegradable polylactic acid (PLA) plastics have been praised as an effective solution to the global pollution caused by petroleum-based plastics, and their widespread use in food packaging and disposable tableware has resulted in increased oral exposure to PLA microplastics (PLA-MPs). Despite their eco-friendly and biodegradable reputation, the in vivo behaviors of PLA-MPs concerning fermentation, carbon cycle, and adverse effects remain unknown. Here, we showed that gut microbiota from the colon can effectively degrade the PLA-MPs by secreting esterase FrsA, whereas esterase FrsA-producing bacteria were identified to dominate this behavior in male C57BL/6 mice. Using isotope tracing and multiomics techniques, we uncovered that 13C-labeled PLA-MPs were incorporated into the carbon cycle of gut microbiota as a carbon source. Meanwhile, these degraded PLA-MPs fragments entered the succinate pathway of the tricarboxylic acid cycle within gut epithelial cells. These processes altered the metabolic phenotype of the gut, resulting in the decreased linear short-chain fatty acids that are primary energy sources of the gut epithelium. Furthermore, we found that exposure of PLA-MPs significantly reduced the appetite and body weight of mice. Our findings present an overall process of biodegradable plastics within hosts, with the focus on the entire double carbon cycle of PLA-MPs in the gut, which offers indispensable insights into the potential impact of exposure to PLA-MPs.
Collapse
Affiliation(s)
- Lin Bao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejing Cui
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Tao Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guanyu Liu
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjia Lai
- Division of Nanotechnology Development, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Hao Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Fene Gao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Junguang Wu
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Chunying Chen
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
32
|
Mukhopadhya I, Louis P. Gut microbiota-derived short-chain fatty acids and their role in human health and disease. Nat Rev Microbiol 2025:10.1038/s41579-025-01183-w. [PMID: 40360779 DOI: 10.1038/s41579-025-01183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/15/2025]
Abstract
Short-chain fatty acids (SCFAs) are a group of organic compounds produced by the fermentation of dietary fibre by the human gut microbiota. They play diverse roles in different physiological processes of the host with implications for human health and disease. This Review provides an overview of the complex microbial metabolism underlying SCFA formation, considering microbial interactions and modulating factors of the gut environment. We explore the multifaceted mechanistic interactions between SCFAs and the host, with a particular focus on the local actions of SCFAs in the gut and their complex interactions with the immune system. We also discuss how these actions influence intestinal and extraintestinal diseases and emerging therapeutic strategies using SCFAs.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Petra Louis
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
33
|
Ghyselinck J, Verstrepen L, Rakebrandt M, Marynissen S, Daminet S, Marzorati M. In vitro fermentation of yeast cell walls (mannan-oligosaccharide) and purified β-glucans modulates the colonic microbiota of dogs with inflammatory bowel disease and demonstrates protective effects on barrier integrity and anti-inflammatory properties. PLoS One 2025; 20:e0322877. [PMID: 40359204 PMCID: PMC12074461 DOI: 10.1371/journal.pone.0322877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by a disruption of intestinal homeostasis, chronic inflammation, and dysbiosis. Prebiotic supplementation may be useful for managing IBD in dogs. The aim of the study is to investigate the effects of two prebiotics, Biolex MB40 or Leiber Beta-S, on the gut microbiota isolated from three dogs with IBD, using the Colon-on-a-plate technology. Biolex MB40 and Leiber Beta-S contain concentrated 1,3-1,6- β-D-glucan isolated from the Saccharomyces cerevisiae cell walls. Biolex MB40 also contains mannan-oligosaccharide (MOS). Wells of the Colon-on-a-plate set up were inoculated with fecal suspensions and supplemented with either Biolex MB40 and Leiber Beta-S, or no test product (blank). Following 48h incubation, bacterial metabolites were measured and 16S rRNA targeted gene sequencing was performed. Colonic supernatants were added to a Caco-2/THP1 co-culture model to evaluate their effects on barrier integrity upon inflammation-induced barrier disruption and interleukin (IL)-10 production. Acetate and propionate concentrations were significantly increased versus blank with Biolex MB40, and biologically relevant numerical increases were observed with Leiber Beta-S supplementation. A donor-dependent, biologically relevant increase in butyrate was observed with both test products versus blank. Alpha diversity and microbiota biomass were increased, as well as the abundance of the five predominant phyla with both test products relative to blank. The greatest increases in abundance were observed for the Bacteroidetes and Firmicutes phyla. Fermentation of both test products had a protective effect on the gut epithelial barrier (measured by transepithelial electrical resistance) that was donor dependent. IL-10 production was significantly increased with Biolex MB40 supplementation for all donors, and with Leiber Beta-S supplementation for one donor. These in vitro findings confirm a prebiotic effect for both products and suggest that supplementation with either Biolex MB40 or Leiber Beta-S may have beneficial effects on the gut microbiota of dogs with IBD.
