1
|
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
|
2
|
Akagbosu CO, McCauley KE, Namasivayam S, Romero-Soto HN, O’Brien W, Bacorn M, Bohrnsen E, Schwarz B, Mistry S, Burns AS, Perez-Chaparro PJ, Chen Q, LaPoint P, Patel A, Krausfeldt LE, Subramanian P, Sellers BA, Cheung F, Apps R, Douagi I, Levy S, Nadler EP, Hourigan SK. Gut microbiome shifts in adolescents after sleeve gastrectomy with increased oral-associated taxa and pro-inflammatory potential. Gut Microbes 2025; 17:2467833. [PMID: 39971742 PMCID: PMC11845021 DOI: 10.1080/19490976.2025.2467833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Bariatric surgery is highly effective in achieving weight loss in children and adolescents with severe obesity, however the underlying mechanisms are incompletely understood, and gut microbiome changes are unknown. Here, we show that adolescents exhibit significant gut microbiome and metabolome shifts several months after laparoscopic vertical sleeve gastrectomy (VSG), with increased alpha diversity and notably with enrichment of oral-associated taxa. To assess causality of the microbiome/metabolome changes in phenotype, pre-VSG and post-VSG stool was transplanted into germ-free mice. Post-VSG stool was not associated with any beneficial outcomes such as adiposity reduction compared pre-VSG stool. However, post-VSG stool exhibited a potentially inflammatory phenotype with increased intestinal Th17 and decreased regulatory T cells. Concomitantly, we found elevated fecal calprotectin and an enrichment of proinflammatory pathways in a subset of adolescents post-VSG. We show that in some adolescents, microbiome changes post-VSG may have inflammatory potential, which may be of importance considering the increased incidence of inflammatory bowel disease post-VSG.
Collapse
Affiliation(s)
- Cynthia O. Akagbosu
- Department of Gastroenterology, Weill Cornell Medicine, New York, New York, USA
| | - Kathryn E. McCauley
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sivaranjani Namasivayam
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hector N. Romero-Soto
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Wade O’Brien
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Qing Chen
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Phoebe LaPoint
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anal Patel
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lauren E. Krausfeldt
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian A. Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Iyadh Douagi
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Shira Levy
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Suchitra K. Hourigan
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Xu J, Igudesman D, Huckins LM, Bulik CM. Genetics of Anorexia Nervosa: Translation to Future Personalized Therapies. Psychiatr Clin North Am 2025; 48:293-309. [PMID: 40348419 DOI: 10.1016/j.psc.2025.01.007] [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] [Indexed: 05/14/2025]
Abstract
Anorexia nervosa (AN) is a debilitating and often refractory eating disorder that is unique among psychiatric disorders insofar as nutrition is key to recovery. Treatment options and efficacy are limited with no approved medications for AN. Genetic studies are clarifying the etiology of AN, with the goal of eventually informing the development of innovative personalized pharmacologic, nutritional, microbial, and behavioral interventions. We present the current state of genome-wide and epigenome-wide association studies, gut microbiome research, and functional genomics investigations and discuss translating this knowledge into clinical practice.
Collapse
Affiliation(s)
- Jiayi Xu
- Department of Psychiatry, Yale School of Medicine, 60 Temple Street, Suite 7A, New Haven, CT 06510, USA.
| | - Daria Igudesman
- AdventHealth Translational Research Institute, 301 E Princeton Street, Orlando, FL 32804, USA
| | - Laura M Huckins
- Department of Psychiatry, Yale School of Medicine, 60 Temple Street, Suite 7A, New Haven, CT 06510, USA
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, CB #7160, 101 Manning Drive, Chapel Hill, NC 27599-7160, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Zheng L, Lu Z, Ma Y, Cui P, Zhang X, Gan J, Li G. Hawthorn total flavonoids ameliorate hyperlipidemia through AMPK/SREBP1-c and PPARα/PGC-1α/CPT-1A pathway activation and gut microbiota modulation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4326-4337. [PMID: 40013442 DOI: 10.1002/jsfa.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND The increased prevalence of hyperlipidemia significantly affects human health worldwide. Although drug treatment is very effective, the harm to the human body cannot be ignored. Improvement of lipid metabolism by natural medicinal and food homologous products is an effective approach to ameliorate hyperlipidemia and it has gradually become a research focus. In this research, we adopted HepG2 cell models and high-fat-diet-fed C57BL/6j mouse models to explore the effect of hawthorn total flavonoids (HTF) on hyperlipidemia. Moreover, we utilized western blot and gut microbiota analysis to elucidate the specific mechanism of HTF's influence on hyperlipidemia. RESULTS We found that HTF significantly alleviated hyperlipidemia and its complications, as manifested by reduced body weight gain and fat accumulation, and improved the disorder of intestinal microorganisms. HTF protected the liver, reducing aspartate transaminase and lactate dehydrogenase levels, and ameliorating inflammatory infiltration. Fat droplet amounts and necrotic cell numbers in liver cells were also decreased. Mechanistically, HTF promoted AMP-activated protein kinase phosphorylation, inhibited sterol regulatory element binding protein 1c expression, downregulating the expression of lipid synthesis-related proteins (acetyl CoA carboxylase, fatty acid synthase, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase), thus suppressing liver lipid synthesis. HTF also functioned as a natural peroxisome proliferator-activated receptor α (PPARα) agonist. Activated PPARα enhanced mitochondrial oxidation and lipid consumption via upregulating carnitine palmitoyltransferase 1A. Peroxisome proliferator-activated receptor-γ coactivator expression was also elevated, activating mitochondrial activity, increasing cholesterol 7α-hydroxylase activity and cholesterol consumption, and reducing blood lipids. Additionally, HTF regulated intestinal flora abundance, restored the ratio of Firmicutes to Bacteroidetes, balanced gut-liver axis crosstalk, and alleviated hyperlipidemia. CONCLUSION The results demonstrated that HTF alleviated the pathological symptoms caused by hyperlipidemia, and had a certain protective effect on the liver. HTF also stimulated the lipid metabolism pathway and accelerated lipid consumption. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Liping Zheng
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Zhihao Lu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Yurong Ma
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Penglei Cui
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Xinxue Zhang
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| | - Jing Gan
- College of Life Science, Yantai University, Yantai, China
| | - Guoming Li
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing, China
| |
Collapse
|
7
|
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
|
8
|
Metwaly A, Kriaa A, Hassani Z, Carraturo F, Druart C, Arnauts K, Wilmes P, Walter J, Rosshart S, Desai MS, Dore J, Fasano A, Blottiere HM, Maguin E, Haller D. A Consensus Statement on establishing causality, therapeutic applications and the use of preclinical models in microbiome research. Nat Rev Gastroenterol Hepatol 2025; 22:343-356. [PMID: 40033063 DOI: 10.1038/s41575-025-01041-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
The gut microbiome comprises trillions of microorganisms and profoundly influences human health by modulating metabolism, immune responses and neuronal functions. Disruption in gut microbiome composition is implicated in various inflammatory conditions, metabolic disorders and neurodegenerative diseases. However, determining the underlying mechanisms and establishing cause and effect is extremely difficult. Preclinical models offer crucial insights into the role of the gut microbiome in diseases and help identify potential therapeutic interventions. The Human Microbiome Action Consortium initiated a Delphi survey to assess the utility of preclinical models, including animal and cell-based models, in elucidating the causal role of the gut microbiome in these diseases. The Delphi survey aimed to address the complexity of selecting appropriate preclinical models to investigate disease causality and to study host-microbiome interactions effectively. We adopted a structured approach encompassing a literature review, expert workshops and the Delphi questionnaire to gather insights from a diverse range of stakeholders. Experts were requested to evaluate the strengths, limitations, and suitability of these models in addressing the causal relationship between the gut microbiome and disease pathogenesis. The resulting consensus statements and recommendations provide valuable insights for selecting preclinical models in future studies of gut microbiome-related diseases.
Collapse
Affiliation(s)
- Amira Metwaly
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany
| | - Aicha Kriaa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Federica Carraturo
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | | - Kaline Arnauts
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jens Walter
- APC Microbiome Ireland, School of Microbiology, and Department of Medicine, University College Cork, Cork, Ireland
| | - Stephan Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Joel Dore
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
- Department of Paediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center,Massachusetts General Hospital Brigham, Harvard Medical School, Boston, MA, USA
| | - Hervé M Blottiere
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
- Nantes Université, INRAE, UMR1280, PhAN, Nantes, France
| | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| | - Dirk Haller
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany.
