1
|
Minty M, Germain A, Sun J, Kaglan G, Servant F, Lelouvier B, Misselis E, Neagoe RM, Rossella M, Cardellini M, Burcelin R, Federici M, Fernandez-Real JM, Blasco-Baque V. Identifying the location-dependent adipose tissue bacterial DNA signatures in obese patients that predict body weight loss. Gut Microbes 2025; 17:2439105. [PMID: 39714075 DOI: 10.1080/19490976.2024.2439105] [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: 02/01/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Recent sets of evidence have described profiles of 16S rDNA sequences in host tissues, notably in fat pads that are significantly overrepresented and can serve as signatures of metabolic disease. However, these recent and original observations need to be further detailed and functionally defined. Here, using state-of-the-art targeted DNA sequencing and discriminant predictive approaches, we describe, from the longitudinal FLORINASH cohort of patients who underwent bariatric surgery, visceral, and subcutaneous fat pad-specific bacterial 16SrRNA signatures. The corresponding Porphyromonadaceae, Campylobacteraceae, Prevotellaceae, Actimomycetaceae, Veillonellaceae, Anaerivoracaceae, Fusobacteriaceae, and the Clostridium family XI 16SrRNA DNA segment profiles are signatures of the subcutaneous adipose depot while Pseudomonadaceae and Micrococcacecae, 16SrRNA DNA sequence profiles characterize the visceral adipose depot. In addition, we have further identified that a specific pre-bariatric surgery adipose tissue bacterial DNA signature predicts the efficacy of body weight loss in obese patients 5-10 years after the surgery. 16SrRNA signatures discriminate (ROC ~ 1) the patients who did not maintain bodyweight loss and those who did. Second, from the 16SrRNA sequences we infer potential pathways suggestive of catabolic biochemical activities that could be signatures of subcutaneous adipose depots that predict body weight loss.
Collapse
Affiliation(s)
- Matthieu Minty
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Alberic Germain
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Jiuwen Sun
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Gracia Kaglan
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | | | | | - Emiri Misselis
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Radu Mircea Neagoe
- Science and Technology "George Emil Palade" Tîrgu Mures, Second Department of Surgery, Emergency Mureş County Hospital, University of Medicine Pharmacy, Târgu Mureș, Romania
| | - Menghini Rossella
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Marina Cardellini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - José Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, University Hospital of Girona 'Dr Josep Trueta'
- Institut d'Investigacio Biomedica de Girona IdibGi, CIBER Fisiopatologia de la Obesidad y Nutricion, Girona, Spain
| | - Vincent Blasco-Baque
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| |
Collapse
|
2
|
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
|
3
|
Driuchina A, Isola V, Hulmi JJ, Salmi VM, Hintikka J, Ahtiainen JP, Pekkala S. Unveiling the impact of competition weight loss on gut microbiota: alterations in diversity, composition, and predicted metabolic functions. J Int Soc Sports Nutr 2025; 22:2474561. [PMID: 40033182 PMCID: PMC11881659 DOI: 10.1080/15502783.2025.2474561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Competitive sports and sports nutrition, popular among amateur athletes aiming for a lean physique, have limited research on gut microbiota. METHODS We conducted a 46-week study to analyze the consequences of fat loss and diet restrictions in 23 fitness athletes who prepared for a physique competition. Body composition, dietary intakes, serum cytokines and chemokines, and fecal samples were analyzed. RESULTS Fat loss through caloric restriction and aerobic exercise led to an increased phylogenetic diversity of gut microbiota and changes in the composition of gut microbiota, with Faecalibacterium, Lachnospiraceae, Bacteroides, and Intestinimonas showing altered abundances. Fat loss also changed the predicted microbial functions responsible for the metabolism of carbohydrates and amino acids. Consumption of energy, carbohydrates, fiber, vitamins and minerals, and various fatty acids decreased during the preparation for the competition, which was partly associated with changes in gut microbiota. Several cytokine levels decreased (IL1a, IL1b, IL10, and TFNα), and certain chemokine levels increased (GROa and RANTES). During the 23-week regain period after the competition, gut microbiota showed signs of recovery, with increased diversity compared to pre- and post-competition measurements. Most taxonomic changes returned to their baseline levels after the regain period. CONCLUSIONS The study highlights the dynamic nature of gut microbiota and its response to fat loss and regain in non-obese fitness/physique competitors and provides novel insights into how competitive sports and sports nutrition can influence the gut ecosystem.
Collapse
Affiliation(s)
- Anastasiia Driuchina
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Ville Isola
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha J Hulmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Vera M Salmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Jukka Hintikka
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha P Ahtiainen
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Satu Pekkala
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
- Turku University Hospital, Department of Clinical Microbiology, Turku, Finland
| |
Collapse
|
4
|
Aboulalazm FA, Kazen AB, deLeon O, Müller S, Saravia FL, Lozada-Fernandez V, Hadiono MA, Keyes RF, Smith BC, Kellogg SL, Grobe JL, Kindel TL, Kirby JR. Reutericyclin, a specialized metabolite of Limosilactobacillus reuteri, mitigates risperidone-induced weight gain in mice. Gut Microbes 2025; 17:2477819. [PMID: 40190120 PMCID: PMC11980487 DOI: 10.1080/19490976.2025.2477819] [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: 04/26/2024] [Revised: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including Limosilactobacillus reuteri as a candidate for further study. Oral supplementation with L. reuteri protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and L. reuteri restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by L. reuteri as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.
Collapse
Affiliation(s)
- Fatima A. Aboulalazm
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexis B. Kazen
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Orlando deLeon
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susanne Müller
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fatima L. Saravia
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Matthew A. Hadiono
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert F. Keyes
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian C. Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephanie L. Kellogg
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tammy L. Kindel
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John R. Kirby
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
5
|
Martinez-Medina JN, Ghazisaeedi F, Kramer C, Ziegler JF, McParland V, Mönch PW, Siegmund B, Jarquín-Díaz VH, Fulde M, Forslund-Startceva SK. Mucosal washes are useful for sampling intestinal mucus-associated microbiota despite low biomass. Gut Microbes 2025; 17:2464296. [PMID: 39980334 PMCID: PMC11849919 DOI: 10.1080/19490976.2025.2464296] [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: 12/26/2023] [Revised: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Understanding the dynamic relationship between mucus-associated microbiota and host health is critical. However, studies predominantly using stool samples may not accurately represent these bacterial communities. Here, we investigated the mucus-associated microbiota in the gastrointestinal tract of mice and the terminal ileum of humans using different sample types: mucosal washes, brushes, scrapings, and intestinal contents in mice and biopsies, brushes and mucosal washes in humans. We used DNA quantification and 16S rRNA amplicon sequencing to evaluate the comparability of the information yielded from the different sample types under a controlled benchmark. In mice, mucosal washes and brushes had comparative bacterial DNA and host DNA contamination than scraping samples. Similarly, in humans, washes outperformed biopsies in bacterial DNA content. Read counts and microbiota alpha diversity remained remarkably similar in mice and between brushes and washes in humans. The composition of the microbiota varied based on the subsegment and sample type in mice and sample type in humans. We conclude that washes and brushes reduce host contamination without inducing substantial compositional bias when sampling mucosal microbiota. Our findings suggest that mucosal washes and brushes are a viable alternative to biopsies in humans and scrapings in mice, thereby improving the transferability of results across hosts. Our study highlights the importance of focusing on mucus-associated microbiota to better capture host-microbiome interactions at their closer interface.
Collapse
Affiliation(s)
- Jennifer N. Martinez-Medina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Catharina Kramer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Jörn F Ziegler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Victoria McParland
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Paul W. Mönch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Víctor Hugo Jarquín-Díaz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Sofia K. Forslund-Startceva
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
6
|
Du W, Zou ZP, Ye BC, Zhou Y. Gut microbiota and associated metabolites: key players in high-fat diet-induced chronic diseases. Gut Microbes 2025; 17:2494703. [PMID: 40260760 PMCID: PMC12026090 DOI: 10.1080/19490976.2025.2494703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/26/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Excessive intake of dietary fats is strongly associated with an increased risk of various chronic diseases, such as obesity, diabetes, hepatic metabolic disorders, cardiovascular disease, chronic intestinal inflammation, and certain cancers. A significant portion of the adverse effects of high-fat diet on disease risk is mediated through modifications in the gut microbiota. Specifically, high-fat diets are linked to reduced microbial diversity, an overgrowth of gram-negative bacteria, an elevated Firmicutes-to-Bacteroidetes ratio, and alterations at various taxonomic levels. These microbial alterations influence the intestinal metabolism of small molecules, which subsequently increases intestinal permeability, exacerbates inflammatory responses, disrupts metabolic functions, and impairs signal transduction pathways in the host. Consequently, diet-induced changes in the gut microbiota play a crucial role in the initiation and progression of chronic diseases. This review explores the relationship between high-fat diets and gut microbiota, highlighting their roles and underlying mechanisms in the development of chronic metabolic diseases. Additionally, we propose probiotic interventions may serve as a promising adjunctive therapy to counteract the negative effects of high-fat diet-induced alterations in gut microbiota composition.
Collapse
Affiliation(s)
- Wei Du
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhen-Ping Zou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Bang-Ce Ye
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ying Zhou
- Laboratory of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
7
|
Liu J, Chen Y, Sun B, Xu D, Wang J, Sun Z, Liu P, Jing F, Song Y, Xia B. Toxicological effects of micro/nanoplastics and benzo[a]pyrene on cellular and molecular responses of Apostichopus japonicus (Selenka, 1867) during intestinal regeneration. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138003. [PMID: 40120257 DOI: 10.1016/j.jhazmat.2025.138003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Micro(nano)plastics (M/NPs) are pervasive in marine environments. Benzo[a]pyrene (B[a]P), a typical polycyclic aromatic hydrocarbon (PAH), possesses teratogenic, mutagenic, and carcinogenic properties. B[a]P can accumulate on M/NPs, altering their toxicity. This study investigated individual and combined effects of M/NPs and B[a]P on the intestinal regeneration of the benthic invertebrate Apostichopus japonicus. Eviscerated sea cucumbers were exposed to 0.1 mg L-1 M/NPs (80 nm [NP80] or 20 μm [MP20]) and/or 0.03 μg L-1 B[a]P for 28 days. Cell proliferation, antioxidant and immunoenzyme activity, gene expression, and microbial community in the regenerated intestine were assessed. It demonstrated that combined exposure prolonged regeneration process, leading to increased oxidative stress and intestinal damage. Differential gene expression analysis revealed that co-exposure and single NP80 exposure both significantly changed translation-related processes, while single MP20 exposure primarily affected lipid metabolism. All treatments significantly altered the intestinal microbiota. Under the MP20+B[a]P treatment, Ralstonia abundance significantly increased, while Cobetia and Paracoccus abundances decreased. In general, co-exposure exerted more detrimental effects on intestinal regeneration than any single exposure, with MP20+B[a]P demonstrating more severe impacts. This study provides novel insights into the biotoxicity of M/NPs and B[a]P, contributing to better understanding of the detriments of microplastics and PAHs on marine benthic invertebrates.
Collapse
Affiliation(s)
- Ji Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Yanru Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Baiqin Sun
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Dongxue Xu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Jinye Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Zhenlong Sun
- Jiangsu Zhongyang Group Co., Ltd., Nantong, Jiangsu Province 226600, China.
| | - Peng Liu
- Shandong Fisheries Development and Resources Conservation Center, Jinan, Shandong Province 250013, China
| | - Futao Jing
- Shandong Fisheries Development and Resources Conservation Center, Jinan, Shandong Province 250013, China
| | - Yize Song
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Bin Xia
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China.
| |
Collapse
|
8
|
Schoonakker MP, van Peet PG, van den Burg EL, Numans ME, Ducarmon QR, Pijl H, Wiese M. Impact of dietary carbohydrate, fat or protein restriction on the human gut microbiome: a systematic review. Nutr Res Rev 2025; 38:238-255. [PMID: 38602133 DOI: 10.1017/s0954422424000131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Restriction of dietary carbohydrates, fat and/or protein is often used to reduce body weight and/or treat (metabolic) diseases. Since diet is a key modulator of the human gut microbiome, which plays an important role in health and disease, this review aims to provide an overview of current knowledge of the effects of macronutrient-restricted diets on gut microbial composition and metabolites. A structured search strategy was performed in several databases. After screening for inclusion and exclusion criteria, thirty-six articles could be included. Data are included in the results only when supported by at least three independent studies to enhance the reliability of our conclusions. Low-carbohydrate (<30 energy%) diets tended to induce a decrease in the relative abundance of several health-promoting bacteria, including Bifidobacterium, as well as a reduction in short-chain fatty acid (SCFA) levels in faeces. In contrast, low-fat diets (<30 energy%) increased alpha diversity, faecal SCFA levels and abundance of some beneficial bacteria, including Faecalibacterium prausnitzii. There were insufficient data to draw conclusions concerning the effects of low-protein (<10 energy%) diets on gut microbiota. Although the data of included studies unveil possible benefits of low-fat and potential drawbacks of low-carbohydrate diets for human gut microbiota, the diversity in study designs made it difficult to draw firm conclusions. Using a more uniform methodology in design, sample processing and sharing raw sequence data could foster our understanding of the effects of macronutrient restriction on gut microbiota composition and metabolic dynamics relevant to health. This systematic review was registered at https://www.crd.york.ac.uk/prospero as CRD42020156929.