Collapse
Affiliation(s)
| | | | | | - Sofie Marynissen
- Small Animal Department, Faculty of Veterinary Medicine, Merelbeke, Belgium
| | - Sylvie Daminet
- Small Animal Department, Faculty of Veterinary Medicine, Merelbeke, Belgium
| | - Massimo Marzorati
- ProDigest, Zwijnaarde, Belgium
- Center of Microbial Ecology and Technology (CMET), Ghent University, Ghent, Belgium
| |
Collapse
|
34
|
Ramar M, Wiscovitch-Russo R, Yano N, Singh H, Lamere E, Short M, Gonzalez-Juarbe N, Fedulov AV. Live bacteria in gut microbiome dictate asthma onset triggered by environmental particles via modulation of DNA methylation in dendritic cells. Cell Rep 2025; 44:115684. [PMID: 40372916 DOI: 10.1016/j.celrep.2025.115684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/24/2025] [Accepted: 04/18/2025] [Indexed: 05/17/2025] Open
Abstract
Despite broad knowledge of the pathogenesis, our understanding of the origin of allergy and asthma remains poor, preventing etiotropic treatments. The gut microbiome is seen to be altered in asthmatics; however, proof of causality of the microbiome alterations is lacking. We report on gut microbiome transplantation (GMT) from mice predisposed to asthma by maternal exposure to pro-allergy environmental particles into naive recipients. This GMT confers asthma predisposition, and the effect is abrogated by gamma sterilization of the transplant material or by co-administration of antibacterials, indicating that viable bacteria are mediating the effect. Metagenomics identifies key changes in the "pro-asthma" microbiome, and metabolomics links the identified species to altered production of butyrate known to act on immune cells and epigenetic mechanisms. We further show that transplant recipients develop DNA methylation alterations in dendritic cells. Finally, dendritic cells with an altered methylome present allergen to T cells, and this effect is abrogated by an epigenetically acting drug in vitro.
Collapse
Affiliation(s)
- Mohankumar Ramar
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Rosana Wiscovitch-Russo
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA
| | - Naohiro Yano
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Harinder Singh
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA
| | - Edward Lamere
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael Short
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Norberto Gonzalez-Juarbe
- Department of Infectious Diseases and Genomic Medicine, J. Craig Venter Institute, Rockville, MD, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA.
| | - Alexey V Fedulov
- Department of Surgery, Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
35
|
Mametja PM, Motshudi MC, Naidoo CM, Rakau K, Seheri LM, Mkolo NM. Tapping into Metabolomics for Understanding Host and Rotavirus Group A Interactome. Life (Basel) 2025; 15:765. [PMID: 40430193 PMCID: PMC12113392 DOI: 10.3390/life15050765] [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: 02/25/2025] [Revised: 04/27/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Group A rotavirus continues to be a leading global etiological agent of severe gastroenteritis in young children under 5 years of age. The replication of this virus in the host is associated with the occurrence of Lewis antigens and the secretor condition. Moreover, histo-blood group antigens (HBGAs) act as attachment factors to the outer viral protein of VP4 for rotavirus. Therefore, in this study, we employed a metabolomic approach to reveal potential signature metabolic molecules and metabolic pathways specific to rotavirus P[8] strain infection (VP4 genotype), which is associated with the expression of HBGA combined secretor and Lewis (Le) phenotypes, specifically secretor/Le(a+b+). Further integration of the achieved metabolomics results with lipidomic and proteomics metadata analyses was performed. Saliva samples were collected from children diagnosed as negative or positive for rotavirus P[8] strain infection (VP4 genotype), which is associated with the HBGA combined secretor/Le(a+b+). A total of 22 signature metabolic molecules that were downregulated include butyrate, putrescine, lactic acid, and 7 analytes. The upregulated metabolic molecule was 2,3-Butanediol. Significant pathway alterations were also specifically observed in various metabolism processes, including galactose and butanoate metabolisms. Butyrate played a significant role in viral infection and was revealed to exhibit different reactions with glycerolipids, glycerophospholipids, sphingolipids, sterol lipids, and fatty acyls. Moreover, butyrate might interact with protein receptors of free fatty acid receptor 2 (FFAR2) and free fatty acid receptor 3 (FFAR3). The revealed metabolic pathways and molecule might provide fundamental insight into the status of rotavirus P[8] strain infection for monitoring its effects on humans.
Collapse
Affiliation(s)
- Phiona Moloi Mametja
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Mmei Cheryl Motshudi
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Clarissa Marcelle Naidoo
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Kebareng Rakau
- Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (K.R.); (L.M.S.)
| | - Luyanda Mapaseka Seheri
- Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (K.R.); (L.M.S.)
| | - Nqobile Monate Mkolo
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| |
Collapse
|
36
|
Chen Y, Xie Y, Yu X. Progress of research on the gut microbiome and its metabolite short-chain fatty acids in postmenopausal osteoporosis: a literature review. Front Med 2025:10.1007/s11684-025-1129-3. [PMID: 40347368 DOI: 10.1007/s11684-025-1129-3] [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: 07/14/2024] [Accepted: 12/16/2024] [Indexed: 05/12/2025]
Abstract
Postmenopausal osteoporosis (PMOP) is a systemic metabolic bone disease caused by the decrease in estrogen levels after menopause. It leads to bone loss, microstructural damage, and an increased risk of fractures. Studies have found that the gut microbiota and its metabolites can regulate bone metabolism through the gut-bone axis and the gut-brain axis. As research progresses, PMOP has been found to be associated with gut microbiota dysbiosis and Th17/Treg imbalance. The gut microbiota is closely related to the development and differentiation of Treg and Th17 cells. Among them, the metabolites of the gut microbiota such as short-chain fatty acids (SCFAs) can regulate the differentiation of effector T cells by acting on molecular receptors on immune cells, thereby regulating the bone immune process. The multifaceted relationship among the gut microbiota, SCFAs, Th17/Treg cell-mediated bone immunity, and bone metabolism is eliciting attention from researchers. Through a review of existing literature, we have comprehensively summarized the effects of the gut microbiota and SCFAs on PMOP, especially from the perspective of Th17/Treg balance. Regulating this balance may provide new opportunities for PMOP treatment.