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany.
| |
Collapse
|
9
|
Yang B, Chen S, Xia X, Tao Z, Liu C, Li S, Zhang S, Huang J, Xia L, Quan W, Yang C, Li J. Mas Signaling Potentiates Neutropil Extracellular Traps Formation Induced by Endothelial Cells Derived S1P in Mice with Acute Liver Failure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411428. [PMID: 40285622 DOI: 10.1002/advs.202411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Mas, a newly identified G-protein-coupled receptor, is prevalent in myeloid-derived immune cells and plays a key role in inflammation. This study investigates Mas signaling and neutrophil extracellular traps (NETs) in acute liver failure (ALF), aiming to elucidate their mechanisms. Male Mas1-/- and wild-type mice, aged 6-8 weeks, receive intraperitoneally injected with lipopolysaccharide (LPS)/D-galactosamine (D-Gal) (L/G) to study NETs formation. Hepatic Mas expression increases in WT-L/G mice, whereas systemic Mas1 knockout significantly reduces L/G-induced NETs and hepatotoxicity. Antibiotics treatment and co-housing (Mas1-/--L/G and WT-L/G mice) experiments show that gut flora influences the disease phenotype in Mas1-/--L/G mice. Fecal metabolite analysis suggests that mice may be protected by reduced deoxycholic acid (DCA) production in Mas1-/- activated hepatic farnesoid X receptor (FXR), suppressing sphingosine-1-phosphate (S1P)-dependent NETs. Additionally, Mas1-/- also activates the FXR-S1P-NETs axis in the liver by inhibiting SHP2. Single-cell sequencing shows decreased interaction between endothelial cells and Cldn1+CD177+ senescent neutrophils through Col4a1-CD44. This inhibits S1P-induced Raf signaling pathway activation and NETs formation. Mas signaling significantly impacts NETs formation, highlighting its potential as an anti-inflammatory therapeutic target for ALF.
Collapse
Affiliation(s)
- Bo Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuai Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoqi Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ziwen Tao
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Chun Liu
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shanshan Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuo Zhang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jiali Huang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lu Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wenqiang Quan
- Department of Laboratory Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Changqing Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
10
|
Kong Y, Yang H, Nie R, Zhang X, Zuo F, Zhang H, Nian X. Obesity: pathophysiology and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:25. [PMID: 40278960 PMCID: PMC12031720 DOI: 10.1186/s43556-025-00264-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Over the past few decades, obesity has transitioned from a localized health concern to a pressing global public health crisis affecting over 650 million adults globally, as documented by WHO epidemiological surveys. As a chronic metabolic disorder characterized by pathological adipose tissue expansion, chronic inflammation, and neuroendocrine dysregulation that disrupts systemic homeostasis and impairs physiological functions, obesity is rarely an isolated condition; rather, it is frequently complicated by severe comorbidities that collectively elevate mortality risks. Despite advances in nutritional science and public health initiatives, sustained weight management success rates and prevention in obesity remain limited, underscoring its recognition as a multifactorial disease influenced by genetic, environmental, and behavioral determinants. Notably, the escalating prevalence of obesity and its earlier onset in younger populations have intensified the urgency to develop novel therapeutic agents that simultaneously ensure efficacy and safety. This review aims to elucidate the pathophysiological mechanisms underlying obesity, analyze its major complications-including type 2 diabetes mellitus (T2DM), cardiovascular diseases (CVD), non-alcoholic fatty liver disease (NAFLD), obesity-related respiratory disorders, obesity-related nephropathy (ORN), musculoskeletal impairments, malignancies, and psychological comorbidities-and critically evaluate current anti-obesity strategies. Particular emphasis is placed on emerging pharmacological interventions, exemplified by plant-derived natural compounds such as berberine (BBR), with a focus on their molecular mechanisms, clinical efficacy, and therapeutic advantages. By integrating mechanistic insights with clinical evidence, this review seeks to provide innovative perspectives for developing safe, accessible, and effective obesity treatments.
Collapse
Affiliation(s)
- Yue Kong
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | - Rong Nie
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuxiang Zhang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fan Zuo
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | - Xin Nian
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
11
|
Creus-Martí I, Moya A, Santonja FJ. Methodology for microbiome data analysis: An overview. Comput Biol Med 2025; 192:110157. [PMID: 40279974 DOI: 10.1016/j.compbiomed.2025.110157] [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: 06/30/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
It is known that microbiome and health are related, in addition, recent research has found that microbiome has potential clinical uses. These facts highlight the importance of the microbiome in actual science. However, microbiome data has some characteristics that makes its statistical study challenging. In recent years, longitudinal and non-longitudinal methods have been designed to analyze the microbiota and knowing more about the bacterial behavior. In this article in the form of a review we summarize the characteristics of microbiome data and the statistical methods most widespread to analyze it. We have taken into account if the strategies are longitudinal or not. We also classify the methods based on their specific analytical objectives and based on their mathematical characteristics. The methods are structured according to their biological goals and mathematical features, ensuring that the insights provided are both relevant and accessible to professionals in biology and statistics. We present this review as a reference for the most widely used methods in microbiome data analysis and as a foundation for identifying potential areas for future research. We want to point out that this review can be particularly useful to remark the importance of the methodology designed in order to study microbiome longitudinal datasets.
Collapse
Affiliation(s)
- Irene Creus-Martí
- Department of Applied Mathematics, Universitat Politècnica de València, Valencia, Spain.
| | - Andrés Moya
- Institute for Integrative Systems Biology (I2Sysbio), Universitat de València and CSIC, València, Spain; The Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO), Valencia, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Francisco J Santonja
- Department of Statistics and Operation Research, Universitat de València, Valencia, Spain
| |
Collapse
|
12
|
Zeng X, Yu P, Li D, Li Y, Wang X, Yang X, Ren D. Structural characterization and alleviative effects of novel polysaccharides from Artemisia sphaerocephala Krasch seed on obese mice by regulating gut microbiota. Int J Biol Macromol 2025; 310:143407. [PMID: 40274139 DOI: 10.1016/j.ijbiomac.2025.143407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
This study aimed to investigate the efficacy of polysaccharides from Artemisia sphaerocephala Krasch (ASK) seed in alleviating high fat diet (HFD) caused obesity. Here, three polysaccharide fractions (ASKP1, ASKP2 and ASKP3) were purified from ASK seed. Chemical characteristic analysis revealed that ASKP1 is a neutral heteropolysaccharide with the average molecular weight of 9.08 × 105 Da, while ASKP2 and ASKP3 are acidic heteropolysaccharides with the molecular weight of 9.39 × 105 and 8.41 × 105 Da, respectively. Animal experiment found that three ASKP fractions obviously relieved obesity and related metabolic disorders induced by HFD, while ASKP1 was more effective in reducing the blood glucose and serum LDL levels. 16S rDNA sequencing showed that ASKP fractions improved the gut microbiota imbalance of obese mice, and ASKP1 promoted the proliferation of beneficial bacterium Akkermansia more effectively than ASKP2 and ASKP3. Furthermore, ASKP fractions facilitated thermogenesis of brown adipose tissue (BAT) of obese mice, as evidenced by increased expression of thermogenic marker genes UCP1 in BAT, and the thermogenesis effect of ASKP1 was the most obvious. Taken together, our results show that ASKP1 is a novel prebiotic that may be used to treat obesity and its related abnormal metabolism.
Collapse
Affiliation(s)
- Xiaoqian Zeng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Pinglian Yu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; Key Laboratory of YunNan University for Plateau Characteristic Functional Food, School of Chemistry and Chemical Engineering, Zhaotong University, 657000, China.
| | - Donglu Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yixiao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xuejie Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
13
|
Corbin KD, Igudesman D, Smith SR, Zengler K, Krajmalnik-Brown R. Targeting the Gut Microbiota's Role in Host Energy Absorption With Precision Nutrition Interventions for the Prevention and Treatment of Obesity. Nutr Rev 2025:nuaf046. [PMID: 40233201 DOI: 10.1093/nutrit/nuaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025] Open
Abstract
The field of precision nutrition aims to develop dietary approaches based on individual biological factors such as genomics or the gut microbiota. The gut microbiota, which is the highly individualized and complex community of microbes residing in the colon, is a key contributor to human physiology. Although gut microbes play multiple roles in the metabolism of nutrients, their role in modulating the absorption of dietary energy from foods that escape digestion in the small intestine has the potential to variably affect energy balance and, thus, body weight. The fate of this energy, and its subsequent impact on body weight, is well described in rodents and is emerging in humans. This narrative review is focused on recent clinical evidence of the role of the gut microbiota in human energy balance, specifically its impact on energy available to the human host. Despite recent progress, remaining gaps in knowledge present opportunities for developing and implementing strategies to understand causal microbial mechanisms related to energy balance. We propose that implementing rigorous microbiota-focused measurements in the context of innovative clinical trial designs will elucidate integrated diet-host-gut microbiota mechanisms. These mechanisms are primed to be targets for precision nutrition interventions to optimize energy balance to achieve desired weight outcomes. Given the magnitude and impact of the obesity epidemic, implementing these interventions within comprehensive weight management paradigms has the potential to be of public health significance.
