1
|
Dongre DS, Saha UB, Saroj SD. Exploring the role of gut microbiota in antibiotic resistance and prevention. Ann Med 2025; 57:2478317. [PMID: 40096354 PMCID: PMC11915737 DOI: 10.1080/07853890.2025.2478317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND/INTRODUCTION Antimicrobial resistance (AMR) and the evolution of multiple drug-resistant (MDR) bacteria is of grave public health concern. To combat the pandemic of AMR, it is necessary to focus on novel alternatives for drug development. Within the host, the interaction of the pathogen with the microbiome plays a pivotal role in determining the outcome of pathogenesis. Therefore, microbiome-pathogen interaction is one of the potential targets to be explored for novel antimicrobials. MAIN BODY This review focuses on how the gut microbiome has evolved as a significant component of the resistome as a source of antibiotic resistance genes (ARGs). Antibiotics alter the composition of the native microbiota of the host by favouring resistant bacteria that can manifest as opportunistic infections. Furthermore, gut dysbiosis has also been linked to low-dosage antibiotic ingestion or subtherapeutic antibiotic treatment (STAT) from food and the environment. DISCUSSION Colonization by MDR bacteria is potentially acquired and maintained in the gut microbiota. Therefore, it is pivotal to understand microbial diversity and its role in adapting pathogens to AMR. Implementing several strategies to prevent or treat dysbiosis is necessary, including faecal microbiota transplantation, probiotics and prebiotics, phage therapy, drug delivery models, and antimicrobial stewardship regulation.
Collapse
Affiliation(s)
- Devyani S. Dongre
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Ujjayni B. Saha
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Sunil D. Saroj
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| |
Collapse
|
2
|
Brito Rodrigues P, de Rezende Rodovalho V, Sencio V, Benech N, Creskey M, Silva Angulo F, Delval L, Robil C, Gosset P, Machelart A, Haas J, Descat A, Goosens JF, Beury D, Maurier F, Hot D, Wolowczuk I, Sokol H, Zhang X, Ramirez Vinolo MA, Trottein F. Integrative metagenomics and metabolomics reveal age-associated gut microbiota and metabolite alterations in a hamster model of COVID-19. Gut Microbes 2025; 17:2486511. [PMID: 40172215 PMCID: PMC11970752 DOI: 10.1080/19490976.2025.2486511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Aging is a key contributor of morbidity and mortality during acute viral pneumonia. The potential role of age-associated dysbiosis on disease outcomes is still elusive. In the current study, we used high-resolution shotgun metagenomics and targeted metabolomics to characterize SARS-CoV-2-associated changes in the gut microbiota from young (2-month-old) and aged (22-month-old) hamsters, a valuable model of COVID-19. We show that age-related dysfunctions in the gut microbiota are linked to disease severity and long-term sequelae in older hamsters. Our data also reveal age-specific changes in the composition and metabolic activity of the gut microbiota during both the acute phase (day 7 post-infection, D7) and the recovery phase (D22) of infection. Aged hamsters exhibited the most notable shifts in gut microbiota composition and plasma metabolic profiles. Through an integrative analysis of metagenomics, metabolomics, and clinical data, we identified significant associations between bacterial taxa, metabolites and disease markers in the aged group. On D7 (high viral load and lung epithelial damage) and D22 (body weight loss and fibrosis), numerous amino acids, amino acid-related molecules, and indole derivatives were found to correlate with disease markers. In particular, a persistent decrease in phenylalanine, tryptophan, glutamic acid, and indoleacetic acid in aged animals positively correlated with poor recovery of body weight and/or lung fibrosis by D22. In younger hamsters, several bacterial taxa (Eubacterium, Oscillospiraceae, Lawsonibacter) and plasma metabolites (carnosine and cis-aconitic acid) were associated with mild disease outcomes. These findings support the need for age-specific microbiome-targeting strategies to more effectively manage acute viral pneumonia and long-term disease outcomes.
Collapse
Affiliation(s)
- Patrícia Brito Rodrigues
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | | | - Valentin Sencio
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Nicolas Benech
- Gastroenterology Department, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- Hospices Civils de Lyon, Lyon GEM Microbiota Study Group, Lyon, France
| | - Marybeth Creskey
- Regulatory Research Division, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, University of Ottawa, Ottawa, Canada
| | - Fabiola Silva Angulo
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Lou Delval
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Cyril Robil
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Philippe Gosset
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Arnaud Machelart
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Joel Haas
- U1011-EGID, University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Amandine Descat
- EA 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, University of Lille, CHU Lille, Lille, France
| | - Jean François Goosens
- EA 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, University of Lille, CHU Lille, Lille, France
| | - Delphine Beury
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Florence Maurier
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - David Hot
- US 41 - UAR 2014 - PLBS, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Isabelle Wolowczuk
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Harry Sokol
- Gastroenterology Department, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Paris, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Xu Zhang
- Regulatory Research Division, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, University of Ottawa, Ottawa, Canada
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | - François Trottein
- U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
3
|
Duan M, Che L, Wu X, Quek SY, Zhang B, Lin H, He N. Incorporation of probiotics with pressure-sensitive pectin-fructooligosaccharide hydrogel for potential intestinal delivery. Carbohydr Polym 2025; 359:123566. [PMID: 40306774 DOI: 10.1016/j.carbpol.2025.123566] [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: 11/13/2024] [Revised: 03/09/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025]
Abstract
Probiotics and prebiotics serve as vital tools in managing gut microecology and enhancing immune responses. However, the effectiveness of non-encapsulated probiotics often diminishes during processing, storage, and transport to the gastrointestinal tract, especially at elevated temperatures. To address this challenge, a novel loading strategy for Lactobacillus reuteri DPC16 (L. reuteri) is proposed in this work, using pressure-sensitive high-methoxy pectin (HMP)/fructooligosaccharides (FOS) hydrogel. The HMP/FOS hydrogel melted at 600 MPa to form a sol. The resulting sol was mixed with L. reuteri immediately at ambient conditions, which underwent a sol-to-gel transition subsequently to form a composite hydrogel with a continuous porous structure. The resulting HMP/FOS@L. reuteri hydrogel achieved a loading concentration of viable bacteria at 109 CFU/mL. In vitro assessments reveal that the hydrogel demonstrates good biocompatibility and targeted release of probiotics within the intestine. Furthermore, the hydrogel substantially boosted the short-chain fatty acids levels and increased the amounts of acetic and isovaleric acids, respectively. This work underscores the unique advantages of employing a pressure-sensitive HMP/FOS hydrogel for loading and targeted delivery of probiotics and prebiotics to improve intestinal health.
Collapse
Affiliation(s)
- Mengwen Duan
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Liming Che
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China.
| | - Xuee Wu
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Siew Young Quek
- Food Science, School of Chemical Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Bangzhou Zhang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350000, PR China
| | - Hao Lin
- Xiamen Treatgut Biotechnology Co. Ltd., Xiamen 361005, PR China
| | - Ning He
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
4
|
Jalševac F, Segú H, Balaguer F, Ocaña T, Moreira R, Abad-Jordà L, Gràcia-Sancho J, Fernández-Iglesias A, Andres-Lacueva C, Martínez-Huélamo M, Beltran-Debon R, Rodríguez-Gallego E, Terra X, Ardévol A, Pinent M. TAS2R5 and TAS2R38 are bitter taste receptors whose colonic expressions could play important roles in age-associated processes. J Nutr Biochem 2025; 140:109872. [PMID: 39986633 DOI: 10.1016/j.jnutbio.2025.109872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/09/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Ageing disrupts how our bodies process nutrients, leading to deregulation of nutrient-sensing and increased inflammation. Dietary interventions can promote healthy ageing, which demonstrates the importance of both metabolism and the gastrointestinal tract for our health. Bitter taste receptors (TAS2R) present in the intestine are key members of metabolic regulation. TAS2R are involved in controlling enterohormonal secretion, detect phenolic compounds in our diet, and potentially have a great impact on the ageing process. Here, we aimed to analyze the potential role of intestinal TAS2R on the ageing process and establish potential impact of these receptors on the biomarkers. Healthy subjects were divided into two age cohorts: young (38.9±6) and aged (63.6±6). TAS2R expression was analyzed in the colon. Analyses of metabolomics and of phenolic markers were performed in plasma. Best discriminatory parameters were obtained using three machine-learning methods. Finally, Spearman's rank correlation was performed. The best separators of the age cohorts were docosahexaenoic acid and multiple lipoprotein fractions. Two TAS2R were also identified: TAS2R5 and TAS2R38. TAS2R5 correlated with multiple lipoprotein-derived fractions, inflammatory marker IL-6 and polyunsaturated fatty acids. TAS2R38 was much more selective, correlating with a few parameters, including membrane lipid sphingomyelin, ketone body acetone, and omega acids. Both TAS2R5 and TAS2R38 correlated with β-hydroxybutyrate. The parameters that correlated with TAS2R have known effects on the ageing process. This suggests that TAS2R5 and TAS2R38 are the bitter receptors most likely to play a role in the development and progress of ageing.
Collapse
Affiliation(s)
- Florijan Jalševac
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Helena Segú
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain
| | - Francesc Balaguer
- Gastroenterology department, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Teresa Ocaña
- Gastroenterology department, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Rebeca Moreira
- Gastroenterology department, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Laia Abad-Jordà
- Liver Vascular Biology, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Jordi Gràcia-Sancho
- Liver Vascular Biology, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Anabel Fernández-Iglesias
- Liver Vascular Biology, Hospital Clinic Barcelona, IDIBAPS (Institut d´Investigacions Biomédiques August Pi i Sunyer), Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Nutrition and Food Safety Research Institute (INSA), Food Innovation Network (XIA), Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Martínez-Huélamo
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Nutrition and Food Safety Research Institute (INSA), Food Innovation Network (XIA), Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Raul Beltran-Debon
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain; IISPV, Hospital Joan XXIII, Tarragona, Spain
| | - Esther Rodríguez-Gallego
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain; IISPV, Hospital Joan XXIII, Tarragona, Spain
| | - Ximena Terra
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain; IISPV, Hospital Joan XXIII, Tarragona, Spain
| | - Anna Ardévol
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain; IISPV, Hospital Joan XXIII, Tarragona, Spain.
| | - Montserrat Pinent
- Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, MoBioFood Research Group, Tarragona, Spain; IISPV, Hospital Joan XXIII, Tarragona, Spain
| |
Collapse
|
5
|
Sangfuang N, Xie Y, McCoubrey LE, Taub M, Favaron A, Mai Y, Gaisford S, Basit AW. Investigating the bidirectional interactions between senotherapeutic agents and human gut microbiota. Eur J Pharm Sci 2025; 209:107098. [PMID: 40216167 DOI: 10.1016/j.ejps.2025.107098] [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: 11/26/2024] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Biological ageing is a time-dependent process that has implications for health and disease. Cellular senescence is a key driver in ageing and age-related diseases. Senotherapeutic agents have been shown to slow biological ageing by eliminating senescent mammalian cells. Given the increasing awareness of the gut microbiome in regulating human health, this study aimed to investigate the effects of senotherapeutic agents as pharmacological interventions on the human gut microbiota. In this study, the bidirectional effects of four senotherapeutic agents, quercetin, fisetin, dasatinib, and sirolimus, with the gut microbiota sourced from healthy human donors were investigated. The results revealed that quercetin was completely biotransformed by the gut microbiota within six hours, while dasatinib was the most stable of the four compounds. Additionally, metagenomic analysis confirmed that all four compounds increased the abundance of bacterial species associated with healthy ageing (e.g., Bacteroides fragilis, Bifidobacterium longum, and Veillonella parvula), and decreased the abundance of pathogenic bacteria primarily associated with age-related diseases (e.g., Enterococcus faecalis and Streptococcus spp.). The findings from this study provide a comprehensive understanding of the pharmacobiomics of senotherapeutic interventions, highlighting the potential of microbiome-targeted senolytics in promoting healthy ageing.
Collapse
Affiliation(s)
| | - Yuan Xie
- School of Pharmaceutical Sciences (Shenzen), Sun Yat-Sen University, Shenzen 518107, China
| | - Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK; Drug Product Development, GSK R&D, Ware SG12 0GX, UK
| | - Marissa Taub
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Alessia Favaron
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Yang Mai
- School of Pharmaceutical Sciences (Shenzen), Sun Yat-Sen University, Shenzen 518107, China.
| | - Simon Gaisford
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
6
|
Elkrief A, Routy B, Derosa L, Bolte L, Wargo JA, McQuade JL, Zitvogel L. Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship. Am Soc Clin Oncol Educ Book 2025; 45:e472902. [PMID: 40262063 DOI: 10.1200/edbk-25-472902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.