Collapse
Affiliation(s)
- Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Maria Wiese
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
9
|
Luo F, Yang J, Song Z, Zhao Y, Wang P, Liu K, Mou X, Liu W, Li W. Renshen Zhuye decoction ameliorates high-fat diet-induced obesity and insulin resistance by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156655. [PMID: 40120542 DOI: 10.1016/j.phymed.2025.156655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Obesity, characterized by excessive adipose tissue accumulation, has become a global health challenge with rapidly increasing prevalence. It contributes significantly to metabolic disorders including insulin resistance (IR). Renshen-zhuye decoction (RZD), a traditional Chinese medicine formula historically used for diabetes, shows potential for improving metabolic parameters, but its effects and mechanisms in obesity and insulin resistance remain unclear. PURPOSE This study aimed to evaluate the therapeutic benefits of RZD on obesity and insulin resistance, and to elucidate the underlying mechanisms through which it improves glucose and lipid metabolism. METHODS The role of RZD was evaluated in a high-fat diet (HFD) mouse model. The formula was characterized using UPLC-MS. Comprehensive analyses including histopathological staining, immunofluorescence, biochemical assays, 16S rRNA gene sequencing of gut microbiota, and non-targeted metabolomic analysis were performed. To validate the role of gut microbiota, we employed antibiotic treatment (ABX) to deplete intestinal flora and conducted fecal microbiota transplantation (FMT) experiments. RESULTS RZD treatment dose-dependently alleviated HFD-induced dyslipidemia and insulin resistance, improving glucose tolerance, insulin sensitivity, and energy expenditure. Gut microbiota analysis revealed that RZD significantly modulated the composition of intestinal flora and their metabolic profiles. Additionally, RZD reduced intestinal and systemic inflammation by enhancing intestinal barrier integrity, particularly through increased expression of tight junction proteins such as Occludin. Importantly, the beneficial effects of RZD on weight management and glucose homeostasis were antagonized by antibiotic intervention, while FMT experiments confirmed that these improvements were mediated through gut microbiota modulation. CONCLUSION This study provides new insights into RZD's modulatory effects on gut microbiota and subsequent improvements in obesity-related metabolic parameters. RZD alleviates HFD-induced obesity and insulin resistance in mice by modulating gut microbiota composition and function, which subsequently improves intestinal barrier integrity, reduces inflammation, and enhances metabolic homeostasis.
Collapse
Affiliation(s)
- Fei Luo
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jie Yang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiping Song
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yuan Zhao
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Panpan Wang
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou 310000, PR China
| | - Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China
| | - Xin Mou
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China.
| | - Wenhong Liu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Wei Li
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
10
|
Kahe K, Laferrère B, Castellanos FX, Zhang Y, Mozaffarian D. Monosodium glutamate: A hidden risk factor for obesity? Obes Rev 2025; 26:e13903. [PMID: 39914377 DOI: 10.1111/obr.13903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/18/2024] [Accepted: 11/17/2024] [Indexed: 05/14/2025]
Abstract
Monosodium glutamate (MSG) has become one of the most widely used food additives in the global food supply. Although it has been classified for decades as a food ingredient that is generally recognized as safe, concerns about the health impacts of chronic MSG use, especially its potential effect on weight, are still ongoing. This comprehensive review summarizes the available human and animal evidence, highlighting potential mechanisms linking MSG use to weight gain or obesity, and discusses challenges and future research directions. Because of MSG intake worldwide as well as hidden MSG in food labeling, there is a pressing need for a mechanistic understanding of the health impacts of MSG use especially on weight. To generate robust scientific evidence and to clarify public concerns, rigorous mechanistic studies and randomized controlled clinical trials are warranted.
Collapse
Affiliation(s)
- Ka Kahe
- Department of Obstetrics and Gynecology and Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Blandine Laferrère
- Division of Endocrinology, New York Nutrition Obesity Research Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Francisco X Castellanos
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Yijia Zhang
- Department of Obstetrics and Gynecology and Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
11
|
Wang L, He X, Zhang Z, Chen N. Distinct gut microbiota signatures in older people with sarcopenic obesity and sarcopenia without obesity. Clin Nutr 2025; 49:77-89. [PMID: 40252601 DOI: 10.1016/j.clnu.2025.04.004] [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: 06/11/2024] [Revised: 03/15/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Previous evidence suggests that gut dysbiosis plays an important role in the development and progression of sarcopenia and sarcopenic obesity (SO), but evidence supporting this association is lacking. Thus, this study aimed to investigate the characteristics of gut microbiota in older people with sarcopenia and SO. METHODS A total of 1558 older adults (age ≥65 years) from a community-based cohort in Shanghai, China, underwent sarcopenia screening using the SARC-F questionnaire, with 351 participants completing further assessment. On the basis of the Asian Working Group for Sarcopenia 2019 and the World Health Organization obesity criteria, 60 participants were categorized into three groups: SO (n = 20), sarcopenia without obesity (Sar, n = 18), and controls (Con, n = 22). Gut microbiota composition was analyzed using 16S rRNA sequencing (V3-V4 regions). RESULTS Significant differences in the diversity and composition of the gut microbiota were observed in the Sar and SO groups. A reduction in alpha diversity (Chao1 and ACE indices) was found in the SO group. Beta diversity based on unweighted Unifrac PCoA was significantly different among the three groups. LEfSe analysis identified 39 taxa with significant differential abundances across groups. The Sar group exhibited enrichment of Christensenellaceae_R-7_group, Alistipes, Ruminococcus, Odoribacter, Prevotellaceae_UCG-001, Hungatella, Family_XIII_AD3011_group, Anaerotruncus, Ruminiclostridium, and Oxalobacter, along with their high taxonomic classifications. Meanwhile, Enterobacteriaceae, Allisonella, and Peptoclostridium were enriched in the SO group. Feature selection via Boruta algorithm identified five and four discriminatory taxa to construct random forest models, effectively distinguishing individuals with Sar and SO from Con. Key predictors for Sar included reduced Enterococcus, Enterobacter, and Hungatella and increased Odoribacter and Christensenellaceae_R-7_group. Conversely, SO was characterized by decreased Enterobacter, Alloprevotella, and Enterococcus and increased Allisonella. Five-fold cross-validation confirmed robust diagnostic efficacy, achieving AUCs of 0.860 (95 % CI: 0.786-0.996) for Sar and 0.826 (95 % CI: 0.735-0.970) for SO. CONCLUSION This study demonstrated that the gut microbiota of SO and Sar have distinct diversity and composition profiles. The results provide new insights into the role of gut microbiota in SO, highlighting its potential as a therapeutic target in this condition.
Collapse
Affiliation(s)
- Ling Wang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China; School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiangfeng He
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhen Zhang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Nan Chen
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China; Department of Rehabilitation, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Budin M, Sandiford NA, Gehrke T, Citak M. Body mass index matters: morbid obese patients have different microorganism profiles in the setting of periprosthetic hip joint infections. INTERNATIONAL ORTHOPAEDICS 2025; 49:1309-1317. [PMID: 40183945 DOI: 10.1007/s00264-025-06513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE This study investigated the relationship between BMI and microorganism profiles, with a particular focus on gut microorganisms in patients with PJI following total hip arthroplasty (THA). It also explored comorbidities, that may contribute to these variations. METHODS This study included all patients treated at our institution for a PJI of a THA between 1996 and 2021. Patients were categorized into four distinct BMI groups: <30; 30-34.9; 35-39.9; ≥ 40. Bivariate and logistic regression analysis were conducted, with presentation of odds ratio (OR) and 95% confidence interval (CI). RESULTS A total of 3645 hip PJI cases were recruited for the final analysis. Patients with a BMI ≥ 40 had approximately a ten fold higher risk for Streptococcus dysgalactiae (p < 0.001; OR = 9.92; 95% CI 3.87-25.44) and a seven fold higher risk for Proteus mirabilis (p < 0.001; OR = 7.43; 95% CI 3.13-17.67) and Klebsiella pneumoniae (p < 0.001; OR = 6.9; 95% CI 2.47-19.31). Furthermore, polymicrobial infections (p < 0.001; OR = 2.17; 95% CI 1.50-3.15) were found to be significantly more prevalent in patients with a BMI ≥ 40. CONCLUSION Obese patients (BMI ≥ 30) displayed a distinct microorganism profile in hip PJIs, mainly dominated by Firmicutes and Proteobacteria. Comorbidities such as diabetes, hypertension, and hyperlipidaemia may contribute to a leaky gut syndrome, increasing PJI risk caused by gut microorganisms. Optimizing comorbidities may help reduce the risk of hip PJI. Further research is needed to clarify the relationship between obesity, gut microbiome alterations and hip PJI development.
Collapse
|
13
|
Liu P, Ma S, Chen J, Duan C, Wang L, Chen D, Lv S, Li Y, Yan X. Fermented sheep milk supplemented with Lactobacillus rhamnosus NM-94: Enhancing fermented milk quality and enriching microbial community in mice. J Dairy Sci 2025; 108:5530-5542. [PMID: 40254162 DOI: 10.3168/jds.2025-26460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/22/2025] [Indexed: 04/22/2025]
Abstract
Currently, many research efforts and product developments focus on goat milk, while studies on the development and probiotic function of sheep milk remain limited. In this study, the effects of Lactobacillus rhamnosus NM-94 on the physicochemical properties, viable count, texture, microrheology, flavor, and quality detection and intestinal flora of fermented milk were analyzed by adding or not adding L. rhamnosus NM-94 to 2 starter cultures. The results showed that the addition of L. rhamnosus NM-94 to sheep milk significantly reduced fermentation time by 2 h (12 h vs. 14 h). At 21 d of storage, the count of viable bacteria in fermented sheep milk supplemented with L. rhamnosus NM-94 (0.61 ± 0.06 × 109 cfu/mL) was surprisingly higher than that of the control milk (0.35 ± 0.03 × 109 cfu/mL). During the microrheological analysis of the fermentation stage, the elastic index, macroscopic viscosity index, and solid-liquid balance values of fermented sheep milk supplemented with L. rhamnosus NM-94 were greater than those of the control milk, indicating that the fermented sheep milk supplemented with L. rhamnosus NM-94 had a more stable gel structure and viscosity. In mice, fermented sheep milk supplemented with L. rhamnosus NM-94 has a variety of beneficial intestinal microorganisms and potential probiotic functions. This study provides new ideas for improving the function of sheep milk and expanding the sheep milk industry.
Collapse
Affiliation(s)
- Pufang Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shaoying Ma
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jia Chen
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Chao Duan
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lixing Wang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Dan Chen
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Siying Lv
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yuanzhu Li
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xinlei Yan
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China.
| |
Collapse
|
14
|
Kumar M, Muthurayar T, Karthika S, Gayathri S, Varalakshmi P, Ashokkumar B. Anti-Diabetic Potentials of Lactobacillus Strains by Modulating Gut Microbiota Structure and β-Cells Regeneration in the Pancreatic Islets of Alloxan-Induced Diabetic Rats. Probiotics Antimicrob Proteins 2025; 17:1096-1116. [PMID: 38329697 DOI: 10.1007/s12602-024-10221-7] [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] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Diabetes mellitus, a most common endocrine disorder of glucose metabolism, has become a global epidemic and poses a serious public health threat with an increased socio-economic burden. Escalating incidence of diabetes is correlated with changes in lifestyle and food habits that cause gut microbiome dysbiosis and β-cells damage, which can be addressed with dietary interventions containing probiotics. Hence, the search for probiotics of human origin with anti-diabetic, anti-AGE, and anti-ACE potentials has gained renewed interest for the effective management of diabetes and its associated complications. The present study used an alloxan (AXN)-induced diabetic rat model to investigate the effects of potential probiotic Lacticaseibacillus casei MKU1, Lactiplantibacillus pentosus MKU3, and Lactiplantibacillus plantarum MKU7 administration individually on physiochemical parameters related to diabetic pathogenesis. Experimental animals were randomly allotted into six groups viz. NCG (control), DCG (AXN), DGM (metformin), DGP1 (MKU1), DGP2 (MKU3), and DGP3 (MKU7), and biochemical data like serum glucose, insulin, AngII, ACE, HbA1c, and TNF-α levels were measured until 90 days. Our results suggest that oral administration with MKU1, MKU3, or MKU7 significantly improved serum insulin levels, glycemic control, glucose tolerance, and body weight. Additionally, β-cell mass was increased by preserving islet integrity in Lactobacillus-treated diabetic rats, whereas TNF-α (~40%), AngII (~30%), and ACE levels (~50%) were strongly inhibited and enhanced sIgA production (5.8 folds) abundantly. Furthermore, Lactobacillus administration positively influenced the gut microbiome with a significant increase in the abundance of Lactobacillus species and the beneficial Bacteroides uniformis and Bacteroides fragilis, while decreased the pathogenic Proteus vulgaris and Parabacteroides distasonis. Among the probiotic treatment groups, L. pentosus MKU3 performed greatly in almost all parameters, indicating its potential use for alleviating diabetes-associated complications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Tharmar Muthurayar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Sukumaran Karthika
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Santhalingam Gayathri
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Balasubramaniem Ashokkumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
15
|
Anvarbatcha R, Kunnathodi F, Arafat AA, Azmi S, Mustafa M, Ahmad I, Alotaibi HF. Harnessing Probiotics: Exploring the Role of the Gut Microbiome in Combating Obesity. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10605-3. [PMID: 40434504 DOI: 10.1007/s12602-025-10605-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Obesity has become a global health crisis driven by genetic, environmental, and lifestyle factors, often linked to gut microbiome imbalances. Probiotics, particularly Lactobacillus and Bifidobacterium strains, have shown promise in clinical trials by promoting weight loss, improving lipid profiles, and addressing gut dysbiosis associated with obesity. This review surveys the literature on the microbiome and obesity, emphasizing the clinical relevance of probiotics in treatment strategies. Our comprehensive PubMed search highlights the mechanisms through which probiotics influence metabolic health, including their effects on inflammation and appetite regulation. We also explore promising future research directions and the potential for integrating probiotics into clinical practice. While results are encouraging, the evidence is limited by strain variability, small sample sizes, short trial durations, and individual differences in microbiota composition. More extensive, long-term studies with standardized methods are crucial to confirm the effectiveness of probiotics as viable anti-obesity treatments.