Collapse
Affiliation(s)
- Karen D Corbin
- AdventHealth Translational Research Institute, Orlando, FL 32804, United States
| | - Daria Igudesman
- AdventHealth Translational Research Institute, Orlando, FL 32804, United States
| | - Steven R Smith
- AdventHealth Translational Research Institute, Orlando, FL 32804, United States
| | - Karsten Zengler
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, United States
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA 92093, United States
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health through Microbiomes, Arizona State University, Tempe, AZ 85281, United States
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ 85281, United States
| |
Collapse
|
14
|
Boicean A, Ichim C, Sasu SM, Todor SB. Key Insights into Gut Alterations in Metabolic Syndrome. J Clin Med 2025; 14:2678. [PMID: 40283508 PMCID: PMC12028006 DOI: 10.3390/jcm14082678] [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: 03/20/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Over time, extensive research has underscored the pivotal role of gut microbiota in the onset and progression of various diseases, with a particular focus on fecal microbiota transplantation (FMT) as a potential therapeutic approach. The practice of transferring fecal matter from a healthy donor to a patient provides valuable insights into how alterations in gut microbiota can impact disease development and how rectifying dysbiosis may offer therapeutic benefits. Re-establishing a balanced symbiotic relationship in the gastrointestinal tract has shown positive results in managing both intestinal and systemic conditions. Currently, one of the most pressing global health issues is metabolic syndrome-a cluster of conditions that includes insulin resistance, lipid imbalances, central obesity and hypertension. In this context, FMT has emerged as a promising strategy for addressing key components of metabolic syndrome, such as improving insulin sensitivity, body weight and lipid profiles. However, further well-structured studies are needed to refine treatment protocols and establish the long-term safety and efficacy of this intervention.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | - Sabina-Maria Sasu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | | |
Collapse
|
15
|
Poulsen CE, Vinding R, Rasmussen MA, Shah S, Trivedi U, Rodriguez CL, Widdowson ML, Jiang J, Poulsen CS, Eliasen A, Chawes B, Bønnelykke K, Hansen CHF, Sørensen SJ, Thorsen J, Stokholm J. No association between the early-life gut microbiota and childhood body mass index and body composition. MED 2025; 6:100538. [PMID: 39536756 DOI: 10.1016/j.medj.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The gut microbiota has been implicated in adult obesity, but the causality is still unclear. It has been hypothesized that an obesity-prone gut microbiota can be established in infancy, but only few studies have examined the early-life gut microbiota in relation to obesity in childhood, and no consistent associations have been reported. Here, we examine the association between the early-life gut microbiota and body mass index (BMI) development and body composition throughout childhood. METHODS Gut microbiota from stool were collected from 700 children in the Copenhagen Prospective Studies on Asthma in Childhood2010 (COPSAC2010) cohort at ages of 1 week, 1month, 1 year, 4 years, and 6 years and analyzed by 16S rRNA gene sequencing. Outcomes included BMI World Health Organization (WHO) Z scores (zBMI), overweight (zBMI > 1.04) and obesity (zBMI > 1.64) (0-10 years), and adiposity rebound and body composition from dual-energy X-ray absorptiometry at 6 years. FINDINGS The early-life gut microbiota diversity, overall composition, and individual taxon abundances in unsupervised and supervised models were not consistently associated with either current or later BMI Z scores, overweight, obesity, adiposity rebound, or body composition in childhood. CONCLUSIONS In a deeply characterized longitudinal birth cohort, we did not observe any consistent associations between the early-life gut microbiota and BMI or risk of obesity in later childhood. While this does not conclusively rule out a relationship, it suggests that if such associations exist, they may be more complex and potentially influenced by factors emerging later in life, including lifestyle changes. FUNDING COPSAC is funded by private and public research funds (all listed on www.copsac.com).
Collapse
Affiliation(s)
- Christina Egeø Poulsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Vinding
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Morten A Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | - Shiraz Shah
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Urvish Trivedi
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Leal Rodriguez
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Michael L Widdowson
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jie Jiang
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Casper S Poulsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Eliasen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Camilla H F Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Søren J Sørensen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| |
Collapse
|
16
|
Yang SJ, Yu XK, Zuo Q. Branched- Chain Fatty Acids and Obesity: A Narrative Review. Nutr Rev 2025:nuaf022. [PMID: 40207993 DOI: 10.1093/nutrit/nuaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Branched- chain fatty acids (BCFAs) are a category of saturated fatty acids that are commonly present in various organisms and play a crucial role in a variety of metabolic reactions, including anticancer, lipid-lowering, anti-inflammatory, and neuroprotective actions. Currently, there is growing interest in the relationship between BCFAs and obesity. Branched- chain fatty acids regulate the gene expression of related enzymes by activating PPARα and sterol regulatory element-binding protein-1c, thereby reducing triglyceride synthesis in the body. Additionally, BCFAs reduce inflammation by decreasing the expression of pro-inflammatory factors in obesity such as cyclooxygenase-2, interleukin-6, and lipoxygenase-15 genes. Branched- chain fatty acids can also expedite the conversion of branched chain amino acids to BCFAs to regulate obesity-induced insulin resistance. In this article we provide a comprehensive review of research progress on how BCFAs affect obesity from the perspectives of lipid metabolism, inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Shi-Jiao Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xin-Kai Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Qun Zuo
- School of Sports Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
17
|
Zhang Z, Jiang C, Xing YQ, Yang T, Zou L, Jia Z, Zhao L, Han X, Qu X, Zhang Z, Zong J, Wang S. Unveiling the interplay among skin microbiota, cytokines, and T2DM: an insightful Mendelian randomization study. Nutr Metab (Lond) 2025; 22:29. [PMID: 40211330 PMCID: PMC11987181 DOI: 10.1186/s12986-025-00922-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/26/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Previous observational studies have indicated a correlation between the skin microbiome and Type 2 diabetes (T2DM). It is hypothesized that this causal relationship may be influenced by inflammatory responses. However, these factors as determinants of T2DM remain largely unexplored. METHOD This study incorporated data from the GWAS database on the skin microbiome, 91 types of inflammatory cytokines, and T2DM. We employed two-sample MR and multivariable MR methods to assess the correlation between the skin microbiome and T2DM, and to investigate whether this correlation is affected by inflammatory cytokines. RESULTS The results of the two-sample MR analysis indicate that within the skin microbiome, genetically predicted genus: Acinetobacter, class: Alphaproteobacteria, genus: Bacteroides, ASV005[Propionibacterium granulosum], and ASV072[Rothia mucilaginosa] are associated with an increased risk of T2DM, while phylum: Proteobacteria, genus: Enhydrobacter, family: Clostridiales, ASV006[Staphylococcus hominis] serve as protective factors against T2DM. Among the inflammatory cytokines, levels of Macrophage colony-stimulating factor 1, Tumor necrosis factor receptor superfamily member 9, Urokinase-type plasminogen activator, and C-C motif chemokine 28 are associated with an increased risk of T2DM. Multivariable MR analysis further revealed that Macrophage colony-stimulating factor 1 levels act as a mediating factor between ASV072[Rothia mucilaginosa] and T2DM. CONCLUSION In this study, we found a connection between the skin microbiome and T2DM, with inflammatory cytokines playing a key role in this relationship. This research helps us better understand this complex link and shows that addressing inflammation is important for preventing and treating diabetes. This could greatly benefit public health by reducing the impact of diabetes and its complications. Our results suggest that future studies should explore the specific biological interactions between the skin microbiome and diabetes to develop more effective risk management and treatment strategies from a microbial perspective.
Collapse
Grants
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 82074426, 82104864, 82204822 National Natural Science Foundation of China
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2023JH2/101300096 Applied Basic Research Project of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- 2021-BS-215, 2022-MS-25, 2023-MS-13 Natural Science Foundation of Liaoning Province
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- XLYC1802014 Liaoning Revitalization Talents Program
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- 2017226015 Liaoning Key Research and Development Planning Project
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- LJKMZ20221286 Basic Research Projects of Liaoning Provincial Department of Education
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
- XZ202301ZR0030G, XZ2023ZR-ZY82(Z) Natural Science Foundation of Tibet Autonomous Region
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
- College of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Chunyu Jiang
- Department of Trauma Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi-Qi Xing
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianke Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Linxuan Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhuqiang Jia
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Quality Management, Dalian Municipal Central Hospital, Dalian, China
| | - Xin Han
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xueling Qu
- Pelvic Floor Repair Center, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Zhen Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Junwei Zong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Shouyu Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
18
|
Boshuizen B, De Maré L, Oosterlinck M, Van Immerseel F, Eeckhaut V, De Meeus C, Devisscher L, Vidal Moreno de Vega C, Willems M, De Oliveira JE, Hosotani G, Gansemans Y, Meese T, Van Nieuwerburgh F, Deforce D, Vanderperren K, Verdegaal EL, Delesalle C. Aleurone supplementation enhances the metabolic benefits of training in Standardbred mares: impacts on glucose-insulin dynamics and gut microbiome composition. Front Physiol 2025; 16:1565005. [PMID: 40276369 PMCID: PMC12018385 DOI: 10.3389/fphys.2025.1565005] [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: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Aleurone, derived from the bran layer of grains like wheat and barley, has demonstrated positive effects on energy metabolism in pigs, mice, and untrained horses, influencing glucose-insulin dynamics and gut microbiome composition. Training itself enhances insulin sensitivity in horses, similar to the improvements in performance capacity observed in human athletes. This study aimed to investigate whether aleurone supplementation provides additional benefits to training by modulating insulin metabolism and gut microbiota in Standardbred mares. Methods Sixteen Standardbred mares (aged 3-5 years) participated in a cross-over study with two 8-week training periods separated by 8 weeks of detraining. Each horse received either 200 g/day aleurone supplementation or a control diet. Insulin metabolism was evaluated using oral (OGTT) and intravenous (FSIGTT) glucose tolerance tests, measuring parameters such as Maximumglucose, AUCglucose, Maximuminsulin, AUCinsulin, Time to peakinsulin (OGTT), Acute Insulin Response to Glucose (AIRg), glucose effectiveness (Sg), and disposition index (DI) (FSIGTT). Fecal samples underwent metagenomic analysis to assess alpha and beta diversity and microbial composition. Results Training alone: Training significantly improved OGTT parameters by decreasing Maximuminsulin (P = 0.005) and AUCinsulin (P = 0.001), while increasing Time to peakinsulin (P = 0.03), indicating enhanced insulin sensitivity. FSIGTT results also showed a decrease in logAIRg (P = 0.044). Training with Aleurone: Aleurone supplementation further reduced FSIGTT AIRg (P = 0.030), logAIRg (P = 0.021) while increasing glucose effectiveness (Sg; P = 0.031). These findings suggest aleurone improves insulin sensitivity, glucose disposal, and fasting glucose regulation beyond training. Microbiome analysis revealed training decreased Pseudomonas, associated with dysbiosis, while aleurone reduced inflammation-associated Desulfovibrio. Beta diversity metrics showed no significant changes. Conclusion Aleurone supplementation enhances training-induced improvements in glucose metabolism and fecal microbiota composition, which could offer potential benefits for equine athletes by optimizing metabolic flexibility. It also supports improvements in glucose and insulin dynamics, particularly by further enhancing insulin sensitivity and glucose-mediated disposal. Future studies should investigate the mechanisms of aleurone at the muscle and gut level and explore its potential applications for metabolic disorders such as Equine Metabolic Syndrome.