Collapse
Affiliation(s)
- Arielle Elkrief
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Laura Bolte
- Department of Medical Oncology, University Groningen and University Medical Center, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Groningen and University Medical Center, Groningen, the Netherlands
| | | | | | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
7
|
Schoultz I, Claesson MJ, Dominguez‐Bello MG, Fåk Hållenius F, Konturek P, Korpela K, Laursen MF, Penders J, Roager H, Vatanen T, Öhman L, Jenmalm MC. Gut microbiota development across the lifespan: Disease links and health-promoting interventions. J Intern Med 2025; 297:560-583. [PMID: 40270478 PMCID: PMC12087861 DOI: 10.1111/joim.20089] [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] [Indexed: 04/25/2025]
Abstract
The gut microbiota plays a pivotal role in human life and undergoes dynamic changes throughout the human lifespan, from infancy to old age. During our life, the gut microbiota influences health and disease across life stages. This review summarizes the discussions and presentations from the symposium "Gut microbiota development from infancy to old age" held in collaboration with the Journal of Internal Medicine. In early infancy, microbial colonization is shaped by factors such as mode of delivery, antibiotic exposure, and milk-feeding practices, laying the foundation for subsequent increased microbial diversity and maturation. Throughout childhood and adolescence, microbial maturation continues, influencing immune development and metabolic health. In adulthood, the gut microbiota reaches a relatively stable state, influenced by genetics, diet, and lifestyle. Notably, disruptions in gut microbiota composition have been implicated in various inflammatory diseases-including inflammatory bowel disease, Type 1 diabetes, and allergies. Furthermore, emerging evidence suggests a connection between gut dysbiosis and neurodegenerative disorders such as Alzheimer's disease. Understanding the role of the gut microbiota in disease pathogenesis across life stages provides insights into potential therapeutic interventions. Probiotics, prebiotics, and dietary modifications, as well as fecal microbiota transplantation, are being explored as promising strategies to promote a healthy gut microbiota and mitigate disease risks. This review focuses on the gut microbiota's role in infancy, adulthood, and aging, addressing its development, stability, and alterations linked to health and disease across these critical life stages. It outlines future research directions aimed at optimizing the gut microbiota composition to improve health.
Collapse
Affiliation(s)
- Ida Schoultz
- School of Medical SciencesFaculty of Medicine and Health Örebro UniversityOrebroSweden
| | | | - Maria Gloria Dominguez‐Bello
- Department of Biochemistry & Microbiology and of AnthropologyRutgers University–New BrunswickNew BrunswickNew JerseyUSA
| | - Frida Fåk Hållenius
- Department of Food Technology, Engineering and NutritionLund UniversityLundSweden
| | - Peter Konturek
- Department of Medicine, Thuringia Clinic SaalfeldTeaching Hospital of the University JenaJenaGermany
| | - Katri Korpela
- Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtthe Netherlands
| | - H. Roager
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksbergDenmark
| | - Tommi Vatanen
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFinland
- Department of Microbiology, Faculty of Agriculture and ForestryUniversity of HelsinkiHelsinkiFinland
- Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Liggins InstituteUniversity of AucklandAucklandNew Zealand
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria C. Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| |
Collapse
|
8
|
Li J, Wu Y, Yang Y, Chen L, He C, Zhou S, Huang S, Zhang X, Wang Y, Gui Q, Lu H, Zhang Q, Yang Y. Metagenomics reveals an increased proportion of an Escherichia coli-dominated enterotype in elderly Chinese people. J Zhejiang Univ Sci B 2025; 26:477-492. [PMID: 40436643 PMCID: PMC12119184 DOI: 10.1631/jzus.b2400341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 06/01/2025]
Abstract
Gut microbial communities are likely remodeled in tandem with accumulated physiological decline during aging, yet there is limited understanding of gut microbiome variation in advanced age. Here, we performed a metagenomics-based enterotype analysis in a geographically homogeneous cohort of 367 enrolled Chinese individuals between the ages of 60 and 94 years, with the goal of characterizing the gut microbiome of elderly individuals and identifying factors linked to enterotype variations. In addition to two adult-like enterotypes dominated by Bacteroides (ET-Bacteroides) and Prevotella (ET-Prevotella), we identified a novel enterotype dominated by Escherichia (ET-Escherichia), whose prevalence increased in advanced age. Our data demonstrated that age explained more of the variance in the gut microbiome than previously identified factors such as type 2 diabetes mellitus (T2DM) or diet. We characterized the distinct taxonomic and functional profiles of ET-Escherichia, and found the strongest cohesion and highest robustness of the microbial co-occurrence network in this enterotype, as well as the lowest species diversity. In addition, we carried out a series of correlation analyses and co-abundance network analyses, which showed that several factors were likely linked to the overabundance of Escherichia members, including advanced age, vegetable intake, and fruit intake. Overall, our data revealed an enterotype variation characterized by Escherichia enrichment in the elderly population. Considering the different age distribution of each enterotype, these findings provide new insights into the changes that occur in the gut microbiome with age and highlight the importance of microbiome-based stratification of elderly individuals.
Collapse
Affiliation(s)
- Jinyou Li
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yue Wu
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yichen Yang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lufang Chen
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Caihong He
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shixian Zhou
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shunmei Huang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xia Zhang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuming Wang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qifeng Gui
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haifeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qin Zhang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China. ,
| | - Yunmei Yang
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
9
|
Han M, Han P, Wang Z, Kong L, Xu Q, Liu Q, Sun Y. Alternative splicing in aging and aging-related diseases: From pathogenesis to therapy. Pharmacol Ther 2025; 272:108887. [PMID: 40414568 DOI: 10.1016/j.pharmthera.2025.108887] [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: 10/13/2024] [Revised: 04/10/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Aging is a complex biological process associated with nearly all diseases. Alternative splicing is increasingly recognized as an important contributor to aging and a key research pathway for extending human lifespan. In this review, we highlight the findings of alternative splicing in the hallmarks of aging including key processes such as genomic instability, telomere length, protein stability, autophagy processes, etc., as well as antagonistic hallmarks of aging such as various metabolic signals, energy metabolism, clearance of senescent cells, stem cell self-renewal, cell communication and inflammatory process, etc. We also discuss the roles of alternative splicing in age-related diseases, including neurodegenerative diseases, cardiovascular diseases, skeletal muscle-related diseases, metabolic disorders, cancer, sensory degeneration, and chronic inflammation, etc. These studies suggest that new anti-aging therapies could be developed by regulating key splicing proteins or specific splicing events.
Collapse
Affiliation(s)
- Mingrui Han
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Peiru Han
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zihui Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Qianqian Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, China.
| |
Collapse
|
10
|
Sen I, Trzaskalski NA, Hsiao YT, Liu PP, Shimizu I, Derumeaux GA. Aging at the Crossroads of Organ Interactions: Implications for the Heart. Circ Res 2025; 136:1286-1305. [PMID: 40403108 DOI: 10.1161/circresaha.125.325637] [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: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/24/2025]
Abstract
Aging processes underlie common chronic cardiometabolic diseases such as heart failure and diabetes. Cross-organ/tissue interactions can accelerate aging through cellular senescence, tissue wasting, accelerated atherosclerosis, increased vascular stiffness, and reduction in blood flow, leading to organ remodeling and premature failure. This interorgan/tissue crosstalk can accelerate aging-related dysfunction through inflammation, senescence-associated secretome, and metabolic and mitochondrial changes resulting in increased oxidative stress, microvascular dysfunction, cellular reprogramming, and tissue fibrosis. This may also underscore the rising incidence and co-occurrence of multiorgan dysfunction in cardiometabolic aging in the population. Examples include interactions between the heart and the lungs, kidneys, liver, muscles, and brain, among others. However, this phenomenon can also present new translational opportunities for identifying diagnostic biomarkers to define early risks of multiorgan dysfunction, gain mechanistic insights, and help to design precision-directed therapeutic interventions. Indeed, this opens new opportunities for therapeutic development in targeting multiple organs simultaneously to disrupt the crosstalk-driven process of mutual disease acceleration. New therapeutic targets could provide synergistic benefits across multiple organ systems in the same at-risk patient. Ultimately, these approaches may together slow the aging process itself throughout the body. In the future, with patient-centered multisystem coordinated approaches, we can initiate a new paradigm of multiorgan early risk prediction and tailored intervention. With emerging tools including artificial intelligence-assisted risk profiling and novel preventive strategies (eg, RNA-based therapeutics), we may be able to mitigate multiorgan cardiometabolic dysfunction much earlier and, perhaps, even slow the aging process itself.
Collapse
Affiliation(s)
- Ilke Sen
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| | - Natasha A Trzaskalski
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Yung-Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
| | - Peter P Liu
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (I. Shimizu)
| | - Geneviève A Derumeaux
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| |
Collapse
|
11
|
Snelson M, Muralitharan RR, Liu CF, Markó L, Forslund SK, Marques FZ, Tang WHW. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ Res 2025; 136:1382-1406. [PMID: 40403109 PMCID: PMC12101525 DOI: 10.1161/circresaha.125.325516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 05/24/2025]
Abstract
Heart failure is a global health issue with significant mortality and morbidity. There is increasing evidence that alterations in the gastrointestinal microbiome, gut epithelial permeability, and gastrointestinal disorders contribute to heart failure progression through various pathways, including systemic inflammation, metabolic dysregulation, and modulation of cardiac function. Moreover, several medications used to treat heart failure directly impact the microbiome. The relationship between the gastrointestinal tract and the heart is bidirectional, termed the gut-heart axis. It is increasingly understood that diet-derived microbial metabolites are key mechanistic drivers of the gut-heart axis. This includes, for example, trimethylamine N-oxide and short-chain fatty acids. This review discusses current insights into the interplay between heart failure, its associated risk factors, and the gut microbiome, focusing on key metabolic pathways, the role of dietary interventions, and the potential for gut-targeted therapies. Understanding these complex interactions could pave the way for novel strategies to mitigate heart failure progression and improve patient outcomes.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Rikeish R. Muralitharan
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| | - Lajos Markó
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
| | - Sofia K. Forslund
- Charité – Universitätsmedizin Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center ( ECRC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Francine Z. Marques
- Hypertension Research Laboratory, Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - W. H. Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
12
|
Le Cosquer G, Pannier M, Meunier E, Thevenin J, Pyhourquet E, Guyonnet S, Vellas B, Santin Y, Guiard B, Parini A, Buscail L, Bournet B, Guillemet D, Deraison C, Vergnolle N, Motta JP, IHU HealthAge INSPIRE/Open Science study group. Pathogenicity of commensal gut biofilm in prefrail aging. NPJ Biofilms Microbiomes 2025; 11:84. [PMID: 40404666 PMCID: PMC12098755 DOI: 10.1038/s41522-025-00716-8] [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: 08/01/2024] [Accepted: 05/01/2025] [Indexed: 05/24/2025] Open
Abstract
Pathophysiological mechanisms of unhealthy aging, particularly the transition from robustness to frailty, remain poorly understood. Despite extensive microbiome research on taxonomy, the behavior of early prefrail gut bacteria in their natural community-host mucosal tissue context remains unexplored. Using fecal samples from the INSPIRE-T aging human cohort, we characterized gut microbiota phenotype during prefrailty stages using a polymicrobial biofilm model. Results revealed that prefrail-derived biofilms exhibited distinct taxonomic and physical alterations, enhanced dispersal, and increased epithelial virulence compared to robust counterparts. Multiparametric analyses linked biofilm characteristics to clinical traits, suggesting their potential as aging status indicators. Polyphenol-rich grape pomace extract partially reversed prefrail biofilm alterations and reduced proinflammatory prefrail biofilm responses in vitro. Microbiota from prefrail-aged mice induced colon damage in antibiotic-treated recipients, establishing a prefrail microbiome-inflammation causality. Overall, the findings identified novel prefrail microbiome characteristics, established causal inflammatory links, and supported microbiota-targeted geroprotective interventions for the prefrail populations.
Collapse
Affiliation(s)
- Guillaume Le Cosquer
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
- Department of Gastroenterology and Pancreatology, Toulouse University Hospital (CHU Toulouse), Toulouse, France and Toulouse University, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Melissa Pannier
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Elodie Meunier
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Julie Thevenin
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Elise Pyhourquet
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Sophie Guyonnet
- Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France and CERPOP, Toulouse University, INSERM, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Bruno Vellas
- Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France and CERPOP, Toulouse University, INSERM, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Yohan Santin
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, Toulouse University, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Bruno Guiard
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), Université de Toulouse, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM, Toulouse University, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Louis Buscail
- Department of Gastroenterology and Pancreatology, Toulouse University Hospital (CHU Toulouse), Toulouse, France and Toulouse University, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Barbara Bournet
- Department of Gastroenterology and Pancreatology, Toulouse University Hospital (CHU Toulouse), Toulouse, France and Toulouse University, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | | | - Celine Deraison
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
| | - Nathalie Vergnolle
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, 3330 Hospital Drive, NW Calgary, AB, Canada
| | - Jean-Paul Motta
- Institute of Digestive Health Research (IRSD), INSERM, Toulouse University, INRAe, ENVT, University Toulouse III Paul Sabatier (UPS), Toulouse, France.
| | | |
Collapse
|
13
|
Liu X, Guan K, Ma Y, Jiang L, Li Q, Liu Y, Mao K, Wang R. Probiotic Combination of Limosilactobacillus fermentum HF07 and Lactococcus lactis HF08 Targeting Gut Microbiota-Secondary Bile Acid Metabolism Ameliorates Inflammation and Intestinal Barrier Dysfunction in Aging Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40391947 DOI: 10.1021/acs.jafc.4c12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
This study investigated the intestinal protective effects of a probiotic mixture (PM) composed of Limosilactobacillus fermentum HF07 and Lactococcus lactis HF08 on d-gal/DSS-induced aging colitis in mice. The PM alleviated age-related colitis symptoms including weight loss, increased disease activity index scores, colonic shortening, and tissue damage. PM supplementation reshaped the gut microbiota by restoring the relative abundances of Lactobacillus, Dubosiella, Odoribacter, and Clostridia_UCG-014, thereby enhancing levels of bile acids (BAs) such as alpha-muricholic acid, isolithocholic acid, and ursodeoxycholic acid. Moreover, transcriptomic analysis revealed that PM administration activated the cAMP pathway through the gut microbiota-secondary BAs axis. Western blot analysis further demonstrated that the effects of anti-inflammatory and intestinal barrier repair induced by PM were associated with downregulation of key proteins in the NLRP3 and RhoA/ROCK pathways, both of which are downstream of the cAMP pathway. Additionally, the role of gut metabolites in mediating these effects via G protein-coupled receptor 5 (TGR5) activation was confirmed through in vitro experiments using Caco-2 cells. These findings provided a comprehensive understanding of how probiotics target intestinal metabolites and leverage the gut microbiota-BAs axis to mitigate age-related gastrointestinal diseases.