Collapse
Affiliation(s)
- Yao Chen
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Xie
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xijie Yu
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
37
|
Cheong KL, Sabir A, Wang M, Zhong S, Tan K. Advancements in the Extraction, Characterization, and Bioactive Potential of Laminaran: A Review. Foods 2025; 14:1683. [PMID: 40428463 PMCID: PMC12110979 DOI: 10.3390/foods14101683] [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: 04/13/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Laminaran, a bioactive β-glucan derived from brown algae, has garnered significant attention due to its diverse pharmacological properties, including antioxidant, immunomodulatory, and mucosal protective effects. Despite promising research highlighting its potential applications in functional foods, nutraceuticals, and pharmaceuticals, the commercial utilization of laminaran remains limited, primarily due to challenges in extraction efficiency, structural complexity, and a lack of standardized methodologies. This review critically examines recent advancements in the extraction, purification, structural characterization, and biological evaluation of laminaran. Both conventional and emerging extraction methods-including ultrasound-assisted extraction, microwave-assisted extraction, and enzymatic techniques-are evaluated for their efficiency, scalability, and sustainability. Analytical tools, such as high-performance liquid chromatography, nuclear magnetic resonance, and mass spectrometry, are discussed for their roles in elucidating key structural features, such as molecular weight, degree of polymerization, and glycosidic linkage patterns, which are closely tied to laminaran's biological activity. Innovative extraction technologies have improved yield and purity, while structural insights have deepened the understanding of structure-function relationships. Interdisciplinary collaboration will be critical to advance laminaran from a marine-derived polysaccharide to a commercially viable bioactive compound for health, nutrition, and biomaterial applications.
Collapse
Affiliation(s)
- Kit-Leong Cheong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.); (A.S.)
| | - Amanullah Sabir
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.); (A.S.)
| | - Min Wang
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (K.-L.C.); (A.S.)
| | - Karsoon Tan
- Guangxi Key Laboratory of Beibu Gulf Biodiversity Conservation, Beibu Gulf University, Qinzhou 535011, China
| |
Collapse
|
38
|
Jia Y, Liu X, Gao X, Yin S, Wu K, Meng X, Ren H, Liu J, Liu Z, Li H, Jiang Y. Plantamajoside alleviates DSS-induced ulcerative colitis by modulating gut microbiota, upregulating CBS, and inhibiting NF-κB. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156827. [PMID: 40381501 DOI: 10.1016/j.phymed.2025.156827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/18/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Plantamajoside (PMS) is a natural bioactive compound derived from medicinal, food homologous plants of the genus Plantago. PURPOSE AND METHODS This study aimed to investigate the protective effects of PMS on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice and explore the associated mechanisms. RESULTS We found that PMS treatment significantly alleviated UC symptoms in mice by preventing body weight loss, increasing colon length, and reducing disease activity index scores. Moreover, PMS alleviated colonic lesions, increased the number of goblet cells, upregulated the expression of intestinal barrier proteins (ZO-1, occludin, and claudin-3), and decreased the levels of pro-inflammatory factors. PMS treatment modulated the gut microbiota by increasing the relative abundance of Bacteroidota and Verrucomicrobiota and decreasing that of Firmicutes and Proteobacteria at the phylum level. At the genus level, PMS suppressed the abundance of pathogenic bacteria, such as Turicibacter and upregulated the abundance of [Eubacterium]_xylanophilum_group. Fecal microbiota transplantation experiments further confirmed that PMS treatment alleviated UC by modulating the gut microbiota. Transcriptomic analysis of colon tissues, coupled with reverse transcription-quantitative polymerase chain reaction and western blotting, showed that PMS treatment upregulated cystathionine beta-synthase (CBS) expression and inhibited NF-κB pathway activation. In a lipopolysaccharide-induced inflammation model in RAW264.7 cells, PMS treatment inhibited the secretion of pro-inflammatory cytokines, upregulated CBS expression, and prevented NF-κB pathway activation. CONCLUSION PMS protects against UC in mice via multiple mechanisms, including modulating the gut microbiota, increasing the expression levels of CBS, and inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Yongheng Jia
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Xianjun Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, No. 3050 Kaixuan Street, Changchun 130000, China; Postdoctoral Research Workstation, Changchun Gangheng Electronics Company Limited, Changchun 130000, China
| | - Xinyi Gao
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Siyuan Yin
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Kun Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Xianglong Meng
- Department of Gastroenterology, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Hui Ren
- Department of General surgery, the China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jiawei Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Zijing Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, No. 3050 Kaixuan Street, Changchun 130000, China
| | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, the China-Japan Union Hospital of Jilin University, No. 126 Xian Tai Street, Changchun 130000, China.