Collapse
Affiliation(s)
- Berit Boshuizen
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Equine Hospital Wolvega, Oldeholtpade, Netherlands
| | - Lorie De Maré
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Maarten Oosterlinck
- Department of Large Animal Surgery, Anaesthesia and Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Filip Van Immerseel
- Department of Pathobiology, Pharmacology and Special Animals Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Venessa Eeckhaut
- Department of Pathobiology, Pharmacology and Special Animals Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Constance De Meeus
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Lindsey Devisscher
- Gut-Liver ImmunoPharmacology Unit, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Carmen Vidal Moreno de Vega
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Maarten Willems
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Yannick Gansemans
- Department of Pharmaceutics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Tim Meese
- Department of Pharmaceutics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Department of Pharmaceutics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Dieter Deforce
- Department of Pharmaceutics, Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
| | - Katrien Vanderperren
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Elisabeth-Lidwien Verdegaal
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Equine Health and Performance Centre, School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Adelaide, SA, Australia
| | - Cathérine Delesalle
- Department of Translational Physiology, Infectiology and Public Health, Research Group of Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Equine Health and Performance Centre, School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
19
|
Wu Y, Mo J, Wang Q, Li J, Wei J, Zhang N, Dong Y, Zhu X, Lu T, Huang S. Microbiome and metabolome explain the high-fat diet-induced diabetes development and diabetes resistance in Guizhou mini-pigs. Front Microbiol 2025; 16:1555069. [PMID: 40291804 PMCID: PMC12023756 DOI: 10.3389/fmicb.2025.1555069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an obesity-related disease claiming substantial global mortality annually. Current animal models of T2DM remain limited, with low success rates in establishing porcine models of high-fat diet (HFD)-induced T2DM. Our experimental design employed 35 Guizhou mini-pigs to develop a T2DM model via HFD induction, aiming to identify microbial and metabolic signatures associated with disease pathogenesis and resistance. At month 10, five individuals from the control (CTR), T2DM (DM), and T2DM resistant (anti-DM) groups were slaughtered, samples were collected, and relevant indices were measured. Metagenomics, metabolomics, and 16S rRNA sequencing were performed to identify microbes and metabolites linked to T2DM progression and resistance. Key findings demonstrated anti-DM group parameters-including metabolic indices (fasting blood glucose, insulin levels, HbA1c, IVGTT), histopathology (HE-stained pancreatic/hepatic tissues), microbial profiles (structural, compositional, functional), and metabolomic signatures-occupied intermediate positions between CTR and DM groups. Network analyses revealed: (1) Lactobacillus, L. amylovorus, fingolimod, polyoxyethylene sorbitan monooleate, thiamine, and atrazine in HFD-associated networks; (2) Limosilactobacillus reuteri, N-oleoyl-L-serine, tolbutamide, tetradecanoyl carnitine, 3'-sulfogalactosylceramide, and guggulsterone in T2DM resistance networks; (3) Ruminococcaceae NK4A214 group, diethyl phthalate, zingerone, enalapril, 5-hydroxytryptophol, 2'-deoxyinosine, icariin, and emetine in T2DM progression networks. These results further clarify the role of the gut microbiota and serum metabolites in the development of T2DM in the Guizhou mini-pig model.
Collapse
Affiliation(s)
- Yanjun Wu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jiayuan Mo
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Engineering Technology Research Center of Pork Quality Control and Enhance, Anhui Science and Technology University, Chuzhou, China
| | - Qianguang Wang
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jialong Li
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jia Wei
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Nuo Zhang
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yuanqiu Dong
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiang Zhu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Taofeng Lu
- The Provincial Key Miao Medicine Laboratory of Guizhou, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Sicheng Huang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University/ Guizhou Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guiyang, China
- Department of Abdominal Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
20
|
Schell LD, Carmody RN. An energetic framework for gut microbiome-mediated obesity induced by early-life exposure to antibiotics. Cell Host Microbe 2025; 33:470-483. [PMID: 40209676 DOI: 10.1016/j.chom.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/15/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
Early-life antibiotic (ELA) exposure has garnered attention for its potential role in modulating obesity risk, although outcomes from mouse experiments and human epidemiological studies often vary based on dosage and sex. Low-dose (subtherapeutic) antibiotics can enhance energy availability through moderate alterations in gut microbiome profile, while high-dose (therapeutic) antibiotics substantially deplete the gut microbiota, thereby contributing to short-term negative energy balance. In this perspective, we propose a framework to understand how these distinct impacts of antibiotics on the gut microbiome during critical developmental windows shape long-term obesity risk through their influence on host energy balance. Using this framework, we then propose several hypotheses to explain variation in ELA-induced obesity outcomes across males and females. We conclude by discussing the evolutionary implications of ELAs, positing that the response of the gut microbiome to ELAs may signal energy availability and environmental volatility, influencing metabolic programming and adaptive traits across generations.
Collapse
Affiliation(s)
- Laura D Schell
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
21
|
Xiong S. Gut-Microbiota-Driven Lipid Metabolism: Mechanisms and Applications in Swine Production. Metabolites 2025; 15:248. [PMID: 40278377 PMCID: PMC12029090 DOI: 10.3390/metabo15040248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: The gut microbiota plays a pivotal role in host physiology through metabolite production, with lipids serving as essential biomolecules for cellular structure, metabolism, and signaling. This review aims to elucidate the interactions between gut microbiota and lipid metabolism and their implications for enhancing swine production. Methods: We systematically analyzed current literature on microbial lipid metabolism, focusing on mechanistic studies on microbiota-lipid interactions, key regulatory pathways in microbial lipid metabolism, and multi-omics evidence (metagenomic/metabolomic) from swine models. Results: This review outlines the structural and functional roles of lipids in bacterial membranes and examines the influence of gut microbiota on the metabolism of key lipid classes, including cholesterol, bile acids, choline, sphingolipids, and fatty acids. Additionally, we explore the potential applications of microbial lipid metabolism in enhancing swine production performance. Conclusions: Our analysis establishes a scientific framework for microbiota-based strategies to optimize lipid metabolism. The findings highlight potential interventions to improve livestock productivity through targeted manipulation of gut microbial communities.
Collapse
Affiliation(s)
- Shuqi Xiong
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
22
|
Roessler J, Zimmermann F, Heidecker B, Landmesser U, Haghikia A. Gut microbiota-related modulation of immune mechanisms in post-infarction remodelling and heart failure. ESC Heart Fail 2025; 12:942-954. [PMID: 39385474 PMCID: PMC11911630 DOI: 10.1002/ehf2.14991] [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: 05/03/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
The immune system has long been recognized as a key driver in the progression of heart failure (HF). However, clinical trials targeting immune effectors have consistently failed to improve patient outcome across different HF aetiologies. The activation of the immune system in HF is complex, involving a broad network of pro-inflammatory and immune-modulating components, which complicates the identification of specific immune pathways suitable for therapeutic targeting. Increasing attention has been devoted to identifying gut microbial pathways that affect cardiac remodelling and metabolism and, thereby impacting the development of HF. In particular, gut microbiota-derived metabolites, absorbed by the host and transported to the peripheral circulation, can act as signalling molecules, influencing metabolism and immune homeostasis. Recent reports suggest that the gut microbiota plays a crucial role in modulating immune processes involved in HF. Here, we summarize recent advances in understanding the contributory role of gut microbiota in (auto-)immune pathways that critically determine the progression or alleviation of HF. We also thoroughly discuss potential gut microbiota-based intervention strategies to treat or decelerate HF progression.