Collapse
Affiliation(s)
- Xiaolin Liu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Kaifang Guan
- School of Medicine and Health, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lin Jiang
- Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Qiming Li
- Dairy Nutrition and Function, Key Laboratory of Sichuan Province, New Hope Dairy Company Limited, Chengdu 610023, China
- Sichuan Engineering Laboratory for High-quality Dairy Product Preparation and Quality Control Technology, Chengdu, Sichuan 610000, China
| | - Yuxuan Liu
- Dairy Nutrition and Function, Key Laboratory of Sichuan Province, New Hope Dairy Company Limited, Chengdu 610023, China
| | - Kaidong Mao
- Jiangsu HOWYOU Biotechnology Company Limited, Shanghai 310000, China
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| |
Collapse
|
14
|
Chen Z, Zhang Z, Nie BN, Huang W, Zhu Y, Zhang L, Xu M, Wang M, Yuan C, Liu N, Wang X, Tian J, Ba Q, Wang Z. Temporal network analysis of gut microbiota unveils aging trajectories associated with colon cancer. mSystems 2025; 10:e0118824. [PMID: 40298386 PMCID: PMC12090783 DOI: 10.1128/msystems.01188-24] [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] [Received: 08/31/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The human gut microbiome's role in colorectal cancer (CRC) pathogenesis has gained increasing recognition. This study aimed to delineate the microbiome characteristics that distinguish CRC patients from healthy individuals, while also evaluating the influence of aging, through a comprehensive metagenomic approach. The study analyzed a cohort of 80 CRC patients and 80 matched healthy controls, dividing participants into a normal and a CRC group, further categorized by age into young, middle-aged, and old-aged subgroups. Extensive metagenomic sequencing of fecal samples allowed for the exploration of both the structural and functional profiles of the microbiome, with findings validated in an independent cohort to ensure robustness. Our results highlight notable differences in microbiome composition between CRC patients and healthy individuals, which exhibit age-dependent variations. Specifically, a higher prevalence of pathogenic bacteria, such as Bacteroides vulgatus, known to drive inflammation and carcinogenesis, was observed in CRC patients, alongside a reduction in beneficial microbes, including Lactobacillus. Functionally, the CRC-associated microbiome showed an increase in pathways related to DNA repair, cell cycle regulation, and metabolic activities, such as the Citrate cycle and Galactose metabolism, underscoring distinct microbial alterations in CRC patients that could influence disease onset and progression. These insights lay a foundation for future research into microbiome-based diagnostics and treatments for CRC. IMPORTANCE This study underscores the critical role of the gut microbiome in colorectal cancer (CRC) pathogenesis, particularly in the context of aging. By identifying age-specific microbial biomarkers and functional pathways associated with CRC, our findings provide novel insights into how microbiome composition and metabolic activities influence disease progression. These discoveries pave the way for developing personalized microbiome-based diagnostic tools and therapeutic strategies, potentially improving CRC prevention and treatment outcomes across different age groups. Understanding these microbial dynamics could also inform interventions targeting gut microbiota to mitigate CRC risk and progression.
Collapse
Affiliation(s)
- Ziqi Chen
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhipeng Zhang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bei Ning Nie
- Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Wei Huang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Zhu
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Zhang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meng Xu
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengfei Wang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyue Yuan
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Wang
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Jianhui Tian
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Ba
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziliang Wang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Wellens J, Vissers E, Dumoulin A, Hoekx S, Vanderstappen J, Verbeke J, Vangoitsenhoven R, Derrien M, Verstockt B, Ferrante M, Matthys C, Raes J, Verbeke K, Vermeire S, Sabino J. Cooking methods affect advanced glycation end products and lipid profiles: A randomized cross-over study in healthy subjects. Cell Rep Med 2025; 6:102091. [PMID: 40280130 DOI: 10.1016/j.xcrm.2025.102091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/13/2024] [Accepted: 03/28/2025] [Indexed: 04/29/2025]
Abstract
Thermal treatments used in ultra-processed foods (UPFs) lead to advanced glycation end products (AGEs). UPFs and serum AGEs are associated with cardiometabolic disease. We explore differential cooking methods as a mechanistic link between UPFs and detrimental health outcomes through a randomized cross-over cooking method trial in healthy subjects using identical ingredients and a deep profiling analysis. We show that low-AGE-generating cooking methods such as boiling and steaming decrease serum AGEs, improve lipid profiles, and increase serum protein 4E-BP1. In contrast, high-AGE-generating cooking methods such as grilling and baking increase fecal butyrate. In sum, this suggests that low-AGE-generating cooking methods should be considered in cardiovascular risk prevention. Since current dietary guidelines focus on ingredients, but not cooking methods, our results suggest that culinary techniques should be considered as an important factor in cardiometabolic preventive strategies and future dietary trial design. This study was registered at ClinicalTrials.gov (NCT06547190).
Collapse
Affiliation(s)
- Judith Wellens
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Eva Vissers
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Anaïs Dumoulin
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Sien Hoekx
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Julie Vanderstappen
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Joke Verbeke
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Roman Vangoitsenhoven
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium; Department of Endocrinology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Muriel Derrien
- Microbiology, Immunology and Transplantation Department, Rega Institute, KU Leuven, 3000 Leuven, Belgium; VIB Center for Microbiology, 3000 Leuven, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Marc Ferrante
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Christophe Matthys
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium; Department of Endocrinology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jeroen Raes
- Microbiology, Immunology and Transplantation Department, Rega Institute, KU Leuven, 3000 Leuven, Belgium; VIB Center for Microbiology, 3000 Leuven, Belgium
| | - Kristin Verbeke
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - João Sabino
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
16
|
Carr L, Mustafa S, Collins-Praino LE. The Hallmarks of Ageing in Microglia. Cell Mol Neurobiol 2025; 45:45. [PMID: 40389766 PMCID: PMC12089641 DOI: 10.1007/s10571-025-01564-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: 01/30/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025]
Abstract
As ageing is linked to the development of neurodegenerative diseases (NDs), such as Alzheimer's Disease and Parkinson's Disease, it is important to disentangle the independent effect of age-related changes from those due to disease processes. To do so, changes to central nervous system (CNS) cells as a function of advanced age need better characterisation. Microglia are of particular interest due to their proposed links with the development and progression of NDs through control of the CNS immune response. Therefore, understanding the extent to which microglial dysfunction is related to phyisological ageing, rather than a disease process, is critical. As microglia age, they are believed to take on a pro-inflammatory phenotype with a distinct dystrophic morphology. Nevertheless, while established hallmarks of ageing have been investigated across a range of other cell types, such as macrophages, a detailed consideration of functional changes that occur in aged microglia remains elusive. Here, we describe the dynamic phenotypes of microglia and evaluate the current state of understanding of microglial ageing, focusing on the recently updated twelve hallmarks of ageing. Understanding how these hallmarks present in microglia represents a step towards better characterisation of microglial ageing, which is essential in the development of more representative models of NDs.
Collapse
Affiliation(s)
- Laura Carr
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Sanam Mustafa
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- School of Biomedicine, The University of Adelaide, Adelaide, Australia.
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.
| |
Collapse
|
17
|
Yadav SK, Chen C, Dhib-Jalbut S, Ito K. The mechanism of disease progression by aging and age-related gut dysbiosis in multiple sclerosis. Neurobiol Dis 2025; 212:106956. [PMID: 40383164 DOI: 10.1016/j.nbd.2025.106956] [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: 02/14/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease caused by a multifaceted interplay of genetic predispositions and environmental factors. Most patients initially experience the relapsing-remitting form of the disease (RRMS), which is characterized by episodes of neurological deficits followed by periods of symptom resolution. However, over time, many individuals with RRMS advance to a progressive form of the disease, known as secondary progressive MS (SPMS), marked by a gradual worsening of symptoms without periods of remission. The mechanisms underlying this transition remain largely unclear, and current disease-modifying therapies (DMTs) are partially effective in treating SPMS. Age is widely acknowledged as a risk factor for the transition from RRMS to SPMS. One factor associated with aging that may influence the progression of MS is gut dysbiosis. This review discusses how aging and age-related gut dysbiosis affect the progression of MS.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Claire Chen
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, United States of America.
| |
Collapse
|
18
|
Ye H, Meehan D, Timmons S, O'Toole PW. Effects of Prebiotics and a Synthetic Microbiome Consortium on the Composition and Metabolites of the Elderly Gut Microbiota In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11720-11729. [PMID: 40324006 DOI: 10.1021/acs.jafc.5c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
A 24 h artificial colon fermentation was performed to assess the effects of a prebiotic MIX and synthetic consortium (S7) on gut microbiota composition and microbial metabolite production in clinically stratified older adults (healthy and frail). Treatments included supplementation of the donor microbiota with a synthetic microbial consortium (S7), a prebiotic mix (MIX), and a combination of MIX and S7 (MIX+S7). The S7 treatment decreased the alpha diversity of long-stay-dwelling donor (LS, "frail") microbiota and increased the relative abundance of S7 taxa at 16 h. MIX alone caused the enrichment of reportedly beneficial genera such as Coprobacillus and Eubacterium in community-dwelling donor (CM, "healthy") microbiota and Citrobacter and Faecalibacterium in LS microbiota. The MIX+S7 treatment sustained higher overall S7 species richness and consortium taxon abundance at 24 h. Both MIX and MIX+S7 treatments enhanced short-chain acid production compared to control. These findings highlight the differential responses of microbiota from distinct elderly health strata to prebiotic and microbial consortia supplementation.
Collapse
Affiliation(s)
- Huimin Ye
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Dara Meehan
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Suzanne Timmons
- Centre for Gerontology and Rehabilitation, School of Medicine, University College Cork, Western Road, T12 Y337 Cork, Ireland
| | - Paul W O'Toole
- School of Microbiology, and APC Microbiome Ireland, University College Cork, Western Road, T12 Y337 Cork, Ireland
| |
Collapse
|
19
|
Goepp M, Milburn JV, Zhang B, Dong Y, Tyrrell V, Zheng X, Marshall JM, Bolsega S, Basic M, Glendinning L, Ho GT, Satsangi J, Breyer RM, Narumiya S, McSorley HJ, Schwarze JKJ, Anderson CJ, Dockrell DH, Rossi AG, Bleich A, Lucas CD, O'Donnell VB, Mole D, Arends MJ, Zhou Y, Yao C. Age-related impairment of intestinal inflammation resolution through an eicosanoid-immune-microbiota axis. Cell Host Microbe 2025; 33:671-687.e6. [PMID: 40373750 DOI: 10.1016/j.chom.2025.04.014] [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: 07/09/2024] [Revised: 02/27/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
Aging manifests a decline of immune function, induces microbiome dysbiosis, drives organ inflammation, and impedes the resolution of inflammation. However, the mechanisms underlying age-related intestinal inflammation remain poorly described. Here, we find that the resolution of T cell-initiated intestinal inflammation is impaired with aging. This impairment is mediated by disrupting the immune-microbiota interplay, controlled by intestinal eicosanoid metabolism. Pharmacologically inhibiting eicosanoid biosynthesis, blocking the prostaglandin E receptor subtype 4 (EP4), or genetically ablating EP4 diminishes age-related impairment of intestinal inflammation resolution. Mechanistically, mononuclear phagocyte-intrinsic eicosanoid-EP4 signaling impedes the resolution of intestinal inflammation through fostering gut microbial dysbiosis and, more importantly, interrupting segmented filamentous bacterial adhesion to the intestinal epithelium. Colonization with EP4-ablated mouse microbiota or segmented filamentous bacteria improves the resolution of intestinal inflammation. These findings reveal that eicosanoid-dependent immune-microbiota interactions impair inflammation resolution in the aged intestine, highlighting potential intervention strategies for improving age-related gut health.