| |
Collapse
|
39
|
Hitch TCA, Masson JM, Pauvert C, Bosch J, Nüchtern S, Treichel NS, Baloh M, Razavi S, Afrizal A, Kousetzi N, Aguirre AM, Wylensek D, Coates AC, Jennings SAV, Panyot A, Viehof A, Schmitz MA, Stuhrmann M, Deis EC, Bisdorf K, Chiotelli MD, Lissin A, Schober I, Witte J, Cramer T, Riedel T, Wende M, Winter KA, Amend L, Riva A, Trinh S, Mitchell L, Hartman J, Berry D, Seitz J, Bossert LC, Grognot M, Allers T, Strowig T, Pester M, Abt B, Reimer LC, Overmann J, Clavel T. HiBC: a publicly available collection of bacterial strains isolated from the human gut. Nat Commun 2025; 16:4203. [PMID: 40328737 PMCID: PMC12056005 DOI: 10.1038/s41467-025-59229-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Numerous bacteria in the human gut microbiome remain unknown and/or have yet to be cultured. While collections of human gut bacteria have been published, few strains are accessible to the scientific community. We have therefore created a publicly available collection of bacterial strains isolated from the human gut. The Human intestinal Bacteria Collection (HiBC) ( https://www.hibc.rwth-aachen.de ) contains 340 strains representing 198 species within 29 families and 7 phyla, of which 29 previously unknown species are taxonomically described and named. These included two butyrate-producing species of Faecalibacterium and new dominant species associated with health and inflammatory bowel disease, Ruminococcoides intestinale and Blautia intestinihominis, respectively. Plasmids were prolific within the HiBC isolates, with almost half (46%) of strains containing plasmids, with a maximum of six within a strain. This included a broadly occurring plasmid (pBAC) that exists in three diverse forms across Bacteroidales species. Megaplasmids were identified within two strains, the pMMCAT megaplasmid is globally present within multiple Bacteroidales species. This collection of easily searchable and publicly available gut bacterial isolates will facilitate functional studies of the gut microbiome.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Johannes M Masson
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Johanna Bosch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Selina Nüchtern
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Nicole S Treichel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Marko Baloh
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Soheila Razavi
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Afrizal Afrizal
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Ntana Kousetzi
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Andrea M Aguirre
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Amy C Coates
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Susan A V Jennings
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Atscharah Panyot
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Alina Viehof
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Matthias A Schmitz
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Maximilian Stuhrmann
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Evelyn C Deis
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Kevin Bisdorf
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Maria D Chiotelli
- Biophysics of Host-Microbe Interactions Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Artur Lissin
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Isabel Schober
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Julius Witte
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Thorsten Cramer
- Molecular Tumor Biology Research Group, Department of General, Visceral, Children and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | - Thomas Riedel
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katrin A Winter
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alessandra Riva
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Stefanie Trinh
- Institute of Neuroanatomy, University Hospital of RWTH Aachen, Aachen, Germany
| | - Laura Mitchell
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - David Berry
- Center for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, University of Vienna, Vienna, Austria
| | - Jochen Seitz
- Clinic for Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, LVR-University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Marianne Grognot
- Biophysics of Host-Microbe Interactions Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Thorsten Allers
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Till Strowig
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Michael Pester
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Technical University Braunschweig, Braunschweig, Germany
| | - Birte Abt
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Lorenz C Reimer
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Jörg Overmann
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
- Technical University Braunschweig, Braunschweig, Germany
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
40
|
Zhang B, Lin Y, Song S, Guo H. Exploring the Vital Role of Microbiota Metabolites in Early-Life Health. J Nutr 2025:S0022-3166(25)00270-6. [PMID: 40324527 DOI: 10.1016/j.tjnut.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
Early-life gut microbiota metabolites profoundly influence gut homeostasis, neurodevelopment, metabolic regulation, and immune system maturation. However, there is still a lack of comprehensive summaries and discussions regarding gut microbiota metabolites during early-life stages. This review systematically analyzes microbiota metabolites, including short-chain fatty acids, secondary bile acids, tryptophan metabolites, and branched-chain fatty acids, and delves into their production mechanisms and temporal dynamics. Additionally, the review highlights how maternal factors, breastfeeding, complementary feeding, and environmental influences shape the composition and metabolic functions of the early-life gut microbiota, emphasizing that early life is a crucial window for lifelong health interventions. By integrating the latest research findings and identifying knowledge gaps, this review emphasizes the molecular mechanisms of gut microbiota metabolites and their role in addressing common early-life diseases, including their potential as early biomarkers for screening, prevention, improvement, and even treatment of early diseases, as well as predicting their potential related diseases. On the basis of these insights, this review lays the foundation for future research and practical applications, aiming to promote optimal health from infancy to childhood.
Collapse
Affiliation(s)
- Baoyi Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yingying Lin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Sijia Song
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China; Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| |
Collapse
|
41
|
Chaves LS, Oliveira ACP, Oliveira AP, Lopes ALF, Araujo AKS, Pacheco G, Silva KC, Martins FEC, Gomes IAB, Ramos SVS, Viana HTMC, Batista AVF, Oliveira BC, Nicolau LAD, Ribeiro FOS, Castro AV, de Araujo-Nobre AR, Silva DA, Cordeiro LMC, Góis MB, Medeiros JVR. Cashew gum fractions protect intestinal mucosa against shiga toxin-producing Escherichia coli infection: Characterization and insights into microbiota modulation. Int J Biol Macromol 2025; 311:143916. [PMID: 40324507 DOI: 10.1016/j.ijbiomac.2025.143916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Diarrheal diseases remain a major public health concern, particularly in regions with poor sanitation. Polysaccharides extracted from natural gums have been investigated as functional agents for intestinal health, and their fractionation enables the production of oligosaccharides with potential prebiotic activity. This study aimed to produce cashew gum (CG) fractions through Smith degradation (CGD48) and partial hydrolysis (CGD24) and to evaluate their ability to modulate and protect the intestinal microbiota. Balb/c mice were administered CG (1200 mg/kg), CGD24 (800 mg/kg), or CGD48 (800 mg/kg) for 10 and 26 days, followed by infection with Shiga toxin-producing Escherichia coli (STEC) (5 × 1010 CFU/mL) for three days. Characterization assays confirmed the fragmentation of CG. Both CGD24 and CGD48 promoted the growth of beneficial bacteria with and without infection and reduced STEC colonization. Furthermore, they preserved mucin levels in the cecum and large intestine and maintained baseline levels of superoxide dismutase (SOD), suggesting protection of the intestinal mucosa. These findings indicate that CG fractions exhibit microbiota-modulating and protective effects against STEC, highlighting their therapeutic potential and the need for further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Letícia S Chaves
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Antonio C P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - André L F Lopes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Andreza K S Araujo
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Gabriella Pacheco
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Katriane C Silva
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Francisco E C Martins
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Isaac A B Gomes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Sabrine V S Ramos
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Hémilly T M C Viana
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana V F Batista
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Beatriz C Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucas A D Nicolau
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Fábio O S Ribeiro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Auricélia V Castro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Alyne Rodrigues de Araujo-Nobre
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Durcilene A Silva
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo B Góis
- Post-Graduation Program in Biosciences and Health, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Jand V R Medeiros
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil; Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil.