Collapse
Affiliation(s)
- Riyasdeen Anvarbatcha
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Faisal Kunnathodi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Amr A Arafat
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
- Departments of Adult Cardiac Surgery Department, Kingdom of Saudi Arabia, Prince Sultan Cardiac Center, Riyadh, Kingdom of Saudi Arabia
| | - Sarfuddin Azmi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Mustafa
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Ishtiaque Ahmad
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Haifa F Alotaibi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia.
- Department of Family Medicine, Kingdom of Saudi Arabia, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
16
|
Wang H, Zhan J, Zhao S, Jiang H, Jia H, Pan Y, Zhong X, Huo J. Sex-induced alterations in rumen microbial communities and metabolite profiles: implications for lamb body weight. BMC Microbiol 2025; 25:328. [PMID: 40426040 PMCID: PMC12107992 DOI: 10.1186/s12866-025-04049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Microbiota-metabolome interactions play a crucial role in host physiological regulation and metabolic homeostasis. The aim of this study was to investigate that sex induces alterations in rumen microbial community composition and metabolite profiles in lambs and the influence on body weight. This study aimed to demonstrate that sex- induced alterations in rumen microbial community and metabolite profiles and blood indices and their linkage to growth performance in lambs. RESULTS This study examined (growth indices, serum indices, rumen fermentation parameters, rumen fluid microbiota community and metabolome profiles) in 180 Hu lambs (90 males, and 90 females) with the same age and diet. At six months, male lambs showed significantly greater body weight, serum indices (glutamic pyruvic transaminase, glutamic oxalacetic transaminase, growth hormone, glucagon-like peptide 1, and ghrelin), and molar percentage of propionic acid, isobutyric acid, butyric acid, isovaleric acid and valeric acid compared to female. However, male had lower VFA molar concentrations (acetic acid, propionic acid, butyric acid, and TVFAs), acetic acid/propionic acid, and VFA molar percentage (acetic acid) than female. Significant sex-related differences were observed in rumen microbiota and metabolic enrichment between genders. Moreover, compared with the females lambs, the relative abundance of Succiniclasticum, uncultured_rumen_bacterium, NK4 A214_group, Veillonellaceae_UCG_001 and Butyrivibrio in the male lambs has been significantly increased, while the relative abundance of Prevotella has been significantly decreased (P < 0.05). Notably, there were significant rumen microbiota-metabolite interactions, especially Firmicutes and Bacteroidota as dominant phyla in the sheep rumen with significant differences in correlation with rumen metabolic modules. Additionally, there are pronounced correlations among the microbiota, particularly within the Firmicutes phylum. Furthermore, the up-regulated metabolites in the rumen fluid of male lambs were predominantly enriched in the amino acid metabolite pathway, and these metabolites exhibited a significant positive correlation with body weight. However, the metabolites that were up-regulated in ewe lambs were predominantly enriched in the lipid metabolic pathway, and these metabolites exhibited a significant negative correlation with body weight. Moreover, lamb rumen microbial markers (Lachnospiraceae_UCG_008, Saccharofermentans, unclassified_Clostridia, Christensenellaceae_R_7_group, Anaerovorax, Mogibacterium, and unclassified_Erysipelotrichaceae) and metabolic markers (C75, 4-Coumarate, Flibanserin,3-Amino-5-mercapto-1,2,4-triazole, 1,3-Propane sultone, Fingolimod phosphate ester, S-,) were significantly positively correlated with body weight, but lamb rumen microbial markers (Anaeroplasma, unclassified_Acholeplasmataceae, uncultured_rumen_bacterum_4c28 d_15) and metabolic markers (Mozenavir, Reduced riboflavin, PG(18:2(9Z,12Z)/0:0), Cowanin) were significantly negatively correlated body weight. CONCLUSIONS This study shows that sex-induced alterations in rumen microbial communities and metabolite profiles, adapting to the growth and development of lambs. The findings may help develop targeted strategies to optimize sheep rumen microbiota and improve productivity.
Collapse
Affiliation(s)
- Haibo Wang
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- Provincial Development and Research Institute of Ruminants in Gansu, Lanzhou, 730070, China
| | - Jinshun Zhan
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Shengguo Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- Provincial Development and Research Institute of Ruminants in Gansu, Lanzhou, 730070, China
| | - Haoyun Jiang
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Haobin Jia
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Yue Pan
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300384, China
| | - Xiaojun Zhong
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China
| | - Junhong Huo
- Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China.
- Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Institute of Animal Husbandry and Veterinary, Jiangxi Academy of Agricultural Science, Nanchang, 330200, China.
| |
Collapse
|
17
|
Wang R, Ma F, Yin D, Wang H, Wei X. Intestinal Microbes, Metabolites, and Hormones in Alcohol-Associated Liver Disease. Semin Liver Dis 2025. [PMID: 40334703 DOI: 10.1055/a-2601-9480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Alcohol-associated liver disease (ALD)-encompassing conditions including steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma-refers to hepatic damage arising from excessive or hazardous alcohol consumption, and is now recognized as a significant global health burden. Although the mechanisms underlying ALD remain incompletely understood, several pathways have been substantiated over the last five decades, notably the involvement of intestinal microorganisms and the involvement of the gut-liver axis in alcohol metabolism and ALD pathogenesis. Ethanol intake disrupts the intestinal microbial balance and compromises the gut barrier, resulting in increased permeability to microbial products. The subsequent translocation of microbial metabolites and other antigenic substances to the liver activates hepatic immune responses, thereby contributing to liver injury. In addition, gastrointestinal hormones are also implicated in ALD progression through various mechanisms. Although no therapies for ALD have been approved by the Food and Drug Administration, various therapeutic strategies targeting the intestinal microbiota and gut barrier have been identified. In conclusion, this review discusses the role of the gut-liver axis in alcohol metabolism and ALD pathogenesis and explores the emerging therapeutic strategies.
Collapse
Affiliation(s)
- Ruimeng Wang
- Second Clinical Medical College, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Dou Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaohui Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
de Moraes Arnoso BJ, de Araújo CA, Ramundo GD, de Bem GF, Ognibene DT, Fontes-Dantas FL, Martins BC, Daleprane JB, de Souza MO, Resende AC, da Costa CA. Açaí seed extract mitigates intestinal and hypothalamic alterations in obese mice. Mol Cell Endocrinol 2025; 606:112574. [PMID: 40409530 DOI: 10.1016/j.mce.2025.112574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/16/2025] [Accepted: 05/18/2025] [Indexed: 05/25/2025]
Abstract
Obesity is a significant health concern, significantly contributing to increased morbidity and mortality by disrupting multiple physiological systems. It is strongly associated with metabolic dysfunctions, including impaired glycemic homeostasis, compromised intestinal barrier integrity, and gut microbiota imbalances, all exacerbating the risk of chronic diseases. The hydroalcoholic extract of açaí seeds (ASE), rich in phenolic compounds, has demonstrated beneficial effects on obesity and hyperglycemia; however, its impacts on gut health and gut-hypothalamus communication remain unclear. This study aimed to investigate the therapeutic effect of ASE in intestinal and hypothalamic alterations associated with obesity and compare it with Metformin. Male C57BL/6 mice were fed a high-fat or standard diet for 14 weeks. The ASE (300 mg/kg/day) and Metformin (300 mg/kg/day) treatments started in the tenth week until the fourteenth week, totaling four weeks of treatment. Our data show that the treatment with ASE and Metformin reduced body weight, ameliorated lipid profile, hyperglycemia, and plasma hyperleptinemia, and decreased the oxidative damage in the gut by reducing immunostaining of 8-isoprostane and NOX-4 expression, and improved the intestinal parameters and hypothalamic gene expression. Obesity-induced dysbiosis in the HF group was marked by reduced Proteobacteria and elevated LPS plasma levels, which were improved by treatments with ASE and Metformin. These findings suggest that ASE and Metformin are promising strategies to counteract the adverse effects of obesity on intestinal health and gut-hypothalamus communication, though they act through distinct mechanisms. Therefore, we can suggest that ASE is a promising natural product for treating the intestinal alterations associated with obesity.
Collapse
Affiliation(s)
| | - Caroline Alves de Araújo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Giovana Dias Ramundo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Fabricia Lima Fontes-Dantas
- Neurogenetics Laboratory, Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Bruna Cadete Martins
- Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
19
|
Tang J, Dong L, Tang M, Arif A, Zhang H, Zhang G, Zhang T, Xie K, Su S, Zhao Z, Dai G. Metagenomic Analysis Reveals the Characteristics of Cecal Microbiota in Chickens with Different Levels of Resistance During Recovery from Eimeria tenella Infection. Animals (Basel) 2025; 15:1500. [PMID: 40427376 PMCID: PMC12108197 DOI: 10.3390/ani15101500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2025] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Coccidiosis, caused by Eimeria protozoa, is a severe intestinal parasitic disease that results in substantial economic losses to the global poultry industry annually. The gut microbiota plays a crucial role in host health, metabolism, immune function, and nutrient absorption in chickens. Recent studies have focused on the effects of Eimeria tenella's (E. tenella) acute infection period on host health. However, recovery conditions, cecal microbiota composition, and functional differences in the ceca of chickens with varying resistance to E. tenella remain poorly understood during the recovery period after infection. This study aimed to compare growth performance, cecal histopathology, and the cecal microbiota characteristics in control (R_JC), resistant (R_JR), and susceptible (R_JS) chickens during recovery, using metagenomic sequencing. The results revealed significant differences in both cecal tissue structure and growth performance between the different groups during recovery. Although no significant differences were observed in microbial alpha diversity among the groups, sequencing analysis highlighted notable changes in microbial composition and abundance. Bacteroidetes, Firmicutes, and Proteobacteria were the predominant phyla in chicken cecal contents; however, Firmicutes abundance was lower in the R_JS group than in the R_JC and R_JR groups. Further analysis, combining linear discriminant analysis effect size (LEfSe) and differential heatmap analysis, identified Bacteroides_fluxus, Ruminococcus_flavefaciens, and Bacteroides_sp_CACC_737 as dominant microorganisms in the R_JR group (p < 0.05) compared to both the R_JC and R_JS groups. In contrast, Sutterella_sp_AM11-39, Bacteroides_sp_43_108, Mycobacterium, Mycoplasma_arginini, and Chlamydia dominated in the R_JS group, while Butyricimonas, Butyricimonas_sp_Marseille-P3923, and Flavonifractor_plautii were significantly reduced in the R_JS group (p < 0.05). Additionally, beneficial cecal microorganisms such as Flavonifractor_sp__An10, Pseudoflavonifractor, and Faecalicoccus were significantly decreased in both the R_JR and R_JS groups (p < 0.05) compared to the R_JC group. Predictive functional analysis using the KEGG and CAZy databases further indicated that the cecal microbiota in the R_JR group exhibited enhanced metabolism-related pathways, whereas these pathways were significantly diminished in the R_JS group, potentially influencing the recovery process from coccidial infection. These findings provide valuable insights into the cecal microbiota's role during recovery from E. tenella infection and deepen our understanding of the impact of coccidial infections on host health.
Collapse
Affiliation(s)
- Jianqiang Tang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Liyue Dong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Meihui Tang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Areej Arif
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Honghong Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Shijie Su
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| | - Zhenhua Zhao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou 225125, China
| | - Guojun Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225000, China; (J.T.)
| |
Collapse
|
20
|
Mugo CW, Church E, Horniblow RD, Mollan SP, Botfield H, Hill LJ, Sinclair AJ, Grech O. Unravelling the gut-brain connection: a systematic review of migraine and the gut microbiome. J Headache Pain 2025; 26:125. [PMID: 40399789 PMCID: PMC12096802 DOI: 10.1186/s10194-025-02039-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/16/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND There is substantial evidence linking migraines to gastrointestinal (GI) issues. Conditions such as irritable bowel syndrome and colitis often co-occur with migraines and GI symptoms are common among migraine patients. However, the evidence supporting the efficacy of gut microbiome-targeted therapies for managing migraines is limited. This systematic review aimed to describe the existing evidence of the gut microbiome in patients with migraine compared to healthy individuals. Additionally, it sought to examine how therapies targeting the gut microbiome including prebiotics, probiotics and synbiotics, might influence clinical outcomes. METHODS We performed searches on Embase, PubMed, and the Cochrane Library to identify studies in migraines and the gut microbiome, focusing on those which investigated the gut microbiome composition and gut microbiome-targeted therapies. Key data was extracted and analysed including study details, patient demographics, migraine type, comorbidities, and clinical outcomes. For gut microbiome composition studies, bacterial diversity and abundance was noted. For gut microbiome-targeted therapies studies, treatment types, dosages, and patient outcomes was recorded. RESULTS A significant difference between various genera of microbes was reported between migraine patients and controls in several studies. Bacteroidetes (also named Bacteroidota), proteobacteria, and firmicutes (also named Bacillota) phyla groups were found significantly abundant in migraine, while studies were conflicted in the abundance of Actinobacteria and Clostridia with regards to increased migraine risk in migraine patients. Patients with migraine had a gut microbiome with reduced species number and relative abundance, as well as a distinct bacterial composition compared to controls. Synbiotic and synbiotic/probiotic combination treatments have been shown in five randomised controlled trials and one open label pilot study to significantly decrease migraine severity, frequency, duration and painkiller consumption. CONCLUSIONS The significant alterations in microbial phyla observed in migraine patients suggest a potential microbial signature that may be associated with migraine risk or chronic progression. However, the mechanistic underpinnings of these associations remain unclear. This systemic review found that probiotic and synbiotic/probiotic combination therapies may be promising interventions for migraine management, offering significant reductions in migraine frequency and painkiller use. Future randomised controlled studies are needed to evaluate the optimal length of treatment and impact on patient related quality of life.