Collapse
Affiliation(s)
- Marie Goepp
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jemma V Milburn
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Birong Zhang
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Yijia Dong
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria Tyrrell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Xiaozhong Zheng
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jennifer M Marshall
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Laura Glendinning
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Gwo-Tzer Ho
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, The University of Oxford, Oxford OX3 9DU, UK
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Shuh Narumiya
- Alliance Laboratory for Advanced Medical Research and Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Henry J McSorley
- Division of Cell Signaling and Immunology, School of Life Sciences, Wellcome Trust Building, The University of Dundee, Dundee DD1 4HN, UK
| | - Jürgen K J Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Christopher J Anderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - David H Dockrell
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Christopher D Lucas
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Valerie B O'Donnell
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Damian Mole
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, Institute of Genetics & Cancer, The University of Edinburgh, Institute of Genetics & Cancer, Edinburgh EH4 2XR, UK
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff CF14 4XN, UK
| | - Chengcan Yao
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
20
|
Sun H, Zhai Q, Liu J, Shi K, Fan W. Interplay between the gut microbiota, its metabolites and carcinogens. Clin Transl Oncol 2025:10.1007/s12094-025-03920-2. [PMID: 40358880 DOI: 10.1007/s12094-025-03920-2] [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: 02/11/2025] [Accepted: 03/30/2025] [Indexed: 05/15/2025]
Abstract
The gut microbiota is a complex and dynamic community of microorganisms that reside in the gastrointestinal tract, playing a critical role in the host. It produces many metabolites, such as bile acids, which play an important role in the metabolism of the host. One area of particular interest is its involvement in the development and treatment of cancer. Carcinogens, which are substances known to promote cancer formation and development, are present in various sources in our daily lives, including cigarettes, barbecues, and moldy foods. The types, amounts, and metabolism of carcinogens have been closely linked to cancer risk, underscoring the importance of understanding their interplay with the gut microbiota. Numerous studies have demonstrated significant differences in the composition and function of the gut microbiota in individuals with cancer compared to healthy individuals. The gut microbiota and its metabolites have been shown to influence the metabolism of various carcinogens, thereby affecting cancer progression. While much attention has been paid to the relationship between the gut microbiota and cancer risk, the potential interplay between the gut microbiota and carcinogens has received less focus. This review aims to emphasize the importance of exploring the interplay between the gut microbiota with its metabolites and carcinogens in cancer development and therapy. By uncovering the mechanisms of the interplay, new approaches for cancer prevention and treatment can be developed.
Collapse
Affiliation(s)
- Huan Sun
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Qiaoli Zhai
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Juan Liu
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Kourong Shi
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| |
Collapse
|
21
|
Escorcia Mora P, Valbuena D, Diez-Juan A. The Role of the Gut Microbiota in Female Reproductive and Gynecological Health: Insights into Endometrial Signaling Pathways. Life (Basel) 2025; 15:762. [PMID: 40430189 PMCID: PMC12113314 DOI: 10.3390/life15050762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Fertility is a dynamic, multifactorial process governed by hormonal, immune, metabolic, and environmental factors. Recent evidence highlights the gut microbiota as a key systemic regulator of reproductive health, with notable impacts on endometrial function, implantation, pregnancy maintenance, and the timing of birth. This review examines the gut-endometrial axis, focusing on how gut microbial communities influence reproductive biology through molecular signaling pathways. We discuss the modulatory roles of microbial-derived metabolites-including short-chain fatty acids, bile acids, and tryptophan catabolites-in shaping immune tolerance, estrogen metabolism, and epithelial integrity at the uterine interface. Emphasis is placed on shared mechanisms such as β-glucuronidase-mediated estrogen recycling, Toll-like receptor (TLR)-driven inflammation, Th17/Treg cell imbalance, and microbial translocation, which collectively implicate dysbiosis in the etiology of gynecological disorders including endometriosis, polycystic ovary syndrome (PCOS), recurrent implantation failure (RIF), preeclampsia (PE), and preterm birth (PTB). Although most current evidence remains correlational, emerging insights from metagenomic and metabolomic profiling, along with microbiota-depletion models and Mendelian randomization studies, underscore the biological significance of gut-reproductive crosstalk. By integrating concepts from microbiology, immunology, and reproductive molecular biology, this review offers a systems-level perspective on host-microbiota interactions in female fertility.
Collapse
Affiliation(s)
| | | | - Antonio Diez-Juan
- R&D Department, Igenomix (Part of Vitrolife Group), Ronda de Narcís Monturiol, nº11, B, Edificios Europark, Parque Tecnológico, 46980 Paterna, Valencia, Spain; (P.E.M.); (D.V.)
| |
Collapse
|
22
|
Nohesara S, Mostafavi Abdolmaleky H, Dickerson F, Pinto-Tomas AA, Jeste DV, Thiagalingam S. Associations of microbiome pathophysiology with social activity and behavior are mediated by epigenetic modulations: Avenues for designing innovative therapeutic strategies. Neurosci Biobehav Rev 2025; 174:106208. [PMID: 40350003 DOI: 10.1016/j.neubiorev.2025.106208] [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: 08/20/2024] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
A number of investigations have shown that gut microbiome influences humans' ability to communicate with others, and impairments in social interactions are linked to alterations in gut microbiome composition and diversity, via epigenetic mechanisms. This article reviews the links among gut microbiome, social behavior, and epigenetic shifts relevant to gut microbiome-derived metabolites. First, we discuss how different social determinants of health, such as socioeconomic status, diet, environmental chemicals, migration, ecological conditions, and seasonal changes may influence gut microbiome composition, diversity, and functionality, along with epigenetic alterations and thereby affect social behavior. Next, we consider how gut microbiome-derived metabolites, diet, probiotics, and fecal microbiome transplantation may reduce impairments in social interactions through the adjustment of epigenetic aberrations (e.g., DNA methylation, histone modifications, and microRNAs expression) which may suppress or increase gene expression patterns. Finally, we present the potential benefits and unresolved challenges with the use of gut microbiome-targeted therapeutics in reducing social deficits.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Faith Dickerson
- Stanley Research Program, Sheppard Pratt, Baltimore, MD, USA
| | - Adrian A Pinto-Tomas
- University of Costa Rica, Center for Research in Microscopic Structures and Biochemistry Department, School of Medicine, San Jose, Costa Rica
| | - Dilip V Jeste
- Global Research Network on Social Determinants of Mental Health and Exposomics, La Jolla, CA 92037, USA.
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02218, USA; Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
23
|
Zhang C, Gong L, Luo S, Yang L, Yan X. Analysis of alterations in the composition of the intestinal microbiota in frail older individuals. PLoS One 2025; 20:e0320918. [PMID: 40338858 PMCID: PMC12061151 DOI: 10.1371/journal.pone.0320918] [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: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Frailty is an ageing-associated geriatric syndrome that severely affects the functional status, quality of life and life expectancy of older adults. Immune dysfunction and chronic inflammation play crucial roles in frailty, and this study aimed to explore the correlation between the intestinal microbiota and frailty. METHODS A cross-sectional survey was conducted using a comprehensive geriatric assessment of older individuals who underwent medical checkups at the Health Management Center from April 2023 to May 2024. A total of 672 older individuals who met the inclusion criteria were included and divided into a healthy control group and a frail case group. Clinical data, as well as blood and stool samples, were collected. The data from the two groups were analysed with 16S rRNA sequencing in 20 and 30 cases, respectively. SPSS 25.0 was used for statistical analysis. RESULTS There were significant differences in income, smoking, and globulin levels between the two groups, while there were no differences in age or sex. There was no significant difference in the abundance or species evenness of intestinal bacteria between the two groups. However, the abundance of accessory bacteria, bifidobacteria, and Escherichia coli in the frail group was greater than that in the control group. Specifically, Escherichia-Shigella was significantly upregulated and fit well into the prediction model of frailty. CONCLUSION The gut microbiota of frail older individuals underwent significant changes in structural composition, and the presence of Escherichia-Shigella may be a diagnostic marker for debilitating diseases. These findings provide an essential clinical reference value for developing methods for preventing or alleviating frailty based on specific microbial communities.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lu Gong
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shilan Luo
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lamei Yang
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoli Yan
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Humińska-Lisowska K, Michałowska-Sawczyn M, Kosciolek T, Łabaj PP, Kochanowicz A, Mieszkowski J, Proia P, Cięszczyk P, Zielińska K. Gut microbiome and blood biomarkers reveal differential responses to aerobic and anaerobic exercise in collegiate men of diverse training backgrounds. Sci Rep 2025; 15:16061. [PMID: 40341642 PMCID: PMC12062308 DOI: 10.1038/s41598-025-99485-9] [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: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
The gut microbiome influences physiological responses to exercise by modulating inflammatory markers and metabolite production. Athletes typically exhibit greater microbial diversity, which may be associated with improved performance, but the mechanisms linking different exercise modalities to the gut microbiome are not fully understood. In this study, blood and stool samples were collected from endurance athletes, strength athletes, and non-athletic controls performing two maximal exercise tests (the anaerobic Wingate test and the aerobic Bruce Treadmill Test) to integrate serum biomarker data with gut bacterial metagenomic profiles. While most biochemical markers showed similar post-exercise trends across groups, SPARC (secreted protein acidic and rich in cysteine) and adiponectin levels showed modality-specific responses. Strength-trained participants showed unique microbiome-biomarker associations after the Wingate test. In addition, baseline enrichment of certain bacterial taxa, including Clostridium phoceensis and Catenibacterium spp., correlated with reduced Bruce Treadmill test response in strength-trained individuals. These findings, while requiring further validation, indicate the complex interplay between exercise type, training background, and the gut microbiome, and suggest that specific microbial species may help shape recovery and adaptation.
Collapse
Affiliation(s)
- Kinga Humińska-Lisowska
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland.
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy.
| | | | | | - Paweł P Łabaj
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | | | - Jan Mieszkowski
- Faculty of Health Sciences, University of Lomza, Lomza, Poland
| | - Patrizia Proia
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, Palermo, Italy
| | - Paweł Cięszczyk
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Kinga Zielińska
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
25
|
Singh S, Saini V, Jha HC. The role of secondary genomes in neurodevelopment and co-evolutionary dynamics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:245-297. [PMID: 40414634 DOI: 10.1016/bs.irn.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
This chapter examines how human biology and microbial "secondary genomes" have co-evolved to shape neurodevelopment through the gut-brain axis. Microbial communities generate metabolites that cross blood-brain and placental barriers, influencing synaptogenesis, immune responses, and neural circuit formation. Simultaneously, Human Accelerated Regions (HARs) and Endogenous Retroviruses (ERVs) modulate gene expression and immune pathways, determining which microbes thrive in the gut and impacting brain maturation. These factors converge to form a dynamic host-microbe dialogue with significant consequences for neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and schizophrenia. Building on evolutionary perspectives, the chapter elucidates how genetic and immune mechanisms orchestrate beneficial and pathological host-microbe interactions in early brain development. It then explores therapeutic strategies, such as probiotics, prebiotics, fecal microbiota transplantation, and CRISPR-driven microbial engineering, targeting gut dysbiosis to mitigate or prevent neurodevelopmental dysfunctions. Furthermore, innovative organ-on-chip models reveal mechanistic insights under physiologically relevant conditions, offering a translational bridge between in vitro experiments and clinical applications. As the field continues to evolve, this work underscores the translational potential of manipulating the microbiome to optimize neurological outcomes. It enriches our understanding of the intricate evolutionary interplay between host genomes and the microbial world.
Collapse
Affiliation(s)
- Siddharth Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| |
Collapse
|
26
|
Ishihara T, Tsugawa H, Iwanami S, Chang JC, Minoda A, Arita M. Transcriptomic and lipidomic analysis of aging-associated inflammatory signature in mouse liver. Inflamm Regen 2025; 45:13. [PMID: 40319315 PMCID: PMC12049063 DOI: 10.1186/s41232-025-00377-2] [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: 01/10/2025] [Accepted: 04/20/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Aging-associated dysbiosis leads to chronic inflammation and the development of a range of aging-related diseases. The gut microbiota crosstalks with the host by providing lipid metabolites and modulating metabolic functions. However, the precise mechanism by which the gut microbiota regulates aging is unknown. The objective of this study was to examine the impact of the gut microbiota on the transcriptome and lipidome associated with aging in mouse liver. METHODS RNA-sequencing was conducted on the livers of young and aged male and female-specific pathogen-free (SPF) and germ-free (GF) mice to comprehensively analyze transcriptomic alterations with aging. We also reanalyzed our previously reported results on aging-associated changes in the hepatic lipidome to investigate the gut microbiota-dependent hepatic lipidome signatures associated with aging. RESULTS In contrast to the findings in male mice, the changes in hepatic transcriptome associated with aging were attenuated in female GF mice compared with those in SPF mice. In particular, the gene sets associated with inflammatory signatures (i.e., inflammation and tissue remodeling) were found to be suppressed in female GF mice. The ChIP-Atlas database predicted that transcription factors associated with sex differences may be involved in the gene signature of aged female GF mice. Significant differences in the lipid profile were observed between aged SPF and GF female mice, including in bile acids, sterol sulfates, lysophospholipids, oxidized triacylglycerols, vitamin D, and phytoceramides. Moreover, notable alterations were identified in the quality of phospholipids and sphingolipids. Integrated transcriptomic and lipidomic analysis identified candidate enzymes responsible for the change of lipid profiles in aged female mice. CONCLUSIONS The findings of this study offer new insights into the molecular mechanisms through which the gut microbiota regulates aging-related phenotypes such as inflammation in the liver, possibly through modulating lipid metabolism in a sex-dependent manner.