Collapse
Affiliation(s)
- Johann Roessler
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Friederike Zimmermann
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| | - Arash Haghikia
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| |
Collapse
|
23
|
Khavani M, Mehranfar A, Mofrad MRK. Unravelling the Glycan Code: Molecular Dynamics and Quantum Chemistry Reveal How O-Glycan Functional Groups Govern OgpA Selectivity in Mucin Degradation by Akkermansia muciniphila. Microb Biotechnol 2025; 18:e70091. [PMID: 40181232 PMCID: PMC11968330 DOI: 10.1111/1751-7915.70091] [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: 10/19/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 04/05/2025] Open
Abstract
Mucins, heavily O-glycosylated glycoproteins, are a key component of mucus, and certain gut microbiota, including Akkermansia muciniphila, can utilise mucin glycans as a carbon source. Akkermansia muciniphila produces the O-glycopeptidase enzyme OgpA, which cleaves peptide bonds at the N-terminus of serine (Ser) or threonine (Thr) residues carrying O-glycan substitutions, with selectivity influenced by the O-glycan functional groups. Using molecular dynamics (MD) simulations and quantum chemistry calculations, we explored how different O-glycan groups affect OgpA's selectivity. Our results show that peptides bind to the enzyme via hydrogen bonds, π-π interactions, van der Waals forces and electrostatic interactions, with key residues, including Tyr90, Val138, Gly176, Tyr210 and Glu91, playing important roles. The primary determinant of selectivity is the interaction between the peptide's functional group and the enzyme's binding cavity, while peptide-enzyme interface interactions are secondary. Quantum chemistry calculations reveal that OgpA prefers peptides with a lower electrophilic character. This study provides new insights into mucin degradation by gut microbiota enzymes, advancing our understanding of this critical biological process.
Collapse
Affiliation(s)
- Mohammad Khavani
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Aliyeh Mehranfar
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Mohammad R. K. Mofrad
- Molecular Cell Biomechanics Laboratory, Department of Bioengineering and Mechanical EngineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrative Bioimaging DivisionLawrence Berkeley National LabBerkeley, CAUSA
| |
Collapse
|
24
|
Amato KR, Back JP, Sardaro MLS, Bicca‐Marques JC. Supplementation With Human Foods Affects the Gut Microbiota of Wild Howler Monkeys. Am J Primatol 2025; 87:e70029. [PMID: 40159691 PMCID: PMC11955745 DOI: 10.1002/ajp.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025]
Abstract
Wild primates face a wide range of anthropogenic influences globally that impact their health, fitness, and survival. One area of potential impact that has been particularly understudied is the supplementation of wild primate diets with human foods. Although the consumption of human foods represents a substantial dietary change for wild primates, knowledge of how it impacts their physiology and behavior is limited. Here we explore how human food supplementation impacts wild primates by comparing the gut microbiomes of free-ranging brown howler monkeys (Alouatta guariba) in periurban Brazil that do or do not have access to human foods. We found that howler monkeys consuming human foods had reduced gut microbial diversity and reduced relative abundances of fiber degrading microbial taxa, which has been associated with negative health consequences in other animals, including humans. However, the effect size of these differences was relatively small and varied over time. Additionally, the composition of the gut microbiome varied significantly across months, regardless of the access to human foods. We suggest that the biology of this howler monkey population is minimally impacted by human foods. Further empirical research will help clarify the relationship between human food supplementation and health across primate populations, facilitating conservation applications.
Collapse
Affiliation(s)
| | - Janaína P. Back
- Laboratório de Primatologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do SulPorto AlegreBrazil
| | - Maria Luisa Savo Sardaro
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
- Department of Human Science and Promotion of the Quality of LifeUniversity of San RaffaeleRomeItaly
| | - Júlio César Bicca‐Marques
- Laboratório de Primatologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do SulPorto AlegreBrazil
| |
Collapse
|
25
|
Wesener DA, Beller ZW, Hill MF, Yuan H, Belanger DB, Frankfater C, Terrapon N, Henrissat B, Rodionov DA, Leyn SA, Osterman A, van Hylckama Vlieg JET, Gordon JI. In vivo manipulation of human gut Bacteroides fitness by abiotic oligosaccharides. Nat Chem Biol 2025; 21:544-554. [PMID: 39443715 PMCID: PMC11949833 DOI: 10.1038/s41589-024-01763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/27/2024] [Indexed: 10/25/2024]
Abstract
Synthetic glycans (SGs) containing glycosidic linkages and structures not identified in nature offer a means for deliberately altering microbial community properties. Here pools of SG oligosaccharides were generated via polymerization of monosaccharides and screened for their ability to increase saccharolytic Bacteroides in ex vivo cultures of human fecal samples. A lead SG preparation was orally administered to gnotobiotic mice harboring a consortium of 56 cultured, phylogenetically diverse human gut bacteria and fed a Western diet. The abundances of 3 of 15 Bacteroides strains increased, most prominently B. intestinalis. Underlying mechanisms were characterized by analyzing in vivo expression of the carbohydrate utilization machinery, using retrievable microscopic paramagnetic particles with bound SG oligosaccharides and assaying SG degradation by individual purified B. intestinalis glycoside hydrolases. The results reveal that SGs can selectively co-opt carbohydrate utilization machinery in different human gut Bacteroides and demonstrate a means for identifying artificial carbohydrate structures for targeted bacterial manipulation.
Collapse
Affiliation(s)
- Darryl A Wesener
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Microbiology, The Ohio State University, Columbus, OH, USA.
| | - Zachary W Beller
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan F Hill
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Han Yuan
- Kaleido Biosciences, Lexington, MA, USA
| | | | - Cheryl Frankfater
- Biomedical Mass Spectrometry Resource, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas Terrapon
- Architecture et Fonction des Macromolecules Biologiques, CNRS, Aix-Marseille University, Marseille, France
| | - Bernard Henrissat
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, Lyngby, Denmark
| | - Dmitry A Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Semen A Leyn
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Jiang Z, He L, Li D, Zhuo L, Chen L, Shi RQ, Luo J, Feng Y, Liang Y, Li D, Congmei X, Fu Y, Chen YM, Zheng JS, Tao L. Human gut microbial aromatic amino acid and related metabolites prevent obesity through intestinal immune control. Nat Metab 2025; 7:808-822. [PMID: 40087408 PMCID: PMC12021661 DOI: 10.1038/s42255-025-01246-5] [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: 12/08/2023] [Accepted: 02/14/2025] [Indexed: 03/17/2025]
Abstract
Obesity affects millions of people in the world. The gut microbiome influences body fat accumulation, but the mechanisms remain to be investigated. Here, we show an association between microbial aromatic amino acid metabolites in serum and body fat accumulation in a large Chinese longitudinal cohort. We next identify that 4-hydroxyphenylacetic acid (4HPAA) and its analogues effectively protect male mice from high-fat-diet-induced obesity. These metabolites act on intestinal mucosa to regulate the immune response and control lipid uptake, which protects against obesity. We further demonstrate that T cells and B cells are not vital for 4HPAA-mediated obesity prevention, and innate lymphoid cells have antagonistic roles. Together, these findings reveal specific microbial metabolites as pivotal molecules to prohibit obesity through immune control, establishing mechanisms of host modulation by gut microbial metabolites.
Collapse
Affiliation(s)
- Zengliang Jiang
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, China
- Innovation Center of Yangtze River Delta, Jiaxing, Zhejiang, China
| | - Liuqing He
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Diyin Li
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Laibao Zhuo
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Lingjun Chen
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Rui-Qi Shi
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jianhua Luo
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yuhui Feng
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yuhui Liang
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Danyang Li
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Xiao Congmei
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Yuanqing Fu
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Ju-Sheng Zheng
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China.
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
| | - Liang Tao
- Research Center for Industries of the Future, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, China.
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
27
|
Chae S, Chae S, Kang TG, Kim SJ, Choi A. Optimization-Incorporated Deep Learning Strategy to Automate L3 Slice Detection and Abdominal Segmentation in Computed Tomography. Bioengineering (Basel) 2025; 12:367. [PMID: 40281727 PMCID: PMC12025211 DOI: 10.3390/bioengineering12040367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
This study introduces a deep learning-based strategy to automatically detect the L3 slice and segment abdominal tissues from computed tomography (CT) images. Accurate measurement of muscle and fat composition at the L3 level is critical as it can serve as a prognostic biomarker for cancer diagnosis and treatment. However, current manual approaches are time-consuming and prone to class imbalance, since L3 slices constitute only a small fraction of the entire CT dataset. In this study, we propose an optimization-incorporated strategy that integrates augmentation ratio and class weight adjustment as correction design variables within deep learning models. In this retrospective study, the CT dataset was privately collected from 150 prostate cancer and bladder cancer patients at the Department of Urology of Gangneung Asan Hospital. A ResNet50 classifier was used to detect the L3 slice, while standard Unet, Swin-Unet, and SegFormer models were employed to segment abdominal tissues. Bayesian optimization determines optimal augmentation ratios and class weights, mitigating the imbalanced distribution of L3 slices and abdominal tissues. Evaluation of CT data from 150 prostate and bladder cancer patients showed that the optimized models reduced the slice detection error to approximately 0.68 ± 1.26 slices and achieved a Dice coefficient of up to 0.987 ± 0.001 for abdominal tissue segmentation-improvements over the models that did not consider correction design variables. This study confirms that balancing class distribution and properly tuning model parameters enhances performance. The proposed approach may provide reliable and automated biomarkers for early cancer diagnosis and personalized treatment planning.