| |
Collapse
|
42
|
van Deuren T, Umanets A, Venema K, Moreno LL, Zoetendal EG, Canfora EE, Blaak EE. Specific dietary fibers steer toward distal colonic saccharolytic fermentation using the microbiota of individuals with overweight/obesity. Food Res Int 2025; 209:116271. [PMID: 40253188 DOI: 10.1016/j.foodres.2025.116271] [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: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Evidence suggests that increased distal short-chain fatty acid (SCFA) production beneficially impacts metabolic health. However, indigestible carbohydrate availability is limited in the distal colon; consequently, microbes shift toward protein fermentation, often linked to adverse metabolic health effects. We aimed to identify specific fiber(s) that promote saccharolytic fermentation in the distal colon and thereby may (partially) inhibit proteolytic fermentation. METHODS Potato-fiber, pectin, and inulin were studied individually and in combination against a high (predigested) protein background using an in vitro model of the colon (TIM-2) inoculated with pooled, standardized fecal microbiota from individuals with overweight/obesity. Microbiota composition and activity were assessed at different timepoints to simulate the travel throughout the colon (proximal: 0-8 h, distal: 8-24 h) and compared to a high protein (HP)_control, receiving only proteins. RESULTS Fiber addition increased total SCFA production compared to HP_control (52.11 ± 1.49 vs 27.07 ± 0.26 mmol) whereas total branched-chain fatty acids (BCFA; a marker for protein fermentation) production only slightly decreased (3.31 ± 0.10 vs 4.18 ± 0.40 mmol). Combining potato-fiber and pectin led to the highest total and distal SCFA production and distal SCFA:BCFA. Fiber addition attenuated HP-induced increases in several bacterial taxa including Mogibacterium and Coprococcus, independent of fiber type. Additionally, time- and fiber-specific microbial signatures were identified: inulin increased Bifidobacterium (proximal) relative abundance and pectin and/or potato-fiber increased Prevotella 9 (distal) relative abundance. CONCLUSION The most marked increase in distal colonic SCFA production was induced by combining potato-fiber and pectin. Further research should elucidate whether this switch toward saccharolytic fermentation translates into beneficial metabolic health effects in humans.
Collapse
Affiliation(s)
- Thirza van Deuren
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Alexander Umanets
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University-Campus Venlo, Venlo, the Netherlands; Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Luis L Moreno
- Laboratory of Food Chemistry, Wageningen University and Research, Bornse Weilanden 9, 6708, WG, Wageningen, the Netherlands; Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Emanuel E Canfora
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Ellen E Blaak
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'.
| |
Collapse
|
43
|
Huang YY, Qin SM, Nguyen MT, Chen W, Si XM, Huang YQ, Zhang HY. The protective effects of dietary resistant starch against post-antibiotic bone loss in meat ducks associated with the recovery of caecal microbiota dysbiosis. Poult Sci 2025; 104:105238. [PMID: 40424882 DOI: 10.1016/j.psj.2025.105238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Compromised bone quality increases the risk of fractures in domesticate birds, resulting in pain and altered behaviour. Although dietary resistant starch (RS) supplementation show promise for improving inferior bone mass, the diet-mediated gut microbiota alterations as a potential mechanism underlying RS positive roles in bone remains uncertain. With a post-antibiotic model and faecal microbiota transplantation (FMT), this study investigated the effects of a RS diet on antibiotic-induced bone loss and gut microbial composition in meat ducks. Ducklings were assigned to 4 treatments with 6 replicate pens until 21 d, including the control group (Ctrl, feeding a basal diet) and the RS-fed group, and post-antibiotic treatment following the gavage of phosphate-buffered saline (Post-anti-PBS) or faecal microbiota transplantation (Post-anti-FMT). The RS diet increased the proportion of Firmicutes, improved intestinal integrity, and reduced inflammation-induced bone resorption, all of which contributed to an increase in tibial bone volume (P < 0.05). Post-antibiotic treatment was found to reduce tibial quality by stimulating bone resorption and inhibiting bone formation, accompanied by gut microbiota dysbiosis, increased intestinal permeability (P = 0.059), and inflammatory flare compared to control birds. FMT from RS-fed ducks into the antibiotic-treated birds reversed bone loss by primarily blocking osteoclastic frequency and activity. Furthermore, FMT increased the ratio of Firmicutes to Bacteroidetes (P < 0.05) and suppressed the release of pro-osteoclastogenic cytokines such as tumour necrosis factor-α (P = 0.062) and interleukin-1β (P < 0.05) in the bone marrow. These results demonstrated the involvement of gut microbiota in improving bone quality of meat ducks by RS, and FMT of RS-fed birds corrected the imbalance of ceca microbiota and attenuated bone loss in meat ducks with enhanced bone resorption.