Collapse
Affiliation(s)
- Tomoaki Ishihara
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Faculty of Pharmaceutical Sciences, Nagasaki International University, 2825-7, Huis Ten Bosch, Sasebo, Nagasaki, 859-3298, Japan.
| | - Hiroshi Tsugawa
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi, Tokyo, 184-8588, Japan
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Seigo Iwanami
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Jen-Chien Chang
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Aki Minoda
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Human Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
27
|
Hudobenko J, Di Gesù CM, Mooz PR, Petrosino J, Putluri N, Ganesh BP, Rebeles K, Blixt FW, Venna VR, McCullough LD. Maternal dysbiosis produces long-lasting behavioral changes in offspring. Mol Psychiatry 2025; 30:1847-1858. [PMID: 39443733 PMCID: PMC12014497 DOI: 10.1038/s41380-024-02794-0] [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: 02/27/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Advanced maternal age (AMA) is defined as a pregnancy in a woman older than 35 years of age. AMA increases the risk for both maternal and neonatal complications, including miscarriage and stillbirth. AMA has also been linked to neurodevelopmental and neuropsychiatric disorders in the offspring. Recent studies have found that age-associated compositional shifts in the gut microbiota contribute to altered microbial metabolism and enhanced inflammation in the host. We investigated the specific contribution of the maternal microbiome on pregnancy outcomes and offspring behavior by recolonizing young female mice with aged female microbiome prior to pregnancy. We discovered that pre-pregnancy colonization of young dams with microbiome from aged female donors significantly increased fetal loss. There were significant differences in the composition of the gut microbiome in pups born from dams recolonized with aged female biome that persisted through middle age. Offspring born from dams colonized with aged microbiome also had significant changes in levels of neurotransmitters and metabolites in the blood and the brain. Adult offspring from dams colonized with an aged microbiome displayed persistent depressive- and anxiety-like phenotypes. Collectively, these results demonstrate that age-related changes in the composition of the maternal gut microbiome contribute to chronic alterations in the behavior and physiology of offspring. This work highlights the potential of microbiome-targeted approaches, even prior to birth, may reduce the risk of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jacob Hudobenko
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Claudia M Di Gesù
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Patrick R Mooz
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Dan L. Duncan Comprehensive Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristen Rebeles
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Frank W Blixt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Venugopal R Venna
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA.
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Wu S, Qiao L, Liu H, Li YL, Wang R, Yin Y, Li E, Wang L, Guan X, Yin L, Liu Q, Peng X, Zhang Y, Yang Z, Zuo L, Zhang C. Age related gut microbiota regulates energy-related metabolism to influence natural aging phenotypes in the heart. Exp Gerontol 2025; 203:112734. [PMID: 40118127 DOI: 10.1016/j.exger.2025.112734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
As the population ages, problems pertaining to health and life expectancy due to the aging heart have become increasingly prominent. The gut microbiota has become a potential therapeutic target in several diseases, including cardiovascular diseases. Current studies on the roles of the gut microbiota in the cardiovascular system have focused mainly on cardiovascular diseases; therefore, the effects of the gut microbiota on the natural aging of myocardial tissue remain unclear. The present study aimed to explore the roles and mechanisms of the gut microbiota and related metabolites in the natural aging of the heart. Animal models of fecal microbiota transplantation (FMT) were established in elderly and young rats. 16S rRNA sequencing revealed that the gut microbiota of the recipients shifted toward the profile of the donors, with concomitant cardiac structure and diastolic function changes detected via ultrasound and positron emission tomography-computed tomography (PET-CT). A group of significantly enriched myocardial metabolites detected by LC/MS were involved in the fatty acid β-oxidation process. Together with altered glucose uptake, as revealed by PET-CT, changes in ATP content and mitochondrial structure further verified a metabolic difference related to energy among rats transplanted with the gut microbiota from donors of different ages. This study demonstrated that gut microbes may participate in the physiological aging process of the rat heart by regulating oxidative stress and autophagy. The gut microbiota has been shown to be involved in the natural aging of the heart at multiple levels, from the organ level to the metabolically plastic myocardiocytes and associated molecules.
Collapse
Affiliation(s)
- Shufen Wu
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China; Department of Pediatrics, Shanxi Medical University, Taiyuan, China
| | - Lingran Qiao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan-Li Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yiru Yin
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China; Translational Medicine Research Center of Shanxi Medical University, Taiyuan, China
| | - Enhui Li
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Lele Wang
- Department of Pediatrics, Shanxi Medical University, Taiyuan, China
| | - Xiaoya Guan
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China; Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Litian Yin
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Qinghua Liu
- Translational Medicine Research Center of Shanxi Medical University, Taiyuan, China
| | - Xiaoyang Peng
- Translational Medicine Research Center of Shanxi Medical University, Taiyuan, China
| | - Yutong Zhang
- Basic Medical School, Shanxi Medical University, Taiyuan, China
| | - Zhuanfang Yang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Lin Zuo
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Ce Zhang
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
29
|
Huang R, Zhang J, Sun M, Xu L, Kuang H, Xu C, Guo L. Oat β-glucan enhances gut barrier function and maintains intestinal homeostasis in naturally aging mice. Int J Biol Macromol 2025; 305:141129. [PMID: 39961571 DOI: 10.1016/j.ijbiomac.2025.141129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/21/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
In the process of aging, adverse changes such as weakened intestinal barrier function, increased chronic inflammation, and decreased gut microbiota diversity often occur. We explored the protective effects of Oat β-glucan (BG) on the gut homeostasis of naturally aging mice. The study shows that daily intervention with 400 mg/kg BG effectively modulates the intestinal mucosal structure, mechanical barrier function [Zonula occludens-1 (ZO-1), occludin, and claudin], and anti-inflammatory [Tumor Necrosis Factor-α (TNF-α), Interleukin-6 (IL-6), and IL-1β], as well as antioxidant responses in aging mice. Spearman correlation analyses showed that BG supplementation increased acetate levels by 1.8-fold, propionate levels by 2.5-fold, and butyrate-derived GABA levels by 2.5-fold. Additionally, BG supplementation improved the gut microbiota, increasing the abundance of beneficial bacteria like Bacteroidota, Prevotellaceae, Coprobacillaceae, and Faecalibacterium. These microbes metabolize BG to produce short-chain fatty acids (SCFAs), activating butanoate and propanoate metabolic pathways to maintain intestinal homeostasis. In conclusion, this study identifies the therapeutic effects of BG in regulating intestinal barrier homeostasis and gut microbiota, providing new insights for nutritional intervention strategies in the elderly.
Collapse
Affiliation(s)
- Renzhi Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jia Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Maozhong Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Liguang Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hua Kuang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chuanlai Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lingling Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
30
|
Yu S, Huang F, Huang Y, Yan F, Li Y, Xu S, Zhao Y, Zhang X, Chen R, Chen X, Zhang P. Deciphering the influence of gut and oral microbiomes on menopause for healthy aging. J Genet Genomics 2025; 52:601-614. [PMID: 39577767 DOI: 10.1016/j.jgg.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Menopause is characterized by the cessation of menstruation and a decline in reproductive function, which is an intrinsic component of the aging process. However, it has been a frequently overlooked field of women's health. The oral and gut microbiota, constituting the largest ecosystem within the human body, are important for maintaining human health and notably contribute to the healthy aging of menopausal women. Therefore, a comprehensive review elucidating the impact of the gut and oral microbiota on menopause for healthy aging is of paramount importance. This paper presents the current understanding of the microbiome during menopause, with a particular focus on alterations in the oral and gut microbiota. Our study elucidates the complex interplay between the microbiome and sex hormone levels, explores microbial crosstalk dynamics, and investigates the associations between the microbiome and diseases linked to menopause. Additionally, this review explores the potential of microbiome-targeting therapies for managing menopause-related diseases. Given that menopause can last for approximately 30 years, gaining insights into how the microbiome and menopause interact could pave the way for innovative interventions, which may result in symptomatic relief from menopause and an increase in quality of life in women.
Collapse
Affiliation(s)
- Shuting Yu
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China
| | - Yixuan Huang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Fangxu Yan
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yi Li
- Hunan Agriculture University, Changsha, Hunan 410128, China
| | - Shenglong Xu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yan Zhao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xinlei Zhang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China.
| | - Xingming Chen
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
31
|
Menassa M, Wilmont I, Beigrezaei S, Knobbe A, Arita VA, Valderrama JF, Bridge L, Verschuren WMM, Rennie KL, Franco OH, van der Ouderaa F. The future of healthy ageing: Wearables in public health, disease prevention and healthcare. Maturitas 2025; 196:108254. [PMID: 40157094 DOI: 10.1016/j.maturitas.2025.108254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Wearables have evolved into accessible tools for sports, research, and interventions. Their use has expanded to real-time monitoring of behavioural parameters related to ageing and health. This paper provides an overview of the literature on wearables in disease prevention and healthcare over the life course (not only in the older population), based on insights from the Future of Diagnostics Workshop (Leiden, January 2024). Wearable-generated parameters include blood glucose, heart rate, step count, energy expenditure, and oxygen saturation. Integrating wearables in healthcare is protracted and far from mainstream implementation, but promises better diagnosis, biomonitoring, and assessment of medical interventions. The main lifestyle factors monitored directly with wearables or through smartphone applications for disease prevention include physical activity, energy expenditure, gait, sleep, and sedentary behaviour. Insights on dietary consumption and nutrition have resulted from continuous glucose monitors. These factors are important for healthy ageing due to their effect on underlying disease pathways. Inclusivity and engagement, data quality and ease of interpretation, privacy and ethics, user autonomy in decision making, and efficacy present challenges to but also opportunities for their use, especially by older people. These need to be addressed before wearables can be integrated into mainstream medical and public health strategies. Furthermore, six key considerations need to be tackled: 1) engagement, health literacy, and compliance with personalised feedback, 2) technical and standardisation requirements for scalability, 3) accountability, data safety/security, and ethical concerns, 4) technological considerations, access, and capacity building, 5) clinical relevance and risk of overdiagnosis/overmedicalisation, and 6) the clinician's perspective in implementation.
Collapse
Affiliation(s)
- Marilyne Menassa
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands.
| | - Ilona Wilmont
- Institute for Computing and Information Sciences, Data Science, Radboud University Nijmegen, Nijmegen, the Netherlands; Stichting Je Leefstijl Als Medicijn, the Netherlands
| | - Sara Beigrezaei
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands
| | - Arno Knobbe
- Leiden Institute of Advanced Computer Science, Universiteit Leiden, Leiden, the Netherlands
| | - Vicente Artola Arita
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands
| | - Jose F Valderrama
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands
| | - Lara Bridge
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands
| | - W M Monique Verschuren
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands; National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Kirsten L Rennie
- MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Oscar H Franco
- Department of Global Public Health & Bioethics, Julius Center for Health Science and Primary Care, UMC Utrecht, Utrecht University, the Netherlands
| | | |
Collapse
|
32
|
Piłot M, Dzięgielewska-Gęsiak S, Walkiewicz KW, Bednarczyk M, Waniczek D, Muc-Wierzgoń M. Gut Microbiota and Metabolic Dysregulation in Elderly Diabetic Patients: Is There a Gender-Specific Effect. J Clin Med 2025; 14:3103. [PMID: 40364140 PMCID: PMC12073094 DOI: 10.3390/jcm14093103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: The aim of this study was to qualitatively and quantitatively assess the bacterial domain of the gut microbiome in elderly patients with type 2 diabetes (T2D), with a focus on sex differences, glycemic control, and lipid disorders. Methods: This study included 60 older adults with T2D (38 women and 22 men) treated with metformin or a combination of metformin and insulin. The gut microbiota was profiled using 16S rRNA gene sequencing. Statistical analyses, including correlation analysis and multiple regression, were performed to identify the associations between microbial taxa, sex, and metabolic parameters. Results: No statistically significant differences in alpha or beta diversity were observed between the sexes. Multiple regression analysis indicated a positive relationship between Tenericutes and HbA1c in male participants (β = 2.22931, CI [0.75, 3.70], R = 0.67; R2 = 0.36; unadjusted p = 0.0052; adjusted p = 0.0496). In female participants, G0' (β = -2.24107, CI [-3.19, -1.30], R = 0.78; R2 = 0.58; unadjusted p = 0.00003; adjusted p = 0.0005) and HbA1c (β = -1.86670, CI [-2.61, -1.12], R = 0.78; R2 = 0.58; unadjusted p = 0.00001; adjusted p = 0.0003) correlated negatively with Verrucomicrobia as well G0' (β = -1.90427, CI [-2.95, -0.85], R = 0.46; R2 = 0.17; unadjusted p = 0.0008; adjusted p = 0.007) and HbA1c (β = -1.69561, CI [-2.52, -0.87], R = 0.46; R2 = 0.17; unadjusted p = 0.0002; adjusted p = 0.002) correlated negatively with OD1 bacteria, known as Parcubacteria. Conclusions: In this elderly population with type 2 diabetes, biological sex did not significantly affect the gut microbiota diversity. However, several exploratory associations between microbial taxa and metabolic parameters differed between men and women, suggesting that sex may influence specific aspects of microbiota-metabolism interactions. These preliminary findings underscore the importance of considering both age- and sex-related factors when investigating the gut microbiome in the context of type 2 diabetes.