Collapse
Affiliation(s)
- Caroline W Mugo
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ella Church
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Richard D Horniblow
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Susan P Mollan
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Neuro-Ophthalmology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
| | - Hannah Botfield
- Inflammation and Ageing, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lisa J Hill
- Biomedical Sciences, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham, B15 2TT, UK
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK
| | - Alexandra J Sinclair
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2GW, UK.
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, B15 2TH, UK.
| | - Olivia Grech
- Metabolism and Systems Science, School of Medical Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
21
|
Kaisanlahti A, Turunen J, Hekkala J, Mishra S, Karikka S, Amatya SB, Paalanne N, Kruger J, Portaankorva AM, Koivunen J, Jukkola A, Vihinen P, Auvinen P, Leppä S, Karihtala P, Koivukangas V, Hukkanen J, Vainio S, Samoylenko A, Bart G, Lahti L, Reunanen J, Tejesvi MV, Ruuska-Loewald T. Gut microbiota-derived extracellular vesicles form a distinct entity from gut microbiota. mSystems 2025; 10:e0031125. [PMID: 40298395 PMCID: PMC12090791 DOI: 10.1128/msystems.00311-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Extracellular vesicles (EVs), nanoparticles secreted by both gram-negative and gram-positive bacteria, carry various biomolecules and cross biological barriers. Gut microbiota-derived EVs are currently being investigated as a communication mechanism between the microbiota and the host. Few clinical studies, however, have investigated gut microbiota-derived EVs. Here, we show that machine learning models were able to accurately distinguish gut microbiota and respective microbiota-derived EV samples according to their taxonomic composition both within each data set (area under the curve [AUC] 0.764-1.00) and in a cross-study setting (AUC 0.701-0.997). These results show that gut microbiota-derived EVs form a distinct taxonomic entity from gut microbiota. Thus, conventional gut microbiota composition may not correctly reflect communication between the gut microbiota and the host unless microbiota-derived EVs are reported separately.IMPORTANCEGut microbiota-derived extracellular vesicles (EVs) have been suggested to be a communication mechanism between the gut microbiota and the human body. However, the data on EV secretion from the gut microbiota remain limited. To investigate and compare the composition of gut microbiota-derived EVs to gut microbiota composition, we used a machine learning approach to classify 16S rRNA gene sequencing data in seven clinical data sets incorporating both gut microbiota and gut microbiota-derived EV samples. The results of the study show that microbiota-derived EVs form a separate taxonomic entity from the gut microbiota. Gut microbiota-derived EVs should be included in clinical studies that investigate gut microbiota to gain more comprehensive insight into gut microbiota-host communication.
Collapse
Affiliation(s)
- Anna Kaisanlahti
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Jenni Turunen
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Jenni Hekkala
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Surbhi Mishra
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Sonja Karikka
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
- Disease Networks Research Unit, University of Oulu, Oulu, Finland
| | - Sajeen Bahadur Amatya
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | - Niko Paalanne
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Johanna Kruger
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Anne M. Portaankorva
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Jussi Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Arja Jukkola
- Tampere Cancer Center, Tampere University, Tampere, Finland
| | - Pia Vihinen
- FICAN West Cancer Centre, Turku University Hospital, University of Turku, Turku, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, The Wellbeing services county of North Savo, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, and iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Peeter Karihtala
- Department of Medical Oncology and Radiotherapy and Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland
| | - Vesa Koivukangas
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Janne Hukkanen
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu, Oulu University Hospital, Oulu, Finland
| | - Seppo Vainio
- Disease Networks Research Unit, University of Oulu, Oulu, Finland
- Kvantum Institute, University of Oulu, Oulu, Finland
| | | | - Genevieve Bart
- Disease Networks Research Unit, University of Oulu, Oulu, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Justus Reunanen
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
| | | | - Terhi Ruuska-Loewald
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
22
|
Tian X, Li L, Chen J, Guo X. Incidence, risk factors, and prognosis of constipation in acute ischemic stroke: A prospective, observational study. J Clin Neurosci 2025; 137:111337. [PMID: 40398175 DOI: 10.1016/j.jocn.2025.111337] [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: 01/18/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Constipation is common in patients with acute ischemic stroke, yet its incidence and prognosis remain unknown. Herein, we investigate the incidence of new-onset constipation and its risk factors and relationship with stroke severity and prognosis. METHODS This prospective study enrolled 358 patients diagnosed with acute ischemic stroke. The Constipation Scoring System was used to assess new-onset constipation during hospitalization. Demographics, medical histories, clinical data, laboratory parameters, and medication use were compared between non-constipation and constipation groups. Univariate and multivariate logistic regression and multiple linear regression were used to explore potential influencing factors and prognosis. RESULTS The cumulative incidence of new-onset constipation during was 41.6 %. Its occurrence was closely associated with multiple demographic factors and laboratory markers. Binary logistic regression identified age (P = 0.002), eosinophil count (P = 0.004), and modified Rankin scale (mRS) scores at admission (P = 0.048) as significant predictors. The NIHSS (β = 0.179, P = 0.001) and mRS (β = 0.168, P = 0.016) scores at admission significantly positively predicted the Constipation Assessment Scale. NIHSS scores at admission (β = 0.300, P < 0.001) also positively predicted constipation severity at six-month follow-up. CONCLUSION New-onset constipation is a common complication of acute ischemic stroke hospitalization. NIHSS, mRS, and water swallowing test scores at admission can predict the incidence and severity of constipation during hospitalization and at the 6-month follow-up. These findings provide clinical insights for early risk assessment and intervention to mitigate constipation-related complications in patients with stroke.
Collapse
Affiliation(s)
- Xiuhua Tian
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Taiping Street, Jiangyang District, Luzhou 646000, China
| | - Lan Li
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Taiping Street, Jiangyang District, Luzhou 646000, China
| | - Junfeng Chen
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Taiping Street, Jiangyang District, Luzhou 646000, China
| | - Xiaoyan Guo
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Taiping Street, Jiangyang District, Luzhou 646000, China.
| |
Collapse
|
23
|
Li Z, Chen Y, Shi T, Cao H, Chen G, Yu L. Potential of queen bee larvae as a dietary supplement for obesity management: modulating the gut microbiota and promoting liver lipid metabolism. Food Funct 2025; 16:3848-3861. [PMID: 40131738 DOI: 10.1039/d5fo00166h] [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: 03/27/2025]
Abstract
Queen bee larvae (QBL) have been consumed as both a traditional food and medicine in China for thousands of years; however, their specific benefits for human health, particularly their potential anti-obesity property, remain underexplored. This study investigated the anti-obesity effect of QBL freeze-dried powder (QBLF) on high-fat diet (HFD) induced obesity in mice and explored the underlying mechanisms. Our findings showed that QBLF effectively reduced body weight, fasting blood glucose levels, lipid accumulation, and inflammation in HFD mice. 16S rRNA sequencing revealed that QBLF significantly modulated the gut microbiota disrupted by an HFD, notably increasing the relative abundance of beneficial microbes such as Ileibacterium, Clostridium sensu stricto 1, Incertae sedis, Streptococcus, Lactococcus, Clostridia UCG-014, and Lachnospiraceae UCG-006, which were inversely associated with obesity-related phenotypes in the mice. RNA sequencing analysis further demonstrated that QBLF intervention upregulated the expression of genes involved in liver lipid metabolism, including Pck1, Cyp4a10, Cyp4a14, and G6pc, while downregulating genes associated with the inflammatory response, such as Cxcl10, Ccl2, Traf1, Mapk15, Lcn2, and Fosb. These results suggested that QBLF can ameliorate HFD-induced obesity through regulating the gut microbiota, promoting liver lipid metabolism, and reducing inflammatory response.
Collapse
Affiliation(s)
- Zhuang Li
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Yiang Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Tengfei Shi
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Haiqun Cao
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Guijie Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Linsheng Yu
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| |
Collapse
|
24
|
Rehman A, Habumugisha T, Huang F, Zhang Z, Kiki C, Al MA, Yan C, Shaheen U, Zhang X. Impacts of polystyrene nanoplastics on zebrafish gut microbiota and mechanistic insights. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118332. [PMID: 40393324 DOI: 10.1016/j.ecoenv.2025.118332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
Aquatic environments are frequently contaminated with nanoplastics (NPs) ranging from 1-100 nm generated by plastic aging, but their bio-enrichment and toxicological impacts remain poorly understood. This study investigates how chronic exposure to carboxylated polystyrene nanoplastics (PNPs) alters gut microbiota composition and function in zebrafish (Danio rerio). Adult zebrafish were exposed to 50 nm PNPs at concentrations of 0.1, 1.0, and 10 mg/L for 14 and 28 days, followed by gut microbiota analysis using 16S rRNA gene sequencing. PNP exposure altered gut microbiota composition, including an increase in Proteobacteria abundance and a decrease in Firmicutes, Bacteroidetes, and the inflammation-related genus Alistipes. Beneficial probiotics such as Faecalibacterium, Streptococcus, Bifidobacterium, and Lachnospira were diminished, while pathogenic bacteria proliferated. TEM imaging revealed the internalization of PNP particles within intestinal tissues resulted in vacuolation, suggesting potential epithelial damage. Co-occurrence network patterns of gut microbiota greatly decreased during treatment with NPs. The neutral community model showed that among PNP treatments, 0.1 mg/L led to a less predictable (stochastic assembly process). PNP exposure led to increased predicted microbial functions (via PICRUSt2) related to xenobiotic metabolism, infection pathways, and lipopolysaccharide (LPS) production, while RNA transport and N-glycan biosynthesis were decreased. However, pathways related to microbial antioxidants exhibited significant variation across different PNP levels. These results provide critical insights into the toxicological impacts of chronic PNP exposure on fish gut health, highlighting the potential risks to aquatic ecosystems and human health.
Collapse
Affiliation(s)
- Abdul Rehman
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Théogène Habumugisha
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Fuyi Huang
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Zixing Zhang
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Claude Kiki
- State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China; Key Laboratory of Urban Pollutant Conversion, Fujian Key Laboratory of Watershed Ecology, Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Mamun Abdullah Al
- Marine Synthetic Ecology Research Center, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Science, State Key Laboratory for Biocontrol Sun Yat-sen University, Zhuhai 519082, China
| | - Changzhou Yan
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Uzma Shaheen
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China
| | - Xian Zhang
- State Key Laboratory of Regional and Urban Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, P.R. China; State Key Laboratory of Advanced Environmental Technology,Institute of Urban Environment Chinese Academy of Sciences, Xiamen 361021, P.R. China.
| |
Collapse
|
25
|
Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO. The Gut Microbiome as a Key Determinant of the Heritability of Body Mass Index. Nutrients 2025; 17:1713. [PMID: 40431453 PMCID: PMC12114430 DOI: 10.3390/nu17101713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The pathogenesis of obesity is complex and incompletely understood, with an underlying interplay between our genetic architecture and obesogenic environment. The public understanding of the development of obesity is shrouded in myths with widespread societal misconceptions. Body Mass Index (BMI) is a highly heritable trait. However, despite reports from recent genome-wide association studies, only a small proportion of the overall heritability of BMI is known to be lurking within the human genome. Other non-genetic heritable traits may contribute to BMI. The gut microbiome is an excellent candidate, implicating complex interlinks with hypothalamic control of appetite and metabolism via entero-endocrine, autonomic, and neuro-humeral pathways. The neonatal gut microbiome derived from the mother via transgenerational transmission (vaginal delivery and breastfeeding) tends to have a permanence within the gut. Conversely, non-maternally derived gut microbiota manifest mutability that responds to changes in lifestyle and diet. We should all strive to optimize our lifestyles and ensure a diet that is replete with varied and unprocessed plant-based foods to establish and nurture a healthy gut microbiome. Women of reproductive age should optimize their gut microbiome, particularly pre-conception, ante- and postnatally to enable the establishment of a healthy neonatal gut microbiome in their offspring. Finally, we should redouble our efforts to educate the populace on the pathogenesis of obesity, and the role of heritable (but modifiable) factors such as the gut microbiome. Such renewed understanding and insights would help to promote the widespread adoption of healthy lifestyles and diets, and facilitate a transition from our current dispassionate and stigmatized societal approach towards people living with obesity towards one that is epitomized by understanding, support, and compassion.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| | - Stefan Kabisch
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
26
|
Sun P, Liu J, Chen G, Guo Y. The Role of G Protein-Coupled Receptors in the Regulation of Orthopaedic Diseases by Gut Microbiota. Nutrients 2025; 17:1702. [PMID: 40431441 PMCID: PMC12114226 DOI: 10.3390/nu17101702] [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: 04/27/2025] [Revised: 05/09/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Exercise and diet modulate the gut microbiota, which is involved in the regulation of orthopaedic diseases and synthesises a wide range of metabolites that modulate cellular function and play an important role in bone development, remodelling and disease. G protein-coupled receptors (GPCRs), the largest family of transmembrane receptors in the human body, interact with gut microbial metabolites to regulate relevant pathological processes. This paper provides a review of different dietary and exercise effects on the pathogenic gut microbiota and their metabolites associated with GPCRs in orthopaedic diseases. RESULTS: Generally, metabolites produced by gut microbiota contribute to the maintenance of bone health by activating the corresponding GPCRs, which are involved in bone metabolism, regulation of immune response, and maintenance of gut flora homeostasis. Exercise and diet can influence gut microbiota, and an imbalance in gut microbiota homeostasis can trigger a series of adverse immune and metabolic responses by affecting GPCR function, ultimately leading to the onset and progression of various orthopaedic diseases. Understanding these relationships is crucial for elucidating the pathogenesis of orthopaedic diseases and developing personalised probiotic-based therapeutic strategies. In the future, we should further explore how to prevent and treat orthopaedic diseases through GPCR-based modulation of gut microbes and their interactions. The development of substances that precisely modulate gut microbes through different exercises and diets will provide more effective interventions to improve bone health in patients.