Collapse
Affiliation(s)
- Seungheon Chae
- Department of Bio-Mechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Seongwon Chae
- Department of Biomedical Engineering, College of Medical Convergence, Catholic Kwandong University, Gangneung 25601, Republic of Korea;
| | - Tae Geon Kang
- Institute for Trauma Research, College of Medicine, Korea University, Seoul 02708, Republic of Korea;
| | - Sung Jin Kim
- Department of Urology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung 25440, Republic of Korea
| | - Ahnryul Choi
- Department of Biomedical Engineering, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
28
|
Yang M, Zhang Y, Li Z, Liu T, He J, Li J. Gut Microbiota Regulate Lipid Metabolism via the Bile Acid Pathway: Resistance to Hypoxia in Gansu Zokor (Eospalax cansus). Integr Zool 2025. [PMID: 40156100 DOI: 10.1111/1749-4877.12971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/09/2024] [Accepted: 02/16/2025] [Indexed: 04/01/2025]
Abstract
The Gansu zokor (Eospalax cansus), a subterranean rodent endemic to the Loess Plateau of China, exhibits remarkable adaptability to hypoxic environments. While gut microbiota are known to regulate lipid metabolism through bile acid (BA) pathways, this phenomenon has not been investigated in subterranean rodents exposed to hypoxia. This study employed 16SrRNA sequencing, targeted analysis of BA metabolites in colonic contents, and assessments of BA and lipid metabolites alongside molecular analyses in the liver and ileum under conditions of acute and chronic hypoxia in Gansu zokors. The results revealed that hypoxia altered the composition of gut microbiota and BA pools in Gansu zokors. Hypoxia-induced changes increased the abundance of gut microbiota associated with BA metabolism, thereby modulating lipid metabolism via farnesoid X receptor (FXR) signaling in the distal ileum and liver cells. Under acute hypoxia, FXR upregulated lipid synthesis and suppressed fatty acid β-oxidation by downregulating the carnitine palmitoyl-transferase1A (CPT1A) expression. Conversely, during chronic hypoxia, particularly under long-term exposure, FXR reduced lipid synthesis and enhanced fatty acid β-oxidation by upregulating acyl-CoA oxidase (ACOX1) expression. In both hypoxic conditions, FXR facilitated lipoprotein metabolism. In summary, this study elucidates that gut microbiota-mediated BA metabolic pathways contribute to the Gansu zokor's ability to maintain lipid metabolic homeostasis and adaptation to hypoxia.
Collapse
Affiliation(s)
- Maohong Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, Shaanxi Normal University, Xi'an, China
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an, China
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Yingying Zhang
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Zhuohang Li
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Tianyi Liu
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Jianping He
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, Shaanxi Normal University, Xi'an, China
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an, China
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Jingang Li
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, Shaanxi Normal University, Xi'an, China
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an, China
- College of Life Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
29
|
Jeerawattanawart S, Angkasekwinai P. Intestinal IL-25 prevents high-fat diet-induced obesity by modulating the cholesterol transporter NPC1L1 expression in the intestinal epithelial cells. Sci Rep 2025; 15:10445. [PMID: 40140439 PMCID: PMC11947149 DOI: 10.1038/s41598-025-95516-7] [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: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 03/28/2025] Open
Abstract
The intestine is essential for digestion and nutrient absorption, and its altered function contributes to metabolic dysregulation and obesity-induced intestinal inflammation. Intestinal immune responses have been associated with the regulation of metabolic dysfunction during obesity. Given that the epithelial cell-derived cytokine IL-25 has been demonstrated to regulate metabolic disorders, we sought to examine the role of intestinal IL-25 in modulating a high-fat diet (HFD)-induced obesity. We found that mice on a high-fat diet exhibited decreased IL-25 expression in the small intestine. Intestinal IL-25 mRNA levels displayed an inverse association with plasma triglycerides, total cholesterol, glucose levels, and the expression of the cholesterol transporter Npc1l1 in the intestine. In HFD-induced obesity, transgenic mice overexpressing IL-25 in the intestinal epithelial cells demonstrated diminished mRNA expression of intestinal genes related to glucose, cholesterol, and fat absorption, along with chylomicron production, while also systemically decreasing plasma glucose, total cholesterol, and triglyceride levels, fat accumulation, and weight gain. In vitro, IL-25 treatment of human intestinal Caco-2 cells directly decreased cholesterol uptake and downregulated the expression of NPC1L1 and its transcriptional regulator, SREBP2. These findings highlight IL-25 as a potential modulator in the intestine that regulates intestinal cholesterol absorption and systemic metabolism in obesity.
Collapse
Affiliation(s)
- Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
- Faculty of Medical Technology, Rangsit University, Pathum Thani, 12000, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand.
- Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani, 12120, Thailand.
| |
Collapse
|
30
|
Liu Y, Li X, Chen Y, Yao Q, Zhou J, Wang X, Meng Q, Ji J, Yu Z, Chen X. Fecal microbiota transplantation: application scenarios, efficacy prediction, and factors impacting donor-recipient interplay. Front Microbiol 2025; 16:1556827. [PMID: 40201444 PMCID: PMC11975908 DOI: 10.3389/fmicb.2025.1556827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Fecal microbiota transplantation (FMT) represents a therapeutic approach that directly regulates the gut microbiota of recipients, normalizes its composition and reaping therapeutic rewards. Currently, in addition to its general application in treating Clostridium difficile (C. difficile) infection (CDI), FMT treatment has also been extended to the fields of other gastrointestinal diseases, infections, gut-liver or gut-brain axis disorders, metabolic diseases and cancer, etc. Prior to FMT, rigorous donor screening is essential to reduce the occurrence of adverse events. In addition, it is imperative to evaluate whether the recipient can safely and effectively undergo FMT treatment. However, the efficacy of FMT is influenced by the complex interactions between the gut microbiota of donor and recipient, the degree of donor microbiota engraftment is not necessarily positively related with the success rate of FMT. Furthermore, an increasing number of novel factors affecting FMT outcomes are being identified in recent clinical trials and animal experiments, broadening our understanding of FMT treatment. This article provides a comprehensive review of the application scenarios of FMT, the factors influencing the safety and efficacy of FMT from the aspects of both the donors and the recipients, and summarizes how these emerging novel regulatory factors can be combined to predict the clinical outcomes of patients undergoing FMT.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinru Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuchao Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qinyan Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinjie Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxuan Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Qingguo Meng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxuan Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
31
|
Liu J, Li F, Yang L, Luo S, Deng Y. Gut microbiota and its metabolites regulate insulin resistance: traditional Chinese medicine insights for T2DM. Front Microbiol 2025; 16:1554189. [PMID: 40177494 PMCID: PMC11963813 DOI: 10.3389/fmicb.2025.1554189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
The gut microbiota is closely associated with the onset and development of type 2 diabetes mellitus (T2DM), characterized by insulin resistance (IR) and chronic low-grade inflammation. However, despite the widespread use of first-line antidiabetic drugs, IR in diabetes and its complications continue to rise. The gut microbiota and its metabolic products may promote the development of T2DM by exacerbating IR. Therefore, regulating the gut microbiota has become a promising therapeutic strategy, with particular attention given to probiotics, prebiotics, synbiotics, and fecal microbiota transplantation. This review first examines the relationship between gut microbiota and IR in T2DM, summarizing the research progress of microbiota-based therapies in modulating IR. We then delve into how gut microbiota-related metabolic products contribute to IR. Finally, we summarize the research findings on the role of traditional Chinese medicine in regulating the gut microbiota and its metabolic products to improve IR. In conclusion, the gut microbiota and its metabolic products play a crucial role in the pathophysiological process of T2DM by modulating IR, offering new insights into potential therapeutic strategies for T2DM.
Collapse
Affiliation(s)
- Jing Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fuxing Li
- Ningxiang Traditional Chinese Medicine Hospital, Changsha, China
| | - Le Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shengping Luo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihui Deng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
32
|
Kim KS, Na HS, Oh TJ, Han H, Kim J, Hong JS, Lee HJ, Park YS, Chung J. Oral microbiome changes in subjects with obesity following bariatric surgery compared to lean counterparts. Front Microbiol 2025; 16:1553404. [PMID: 40170925 PMCID: PMC11959278 DOI: 10.3389/fmicb.2025.1553404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction This study aimed to compare oral microbiome profiles between obese and lean individuals without clinical periodontitis, and to assess changes in the oral microbiome of obese subjects following bariatric surgery. Methods Individuals with a body mass index (BMI) > 30 were enrolled in the obese group, whereas those with a BMI < 23 served as controls. The obese surgery group, which consented to bariatric surgery, was followed up at 1, 3, and 6 months with clinical examinations. Oral examinations were conducted and periodontal disease was classified based on probing results. Saliva, buccal and subgingival microbiome samples were analyzed for community diversity, relative bacterial abundance, and differential abundance between control (n = 24) and obese group (n = 31). To evaluate effect size and statistical power, we used micropower, a simulation-based method for Permutational Multivariate Analysis of Variance-based β-diversity comparisons. Results The obese group exhibited distinct alpha diversity (buccal: Chao1 p = 0.0002, Shannon p = 0.0003, supragingival: Shannon p < 0.0001) compared with the control group. Bray-Curtis distance analysis indicated significant disparities in microbiome composition distribution in saliva (p = 0.003), buccal (p = 0.002), and subgingival plaque samples (p = 0.001). Although the obese and normal weight groups exhibited no significant periodontal differences, the obese group showed distinct species associated with periodontal disease, especially in subgingival plaque including Filifactor alocis, Peptostreptococcaceae spp., Prevotella spp., and Treponema maltophilum. Cluster analysis of the obese surgery group indicated the emergence of microbiomes associated with a healthy state that increased over time including Streptococcus salivarious and various Veillonella spp., whereas clusters containing periodontal pathogens including Porphyromonas spp., tended to diminish. Discussion The oral microbiome at 6 months post-bariatric surgery indicates a potential shift toward a healthy periodontal state, suggesting that weight loss interventions may positively impact oral microbial communities even in the absence of clinical periodontitis.