Collapse
Affiliation(s)
- Magdalena Piłot
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Sylwia Dzięgielewska-Gęsiak
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Katarzyna Weronika Walkiewicz
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Martyna Bednarczyk
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Katowice, Poland;
| | - Małgorzata Muc-Wierzgoń
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| |
Collapse
|
33
|
Brasiel PGDA, Potente Dutra Luquetti SC. Effects of Probiotics Supplementation on Short-Chain Fatty Acids: A Systematic Review of Randomized Controlled Trials. Nutr Rev 2025:nuaf047. [PMID: 40265671 DOI: 10.1093/nutrit/nuaf047] [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] [Indexed: 04/24/2025] Open
Abstract
CONTEXT With the increased use of fermented products and probiotic foods, interest in knowing their benefits and safety of their intake is increasing. OBJECTIVE The current systematic review investigated the effects of probiotics supplementation on short-chain fatty acid (SCFA) levels. METHODS A systematic review was conducted according to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Medline/PubMed, Scopus, Web of Science, and Embase databases were searched from inception to February 2024, including only randomized controlled trials. RESULTS A total of 30 studies were included, involving 1499 participants. The vast majority of trials investigated Bifidobacterium and Lactobacillus strains. These were categorized into healthy adults (n = 6), gastrointestinal/inflammatory diseases (n = 7), metabolic diseases (n = 6), elderly individuals (n = 3), children (n = 4), and infant formula (n = 4). Most studies evaluated isolated strain supplementation (n = 12), followed by fermented beverages (n = 11) and probiotic mixes (n = 3). Globally, 16 studies (53.3%) revealed an increase in at least 1 SCFA in participants supplemented with probiotics. In comparison, 5 studies (16.7%) reported a reduction and 9 studies showed no statistically significant impact in their findings (30%). The subgroup evaluation showed heterogeneity in the results, with low to moderate evidence grading levels. CONCLUSION Probiotics supplementation during childhood was the intervention period more effective in increasing fecal SCFAs and immunoglobulin A (IgA) levels (GRADE: moderate/high-certainty evidence). SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42024513221.
Collapse
|
34
|
Cho MY, Eom JH, Choi EM, Yang SJ, Lee D, Kim YY, Kim HS, Hwang I. Recent advances in therapeutic probiotics: insights from human trials. Clin Microbiol Rev 2025:e0024024. [PMID: 40261032 DOI: 10.1128/cmr.00240-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
SUMMARYRecent advances in therapeutic probiotics have shown promising results across various health conditions, reflecting a growing understanding of the human microbiome's role in health and disease. However, comprehensive reviews integrating the diverse therapeutic effects of probiotics in human subjects have been limited. By analyzing randomized controlled trials (RCTs) and meta-analyses, this review provides a comprehensive overview of key developments in probiotic interventions targeting gut, liver, skin, vaginal, mental, and oral health. Emerging evidence supports the efficacy of specific probiotic strains and combinations in treating a wide range of disorders, from gastrointestinal (GI) and liver diseases to dermatological conditions, bacterial vaginosis, mental disorders, and oral diseases. We discuss the expanding understanding of microbiome-organ connections underlying probiotic mechanisms of action. While many clinical trials demonstrate significant benefits, we acknowledge areas requiring further large-scale studies to establish definitive efficacy and optimal treatment protocols. The review addresses challenges in standardizing probiotic research methodologies and emphasizes the importance of considering individual variations in microbiome composition and host genetics. Additionally, we explore emerging concepts such as the oral-gut-brain axis and future directions, including high-resolution microbiome profiling, host-microbe interaction studies, organoid models, and artificial intelligence applications in probiotic research. Overall, this review offers a comprehensive update on the current state of therapeutic probiotics across multiple domains of human health, providing insights into future directions and the potential for probiotics to revolutionize preventive and therapeutic medicine.
Collapse
Affiliation(s)
- Mu-Yeol Cho
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Je-Hyun Eom
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Eun-Mi Choi
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | | | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Young Youn Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Hye-Sung Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Inseong Hwang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| |
Collapse
|
35
|
Kirschner SK, Engelen MP, Haas P, Bischoff SC, Deutz NE. Short-chain fatty acid kinetics and concentrations are higher after inulin supplementation in young and older adults: a randomized trial. Am J Clin Nutr 2025:S0002-9165(25)00235-7. [PMID: 40274191 DOI: 10.1016/j.ajcnut.2025.04.018] [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: 11/01/2024] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Beneficial short-chain fatty acids (SCFAs) are produced through intestinal microbial fiber fermentation. Using stable tracer methodology and compartmental modeling, we observed lower SCFA production in older (OAs) than in young adults (YAs) in both an accessible [that is, systemic circulation; whole-body production] and inaccessible [potentially representing intestine absorbing microbially produced SCFAs (U2)] pool. OBJECTIVES We now investigated whether fiber supplementation increases SCFA production in OAs and whether concentrations reflect production rate changes. METHODS In this randomized, placebo-controlled, double-blind crossover study, 21 YAs (20-29 y) and 40 OAs (59-87 y) adults were supplemented with inulin or placebo (maltodextrin) for 7 d (final intake: 30 g/d). Before and after interventions, participants collected stool and received an intravenous pulse containing [U-13C]-labeled SCFAs followed by blood draws. We measured plasma tracer enrichments, plasma and fecal concentrations by gas chromatography-mass spectrometry and performed compartmental analysis. Data are mean (95% confidence interval). RESULTS Inulin evoked a 44% increase in butyrate production (μmol/min) in the inaccessible pool {YA: 28-44 [+16.2 (4.3, 28.1); P = 0.038], OA: 14-20 [+6.1 (2.2, 9.9); P = 0.011]} and were not different between YAs and OAs. In addition, a 34% increase in propionate production in YA only. We found a 50%-60% increase in fecal acetate, propionate, and butyrate and a 34% increase in plasma butyrate in OA, whereas in YA only 34% increase in fecal acetate. Plasma but not fecal concentrations correlated positively with SCFA production in the inaccessible pool (R2 = 0.20-0.45; P < 0.001). CONCLUSIONS OAs have a lower SCFA production. Inulin intake increases SCFA production. Tracer pulse approach detects SCFA metabolism changes more sensitively than plasma or fecal concentration measurements (NCT04459156).
Collapse
Affiliation(s)
- Sarah K Kirschner
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, United States
| | - Mariëlle Pkj Engelen
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, United States
| | - Paula Haas
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, United States
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Nicolaas Ep Deutz
- Center for Translational Research in Aging & Longevity, Texas A&M University, College Station, TX, United States.
| |
Collapse
|
36
|
Garg G, Trisal A, Singh AK. Unlocking the therapeutic potential of gut microbiota for preventing and treating aging-related neurological disorders. Neuroscience 2025; 572:190-203. [PMID: 40073931 DOI: 10.1016/j.neuroscience.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Billions of microorganisms inhabit the human gut and maintain overall health. Recent research has revealed the intricate interaction between the brain and gut microbiota through the microbiota-gut-brain axis (MGBA) and its effect on neurodegenerative disorders (NDDs). Alterations in the gut microbiota, known as gut dysbiosis, are linked to the development and progression of several NDDs. Studies suggest that the gut microbiota may be a viable target for improving cognitive health and reducing hallmarks of brain aging. Numerous pathways including hypothalamic-pituitary-adrenal axis stimulation, neurotransmitter release disruption, system-wide inflammation, and increased intestinal and blood-brain barrier permeability connect gut dysbiosis to neurological conditions. Metabolites produced by the gut microbiota influence neural processes that affect brain function. Clinical interventions depend on the capacity to understand the equilibrium between beneficial and detrimental gut microbiota, as it affects both neurodegeneration and neuroprotection. The importance of the gut microbiota and its metabolites during brain aging and the development of neurological disorders is summarized in this review. Moreover, we explored the possible therapeutic effects of the gut microbiota on age-related NDDs. Highlighting various pathways that connect the gut and the brain, this review identifies several important domains where gut microbiota-based interventions could offer possible solutions for age-related NDDs. Furthermore, prebiotics and probiotics are discussed as effective alternatives for mitigating indirect causes of gut dysbiosis. These therapeutic interventions are poised to play a significant role in improving dysbiosis and NDDs, paving the way for further research.
Collapse
Affiliation(s)
- Geetika Garg
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576 104, India.
| |
Collapse
|
37
|
Peters BA, Xue X, Hanna DB, Wang Y, Wang Z, Sharma A, Floris-Moore M, Konkle-Parker D, Alcaide ML, Sheth AN, Topper EF, Weber KM, Tien PC, Merenstein D, Vásquez E, Chen Y, Mimiaga MJ, Stosor V, Brown TT, Erlandson KM, Dillon SM, Elsayed NS, Usyk M, Sollecito CC, Kaplan RC, Burk RD, Qi Q. Healthy Aging and the Gut Microbiome in People With and Without HIV. J Infect Dis 2025; 231:981-992. [PMID: 39841165 PMCID: PMC11998545 DOI: 10.1093/infdis/jiae644] [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: 11/15/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Aging-related comorbidities are more common in people with human immunodeficiency virus (HIV) compared to people without HIV. The gut microbiome may play a role in healthy aging; however, this relationship remains unexplored in the context of HIV. METHODS 16S rRNA gene sequencing was conducted on stool from 1409 women (69% with HIV; 2304 samples) and 990 men (54% with HIV; 1008 samples) in the MACS/WIHS Combined Cohort Study. Associations of age with gut microbiome diversity, uniqueness, and genus-level abundance were examined in women and men separately, followed by examining relationships of aging-related genera with frailty (Fried frailty phenotype) and mortality risk (Veterans Aging Cohort Study [VACS] index). RESULTS Older age was associated with greater microbiome diversity and uniqueness, greater abundance of Akkermansia and Streptococcus, and lower abundance of Prevotella and Faecalibacterium, among others; findings were generally consistent by sex and HIV status. An aging-related microbiome score, generated via combination of 18 age-related genera, significantly increased with age in both women and men independently of demographic, behavioral, and cardiometabolic factors. In general, age was more strongly related to microbiome features (eg, diversity, microbiome score) in men without compared to with HIV, but age-microbiome associations were similar in women with and without HIV. Some age-related genera associated with healthy/unhealthy aging, such as Faecalibacterium (related to reduced frailty) and Streptococcus (related to higher VACS index). CONCLUSIONS Age is associated with consistent changes in the gut microbiome in both women and men with or without HIV. Some aging-related microbiota are associated with aging-related declines in health.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yi Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Zheng Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Anjali Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Michelle Floris-Moore
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Deborah Konkle-Parker
- Schools of Nursing, Medicine, and Population Health Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Maria L Alcaide
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anandi N Sheth
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Ponce de Leon Center, Grady Health System, Atlanta, Georgia, USA
| | - Elizabeth F Topper
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Phyllis C Tien
- Medical Service, Department of Veterans Affairs Medical Center, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Daniel Merenstein
- Department of Family Medicine, Georgetown University, Washington, District of Columbia, USA
| | - Elizabeth Vásquez
- Department of Epidemiology and Biostatistics, College of Integrated Health Science, State University of New York, Rensselaer, New York, USA
| | - Yue Chen
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew J Mimiaga
- Department of Epidemiology, University of California Los Angeles, Los Angeles, California, USA
| | - Valentina Stosor
- Department of Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Todd T Brown
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristine M Erlandson
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Stephanie M Dillon
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Noha S Elsayed
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mykhaylo Usyk
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
38
|
Urbani G, Rondini E, Distrutti E, Marchianò S, Biagioli M, Fiorucci S. Phenotyping the Chemical Communications of the Intestinal Microbiota and the Host: Secondary Bile Acids as Postbiotics. Cells 2025; 14:595. [PMID: 40277921 PMCID: PMC12025480 DOI: 10.3390/cells14080595] [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: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
The current definition of a postbiotic is a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics can be mainly classified as metabolites, derived from intestinal bacterial fermentation, or structural components, as intrinsic constituents of the microbial cell. Secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA) are bacterial metabolites generated by the enzymatic modifications of primary bile acids by microbial enzymes. Secondary bile acids function as receptor ligands modulating the activity of a family of bile-acid-regulated receptors (BARRs), including GPBAR1, Vitamin D (VDR) receptor and RORγT expressed by various cell types within the entire human body. Secondary bile acids integrate the definition of postbiotics, exerting potential beneficial effects on human health given their ability to regulate multiple biological processes such as glucose metabolism, energy expenditure and inflammation/immunity. Although there is evidence that bile acids might be harmful to the intestine, most of this evidence does not account for intestinal dysbiosis. This review examines this novel conceptual framework of secondary bile acids as postbiotics and how these mediators participate in maintaining host health.