Collapse
Affiliation(s)
- Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Jinchao Liu
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Guannan Chen
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Yilan Guo
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| |
Collapse
|
27
|
Gong S, Ma W, Hu D, Wang X, Xu H, Li B. A novel pH-responsive berberine-loaded attapulgite microsphere for IBD therapy via the regulation of gut immunity and flora. Int J Pharm 2025; 679:125705. [PMID: 40381667 DOI: 10.1016/j.ijpharm.2025.125705] [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: 03/05/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic, lifelong, and devastating chronic inflammatory disease of the gastrointestinal tract. Berberine, an alkaloid extracted from the Chinese herb Huanglian, is a pharmaceutical ingredient with antibacterial, anti-inflammatory, and antiviral pharmacological effects in Chinese medicine. However, direct administration of berberine requires high doses due to its low bioavailability and high waste in the stomach. In this study, we prepared berberine-loaded pH-responsive Eudragit® FS 30D-attapulgite (FS-ATT) microspheres for the targeted treatment of colitis. The microspheres had pH-responsive release behavior, with minimal berberine release (about 5.97%) in simulated gastric fluid and high berberine release in simulated colonic fluid (about 69.27%). In addition, it was revealed by in vivo biological evaluations that, compared with drugs in an equivalent solution form taken orally, a significant improvement in IBD symptoms was observed after oral administration of drug-loaded FS-ATT microspheres, leading to an increase in the diversity of intestinal microbiota, inhibition of inflammatory cytokines, and enhanced therapeutic effects of berberine. The pH-responsive FS-ATT microspheres could offer a promising drug delivery platform for managing and treating many gastrointestinal diseases.
Collapse
Affiliation(s)
- Shiwen Gong
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology, Wuhan 430070, PR China; State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Wentao Ma
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Die Hu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan 528200, PR China; Hainan Institute, Wuhan University of Technology, Sanya 572000, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China.
| | - Haixing Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology, Wuhan 430070, PR China.
| | - Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Hainan Institute, Wuhan University of Technology, Sanya 572000, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, PR China.
| |
Collapse
|
28
|
Xu H, Lv D, Guan Y. Appeal of Urolithins from Synthesis to Biological Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11477-11494. [PMID: 40300072 DOI: 10.1021/acs.jafc.5c00634] [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: 05/01/2025]
Abstract
Urolithins (Uros), a series of natural polyphenols derived from ellagic acid through gut bacteria metabolism, have gathered significant attention due to their diverse bioactivities such as maintaining mitochondrial health and anti-inflammatory and antioxidative effects. However, the ability to metabolize Uros varies among individuals. This Review provides a comprehensive insight into the synthesis, encapsulation and bioactivities of Uros, focusing on their biotransformation in vivo. We highlight the critical role of gut microbiota in the biotransformation of urolithins, including primary bacterial species such as Gordonibacter urolithinfaciens, Enterocloster bolteae and Enterococcus faecium. Furthermore, the therapeutic potential of Uros in alleviating neurodegenerative diseases, cancer, and Duchenne muscular dystrophy is discussed. Finally, several encapsulation strategies for enhancing the solubility and bioavailability of Uros are summarized. Future research direction includes identifying key genes involved in Uros biotransformation, elucidating the bioactive mechanisms of Uros, and improving their bioavailability. In conclusion, we synthesized biosynthetic pathways and bioactive properties of Uros for better utilization in health management.
Collapse
Affiliation(s)
- Huanyu Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Danyu Lv
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yongguang Guan
- Department of Food Science, Foshan University, Foshan 528000, China
| |
Collapse
|
29
|
Lanou HB, Somé JW, Koumbem MAA, Kouanda S. Microbiome-directed food to promote sustained recovery in children with uncomplicated acute malnutrition: protocol for a randomized controlled trial in Burkina Faso. BMC Nutr 2025; 11:92. [PMID: 40361242 PMCID: PMC12070536 DOI: 10.1186/s40795-025-01045-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/13/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Acute malnutrition still affects millions of children under five years of age globally each year and contributes to approximately half of all annual childhood deaths. A considerable proportion of patients who recover from acute malnutrition experience poor health and nutrition and eventually relapse after they are discharged from community management of acute malnutrition programs. A microbiota-directed complementary food (MDCF) showed a superior effect compared to standard ready-to-use supplementary food (RUSF) in terms of ponderal growth and potential benefit for bacterial taxa that were correlated with weight-for-height z-score (WHZ). This paper describes a protocol for the MDCF phase III trial on a larger African sample for promoting sustained recovery. METHODS This study is an individually controlled open-label phase III trial to determine the efficacy of MDCF on programmatic and sustained recovery compared to standards RUTF and RUSF. Eligible MAM children will be randomly assigned to MDCF or RUSF and those with SAM to MDCF or RUTF. Supplementation and follow-up visits will be performed following national guidelines for acute malnutrition management. Primary outcomes are programmatic recovery at 12 weeks after enrollment and sustained recovery at 12 weeks after recovery. The secondary outcomes included the mean WHZ, weight-for-age z score, height-for-age z score change, average length of stay, nonresponse, failure and dropout. DISCUSSION The present study is designed to investigate the efficacy of a microbiota-targeted food in treating acute uncomplicated malnutrition and preventing relapses. It will provide evidence as a phase III clinical trial. TRIAL REGISTRATION Clinicaltrials.gov Protocol registration and results system (NCT05586139). Registered on 2022-10-14. https://register. CLINICALTRIALS gov/ .
Collapse
Affiliation(s)
- Hermann Biénou Lanou
- Institut de Recherche en Sciences de la Santé (IRSS), National Centre for Scientific and Technologic Research (CNRST), Wemtenga, Rue 29, 39 Porte 74, 03 P.B. 7192, Ouagadougou, Burkina Faso.
| | - Jérôme Winetourefa Somé
- Institut de Recherche en Sciences de la Santé (IRSS), National Centre for Scientific and Technologic Research (CNRST), Wemtenga, Rue 29, 39 Porte 74, 03 P.B. 7192, Ouagadougou, Burkina Faso
| | - Marie Arsène Aristide Koumbem
- Institut de Recherche en Sciences de la Santé (IRSS), National Centre for Scientific and Technologic Research (CNRST), Wemtenga, Rue 29, 39 Porte 74, 03 P.B. 7192, Ouagadougou, Burkina Faso
| | - Seni Kouanda
- Institut de Recherche en Sciences de la Santé (IRSS), National Centre for Scientific and Technologic Research (CNRST), Wemtenga, Rue 29, 39 Porte 74, 03 P.B. 7192, Ouagadougou, Burkina Faso
| |
Collapse
|
30
|
Zhang Y, Bai C, Sha J, Huo X, Qu D, Chen J. Ginseng Soluble Dietary Fiber Reverses Obesity via the PPAR/AMPK Signaling Pathway and Improves Intestinal Flora in Mice. Foods 2025; 14:1716. [PMID: 40428495 PMCID: PMC12111629 DOI: 10.3390/foods14101716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Ginseng soluble dietary fiber (GSDF) has been shown to have good physicochemical properties; however, its in vivo benefits in obesity are yet to be fully elucidated. METHODS To explore this, C57BL/6J obese mice were given metformin hydrochloride and different doses of GSDF for 60 days. The levels of blood lipids and inflammatory factors were detected by ELISA, and the pathological alterations were detected through the application of HE staining. The level of adipose tissue protein in epididymis was detected by Western blotting and through the effects of 16S rRNA sequencing on gut microbiota. RESULTS The results showed that GSDF significantly improved basal physiological indices, lipid levels, and serum cytokine levels in the obese mice. GSDF increased the expression levels of PPAR-γ, AMPK, and P-AMPK proteins, and lowered the expression of IL-1β, TNF-α, and other proteins in the adipose tissues of the epididymis, in turn inhibiting adipogenesis and ameliorating lipid metabolism disorders. By lowering the Firmicutes/Bacteroidetes ratio in the gut and altering the abundance of thick-walled bacteria and mycobacterium, the abundance of species such as Lactobacillus, Alloprevotella, and Faecalibaculum was altered to improve cecum health. CONCLUSIONS These results suggest that GSDF may have a positive effect on growth, obesity, and cecal health in obese mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianbo Chen
- Institute of Special Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun 130112, China; (Y.Z.); (C.B.); (J.S.); (X.H.); (D.Q.)
| |
Collapse
|
31
|
Zeng H, Liu C, Wan L, Peng L, Wang K, Zhou F, Fang W, Wen S, Bai Q, Yang X, Liu L, Zeng J, Huang J, Liu Z. Epigallocatechin gallate prevents and alleviates type 2 diabetes mellitus (T2DM) through gut microbiota and multi-organ interactions in Wistar healthy rats and GK T2DM rats. J Adv Res 2025:S2090-1232(25)00296-6. [PMID: 40349958 DOI: 10.1016/j.jare.2025.05.002] [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: 03/06/2025] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION As the main active ingredient of the first FDA-approved phytochemical drug, epigallocatechin gallate (EGCG) can effectively alleviate glucolipid metabolic disorders. However, existing studies mainly focuses on the treatment of EGCG in disease models, with limited focus on its preventive effect on diseases in healthy models. OBJECTIVES This study investigated how EGCG prevents and alleviates T2DM through gut microbiota and multi-organ interactions in Wistar healthy rats and GK T2DM rats. METHODS The GK T2DM rat strain was established through repeated selective breeding of Wistar rats with glucose intolerance. Whether and how EGCG prevents and alleviates T2DM were evaluated, including glucose production and absorption efficiency, glucose transport, glucose metabolism, glucose excretion, T2DM-related tissue damage, gut microbiota, and liver transcriptome. RESULTS The health benefits of EGCG are primarily reliant on the involvement of the gut microbiota. Our study showed that although the specific microbial communities involved differ, the bidirectional interaction between EGCG and gut microbiota is widespread in healthy rats and T2DM rats. EGCG intervention elevated the relative abundance of specific microbial communities, which in turn promoted the metabolic processing of EGCG in the gut, producing numerous EGCG metabolites that may contribute to preventing and alleviating T2DM. In healthy rats, EGCG intervention selectively enhanced insulin secretion and serum insulin levels to prevent T2DM. In T2DM rats, EGCG intervention selectively lowered blood glucose levels, improved insulin resistance, delayed glucose production and absorption, and promoted urinary glucose excretion to alleviate T2DM. In both healthy and T2DM rats, EGCG intervention universally reduced gut microbiota-derived lipopolysaccharides, maintained systemic oxidative stress homeostasis, alleviated liver and kidney damage, increased muscle glycogen content, and promoted beige thermogenesis in white fat, thus demonstrating potential for preventing and alleviating T2DM. CONCLUSION As a natural active ingredient, EGCG could prevent and alleviate T2DM through gut microbiota and multi-organ interactions.
Collapse
Affiliation(s)
- Hongzhe Zeng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Changwei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Liwei Wan
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Liyuan Peng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Kuofei Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Fang Zhou
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Wenwen Fang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Shuai Wen
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Qixian Bai
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Xiaomei Yang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Linmei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China
| | - Jie Zeng
- Micangshan Tea Industry Research Institute, Sichuan Wangcang Vocational Middle School, Sichuan 628200, China
| | - Jian'an Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China.