Collapse
Affiliation(s)
- Keun-Suh Kim
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hee Sam Na
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyejung Han
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Jiyeon Kim
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jin-Sil Hong
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jin Chung
- Department of Oral Microbiology, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
33
|
Nabakhteh S, Lotfi A, Afsartaha A, Khodadadi ES, Abdolghaderi S, Mohammadpour M, Shokri Y, Kiani P, Ehtiati S, Khakshournia S, Khatami SH. Nutritional Interventions in Amyotrophic Lateral Sclerosis: From Ketogenic Diet and Neuroprotective Nutrients to the Microbiota-Gut-Brain Axis Regulation. Mol Neurobiol 2025:10.1007/s12035-025-04830-8. [PMID: 40097762 DOI: 10.1007/s12035-025-04830-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disease with significant challenges in diagnosis and treatment. Recent research has highlighted the complex nature of ALS, encompassing behavioral impairments in addition to its neurological manifestations. While several medications have been approved to slow disease progression, ongoing research is focused on identifying new therapeutic targets. The current review focuses on emerging therapeutic strategies and personalized approaches aimed at improving patient outcomes. Recent advancements highlight the importance of targeting additional pathways such as mitochondrial dysfunction and neuroinflammation to develop more effective treatments. Personalized medicine, including genetic testing and biomarkers, is proving valuable in stratifying patients and tailoring treatment options. Complementary therapies, such as nutritional interventions like the ketogenic diet and microbiome modulation, also show promise. This review emphasizes the need for a multidisciplinary approach that integrates early diagnosis, targeted treatments, and supportive care to address the multisystemic nature of ALS and improve the quality of life for patients.
Collapse
Affiliation(s)
- Samira Nabakhteh
- Department of Biochemistry, School of Basic Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Anahita Lotfi
- Department of Food Sciences and Industry, School of Agricultural Sciences and Natural Resources, Islamic Azad University, Khorasgan Branch, Isfahan, Iran
| | - Arman Afsartaha
- Department of Nutrition, Faculty of Medical Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elaheh Sadat Khodadadi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, 35122, Italy
| | - Siavash Abdolghaderi
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Mozhdeh Mohammadpour
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, Tehran, Iran
| | - Yasaman Shokri
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Pouria Kiani
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sajad Ehtiati
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Khakshournia
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Fan J, Wu Y, Wang X, Ullah H, Ling Z, Liu P, Wang Y, Feng P, Ji J, Li X. The probiotic enhances donor microbiota stability and improves the efficacy of fecal microbiota transplantation for treating colitis. J Adv Res 2025:S2090-1232(25)00177-8. [PMID: 40089059 DOI: 10.1016/j.jare.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
INTRODUCTION The stability and metabolic functionality of donor microbiota are critical determinants of fecal microbiota transplantation (FMT) efficacy in inflammatory bowel disease (IBD). While probiotics show potential to enhance microbiota resilience, their role in optimizing donor microbiota for FMT remains underexplored. OBJECTIVES This study investigated whether pretreatment of donor microbiota with L. plantarum GR-4 could improve FMT outcomes in a DSS-induced colitis model by modulating microbial stability, metabolic activity, and host-microbiome interactions. METHODS Donor mice received L. plantarum GR-4 for 3 weeks to generate modified FMT (MFMT). DSS-colitis mice were treated with MFMT, conventional FMT, or 5-aminosalicylic acid (5-ASA). Multi-omics analyses and functional assays (stress resistance, engraftment efficiency) were used to evaluate therapeutic mechanisms. RESULTS GR-4 pretreatment conferred three key advantages to donor microbiota: Ecological stabilization: 1. GR-4-driven acidification (pH 3.97 vs. 4.59 for LGG, p < 0.0001) enriched butyrogenic Butyricicoccus (73 % butyrate increase, p < 0.05) and improved stress resistance to bile acids/gastric conditions (1.25 × survival vs. FMT). 2. Metabolic reprogramming: GR-4 metabolized 25.3 % of tryptophan (vs. 10.3 % for LGG) to generate immunomodulatory indoles (ILA, IAA), activating aryl hydrocarbon receptor (AHR) signaling and upregulating anti-inflammatory IL-10/IL-22. 3. Bile acid remodeling: MFMT restored sulfolithocholic acid and β-MCA levels, outperforming FMT in resolving DSS-induced dysregulation. MFMT achieved an 83 % remission rate (vs. 50 % for FMT), enhanced gut barrier integrity, and reversed colitis-associated metabolic dysregulation (e.g., elevated spermidine, 7-sulfocholic acid). Probiotic preconditioning improved donor engraftment by 1.25 × and enriched success-associated taxa (Sporobacter, Butyricimonas), while suppressing pathogens (Clostridium papyrosolvens). CONCLUSIONS L. plantarum GR-4 optimizes donor microbiota via pH-driven niche engineering, immunometabolic reprogramming, and bile acid modulation, addressing key limitations of conventional FMT. The multi-targeted efficacy of MFMT, evidenced by superior remission rates and metabolic restoration, establishes this approach as a translatable strategy for IBD therapy. This study establishes probiotic-enhanced FMT as a paradigm for precision microbiome interventions.
Collapse
Affiliation(s)
- Jingjing Fan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Ying Wu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Xing Wang
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Habib Ullah
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Zhenmin Ling
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Pu Liu
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Yu Wang
- Nutrition and Health Research Center, Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Pengya Feng
- Department of Children Rehabilitation Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jing Ji
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China.
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu 730000, PR China.
| |
Collapse
|
35
|
Hernandez-Leyva AJ, Rosen AL, Tomera CP, Lin EE, Akaho EH, Blatz AM, Otto WR, Logan J, Young LR, Harris RM, Whiteside SA, Kau AL, Odom John AR. Upper and lower airway microbiota across infancy and childhood. Pediatr Res 2025:10.1038/s41390-025-03942-0. [PMID: 40075175 DOI: 10.1038/s41390-025-03942-0] [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] [Received: 08/02/2024] [Revised: 01/22/2025] [Accepted: 02/02/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The upper and lower respiratory tracts feature distinct environments and responses affecting microbial colonization but investigating the relationship between them is technically challenging. We aimed to identify relationships between taxa colonizing the nasopharynx and trachea across childhood. METHODS We employed V4 16S rRNA gene sequencing to profile nasopharyngeal swabs and tracheal aspirates collected from 172 subjects between 20 weeks and 18 years of age. These samples were collected prior to elective procedures over the course of 20 weeks in 2020 from subjects enrolled in a cross-sectional study. After extraction, sequencing, and quality control, we studied the remaining 147 of 172 nasopharyngeal swabs and 95 of 172 tracheal aspirates, including 80 subject-matched pairs of samples. RESULTS Sequencing data revealed that the nasopharynx is colonized by few, often highly abundant taxa, while the tracheal aspirates feature greater diversity. The patterns of colonization identified in the nasopharynx correlate with subject age across childhood. CONCLUSION Our data suggests that there are relatively few species that colonize both the nasopharyngeal tract and the trachea. Furthermore, we observe a pattern of change in the nasopharyngeal microbiota that is correlated with age, suggesting a possible developmental progression of the nasopharyngeal microbiota across childhood. IMPACT The airway microbiota in childhood plays important roles in respiratory health and immune development. In this work, we report on paired nasopharyngeal swab and tracheal aspirate samples from a cross-sectional cohort of children from infancy to 18 years. We find that the upper and lower airway microbiota are unlikely to share taxa and do not correlate in terms of diversity. We show that the composition of the upper airway microbiota is strongly correlated with age, with a stereotypic developmental trajectory during childhood and adolescence. Our results inform our understanding of airway microbiota assembly and may be used to predict airway disease in young children.