Collapse
Affiliation(s)
- Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Elena Rondini
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Silvia Marchianò
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| |
Collapse
|
39
|
Eastwood J, van Hemert S, Stolaki M, Williams C, Walton G, Lamport D. Exploring the acute and chronic effects of a multistrain probiotic supplement on cognitive function and mood in healthy older adults: a randomized controlled trial. Am J Clin Nutr 2025:S0002-9165(25)00188-1. [PMID: 40222448 DOI: 10.1016/j.ajcnut.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Aging is associated with a decline in cognitive function and vulnerability to depression. Probiotic supplements have shown beneficial effects on cognition and mood in clinical populations, but the potential benefit for healthy older adults experiencing age-related decline in cognition remains unclear. OBJECTIVES The primary aim of the present work was to explore the effect of a chronic (long-term) multispecies probiotic intervention on cognition in healthy aging adults. Secondary aims included exploring the chronic effect on mood outcomes and gut microbiota community, as well as a novel investigation into the acute effect of supplementation on cognition and mood. METHODS The study employed a randomized, placebo-controlled, cross-over trial in 30 healthy older adults to explore the acute (1 d) and chronic (8 wk) effects of a probiotic supplement on cognitive domains of memory and executive function, alongside mood measures of stress, anxiety, depression, and cognitive reactivity to sad mood. 16s rRNA sequencing of stool samples was also performed pre- and postchronic intervention to assess potential effects on the gut microbiota. RESULTS Acute probiotic supplementation was associated with faster reaction times on cognitively demanding trials during a task of executive function [-64.91 ms, 95% confidence interval (CI): -115.70, -14.15]. Chronic supplementation was associated with improvement in cognitive biases such as hopelessness (-0.97, 95% CI: -1.72, -0.23), rumination (-1.58, 95% CI: -2.86, -0.29), and aggression (-1.57, 95% CI: -2.63, -0.51) that contribute to reactivity to sad mood and therefore vulnerability to depression, and may improve executive function under higher cognitive demand (0.43%, 95% CI: -0.53%, 1.38%). CONCLUSIONS The current work provides novel evidence for an acute effect of probiotics on reaction times during executive function, which should be replicated in future work. Additionally, this work replicates previous findings of improved cognitive reactivity to sad mood following chronic probiotic supplementation, indicating probiotics may reduce risk of developing depression in a healthy aging population. This study was registered at clinicaltrials.gov as NCT04951687.
Collapse
Affiliation(s)
- Jessica Eastwood
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom.
| | | | | | - Claire Williams
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| | - Gemma Walton
- Food Microbial Sciences Unit, University of Reading, Reading, United Kingdom
| | - Daniel Lamport
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
40
|
Mohammadzadeh R, Mahnert A, Shinde T, Kumpitsch C, Weinberger V, Schmidt H, Moissl-Eichinger C. Age-related dynamics of predominant methanogenic archaea in the human gut microbiome. BMC Microbiol 2025; 25:193. [PMID: 40181255 PMCID: PMC11969853 DOI: 10.1186/s12866-025-03921-9] [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: 05/06/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The reciprocal relationship between aging and alterations in the gut microbiota is a subject of ongoing research. While the role of bacteria in the gut microbiome is well-documented, specific changes in the composition of methanogens during extreme aging and the impact of high methane production in general on health remain unclear. This study was designed to explore the association of predominant methanogenic archaea within the human gut and aging. METHODS Shotgun metagenomic data from the stool samples of young adults (n = 127, Age: 19-59 y), older adults (n = 86, Age: 60-99 y), and centenarians (n = 34, age: 100-109 years) were analyzed. RESULTS Our findings reveal a compelling link between age and the prevalence of high methanogen phenotype, while overall archaeal diversity diminishes. Surprisingly, the archaeal composition of methanogens in the microbiome of centenarians appears more akin to that of younger adults, showing an increase in Methanobrevibacter smithii, rather than Candidatus Methanobrevibacter intestini. Remarkably, Ca. M. intestini emerged as a central player in the stability of the archaea-bacteria network in adults, paving the way for M. smithii in older adults and centenarians. Notably, centenarians exhibit a highly complex and stable network of these two methanogens with other bacteria. The mutual exclusion between Lachnospiraceae and these methanogens throughout all age groups suggests that these archaeal communities may compensate for the age-related drop in Lachnospiraceae by co-occurring with Oscillospiraceae. CONCLUSIONS This study underscores the dynamics of archaeal microbiome in human physiology and aging. It highlights age-related shifts in methanogen composition, emphasizing the significance of both M. smithii and Ca. M. intestini and their partnership with butyrate-producing bacteria for potential enhanced health.
Collapse
Affiliation(s)
- Rokhsareh Mohammadzadeh
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Alexander Mahnert
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Tejus Shinde
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Viktoria Weinberger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Helena Schmidt
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria.
- BioTechMed, Graz, 8010, Austria.
| |
Collapse
|
41
|
Lee DY, Noren Hooten N, O'Connell JF, Lee BY, Kim Y. The Role of Ginseng and Its Bioactive Compounds in Aging: Cells and Animal Studies. Annu Rev Food Sci Technol 2025; 16:333-354. [PMID: 39971378 DOI: 10.1146/annurev-food-111523-121753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Aging is an inevitable process that is characterized by physiological deterioration and increased vulnerability to stressors. Therefore, the interest in hallmarks, mechanisms, and ways to delay or prevent aging has grown for decades. Natural plant products and their bioactive compounds have been studied as a promising strategy to overcome aging. Ginseng, a traditional herbal medicine, and its bioactive compound, the ginsenosides, have increasingly gained attention because of various pharmacological functions. This review introduces the species, useful parts, characteristics, and active components of ginseng. It primarily focuses on the bioconversion of ginsenosides through the unique steaming and drying process. More importantly, this review enumerates the antiaging mechanisms of ginseng, ginsenosides, and other bioactive compounds, highlighting their potential to extend the health span and mitigate age-related diseases based on twelve representative hallmarks of aging.
Collapse
Affiliation(s)
- Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, Maryland, USA
| | - Jennifer F O'Connell
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, USA
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea;
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| |
Collapse
|
42
|
Golshani M, Taylor JA, Woolbright BL. Understanding the microbiome as a mediator of bladder cancer progression and therapeutic response. Urol Oncol 2025; 43:254-265. [PMID: 39117491 DOI: 10.1016/j.urolonc.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/17/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Bladder cancer (BCa) remains a significant source of morbidity and mortality. BCa is one of the most expensive tumors to treat, in part because of a lack of nonsurgical options. The recent advent of immunotherapy, alone or in combination with other compounds, has improved therapeutic options. Resistance to immunotherapy remains common, and many patients do not have durable response. Recent advances indicate immunotherapy efficacy may be tied in part to the endogenous bacteria present in our body, more commonly referred to as the microbiome. Laboratory and clinical data now support the idea that a healthy microbiome is critical to effective response to immunotherapy. At the same time, pathogenic interactions between the microbiome and immune cells can also serve to drive formation of tumors, increasing the complexity of these interactions. Given the rising importance of immunotherapy in BCa, understanding how we might be able to alter the microbiome to improve therapeutic efficacy offers a novel route to improved patient care. The goal of this review is to examine our current understanding of microbial interactions with the immune system and cancer with an emphasis on BCa. We will further attempt to define both current gaps in knowledge and future directions that may yield beneficial results to the field.
Collapse
Affiliation(s)
- Mahgol Golshani
- School of Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS; Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
43
|
Mortimer T, Smith JG, Muñoz-Cánoves P, Benitah SA. Circadian clock communication during homeostasis and ageing. Nat Rev Mol Cell Biol 2025; 26:314-331. [PMID: 39753699 DOI: 10.1038/s41580-024-00802-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 03/28/2025]
Abstract
Maintaining homeostasis is essential for continued health, and the progressive decay of homeostatic processes is a hallmark of ageing. Daily environmental rhythms threaten homeostasis, and circadian clocks have evolved to execute physiological processes in a manner that anticipates, and thus mitigates, their effects on the organism. Clocks are active in almost all cell types; their rhythmicity and functional output are determined by a combination of tissue-intrinsic and systemic inputs. Numerous inputs for a specific tissue are produced by the activity of circadian clocks of other tissues or cell types, generating a form of crosstalk known as clock communication. In mammals, the central clock in the hypothalamus integrates signals from external light-dark cycles to align peripheral clocks elsewhere in the body. This regulation is complemented by a tissue-specific milieu of external, systemic and niche inputs that modulate and cooperate with the cellular circadian clock machinery of a tissue to tailor its functional output. These mechanisms of clock communication decay during ageing, and growing evidence suggests that this decline might drive ageing-related morbidities. Dietary, behavioural and pharmacological interventions may offer the possibility to overcome these changes and in turn improve healthspan.
Collapse
Affiliation(s)
- Thomas Mortimer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Jacob G Smith
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain.
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA, USA.
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
44
|
Coperchini F, Greco A, Teliti M, Croce L, Chytiris S, Magri F, Gaetano C, Rotondi M. Inflamm-ageing: How cytokines and nutrition shape the trajectory of ageing. Cytokine Growth Factor Rev 2025; 82:31-42. [PMID: 39237438 DOI: 10.1016/j.cytogfr.2024.08.004] [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: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
Population ageing is increasing in prevalence in most developed countries. Ageing is the decline of functional properties at the cellular, tissue, and organ level. Biochemical changes that occur in all organisms that experience biological ageing are referred to as the "Hallmarks of ageing". Inflammation is a common denominator of the hallmarks of ageing, being mechanistically involved in most age-related health consequences. Inflamm-ageing refers to age-related changes in the inflammatory and immune systems which somehow drive the ageing process towards healthy or unhealthy ageing. Current evidences, support that, reversing the age-related pro-inflammatory status of inflamm-ageing, is able to modulate most hallmarks of ageing. Inflamm-ageing is associated with increased levels of pro-inflammatory molecules (e.g. cytokines, chemokines), ultimately producing a chronic low-grade inflammatory state typically observed in older individuals. It is commonly accepted that, the balance between pro- and anti-inflammatory cytokines/chemokines is one of the factors determining whether healthy or unhealthy ageing occurs. Malnutrition and nutritional imbalances, are highly prevalent in the elderly, playing a role in driving the balance of pro- and anti-inflammatory immunoactive molecules. In particular, malnutrition is a major risk factor for sarcopenia, a phenomenon characterized by loss of muscle mass, which is often referred to as the biological basis for frailty. Given the close relationship between malnutrition and sarcopenia, there is also evidence for a link between malnutrition and frailty. Indeed, changes in cytokine/chemokine levels in elderly patients with malnutrition were demonstrated. The demonstration that specific cytokines play a role in modulating appetite and nutrient sensing and taste reception, provided further evidence for the existence of a link between inflamm-ageing, nutrition and cytokines in shaping the trajectory of ageing. The present review will overview current evidence supporting the role of specific circulating cytokines and chemokines in the relationship between ageing, inflammation, and malnutrition.
Collapse
Affiliation(s)
- Francesca Coperchini
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Alessia Greco
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy
| | - Marsida Teliti
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Laura Croce
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Spyridon Chytiris
- Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Flavia Magri
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Mario Rotondi
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia 27100, Italy.
| |
Collapse
|
45
|
Cavon J, Basso M, Kadosh KC, Gibbons SM. The human gut microbiome and sleep across adulthood: associations and therapeutic potential. Lett Appl Microbiol 2025; 78:ovaf043. [PMID: 40113228 PMCID: PMC11959190 DOI: 10.1093/lambio/ovaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/25/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025]
Abstract
Sleep is an essential homeostatic process that undergoes dynamic changes throughout the lifespan, with distinct life stages predisposed to specific sleep pathologies. Similarly, the gut microbiome also varies with age, with different signatures associated with health and disease in the latest decades of life. Emerging research has shown significant cross-talk between the gut microbiota and the brain through several pathways, suggesting the microbiota may influence sleep, though the specific mechanisms remain to be elucidated. Here, we critically examine the existing literature on the potential impacts of the gut microbiome on sleep and how this relationship varies across adulthood. We suggest that age-related shifts in gut microbiome composition and immune function may, in part, drive age-related changes in sleep. We conclude with an outlook on the therapeutic potential of microbiome-targeted interventions aimed at improving sleep across adulthood, particularly for individuals experiencing high stress or with sleep complaints.