Collapse
Affiliation(s)
- Y Y Huang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou 450046, PR China
| | - S M Qin
- College of Animal Science and Technology, Southwest University, Chongqing 402460, PR China
| | - M T Nguyen
- Agriculture and Forestry, Hue University, Hue 49000, Vietnam
| | - W Chen
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou 450046, PR China
| | - X M Si
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Y Q Huang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou 450046, PR China
| | - H Y Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, Henan Agricultural University, Zhengzhou 450046, PR China; Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Ghent University, Ghent 9000, Belgium.
| |
Collapse
|
44
|
Mo F, Qian Q, Lu X, Zheng D, Cai W, Yao J, Chen H, Huang Y, Zhang X, Wu S, Shen Y, Bai Y, Wang Y, Jiang W, Fan L. mKmer: an unbiased K-mer embedding of microbiomic single-microbe RNA sequencing data. Brief Bioinform 2025; 26:bbaf227. [PMID: 40407385 PMCID: PMC12100620 DOI: 10.1093/bib/bbaf227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/13/2025] [Accepted: 05/01/2025] [Indexed: 05/26/2025] Open
Abstract
The advanced single-microbe RNA sequencing (smRNA-seq) technique addresses the pressing need to understand the complexity and diversity of microbial communities, as well as the distinct microbial states defined by different gene expression profiles. Current analyses of smRNA-seq data heavily rely on the integrity of reference genomes within the queried microbiota. However, establishing a comprehensive collection of microbial reference genomes or gene sets remains a significant challenge for most real-world microbial ecosystems. Here, we developed an unbiased embedding algorithm utilizing K-mer signatures, named mKmer, which bypasses gene or genome alignment to enable species identification for individual microbes and downstream functional enrichment analysis. By substituting gene features in the canonical cell-by-gene matrix with highly conserved K-mers, we demonstrate that mKmer outperforms gene-based methods in clustering and motif inference tasks using benchmark datasets from crop soil and human gut microbiomes. Our method provides a reference genome-free analytical framework for advancing smRNA-seq studies.
Collapse
Affiliation(s)
- Fangyu Mo
- Hainan Institute, Zhejiang University, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Qinghong Qian
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Xiaolin Lu
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Dihuai Zheng
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Wenjie Cai
- Liangzhu Laboratory (Zhejiang Provincial Laboratory for Systems Medicine and Precision Diagnosis), Zhejiang University, 1369 Wenyi West Road, Yuhang District, Hangzhou 311121, Zhejiang Province, China
| | - Jie Yao
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Hongyu Chen
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Yujie Huang
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Xiang Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Sanling Wu
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Yifei Shen
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Yinqi Bai
- BGI-Sanya, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
| | - Yongcheng Wang
- Liangzhu Laboratory, Zhejiang University, 1369 Wenyi West Road, Yuhang District, Hangzhou 311113, Zhejiang Province, China
| | - Weiqin Jiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Longjiang Fan
- Hainan Institute, Zhejiang University, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
45
|
Alam N, Ding X, Fu Y, Jia L, Ali S, Liu E. Oryzanol ameliorates MCD-induced metabolic dysfunction-associated steatohepatitis in mice via gut microbiota reprogramming and TLR4/NF-κB signaling suppression. Am J Physiol Gastrointest Liver Physiol 2025; 328:G578-G593. [PMID: 40243180 DOI: 10.1152/ajpgi.00190.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/01/2024] [Accepted: 03/03/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) has emerged as a major global health concern that affects about a quarter of the global population. Recently, host-gut microbiota metabolic interactions have emerged as key mechanistic pathways in MASH development. Oryzanol (ORY), a rice bran bioactive compound, exhibits antioxidant, anti-inflammatory, hypolipidemic, and hypoglycemic properties. Here, we investigated the potential of ORY in alleviating MASH and its association with gut microbiota and MASH progression. Male C57BL/6J mice were fed normal chow diet or methionine-choline-deficient diet and received ORY supplementation at 300 mg/kg/day via gavage for 4 wk. Liver injury, inflammation, lipid accumulation, and TLR4/NF-κB signaling protein levels were assessed. In addition, changes in gut microbiota diversity and abundance across groups were evaluated using 16S rDNA sequencing. Our results demonstrated that ORY significantly reduced lipid accumulation and liver enzymes, ameliorated liver and ileum damage, and restored intestinal barrier function in MASH mice. Furthermore, ORY decreased plasma lipopolysaccharide levels, and inflammatory cytokines and downregulated TLR4, MyD88, and NF-κB protein levels in the liver. ORY enhanced tight junction protein level (ZO-1, occludin) in the gut. Microbial analysis revealed that ORY positively impacted Firmicutes and Bacteroidetes abundance, promoted beneficial bacteria like Lactobacillus and Lachnospiraceae_NK4A136_group, and inhibited harmful bacteria such as Mucispirillum, Bacteroides, and Colidextribacter. Notably, ORY increased Akkermansia abundance, potentially modulating metabolic and inflammatory pathways. ORY exerted restorative and reversible effects on the pathophysiological damage within the gut-liver axis in MASH mice. The therapeutic mechanism may be related to the modulation of the gut microbiota and TLR4/NF-κB signaling pathway.NEW & NOTEWORTHY This study demonstrates that oryzanol (ORY), a bioactive rice bran compound, alleviates metabolic dysfunction-associated steatohepatitis (MASH) in mice by reducing lipid accumulation and inflammation. ORY beneficial effects are associated to the modulation of gut microbiota, enhancing gut barrier integrity, and lowering endotoxemia and TLR4/NF-κB signaling pathway. These findings suggest ORY potential in MASH prevention and treatment, highlighting its influence on gut-liver axis dynamics.