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; Yuelushan Laboratory, Changsha 410128, China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Changsha 410128, China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
32
|
Origüela V, Lopez-Zaplana A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025; 13:1084. [PMID: 40431257 PMCID: PMC12113704 DOI: 10.3390/microorganisms13051084] [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: 03/17/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
The importance of the microbiome, particularly the gut microbiota and its implications for health, is well established. However, an increasing number of studies further strengthen the link between an imbalanced gut microbiota and a greater predisposition to different diseases. The gut microbiota constitutes a fundamental ecosystem for maintaining human health. Its alteration, known as dysbiosis, is associated with a wide range of conditions, including intestinal, metabolic, immunological, or neurological pathologies, among others. In recent years, there has been a substantial increase in knowledge about probiotics-bacterial species that enhance health or address various diseases-with numerous studies reporting their benefits in preventing or improving these conditions. This review aims to analyze the most common pathologies resulting from an imbalance in the gut microbiota, as well as detail the most important and known gut probiotics, their functions, and mechanisms of action in relation to these conditions.
Collapse
Affiliation(s)
- Valentina Origüela
- Department of Physiology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain;
| | | |
Collapse
|
33
|
Zhu B, Zhang Q, Chen X, Zheng N, Wang X, Shi X, Yang L, Han J, Liu C, Zhou B. Impact of 1,2-Bis (2,4,6-Tribromophenoxy) Ethane on Liver Metabolism and Intestinal Health in Zebrafish: Role of the Liver X Receptor. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8439-8450. [PMID: 40277015 DOI: 10.1021/acs.est.5c00681] [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: 04/26/2025]
Abstract
1,2-Bis (2,4,6-tribromophenoxy) ethane (BTBPE) has been increasingly detected in environmental and biota samples, primarily accumulating in the liver. However, the mechanism underlying BTBPE-induced metabolic dysregulation remains unclear. In this study, molecular docking and microscale thermophoresis assays indicated that BTBPE binds to zebrafish liver X receptor α (LXRα). Subsequently, zebrafish embryos were exposed to BTBPE, an LXR antagonist (GSK2033), or coexposed to BTBPE with an LXR agonist (GW3965) for 120 h postfertilization (hpf). The results showed that BTBPE induced reduction in body weight and lipid levels, likely via inhibition of the LXR signaling pathway. Exposure of adult female zebrafish to environmentally relevant concentrations of BTBPE (0.01-10 μg/L) for 28 days induced developmental toxicity, evidenced by decreases in body weight, growth rate, and fat accumulation. Metabolomic analysis revealed that BTBPE-induced alterations in liver metabolites were primarily associated with LXR-mediated lipid metabolic pathways such as glycerophospholipid metabolism and primary bile acid biosynthesis. Additionally, BTBPE impaired the physical barrier and induced inflammation, resulting in gut microbiota dysbiosis, which is potentially linked to LXR activation. These effects were validated through the alterations of multiple biomarkers at various levels. Overall, our results suggest that BTBPE disrupts lipid metabolism and gut function via the LXR-mediated pathway.
Collapse
Affiliation(s)
- Biran Zhu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qianqian Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xianglin Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Na Zheng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiaochen Wang
- Ecology and Environment Monitoring and Scientific Research Center, Ecology and Environment Administration of Yangtze River Basin, Ministry of Ecology and Environment, Wuhan 430010, China
| | - Xiongjie Shi
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Chunsheng Liu
- MOE Key Laboratory of Groundwater Quality and Health, School of Environmental Studies, China University of Geosciences, Wuhan 430078, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
34
|
Chi Y, Zhang H, Gao J, Wan L, Jiao Y, Wang H, Liao M, Cuthbert RN. Nanoplastics Elicit Stage-Specific Physiological, Biochemical, and Gut Microbiome Responses in a Freshwater Mussel. TOXICS 2025; 13:374. [PMID: 40423453 DOI: 10.3390/toxics13050374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/03/2025] [Accepted: 05/03/2025] [Indexed: 05/28/2025]
Abstract
Mussels are highly efficient filter feeders, playing a crucial role in managing eutrophication and assessing pollution. Although research on nanoplastic (NP) toxicity in marine organisms is expanding, studies on freshwater species remain limited despite freshwater ecosystems being disproportionately biodiverse and vulnerable to pollutants. Here, we quantified the effects of polystyrene nanoplastics (PS-NPs, 50 nm) at concentrations of 0, 2, 20, and 200 μg/L on different growth stages of the freshwater mussel Cristaria plicata. After a 45-day exposure, PS-NPs at concentrations ≥ 20 μg/L damaged intestinal epithelial cilia in both age groups. Exposure to 200 μg/L PS-NPs significantly increased malondialdehyde levels and decreased superoxide dismutase activity in both groups, with adults showing a significant rise in total protein content and juveniles exhibiting marked increases in respiratory and ammonia excretion rates. Additionally, PS-NP exposure significantly altered the relative abundance of gut microbial phyla, including Proteobacteria, Firmicutes, Verrucomicrobiota, and Bacteroidota, with Fusobacteriota also being affected in adults. Juveniles were more sensitive to physiological changes, whereas adults exhibited greater microbiota shifts in response to PS-NP exposure. Therefore, this study provides new insights into the stage-specific effects of PS-NPs on intestinal integrity and physiological and biochemical health in freshwater mussels, underscoring the need for targeted management strategies to protect freshwater ecosystems.
Collapse
Affiliation(s)
- Yangli Chi
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Hui Zhang
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Jian Gao
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Liang Wan
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Yiying Jiao
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Heyun Wang
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Mingjun Liao
- Key Laboratory of Intelligent Health Perception and Ecological Restoration of Rivers and Lakes, Ministry of Education, Hubei University of Technology, Wuhan 430068, China
- Hubei Key Laboratory of Environmental Geotechnology and Ecological Remediation for Lake and River, Innovation Demonstration Base of Ecological Environment Geotechnical and Ecological Restoration of Rivers and Lakes, Hubei University of Technology, Wuhan 430068, China
| | - Ross N Cuthbert
- Institute for Global Food Security, School of Biological Sciences, Queens University Belfast, Belfast BT9 5DL, UK
| |
Collapse
|
35
|
van Deuren T, Umanets A, Venema K, Moreno LL, Zoetendal EG, Canfora EE, Blaak EE. Specific dietary fibers steer toward distal colonic saccharolytic fermentation using the microbiota of individuals with overweight/obesity. Food Res Int 2025; 209:116271. [PMID: 40253188 DOI: 10.1016/j.foodres.2025.116271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Evidence suggests that increased distal short-chain fatty acid (SCFA) production beneficially impacts metabolic health. However, indigestible carbohydrate availability is limited in the distal colon; consequently, microbes shift toward protein fermentation, often linked to adverse metabolic health effects. We aimed to identify specific fiber(s) that promote saccharolytic fermentation in the distal colon and thereby may (partially) inhibit proteolytic fermentation. METHODS Potato-fiber, pectin, and inulin were studied individually and in combination against a high (predigested) protein background using an in vitro model of the colon (TIM-2) inoculated with pooled, standardized fecal microbiota from individuals with overweight/obesity. Microbiota composition and activity were assessed at different timepoints to simulate the travel throughout the colon (proximal: 0-8 h, distal: 8-24 h) and compared to a high protein (HP)_control, receiving only proteins. RESULTS Fiber addition increased total SCFA production compared to HP_control (52.11 ± 1.49 vs 27.07 ± 0.26 mmol) whereas total branched-chain fatty acids (BCFA; a marker for protein fermentation) production only slightly decreased (3.31 ± 0.10 vs 4.18 ± 0.40 mmol). Combining potato-fiber and pectin led to the highest total and distal SCFA production and distal SCFA:BCFA. Fiber addition attenuated HP-induced increases in several bacterial taxa including Mogibacterium and Coprococcus, independent of fiber type. Additionally, time- and fiber-specific microbial signatures were identified: inulin increased Bifidobacterium (proximal) relative abundance and pectin and/or potato-fiber increased Prevotella 9 (distal) relative abundance. CONCLUSION The most marked increase in distal colonic SCFA production was induced by combining potato-fiber and pectin. Further research should elucidate whether this switch toward saccharolytic fermentation translates into beneficial metabolic health effects in humans.
Collapse
Affiliation(s)
- Thirza van Deuren
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Alexander Umanets
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University-Campus Venlo, Venlo, the Netherlands; Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Venlo, the Netherlands
| | - Luis L Moreno
- Laboratory of Food Chemistry, Wageningen University and Research, Bornse Weilanden 9, 6708, WG, Wageningen, the Netherlands; Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, the Netherlands
| | - Emanuel E Canfora
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'
| | - Ellen E Blaak
- Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, Maastricht, the Netherlands'.
| |
Collapse
|
36
|
Li Z, Jin Y, Zhao H, Gu Y, Zhang Y, Cheng S, Zhang L, He P, Liu X, Jia Y. Aurantio-Obtusin Regulates Gut Microbiota and Serum Metabolism to Alleviate High-Fat Diet-Induced Obesity-Associated Non-Alcoholic Fatty Liver Disease in Mice. Phytother Res 2025; 39:1946-1965. [PMID: 39953693 DOI: 10.1002/ptr.8459] [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: 09/10/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive condition with limited effective treatments. This study investigated the therapeutic effects of Aurantio-obtusin (AO), a bioactive compound from Cassiae Semen, on obesity-associated NAFLD. An obesity-related NAFLD model was established in ApoE -/- mice fed a high-fat diet (HFD) for 24 weeks, with AO administered during the last 16 weeks. Mouse body weight, adipose tissue weights, liver weights, serum lipid levels, hepatic steatosis, inflammatory damage, and colonic tissue barrier integrity were evaluated. Gut microbial communities and serum metabolic profiles were analyzed using 16S rRNA sequencing and untargeted metabolomics. Hepatic lipid metabolism-related gene expression was assessed using molecular biology techniques. AO treatment significantly ameliorated HFD-induced adiposity, hyperlipidemia, and NAFLD symptoms. It preserved intestinal barrier integrity, modulated gut microbial composition by enriching beneficial taxa, and improved serum metabolic profiles. AO favorably adjusted hepatic lipid metabolism by upregulating PPARα and CPT1A while downregulating SREBP1, FASN, and SCD1. Correlation analysis revealed significant associations among gut microbial composition, serum metabolites, and disease indicators. AO's therapeutic benefits in NAFLD might be attributed to its ability to modulate gut microbial community composition and serum metabolic profile, enhance intestinal barrier function, and regulate hepatic lipid metabolism gene expression. AO presents a promising therapeutic agent for obesity-associated NAFLD, warranting further investigation into its potential clinical applications.
Collapse
Affiliation(s)
- Zhaoyong Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Jin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Huashan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyan Gu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yaxin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Saibo Cheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lifang Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Peikun He
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xiaoyu Liu
- Pingshan Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Pingshan District Peoples' Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Yuhua Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Liu WT, Hu XW, Choy YN, Lai W, Xu HY, Zeng YJ, Lan QS, Liu L, Yue RB, Chu ZH. Investigating the role of inflammatory cytokines in mediating the effect of gut microbiota on gastrointestinal cancers: a mendelian randomization study. Gastric Cancer 2025; 28:442-454. [PMID: 39961989 DOI: 10.1007/s10120-025-01587-w] [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: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 04/13/2025]
Abstract
PURPOSE The purpose of this study is to explore the causal relationship between gut microbiota and gastrointestinal (GI) cancers and to investigate the potential mediating factors influencing the development of GI cancers. METHODS Using data from genome-wide association studies (GWAS), we employed two-sample Mendelian randomization (TSMR) to explore the relationship among gut microbiota, inflammatory cytokines and GI cancers. Subsequently, a multivariable Mendelian randomization (MVMR) analysis was meticulously conducted to perform a mediation analysis, thereby estimating the proportion of mediation effects conferred by inflammatory cytokines. RESULTS TSMR analysis established a causal relationship between 23 gut microbiota taxa and 11 inflammatory cytokines with GI cancers. Specifically, 7 gut microbiota taxa were associated with an increased risk of gastric cancer (GC), 6 with small intestine cancer, and 10 with colorectal cancer (CRC). Among the inflammatory cytokines, 4 were linked to GC risk, 3 to small intestine cancer, and to CRC. Mediation analysis further indicatedthat tumor necrosis factor ligand superfamily member 12 (TNFSF12) mediated 9.703% (95% CI 0.108%~15.891%) of the total effect of genus Ruminiclostridium9 on GC. CONCLUSION Our findings support a causal relationship between gut microbiota, inflammatory cytokines, and GI cancers. These biomarkers provide new insights into the mechanisms underlying GI cancers and have the potential to improve strategies forprevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Wen-Tao Liu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Xin-Wen Hu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Yan-Ni Choy
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Wei Lai
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - He-Yang Xu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Yu-Jie Zeng
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Qiu-Sheng Lan
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Lu Liu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Rong-Bin Yue
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China
| | - Zhong-Hua Chu
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan-Jiang Xi Road, Guangzhou, 510120, P.R. China.
| |
Collapse
|
38
|
Shen Y, Fan N, Ma S, Cheng X, Yang X, Wang G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm (Beijing) 2025; 6:e70168. [PMID: 40255918 PMCID: PMC12006732 DOI: 10.1002/mco2.70168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut-brain axis, gut-liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Nairui Fan
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Shu‐xia Ma
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- International SchoolGuangzhou Huali College, ZengchengGuangzhouChina
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryGuangdong Second Provincial General HospitalSchool of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
39
|
Yamamura R, Okubo R, Ukawa S, Nakamura K, Okada E, Nakagawa T, Imae A, Kimura T, Tamakoshi A. Increased fecal glycocholic acid levels correlate with obesity in conjunction with the depletion of archaea: The Dosanco Health Study. J Nutr Biochem 2025; 139:109846. [PMID: 39863085 DOI: 10.1016/j.jnutbio.2025.109846] [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: 07/22/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Recent studies have focused on the relationship between obesity and gut microbiota. This study aims to identify fecal components and gut bacterial species associated with different BMI categories. In this study, 538 participants aged ≥18 years were categorized into underweight, normal, and obese groups based on BMI (cutoffs: 18.5 and 25.0 kg/m²). We compared 30 fecal components among these groups and calculated correlation coefficients between each component and BMI. Participants were then divided into quartiles based on fecal component levels correlated with BMI, and the prevalence ratio (PR) of obesity was calculated, adjusted for confounding factors. We also analyzed the composition and diversity of gut microbiota and bacterial gene expression among the quartiles for each fecal component. Fecal glycocholic acid (GCA) showed a significant positive correlation with BMI. The PR for obesity in the highest quartile of fecal GCA was 3.30 (95% CI, 1.21-9.54), indicating a significantly higher risk of obesity compared to the lowest quartile. Gut microbiota analysis revealed significant differences in the abundance of Ruminococcaceae Incertae Sedis, Faecalibacterium, and Methanobrevibacter, with Methanobrevibacter being absent in the higher quartiles of fecal GCA. Additionally, gene expression for enzymes involved in the deconjugation of conjugated bile acids, including GCA, was downregulated in the highest quartile. Increased fecal GCA levels are positively correlated with obesity, alongside a depletion of archaea.