Collapse
Affiliation(s)
- Jacob Cavon
- Institute for Systems Biology, Seattle, WA 98109, United States
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA 98195, United States
| | - Melissa Basso
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7HX, United Kingdom
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom
| | - Kathrin Cohen Kadosh
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7HX, United Kingdom
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA 98109, United States
- Molecular Engineering Graduate Program, University of Washington, Seattle, WA 98195, United States
- Department of Bioengineering, University of Washington, Seattle, WA 98195, United States
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, United States
- eScience Institute, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
46
|
Jones D, Morrison DJ, Gray SR, Ozanne SE, Celis-Morales C, Jain M, Mattin LR, Gittins M, Alkhedhairi SAA, Dorling JL, Burden S. Dietary intake in healthy older individuals is associated with lipopolysaccharide binding protein a biomarker of gut function: an exploratory cross-sectional study. FRONTIERS IN AGING 2025; 6:1572867. [PMID: 40231185 PMCID: PMC11994966 DOI: 10.3389/fragi.2025.1572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025]
Abstract
Diet, physical function and gut health are important modifiable factors in ageing. However, it is unclear how ageing affects various domains of gut function. Aims of this cross-sectional study were to explore relationships between nutrient intake, physical function, and biomarkers of gut function in older individuals. Healthy participants (n = 94, mean age 71.1 years SD 5.10, 56% female) were recruited to investigate the relationship between nutrient intake (protein, fibre, carbohydrate, fat), physical function (chair rise time, handgrip strength) and lipopolysaccharide (LPS) binding protein (LBP); a marker of gut permeability. Linear regression models, adjusted for age, fat mass/fat free mass ratio, weight and gender, reported LBP changed by; -161.9 ng/mL (95% CI -323.0, -0.8) for every 1 g increase in daily fibre/1,000 kilocalories; 80.5 ng/mL (6.7, 154.2) for 1% increase in daily energy intake as fat; and -88.1 ng/mL (-146.7, -29.6) for 1% increase in daily energy as carbohydrates. When further adjusted for C-reactive protein (CRP), a marker of inflammation, LBP decreased by an additional 6.9 ng/mL for fibre, increased by an additional 4.0 ng/mL for fat and decreased by an additional 3.7 ng/mL for carbohydrate. These findings suggest that in healthy older adults' nutrient intake is associated with LBP, and CRP appears to slightly modify these associations. There were no associations between LBP and handgrip strength or chair rise time. Results suggest that fibre, fat, and carbohydrates are important for maintaining gut function, potentially mediated by inflammation in older adults, although further research is needed to explore the implications for physical function and CRP as a mediator.
Collapse
Affiliation(s)
- Debra Jones
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Douglas J. Morrison
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Stuart R. Gray
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
- Institute of Sports Science and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| | - Susan E. Ozanne
- Institute of Metabolic Science - Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - Carlos Celis-Morales
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Mahek Jain
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, United Kingdom
| | - Lewis R. Mattin
- School of Life Sciences, University of Westminster, London, United Kingdom
| | - Matthew Gittins
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Saleh A. A. Alkhedhairi
- Department of Medical Biosciences, College of Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - James L. Dorling
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sorrel Burden
- School of Health Sciences, University of Manchester, Manchester, United Kingdom
- Salford Care Organisation, Northern Care Alliance NHS Trust, Salford, United Kingdom
| |
Collapse
|
47
|
Chen N, Yu Z, Ji X, Zhang S, Yu C, Valencak TG, Shi F, Ren D. Canine-derived Weissella confusa ZJUIDS-D034 and Enterococcus faecalis ZJUIDS-D016 combat aging by regulating gut microbiota. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100381. [PMID: 40248687 PMCID: PMC12005927 DOI: 10.1016/j.crmicr.2025.100381] [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] [Indexed: 04/19/2025] Open
Abstract
Old age raises the susceptibility of age-related disease in domestic dogs. Discovering effective anti-aging interventions is key for mitigating age-related disease and conserving "healthspan" in pet dogs. In this study, 2 bacterial strains were isolated from canine feces. After screening and identifying the strains, Weissella confusa ZJUIDS-D034 and Enterococcus faecalis ZJUIDS-D016 were chosen to intervene during d-galactose-induced senescence in mice. We found that administering Weissella confusa ZJUIDS-D034 and Enterococcus faecalis ZJUIDS-D016 improved the aging phenotype of mice, including an increase in antioxidant activity, a decrease in pro-inflammatory cytokines, and the restoration of intestinal and liver tissue damage. In addition, Weissella confusa ZJUIDS-D034 and Enterococcus faecalis ZJUIDS-D016 lead to changes in the structure of intestinal microbiota in aging mice. Specifically, there was a decrease in the abundance of the Cyanobacteria and an increase in the abundance of Akkermansia and Lactobacillus. More importantly, there was a significant increase in acetic acid, a short-chain fatty acid, due to intervention with the 2 strains. This increase might be attributed to higher Akkermansia. We show that the modulation of gut microbiota and metabolism in aging mice may be a promising strategy through which Weissella confusa ZJUIDS-D034 and Enterococcus faecalis ZJUIDS-D016 might exert their anti-aging effects.
Collapse
Affiliation(s)
- Nan Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zexu Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuan Ji
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Siyi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chongwei Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | | | - Fushan Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daxi Ren
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Xinjiang Agricultural University-Zhejiang University Joint Research Center for Feed and Quality Livestock and Poultry Products, Xinjiang Agricultural University, Urumqi, 830052, China
| |
Collapse
|
48
|
Porter Starr KN, Connelly MA, Wallis J, North R, Zhang Q, Song K, González-Delgado JM, Brochu HN, Icenhour CR, Iyer LK, Miller MG, Huffman KM, Kraus WE, Bales CW. Effects of Blueberry Consumption on Fecal Microbiome Composition and Circulating Metabolites, Lipids, and Lipoproteins in a Randomized Controlled Trial of Older Adults with Overweight or Obesity: The BEACTIVE Trial. Nutrients 2025; 17:1200. [PMID: 40218958 PMCID: PMC11990464 DOI: 10.3390/nu17071200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Generous consumption of phytonutrient-rich foods, including blueberries, provides benefits to multiple physiologic and metabolic systems. This study explored the potential that regular, generous blueberry intake could favorably modulate fecal microbiome composition in sedentary older (>60 years) men and women with overweight or obesity (BMI ≥ 25 to 32 kg/m2). Methods: Participants (n = 55) were randomized to daily consumption of either lyophilized blueberry powder (equivalent to 1.5 cups of blueberries) or an indistinguishable placebo powder; both groups participated in weekly supervised exercise classes. Fecal samples were collected at 0 and 12 weeks and frozen. Following this, 16S rRNA gene sequencing was used to profile each participant's fecal microbiome. Blood biomarkers of cardiometabolic health were measured via nuclear magnetic resonance spectroscopy (NMR) pre- and post-treatment. Results: Comparing the baseline and endpoint results for the blueberry (n = 15) and placebo (n = 19) groups, there were no significant overall compositional differences or differences in the level of diversity in the fecal microbiome. However, in subjects whose diet included blueberry powder, there was a significant enrichment (p = 0.049) in the relative abundance of Coriobacteriales incertae sedis, a taxonomic group of bacteria that facilitates the metabolism of dietary polyphenols. The placebo group exhibited significant reductions in total cholesterol, LDL-C, non-HDL-C, total LDL-P, large LDL-P, and ApoB, while the blueberry group exhibited significant reductions in total HDL-P and ApoA-I after 12 weeks compared to baseline. Conclusions: Generous blueberry consumption may upregulate the ability of the older human gut to utilize dietary polyphenols by altering the fecal microbiome. Longer, larger-scale studies with blueberries or blueberry powder are needed to observe improvements in cardiometabolic risk factors in older adults with overweight or obesity.
Collapse
Affiliation(s)
- Kathryn N. Porter Starr
- Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27710, USA; (J.W.); (R.N.); (M.G.M.)
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; (K.M.H.); (W.E.K.); (C.W.B.)
- Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC 27710, USA
| | | | - Jessica Wallis
- Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27710, USA; (J.W.); (R.N.); (M.G.M.)
| | - Rebecca North
- Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27710, USA; (J.W.); (R.N.); (M.G.M.)
| | - Qimin Zhang
- Labcorp, Westborough, MA 01581, USA; (Q.Z.); (K.S.); (H.N.B.); (L.K.I.)
| | - Kuncheng Song
- Labcorp, Westborough, MA 01581, USA; (Q.Z.); (K.S.); (H.N.B.); (L.K.I.)
| | | | - Hayden N. Brochu
- Labcorp, Westborough, MA 01581, USA; (Q.Z.); (K.S.); (H.N.B.); (L.K.I.)
| | | | | | - Marshall G. Miller
- Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27710, USA; (J.W.); (R.N.); (M.G.M.)
| | - Kim M. Huffman
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; (K.M.H.); (W.E.K.); (C.W.B.)
- Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - William E. Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; (K.M.H.); (W.E.K.); (C.W.B.)
- Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Connie W. Bales
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; (K.M.H.); (W.E.K.); (C.W.B.)
- Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC 27710, USA
| |
Collapse
|
49
|
Sujaya IN, Mariyatun M, Hasan PN, Manurung NEP, Pramesi PC, Juffrie M, Utami T, Cahyanto MN, Yamamoto S, Takahashi T, Asahara T, Akiyama T, Rahayu ES. Randomized study of Lacticaseibacillus fermented milk in Indonesian elderly houses: Impact on gut microbiota and gut environment. World J Gastroenterol 2025; 31:104081. [PMID: 40182598 PMCID: PMC11962840 DOI: 10.3748/wjg.v31.i12.104081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Health maintenance in elderly houses includes management of the gut microbiota and the environment. Lacticaseibacillus paracasei Shirota (LcS) is a probiotic strain that positively affects the human gut. However, the evidence of its effects on the Indonesian population remains limited. AIM To investigate the effect of LcS-fermented milk on the gut microbiota and environment of Indonesian elderly houses. METHODS This double-blind, randomized, placebo-controlled trial involved 112 participants from Indonesian elderly houses, spanning a 2-week baseline and 24-week treatment. Participants were randomly assigned to probiotic or placebo groups, consuming fermented milk with or without LcS (> 6.5 × 109 colony-forming units). Fecal samples were collected every three months. Gut microbiota analysis was performed using 16S rRNA gene sequencing and reverse transcription quantitative polymerase chain reaction, while gut environment was assessed by measuring fecal organic acids, amino acid metabolites, and stool frequency. RESULTS Analyses of 16S rRNA gene sequence data at the 3-month period revealed increased Bifidobacterium and Succinivibrio and decreased Rikenellaceae RC9 gut group in the probiotic group. These shifts were associated with significant differences in β-diversity metrics. The change in Bifidobacterium was confirmed by reverse transcription quantitative polymerase chain reaction, demonstrating higher abundance in the probiotic group than in the placebo group (8.5 ± 1.1 vs 8.0 ± 1.1, log10 bacterial cells/g; P = 0.044). At 6-month period, the differences in Succinivibrio and Rikenellaceae RC9 gut group persisted. The probiotic group showed higher butyrate levels than the placebo group at the 6-month period (5.04 ± 3.11 vs 3.95 ± 2.89, μmol/g; P = 0.048). The effect on amino acid metabolites and stool frequency was not significant. CONCLUSION Daily intake of LcS positively affects the gut microbiota and environment of people living in Indonesian elderly houses.
Collapse
Affiliation(s)
- I Nengah Sujaya
- School of Public Health, Faculty of Medicine, Udayana University, Denpasar 80230, Bali, Indonesia
| | - Mariyatun Mariyatun
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Pratama Nur Hasan
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Nancy Eka Putri Manurung
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Putrika Citta Pramesi
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Mohammad Juffrie
- Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Tyas Utami
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Muhammad Nur Cahyanto
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| | - Shuta Yamamoto
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Takahashi
- Yakult Honsha European Research Center for Microbiology VOF, Ghent 9052, East Flanders, Belgium
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Takuya Akiyama
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi 186-8650, Tōkyō, Japan
| | - Endang Sutriswati Rahayu
- Center for Food and Nutrition Studies, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Center of Excellence for Research and Application on Integrated Probiotics Industry, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Sleman 55281, Daerah Istimewa Yogyakarta, Indonesia
| |
Collapse
|
50
|
Xu T, Fang D, Xu T, Tao X, Wang Z, Liu Y. Exercise-driven gut microbiota alterations enhance colonization resistance against methicillin-resistant Staphylococcus aureus. Cell Rep 2025; 44:115424. [PMID: 40080501 DOI: 10.1016/j.celrep.2025.115424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Gut microbiota plays a crucial role in resisting the invasion of pathogens, particularly multidrug-resistant (MDR) bacteria, which pose a significant threat to public health. While exercise offers numerous health benefits, its impact on host colonization resistance remains largely unclear. In this study, we demonstrate that moderate exercise significantly reduces gut colonization by methicillin-resistant Staphylococcus aureus (MRSA), a clinically important MDR pathogen. Moreover, we identify an understudied strain of the intestinal probiotic Dubosiella newyorkensis (L8) as a critical factor in mediating exercise-induced colonization resistance against MRSA. Mechanistically, L8 enhances the deprivation of fucose, a crucial carbon source essential for MRSA growth and pathogenicity. This process relies on the high binding affinity of pyruvate to the ILE257 site of the lactate dehydrogenase in L8. Overall, our work highlights the importance of moderate exercise in maintaining host colonization resistance and demonstrates L8 as a probiotic in protecting against MRSA colonization.
Collapse
Affiliation(s)
- Tingting Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Dan Fang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiuying Tao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|