Collapse
Affiliation(s)
- Naqash Alam
- Laboratory of Animal Center, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xinhua Ding
- Laboratory of Animal Center, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yu Fu
- Laboratory of Animal Center, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Linying Jia
- Laboratory of Animal Center, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Sadiq Ali
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Enqi Liu
- Laboratory of Animal Center, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
46
|
Chen YM, Chuang SY, Tsai CY. The Impact of Daily Walnut Consumption on Gastrointestinal Symptoms: A Mixed-Method Study in Healthy Adults. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:332-337. [PMID: 39778130 DOI: 10.1080/27697061.2024.2431287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Common gastrointestinal (GI) symptoms such as abdominal pain, indigestion, and constipation affect a significant portion of the global population and can substantially impair quality of life. Despite these widespread issues, research specifically investigating the effects of walnuts on gut function and GI symptoms remain limited. OBJECTIVE This study investigates the effects of walnuts on gastrointestinal symptoms in healthy adults. DESIGN An experimental baseline-end study with an equivalent group design was employed. SETTING The experimental group consumed 42 grams of walnuts daily, and their gastrointestinal symptoms were compared with those of a control group that did not consume walnuts over a 3-week period. PARTICIPANTS Sixty university students were recruited as volunteer subjects, consisting of 30 males and 30 females. INTERVENTION(S) Participants were randomly assigned to either an experimental group or a control group. MAIN OUTCOME MEASURE(S) The independent variable was walnut consumption, and the dependent variable was gastrointestinal health, assessed using the Gastrointestinal Symptom Rating Scale (GSRS) and a qualitative questionnaire to collect participants' perceived changes in GI symptoms. ANALYSIS A t-test with a p-value of less than 0.05 and verbatim analysis were utilized. RESULTS This mixed-methods study provides evidence for the beneficial effects of walnuts in promoting normal digestive function. CONCLUSIONS AND IMPLICATIONS The study provides alternative evidence for the beneficial effects of walnuts in promoting normal digestive function.
Collapse
Affiliation(s)
- Yi-Mei Chen
- School of Foreign Languages, Jiaying University, Meizhou City, Guangdong, China
| | - Shu-Yu Chuang
- Department of Education, University of Taipei, Taipei, Taiwan
| | - Chih-Yung Tsai
- Department of Education, University of Taipei, Taipei, Taiwan
| |
Collapse
|
47
|
Liao M, Zhu Y, Cui Q, Yue M, Li J, Gao L, He Y, Guo Y, Zhang W, Wei Z, Xia Y, Dai Y. Intestinal Butyrate Ameliorates Rheumatoid Arthritis Through Promoting the Expression of Cortistatin in Ileum via HDAC3-Vitamin D Receptor Pathway. Immunology 2025. [PMID: 40304573 DOI: 10.1111/imm.13939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/17/2025] [Accepted: 04/20/2025] [Indexed: 05/02/2025] Open
Abstract
Butyrate, administered orally or via drinking water, can effectively ameliorate experimental rheumatoid arthritis (RA) in mice despite its limited bioavailability. The discrepancy urges us to explore the involvement and role of intestinal anti-RA factors in the action of butyrate. In this study, we found that substituting drinking water with butyrate (75 mM) could promote the expression of cortistatin (CST) in the ileal epithelium of mice with collagen-induced arthritis (CIA), but butyrate did not alter the expression of other anti-RA neuropeptides in the intestine and the expression of CST in the spleen and brain. The anti-RA efficacy of butyrate was remarkably reduced following adeno-associated virus (AAV)-mediated knockdown of CST. Transcription factor screening revealed that butyrate upregulated CST expression via the vitamin D receptor (VDR). Notably, butyrate-induced VDR and CST expression in intestinal epithelial cells was diminished by α-cyano-4-hydroxycinnamic acid (CHC) rather than siRNA targeting G protein-coupled receptors (GPCRs), suggesting that butyrate functions through an intracellular pathway. Furthermore, butyrate significantly reduced HDAC activity in intestinal epithelial cells and HDAC3 plasmid transfection attenuated the upregulation of butyrate against VDR and CST expression. Chromatin immunoprecipitation assay showed that butyrate selectively enhanced histone acetylation in the P3 and P4 regions of the VDR promoter. In summary, intestinal butyrate exerts an anti-RA effect through selectively promoting the expression of CST in ileal epithelial cells via the HDAC3-VDR pathway.
Collapse
Affiliation(s)
- Minghui Liao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qi Cui
- The Second Affiliated Hospital of Inner Mongolia University for the Nationalities, Inner Mongolia General Forestry Hospital, Inner Mongolia Autonomous Region, China
| | - Mengfan Yue
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jialin Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lei Gao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue He
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjie Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yufeng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
48
|
Kaulpiboon J, Rudeekulthamrong P. Maltotriosyl-erythritol, a transglycosylation product of erythritol by Thermus sp. amylomaltase and its application to prebiotic. Food Chem 2025; 472:142937. [PMID: 39827568 DOI: 10.1016/j.foodchem.2025.142937] [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: 10/30/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
In this study, maltotriosyl-erythritol (EG3) was synthesized as a novel prebiotic candidate via transglycosylation using recombinant amylomaltase (AMase) from Thermus sp. Tapioca starch served as the glucosyl donor, and erythritol as the acceptor. High-performance liquid chromatography (HPLC) revealed an EG3 yield of 14.0 % with a concentration of 2.8 mg/mL. Mass spectrometry confirmed the molecular weight of EG3 as 608 Da, and its strucopture was verified by 1H and 13C NMR analysis. EG3 exhibited greater resistance to acid, heat, and digestive enzymes compared to erythritol glucosides (EG1-2) and significantly promoted the growth of Lactobacillus casei BCC36987. Fermentation of EG3 resulted in the highest levels of lactic acid and total short-chain fatty acids, which may contribute to reduced pH levels. These findings suggest that erythritol-receptor products formed via AMase-catalyzed reactions, particularly EG3, are promising prebiotic ingredients, with the prebiotic activity of erythritol derivatives being influenced by the length of the carbohydrate chain.