Collapse
Affiliation(s)
- Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| | - Ryo Okubo
- Department of Neuropsychiatry, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shigekazu Ukawa
- Osaka Metropolitan University Graduate School of Human Life and Ecology, Sumiyoshi, Osaka, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Koshi Nakamura
- Department of Public Health and Epidemiology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Emiko Okada
- The Health Care Science Institute, Minato-ku, Tokyo, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Akihiro Imae
- The Hokkaido Centre for Family Medicine, Sapporo, Japan
| | - Takashi Kimura
- Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Akiko Tamakoshi
- Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
40
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
41
|
Xu D, Wu M, Gao Z, Zhao Y, Hu M, Wen Y, Wang L, Xu D, Chen L. Seasonal Variation in the Diversity of the Gut Microbiota of Short-Faced Moles Reveals the Associations of Climatic Factors on the Gut Microbiota of Subterranean Mammals. Ecol Evol 2025; 15:e71382. [PMID: 40342707 PMCID: PMC12058457 DOI: 10.1002/ece3.71382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 04/01/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
The composition of animal gut microbiota is significantly affected by a variety of factors. Seasonal variation in environmental factors is believed to have a significant impact on the composition of mammalian gut microbiota. Therefore, studying the seasonal differences in gut microbiota diversity in wildlife is of great importance to explore their ecological adaptability. This study compared the diversity of gut microbiota of the short-faced moles (Scaptochirus moschatus) in spring, summer, and autumn by using 16S rRNA amplification sequencing. Our results reveal significant seasonal differences in the diversity and function of the short-faced moles gut microbiota. Compared to spring, the diversity and function of the gut microbiota in summer and autumn of short-faced moles are more similar to each other. The relative abundance of Firmicutes is higher in spring than in summer and autumn, while the relative abundance of Proteobacteria in summer and autumn is higher than that of spring. There are significant differences in carbohydrate metabolism between spring and summer, and between spring and autumn. The correlation analysis results suggest that climatic factors are strongly associated with seasonal variation in gut microbiota of the short-faced moles, especially temperature and relative humidity. The present study discusses the seasonal variations in the gut microbiota diversity of short-faced moles and the significant impact of climatic factors on gut microbiota diversity. These results will highlight the potential impact of climatic factors on the seasonal changes of the gut microbiota of subterranean mammals and provide a new view for comprehensively understanding the ecological adaptation of subterranean mammals.
Collapse
Affiliation(s)
- Di Xu
- College of Life SciencesQufu Normal UniversityQufuChina
| | - Mengmeng Wu
- Shandong Freshwater Fisheries Research InstituteJinanChina
| | - Zenghao Gao
- College of Life SciencesQufu Normal UniversityQufuChina
| | - Yue Zhao
- College of Life SciencesQufu Normal UniversityQufuChina
| | - Meng Hu
- Forestry Protection and Development Service Center of JiningJiningChina
| | - Yang Wen
- Forestry Protection and Development Service Center of JiningJiningChina
| | - Linlin Wang
- Jining Bureau of Natural Resources and PlanningJiningChina
| | - Deli Xu
- College of Life SciencesQufu Normal UniversityQufuChina
| | - Lei Chen
- College of Life SciencesQufu Normal UniversityQufuChina
| |
Collapse
|
42
|
Rukavina Mikusic NL, Prince PD, Choi MR, Chuffa LGA, Simão VA, Castro C, Manucha W, Quesada I. Microbiota, mitochondria, and epigenetics in health and disease: converging pathways to solve the puzzle. Pflugers Arch 2025; 477:635-655. [PMID: 40111427 DOI: 10.1007/s00424-025-03072-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Dysbiosis, which refers to an imbalance in the composition of the gut microbiome, has been associated with a range of metabolic disorders, including type 2 diabetes, obesity, and metabolic syndrome. Although the exact mechanisms connecting gut dysbiosis to these conditions are not fully understood, various lines of evidence strongly suggest a substantial role for the interaction between the gut microbiome, mitochondria, and epigenetics. Current studies suggest that the gut microbiome has the potential to affect mitochondrial function and biogenesis through the production of metabolites. A well-balanced microbiota plays a pivotal role in supporting normal mitochondrial and cellular functions by providing metabolites that are essential for mitochondrial bioenergetics and signaling pathways. Conversely, in the context of illnesses, an unbalanced microbiota can impact mitochondrial function, leading to increased aerobic glycolysis, reduced oxidative phosphorylation and fatty acid oxidation, alterations in mitochondrial membrane permeability, and heightened resistance to cellular apoptosis. Mitochondrial activity can also influence the composition and function of the gut microbiota. Because of the intricate interplay between nuclear and mitochondrial communication, the nuclear epigenome can regulate mitochondrial function, and conversely, mitochondria can produce metabolic signals that initiate epigenetic changes within the nucleus. Given the epigenetic modifications triggered by metabolic signals from mitochondria in response to stress or damage, targeting an imbalanced microbiota through interventions could offer a promising strategy to alleviate the epigenetic alterations arising from disrupted mitochondrial signaling.
Collapse
Affiliation(s)
- Natalia Lucia Rukavina Mikusic
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Paula Denise Prince
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Químicas, Cátedra de Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina.
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina.
| | - Luiz Gustavo A Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Vinícius Augusto Simão
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Claudia Castro
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
- Laboratorio de Farmacología Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina.
| | - Isabel Quesada
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
43
|
Shi Y, Jiang M, Zhu W, Chang K, Cheng X, Bao H, Peng Z, Hu Y, Li C, Fang F, Song J, Jian C, Chen J, Shu X. Cyclosporine combined with dexamethasone regulates hepatic Abca1 and PPARα expression and lipid metabolism via butyrate derived from the gut microbiota. Biomed Pharmacother 2025; 186:118017. [PMID: 40168721 DOI: 10.1016/j.biopha.2025.118017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
Immunosuppression often leads to drastic metabolic, hormonal, and physiological disorders. Changes in the gut microbiota are believed to be one of the factors contributing to these disorders, but the association remains uncertain. Clinical studies can be complicated by confounding variables, such as diet and other drivers of heterogeneity in human microbiomes. In this study, we identified pronounced gut microbiome signatures in rhesus macaques (RMs) with immunosuppression-induced lipid metabolism disorders following cyclosporine combined with dexamethasone. Furthermore, we observed similar changes in the gut microbiota of mice with immunosuppression-induced lipid metabolism disorders, which were associated with short-chain fatty acid metabolism. ELISA showed that immunosuppression significantly reduced the levels of butyric acid in both feces and serum of mice. Spearman correlation analysis identified a significant correlation between serum butyric acid levels and gut microbial dysbiosis induced by immunosuppression, particularly in relation to f_Lachnospiraceae, g_unidentified_Ruminococcaceae, and s_Clostridium leptum. Additionally, mice transplanted with gut microbiota from immunosuppressed mice exhibited hepatic lipid metabolism disorders, and RNA sequencing revealed significant downregulation of ABC transporters and PPARα in the liver, which was closely associated with lipid transport and metabolism, particularly Abca1. Moreover, butyric acid supplementation alleviated hepatic lipid metabolism disorders and upregulated the expression of Abca1 and PPARα in mice transplanted with immunosuppression-induced gut microbiota. Thus, we propose that the combination of cyclosporine and dexamethasone regulates the expression of hepatic Abca1 and PPARα by modulating the gut microbiota and its derived butyrate, particularly Lachnospiraceae and Clostridium leptum, further regulating hepatic lipid metabolism.
Collapse
Affiliation(s)
- Yongping Shi
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Mi Jiang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wenzhong Zhu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Ke Chang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xukai Cheng
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Haijun Bao
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zuojie Peng
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yuan Hu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chao Li
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feifei Fang
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jia Song
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Chenxing Jian
- Department of anorectal surgery, Affiliated hospital of Putian University, China
| | - Jinhuang Chen
- Department of emergency surgery, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaogang Shu
- Department of ganstroenterology, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
44
|
Gao P, Nie Y, Zhao L, Zhang J, Ge W. Lactococcus lactis Subsp. lactis LL-1 and Lacticaseibacillus paracasei LP-16 Influence the Gut Microbiota and Metabolites for Anti-Obesity and Hypolipidemic Effects in Mice. Antioxidants (Basel) 2025; 14:547. [PMID: 40427429 PMCID: PMC12108308 DOI: 10.3390/antiox14050547] [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: 03/06/2025] [Revised: 04/19/2025] [Accepted: 04/20/2025] [Indexed: 05/29/2025] Open
Abstract
This study utilized a high-fat diet-induced obese male C57BL/6 mice model to investigate the anti-obesity and lipid-lowering effects of Lactococcus lactis subsp. lactis LL-1 and Lacticaseibacillus paracasei LP-16. A gut microbiota analysis via 16S rRNA sequencing, along with measurements of body weight, lipids, inflammation markers, and gut metabolites, revealed that lactic acid bacteria (LAB) significantly reduced body weight, blood lipid levels, and liver oxidative stress. They also enhanced gut microbiota diversity and evenness, potentially by modulating the Firmicutes/Bacteroidetes ratio to limit excess energy absorption. Malondialdehyde (MDA) showed extremely significant positive correlations with Lachnospiraceae, Blautia, and Colidextribacter, and a significant positive correlation with Helicobacter, while superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) exhibited opposite trends. Specifically, Muribaculaceae, Bacteroides, and Lactobacillus showed negative correlations with MDA levels and positive correlations with SOD and GSH-Px. Short-chain fatty acids (SCFAs) positively correlated with Muribaculaceae, Bacteroides, Mucispirillum, and Lactobacillus, but negatively correlated with Lachnospiraceae, Blautia, Colidextribacter, Alistipes, and Helicobacter. They increased SCFA levels by promoting beneficial bacteria and reducing pathogens, alleviating obesity and hyperlipidemia. Additionally, they regulated the gut microbiota, decreasing bile acids and long-chain fatty acids while increasing SCFAs, short peptides, and vitamins, thereby improving gut metabolic disorders and enhancing host gut health.
Collapse
Affiliation(s)
- Peng Gao
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (P.G.); (L.Z.); (J.Z.)
| | - Yuanyang Nie
- School of Food Science, Henan Institute of Science and Technology, Xinxiang 453003, China;
| | - Lili Zhao
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (P.G.); (L.Z.); (J.Z.)
| | - Jing Zhang
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (P.G.); (L.Z.); (J.Z.)
| | - Wupeng Ge
- College of Food Science and Engineering, Northwest A&F University, Xianyang 712100, China; (P.G.); (L.Z.); (J.Z.)
| |
Collapse
|
45
|
Zou M, Li X, Li C, Pei H, Kang R, Liu L, Gao L. Comparative Analysis of Gut Bacteria of Four Waterbirds Species in Taolimiao-Alashan Nur (T-A Nur) in Erdos Relic Gull National Nature Reserve, Inner Mongolia, China. Ecol Evol 2025; 15:e71432. [PMID: 40370353 PMCID: PMC12074897 DOI: 10.1002/ece3.71432] [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: 12/13/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
Taolimiao-Alashan Nur (T-A Nur) is an important breeding site for the Relict Gulls (Larus relictus) and many other waterbirds. To understand the gut health status of rare bird species living there and to protect these bird species, this study analyzed the gut microbiota of four waterbird species, including Relict Gull (L. relictus), Black-necked Grebe (Podiceps nigricollis), Greylag Goose (Anser anser), and Ruddy Shelduck (Tadorna ferruginea), using 16S rRNA high-throughput sequencing. Results showed that the gut microbiota of Ruddy Shelduck had the highest α-diversity, while Greylag Goose had the lowest. The composition of gut microbiota varied significantly among the bird species. The dominant bacterial phylum in the guts of Black-necked Grebe, Greylag Goose, and Ruddy Shelduck was Firmicutes, while it was Pseudomonadota in Relict Gull. At the genus level, the dominant bacteria were Halomonas in Black-necked Grebe, Escherichia-Shigella in Relict Gull, Ligilactobacillus in Greylag Goose, and Enterococcus in Ruddy Shelduck. Correlation analysis revealed significant relationships among gut bacterial communities, suggesting that gut bacteria can regulate host metabolism and physiological state by their interactions. KEGG functional predictions indicated that gut microbiota were primarily involved in metabolism. The abundance of metabolism-related microorganisms in Relict Gull was significantly lower than in Greylag Goose and Ruddy Shelduck, indicating that the gut microbiota of Greylag Goose and Ruddy Shelduck can provide stronger metabolic functions for the hosts. Additionally, microorganisms related to human diseases were more abundant in the gut of Relict Gull compared to Ruddy Shelduck and Black-necked Grebe, and in Greylag Goose compared to Ruddy Shelduck. These findings suggested that the gut microbiota of birds in this area harbor some human pathogens, which warrants attention and preventive measures.