Collapse
Affiliation(s)
- Jarunee Kaulpiboon
- Division of Biochemistry, Department of Pre-Clinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Prakarn Rudeekulthamrong
- Department of Biochemistry, Phramongkutklao College of Medicine, Phramongkutklao Hospital, Bangkok 10400, Thailand.
| |
Collapse
|
49
|
Yi K, Huang Y, Jiang Y, Zhou L. Causal relationship between gut microbiota and laryngeal cancer: a mendelian randomization analysis. Braz J Otorhinolaryngol 2025; 91:101634. [PMID: 40305979 PMCID: PMC12118545 DOI: 10.1016/j.bjorl.2025.101634] [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: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE Laryngeal cancer incidence is rising globally; the role of gut microbiota remains underexplored. This study aimed to establish a causal link between gut microbiota and laryngeal cancer to inform preventive and therapeutic strategies. METHODS Gut microbiota data from GWAS conducted by the MiBioGen consortium served as the exposure variable, with laryngeal cancer as the outcome variable. the exposure variable and the outcome variable were analyzed using Mendelian Randomization. The primary method was Inverse Variance Weighted analysis, with heterogeneity and pleiotropy assessed through Cochran's Q test, MR-Egger regression, and MR-PRESSO. RESULTS In the study, we identified five bacterial taxa with potential causal relationships with laryngeal cancer risk: Higher levels of Clostridiaceae1 (OR = 0.9993, 95% CI 0.9986-0.9999, p = 0.0463) and Turicibacter (OR = 0.9995, 95% CI 0.9989-0.9999, p = 0.0384) were linked to reduced cancer risk, while Mollicutes RF9 (OR = 1.0010, 95% CI 1.0003-1.0016, p = 0.0027), Euryarchaeota (OR = 1.0004, 95% CI 1.0001-1.0007, p = 0.0234), and Cyanobacteria (OR = 1.0005, 95% CI 1.0000-1.0009, p = 0.0464) were associated with increased risk. CONCLUSION Our findings suggest a causal relationship between gut microbiota composition and laryngeal cancer risk. Clostridiaceae1 and Turicibacter may play a protective role, while Mollicutes RF9, Euryarchaeota, and Cyanobacteria could contribute to increased cancer susceptibility. These insights highlight potential microbiome-based strategies for early detection, prevention, and therapeutic intervention in laryngeal cancer. LEVEL OF EVIDENCE Level 5.
Collapse
Affiliation(s)
- Kaiyan Yi
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yun Jiang
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Lingling Zhou
- Department of Otolaryngology-Head and Neck Surgery, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
Ren Y, He X, Wang L, Chen N. Comparison of the gut microbiota in older people with and without sarcopenia: a systematic review and meta-analysis. Front Cell Infect Microbiol 2025; 15:1480293. [PMID: 40357398 PMCID: PMC12066693 DOI: 10.3389/fcimb.2025.1480293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Sarcopenia, an age-related disorder marked by decreased skeletal muscle mass, strength, and function, is associated with negative health impacts in individuals and financial burdens on families and society. Studies have suggested that age-related alterations in gut microbiota may contribute to the development of sarcopenia in older people through the gut-muscle axis, thus modulation of gut microbiota may be a promising approach for sarcopenia treatment. However, the characteristic gut microbiota for sarcopenia has not been consistent across studies. Therefore, the aim of this study was to compare the diversity and compositional differences in the gut microbiota of older people with and without sarcopenia, and to identify gut microbiota biomarkers with therapeutic potential for sarcopenia. Methods The PubMed, Embase, Web of Science, Cochrane Library, China National Knowledge Infrastructure, and Wanfang Database were searched studies about the gut microbiota characteristics in older people with sarcopenia. The quality of included articles was assessed by the Newcastle-Ottawa Scale (NOS). Weighted standardized mean differences (SMDs) and 95% confidence intervals (CIs) for α-diversity index were estimated using a random effects model. Qualitative synthesis was conducted for β-diversity and the correlation between gut microbiota and muscle parameters. The relative abundance of the gut microbiota was analyzed quantitatively and qualitatively, respectively. Results Pooled estimates showed that α-diversity was significantly lower in older people with sarcopenia (SMD: -0.41, 95% CI: -0.57 to -0.26, I²: 71%, P < 0.00001). The findings of β-diversity varied across included studies. In addition, our study identified gut microbiota showing a potential and negative correlation with sarcopenia, such as Prevotella, Slackia, Agathobacter, Alloprevotella, Prevotella copri, Prevotellaceae sp., Bacteroides coprophilus, Mitsuokella multacida, Bacteroides massiliensis, Bacteroides coprocola Conversely, a potential and positive correlation was observed with opportunistic pathogens like Escherichia-Shigella, Eggerthella, Eggerthella lenta and Collinsella aerofaciens. Discussion This study showed that α-diversity is decreased in sarcopenia, probably predominantly due to diminished richness rather than evenness. In addition, although findings of β-diversity varied across included studies, the overall trend toward a decrease in SCFAs-producing bacteria and an increase in conditionally pathogenic bacteria. This study provides new ideas for targeting the gut microbiota for the prevention and treatment of sarcopenia. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42024573090, identifier CRD42024573090.
Collapse
Affiliation(s)
- Yanqing Ren
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Xiangfeng He
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Ling Wang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Nan Chen
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
- Department of Rehabilitation, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|