Collapse
Affiliation(s)
- Mingxin Zou
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Xuanyu Li
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Chunyu Li
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Hongda Pei
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Ruobing Kang
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Li Liu
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Li Gao
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| |
Collapse
|
46
|
Sun X, Yang B, Chen C. Uncovering the heterogeneity of the gut microbial taxa associated with the contents of different fatty acids in muscle with cecum luminal content and fecal samples from two pig populations. Front Microbiol 2025; 16:1575383. [PMID: 40371116 PMCID: PMC12075296 DOI: 10.3389/fmicb.2025.1575383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
Fatty acids in pork are involved in cellular physiological functions and related to meat nutrition, tenderness, and flavor. Increasing evidences have suggested that short-chain fatty acids produced by the gut microbiota may affect host metabolism and energy utilization. However, the association between gut microbiota and long-chain fatty acids (LCFAs) in pork has been largely unknown. In this study, the microbial compositions of 243 cecum content samples from Erhualian pigs and 235 fecal samples from Bamaxiang pigs were determined by high throughput 16S rRNA gene sequencing. The contents of 12 LCFAs in longissimus dorsi (LD) muscle were also determined for all experimental pigs of both pig populations. We systematically evaluated the contribution of gut microbiota to the variations of muscle fatty acid contents from the α-diversity of gut microbiota, co-abundance groups (CAGs) of Amplicon Sequence Variants (ASVs), and fatty acid-associated bacterial taxa. We identified hundred ASVs and > 40 bacterial taxa that were significantly associated with muscle fatty acid contents in two pig populations. Different numbers and bacterial taxa associated with the content of specific LCFAs in muscle were detected between cecum luminal content and fecal samples, suggesting the heterogeneity of the specific LCFA-associated bacterial taxa between two gut locations. We uncovered some interesting associations between bacterial taxa and muscle fatty acid contents. The strongest association was observed between the ASV annotated to Akkermansia and the n-6/n-3 polyunsaturated fatty acid ratio (p = 6.45E-04, Z = -9.65). The gut microbiota could explain 1.47-4.62% variation of muscle contents of twelve fatty acids. The functional prediction analysis identified that the KEGG pathways related to the metabolisms of carbohydrate and lipids, and to fat digestion and absorption were positively associated with the contents of muscle fatty acids. However, adipocytokine signaling pathway and thermogenesis were negatively associated with muscle fatty acid contents. The results from this study provided the basic knowledge for improving the muscle fatty acid contents by regulating the gut microbiome.
Collapse
Affiliation(s)
| | | | - Congying Chen
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
47
|
Wang J, Chen J, Gao M, Ouyang Z, Li Y, Liu D, Zhu M, Sun H. Research Progress on the Mechanism of Action and Screening Methods of Probiotics for Lowering Blood Lipid Levels. Foods 2025; 14:1583. [PMID: 40361665 PMCID: PMC12071596 DOI: 10.3390/foods14091583] [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: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Hyperlipidemia is one of the most prevalent metabolic disorders worldwide. It is a significant risk factor for a range of cardiovascular diseases, including acute pancreatitis, fatty liver disease, atherosclerosis, and coronary heart disease. In clinical practice, the management of hyperlipidemia is hindered by numerous challenges. One of the critical issues is that traditional lipid-lowering drugs often require long-term or even lifelong administration, potentially inducing a range of adverse effects that compromise patient compliance and therapeutic efficacy. Therefore, there is an urgent need to develop safer and more effective strategies for the prevention and adjunctive treatment of hyperlipidemia with the aim of reducing the risk of disease and over-reliance on medication. Recent studies have revealed a close relationship between hyperlipidemia and related metabolic disorders involving gut microbiota dysbiosis, and the administration of probiotics has been shown to improve lipid metabolism homeostasis. This review summarizes the molecular mechanisms of probiotics in hyperlipidemia treatment and the latest advances in probiotic research on lipid metabolism, enumerates the experimental and clinical applications of probiotic-based therapies, introduces methods for screening and identifying probiotics with lipid-lowering functions, and, for the first time, summarizes the roles of emerging technologies such as functional genomics and in vivo zebrafish-on-a-chip models in studying the lipid-lowering efficacy of probiotics, providing insights for researchers. By facilitating a deeper understanding of the mechanisms whereby probiotics reduce blood lipid levels and furthering the development of multifaceted screening methods, we hope that we can achieve high-throughput and efficient screening of probiotics with lipid-lowering functions, thereby promoting the sustainable, high-quality, and rapid development of the probiotics industry.
Collapse
Affiliation(s)
- Jingli Wang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jieyu Chen
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
| | - Mingkun Gao
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
| | - Yanhui Li
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
| | - Dong Liu
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
| | - Mingjun Zhu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China; (J.W.); (J.C.); (M.G.); (Z.O.); (Y.L.); (D.L.)
| |
Collapse
|
48
|
Huang S, Xiao H, Xiao H, Liu L, Cai S. Higher dietary live microbe intake is linked to reduced risk of metabolic syndrome and mortality: a cross-sectional and longitudinal study. Front Nutr 2025; 12:1592969. [PMID: 40365238 PMCID: PMC12069296 DOI: 10.3389/fnut.2025.1592969] [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: 03/13/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Background The association between dietary live microbe intake and metabolic syndrome (MetS) prevalence, as well as its impact on all-cause and cardiovascular disease (CVD) mortality in MetS patients, remains underexplored. Methods A total of 38,462 individuals from the National Health and Nutrition Examination Survey (NHANES) 1999-2018 were analyzed. Based on the live microbial level of the consumed foods, participants were divided into three dietary live microbe intake groups: low, medium, and high. Foods with medium and high live microbe content were aggregated into a medium-high consumption category. MetS was defined based on NCEP-ATP III criteria. Survey-weighted logistic regression assessed the cross-sectional association with MetS prevalence, while Cox proportional hazards models evaluated mortality risks in 12,432 individuals with MetS, among whom 2,641 all-cause and 901 CVD deaths occurred. Results Higher dietary live microbe intake was significantly associated with lower odds of MetS. Compared to the low intake group, participants in the high intake group had a 12% lower risk of MetS in the fully adjusted model (OR: 0.88; 95% CI: 0.80-0.97; p = 0.01). Among MetS components, significant inverse associations were observed for low HDL-C, elevated TG, and elevated BP. Participants with high dietary live microbe intake demonstrated a significantly lower risk of all-cause mortality (HR: 0.85; 95% CI: 0.77-0.94; p = 0.002) and CVD-specific mortality (HR: 0.71; 95% CI: 0.55-0.92; p = 0.001) compared to the low intake group. Kaplan-Meier survival curves revealed better survival probabilities in individuals with medium and high intake levels, with significant differences across groups (log-rank p < 0.005). Quantitatively, each 100g increase in MedHi foods was associated with 6% lower all-cause mortality (HR: 0.94; 95% CI: 0.90-0.99; p = 0.01) and 8% lower CVD mortality (HR: 0.92; 95% CI: 0.84-1.00; p = 0.05). Conclusion Dietary live microbe intake is inversely associated with the prevalence of MetS and its key components, particularly low HDL-C, elevated TG, and elevated BP. In individuals with MetS, higher live microbe intake is associated with reduced all-cause and CVD-specific mortality. These findings suggest that dietary live microbes are a promising modifiable factor for MetS prevention and management, as well as for improving long-term survival outcomes.
Collapse
Affiliation(s)
- Shan Huang
- Department of MICU, Guangdong Women and Children Hospital, Guangzhou, China
| | - Haixia Xiao
- Department of Obstetrics, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huanshun Xiao
- Department of MICU, Guangdong Women and Children Hospital, Guangzhou, China
| | - Lu Liu
- Department of Internal Medicine, Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuangming Cai
- Department of MICU, Guangdong Women and Children Hospital, Guangzhou, China
| |
Collapse
|
49
|
Ren Q, Cui C, Peng Y, Zhou Y, Zhang H, Chen L, Liu Z. Causal relationship between gut microbiota and metabolic syndrome: A bidirectional Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42179. [PMID: 40295236 PMCID: PMC12040044 DOI: 10.1097/md.0000000000042179] [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: 11/09/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic syndromes (MetS) are complex metabolic disorders, the pathogenesis of which has not been fully elucidated. In recent years, the association between the gut microbiota and MetS has attracted widespread attention, but the causal relationship remains unclear. We performed a 2-sample Mendelian randomization analysis (MR) to examine whether the gut microbiota is causally related to MetS and its components to find a basis for potential diagnostic or intervention approaches for MetS. We utilized summary statistics from whole-genome association analyses of gut microbiota from the MiBioGen consortium and obtained MetS-related data from the UK Biobank, IEU Open GWAS project, and The Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC). MR analyses were performed using inverse variance weighted, MR-Egger, and weighted median. Sensitivity analyses were conducted to verify the robustness of the results. Among the 211 gut microbiota, we identified 8 that were significantly associated with the risk of MetS. Specifically, Lachnospiraceae (family), Veillonellaceae (family), Victivallaceae (family), Odoribacter (genus), and Olsenella (genus) may increase the risk of MetS, while Bifidobacteriaceae (family), Ruminococcaceae UCG-010 (genus), Actinobacteria (phylum) may decrease the risk of MetS. Additionally, we discovered that multiple microbiota are associated with various components of MetS, such as BMI, hypertension, and blood lipid levels. This study is the first to use MR methods to reveal the potential causal relationship between specific gut microbiota and MetS, providing a new perspective for understanding the pathogenesis of MetS, and offering important evidence for the development of gut microbiota-based prevention and treatment strategies for MetS.
Collapse
Affiliation(s)
- Qiqi Ren
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Congshong Cui
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - You Peng
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yingling Zhou
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Hang Zhang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Lin Chen
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zhenjie Liu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
50
|
Wang H, Kc P, Zhang K, Materne C, Lhomme M, Galier S, Ichou F, Neves C, Lehuen A, Haas JT, Salem JE, Guerin M, Lesnik P. MAIT Cells Promote Cholesterol Excretion Pathways Mitigating Atherosclerosis. Circ Res 2025; 136:968-981. [PMID: 40135347 DOI: 10.1161/circresaha.124.325841] [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: 11/08/2024] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous clinical studies have indicated reduced circulating mucosal-associated invariant T (MAIT) cells in individuals with coronary artery disease. However, the precise role and underlying mechanisms of MAIT cells in this context remain unclear. Immune homeostasis plays a pivotal role in the development of atherosclerosis. This study explores the impact of MAIT cells on atherosclerosis. METHODS Vα19+/- Ldlr-/- mice, characterized by a high MAIT cell frequency, and MAIT cell deficient MR1-/- (major histocompatibility complex-related molecule 1) Ldlr-/- mice and their respective controls were used. Starting at 6 weeks of age, mice were subjected to a 1% cholesterol diet for 16 weeks. Additionally, the study analyzed circulating MAIT cell frequency and cholesterol levels in 68 patients with hypercholesterolemia. RESULTS In Vα19+/- Ldlr-/- mice, increased MAIT cells demonstrated a protective effect against atherosclerosis by reducing VLDL-C (very-low-density lipoprotein cholesterol) levels through heightened cholesterol excretion. This effect was accompanied by elevated jejunal ABCB1a, ABCG5, and ABCG8 expression, mediated by augmented levels of Liver X receptor transcription and activation, likely through intestinal IL-22 (interleukin-22) signaling. Conversely, cholesterol reduction mediated by intestinal cholesterol excretion was blocked by inhibition of MAIT cells. Moreover, MAIT cell-deficient MR1-/- Ldlr-/- mice exhibited elevated total cholesterol levels and increased atherosclerotic lesions. In patients with hypercholesterolemia, circulating MAIT cell frequency displayed negative correlations with VLDL-C levels and positive correlations with HDL-C (high-density lipoprotein cholesterol) levels. CONCLUSIONS Our findings demonstrate a new mechanism for plasma VLDL-C clearance by MAIT cell-mediated cholesterol excretion. The results provide further evidence that immunity is involved in cholesterol homeostasis. Targeting intestinal immunity to regulate cholesterol homeostasis holds promise as a new cholesterol-lowering modality to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hua Wang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Pukar Kc
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Kaidi Zhang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Clément Materne
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Sophie Galier
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Carolina Neves
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Agnès Lehuen
- Université Paris Cité, Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR 8104, Inflamex Laboratory, Paris, France (A.L.)
| | - Joel T Haas
- Université de Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, Lille, France (J.T.H.)
| | - Joe-Elie Salem
- INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France (J.-E.S.)
| | - Maryse Guerin
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Philippe Lesnik
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| |
Collapse
|