Collapse
Affiliation(s)
- Ariel J Hernandez-Leyva
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne L Rosen
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher P Tomera
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Elaina E Lin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elikplim H Akaho
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, USA
| | - Allison M Blatz
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Nemours Children's Hospital, Wilmington, DE, USA
| | - William R Otto
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Infectious Disease, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joey Logan
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa R Young
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca M Harris
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samantha A Whiteside
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew L Kau
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Audrey R Odom John
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Infectious Diseases, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Sun J, Song S, Liu J, Chen F, Li X, Wu G. Gut microbiota as a new target for anticancer therapy: from mechanism to means of regulation. NPJ Biofilms Microbiomes 2025; 11:43. [PMID: 40069181 PMCID: PMC11897378 DOI: 10.1038/s41522-025-00678-x] [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: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
In order to decipher the relationship between gut microbiota imbalance and cancer, this paper reviewed the role of intestinal microbiota in anticancer therapy and related mechanisms, discussed the current research status of gut microbiota as a biomarker of cancer, and finally summarized the reasonable means of regulating gut microbiota to assist cancer therapy. Overall, our study reveals that the gut microbiota can serve as a potential target for improving cancer management.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shiyan Song
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiahua Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
37
|
Turjeman S, Rozera T, Elinav E, Ianiro G, Koren O. From big data and experimental models to clinical trials: Iterative strategies in microbiome research. Cell 2025; 188:1178-1197. [PMID: 40054445 DOI: 10.1016/j.cell.2025.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/02/2024] [Accepted: 01/27/2025] [Indexed: 05/13/2025]
Abstract
Microbiome research has expanded significantly in the last two decades, yet translating findings into clinical applications remains challenging. This perspective discusses the persistent issue of correlational studies in microbiome research and proposes an iterative method leveraging in silico, in vitro, ex vivo, and in vivo studies toward successful preclinical and clinical trials. The evolution of research methodologies, including the shift from small cohort studies to large-scale, multi-cohort, and even "meta-cohort" analyses, has been facilitated by advancements in sequencing technologies, providing researchers with tools to examine multiple health phenotypes within a single study. The integration of multi-omics approaches-such as metagenomics, metatranscriptomics, metaproteomics, and metabolomics-provides a comprehensive understanding of host-microbe interactions and serves as a robust hypothesis generator for downstream in vitro and in vivo research. These hypotheses must then be rigorously tested, first with proof-of-concept experiments to clarify the causative effects of the microbiota, and then with the goal of deep mechanistic understanding. Only following these two phases can preclinical studies be conducted with the goal of translation into the clinic. We highlight the importance of combining traditional microbiological techniques with big-data approaches, underscoring the necessity of iterative experiments in diverse model systems to enhance the translational potential of microbiome research.
Collapse
Affiliation(s)
- Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | - Tommaso Rozera
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Microbiome & Cancer Division, DKFZ, Heidelberg, Germany
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Zhang Y, Liu R, Chen Y, Cao Z, Liu C, Bao R, Wang Y, Huang S, Pan S, Qin L, Wang J, Ning G, Wang W. Akkermansia muciniphila supplementation in patients with overweight/obese type 2 diabetes: Efficacy depends on its baseline levels in the gut. Cell Metab 2025; 37:592-605.e6. [PMID: 39879980 DOI: 10.1016/j.cmet.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Akkermansia muciniphila is a promising target for managing obesity and type 2 diabetes (T2D), but human studies are limited. We conducted a 12-week randomized, double-blind, placebo-controlled trial involving 58 participants with overweight or obese T2D, who received A. muciniphila (AKK-WST01) or placebo, along with routine lifestyle guidance. Both groups showed decreases in body weight and glycated hemoglobin (HbA1c), without significant between-group differences. In participants with low baseline A. muciniphila, AKK-WST01 supplementation showed high colonization efficiency and significant reductions in body weight, fat mass, and HbA1c, which were not found in the placebo group. However, AKK-WST01 supplementation showed poor colonization and no significant clinical improvements in participants with high baseline A. muciniphila. These findings were verified in germ-free mice receiving feces with low or high A. muciniphila. Our study indicates that metabolic benefits of A. muciniphila supplementation could depend on its baseline intestinal levels, supporting the potential for gut microbiota-guided probiotic supplementation. (ClinicalTrials.gov number, NCT04797442).
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cong Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Riqiang Bao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shan Huang
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Shijia Pan
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Li Qin
- Department of Endocrinology, Chongming hospital affiliated to Shanghai University of Health & Medicine Sciences, Shanghai 202150, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
39
|
Chavarria X, Park HS, Oh S, Kang D, Choi JH, Kim M, Cho YH, Yi MH, Kim JY. Using gut microbiome metagenomic hypervariable features for diabetes screening and typing through supervised machine learning. Microb Genom 2025; 11:001365. [PMID: 40063675 PMCID: PMC11893737 DOI: 10.1099/mgen.0.001365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetes mellitus is a complex metabolic disorder and one of the fastest-growing global public health concerns. The gut microbiota is implicated in the pathophysiology of various diseases, including diabetes. This study utilized 16S rRNA metagenomic data from a volunteer citizen science initiative to investigate microbial markers associated with diabetes status (positive or negative) and type (type 1 or type 2 diabetes mellitus) using supervised machine learning (ML) models. The diversity of the microbiome varied according to diabetes status and type. Differential microbial signatures between diabetes types and negative group revealed an increased presence of Brucellaceae, Ruminococcaceae, Clostridiaceae, Micrococcaceae, Barnesiellaceae and Fusobacteriaceae in subjects with diabetes type 1, and Veillonellaceae, Streptococcaceae and the order Gammaproteobacteria in subjects with diabetes type 2. The decision tree, elastic net, random forest (RF) and support vector machine with radial kernel ML algorithms were trained to screen and type diabetes based on microbial profiles of 76 subjects with type 1 diabetes, 366 subjects with type 2 diabetes and 250 subjects without diabetes. Using the 1000 most variable features, tree-based models were the highest-performing algorithms. The RF screening models achieved the best performance, with an average area under the receiver operating characteristic curve (AUC) of 0.76, although all models lacked sensitivity. Reducing the dataset to 500 features produced an AUC of 0.77 with sensitivity increasing by 74% from 0.46 to 0.80. Model performance improved for the classification of negative-status and type 2 diabetes. Diabetes type models performed best with 500 features, but the metric performed poorly across all model iterations. ML has the potential to facilitate early diagnosis of diabetes based on microbial profiles of the gut microbiome.
Collapse
Affiliation(s)
- Xavier Chavarria
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyun Seo Park
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Singeun Oh
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Dongjun Kang
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jun Ho Choi
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Myungjun Kim
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yoon Hee Cho
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Myung-hee Yi
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ju Yeong Kim
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
40
|
Shukla A, Sharma C, Malik MZ, Singh AK, Aditya AK, Mago P, Shalimar, Ray AK. Deciphering the tripartite interaction of urbanized environment, gut microbiome and cardio-metabolic disease. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 377:124693. [PMID: 40022791 DOI: 10.1016/j.jenvman.2025.124693] [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: 12/02/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The world is experiencing a sudden surge in urban population, especially in developing Asian and African countries. Consequently, the global burden of cardio-metabolic disease (CMD) is also rising owing to gut microbiome dysbiosis due to urbanization factors such as mode of birth, breastfeeding, diet, environmental pollutants, and soil exposure. Dysbiotic gut microbiome indicated by altered Firmicutes to Bacteroides ratio and loss of beneficial short-chain fatty acids-producing bacteria such as Prevotella, and Ruminococcus may disrupt host-intestinal homeostasis by altering host immune response, gut barrier integrity, and microbial metabolism through altered T-regulatory cells/T-helper cells balance, activation of pattern recognition receptors and toll-like receptors, decreased mucus production, elevated level of trimethylamine-oxide and primary bile acids. This leads to a pro-inflammatory gut characterized by increased pro-inflammatory cytokines such as tumour necrosis factor-α, interleukin-2, Interferon-ϒ and elevated levels of metabolites or metabolic endotoxemia due to leaky gut formation. These pathophysiological characteristics are associated with an increased risk of cardio-metabolic disease. This review aims to comprehensively elucidate the effect of urbanization on gut microbiome-driven cardio-metabolic disease. Additionally, it discusses targeting the gut microbiome and its associated pathways via strategies such as diet and lifestyle modulation, probiotics, prebiotics intake, etc., for the prevention and treatment of disease which can potentially be integrated into clinical and professional healthcare settings.
Collapse
Affiliation(s)
- Avaneesh Shukla
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Chanchal Sharma
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Md Zubbair Malik
- Department of Translational Medicine, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Center, Mathura, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India; Campus of Open Learning, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
41
|
Caesar R. The impact of novel probiotics isolated from the human gut on the gut microbiota and health. Diabetes Obes Metab 2025; 27 Suppl 1:3-14. [PMID: 39726216 PMCID: PMC11894790 DOI: 10.1111/dom.16129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The gut microbiota plays a pivotal role in influencing the metabolism and immune responses of the body. A balanced microbial composition promotes metabolic health through various mechanisms, including the production of beneficial metabolites, which help regulate inflammation and support immune functions. In contrast, imbalance in the gut microbiota, known as dysbiosis, can disrupt metabolic processes and increase the risk of developing diseases, such as obesity, type 2 diabetes, and inflammatory disorders. The composition of the gut microbiota is dynamic and can be influenced by environmental factors such as diet, medication, and the consumption of live bacteria. Since the early 1900s, bacteria isolated from food and have been used as probiotics. However, the human gut also offers an enormous reservoir of bacterial strains, and recent advances in microbiota research have led to the discovery of strains with probiotic potentials. These strains, derived from a broad spectrum of microbial taxa, differ in their ecological properties and how they interact with their hosts. For most probiotics bacterial structural components and metabolites, such as short-chain fatty acids, contribute to the maintenance of metabolic and immunological homeostasis by regulating inflammation and reinforcing gut barrier integrity. Metabolites produced by probiotic strains can also be used for bacterial cross-feeding to promote a balanced microbiota. Despite the challenges related to safety, stability, and strain-specific properties, several newly identified strains offer great potential for personalized probiotic interventions, allowing for targeted health strategies.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
42
|
Zhou X, Ganz AB, Rayner A, Cheng TY, Oba H, Rolnik B, Lancaster S, Lu X, Li Y, Johnson JS, Hoyd R, Spakowicz DJ, Slavich GM, Snyder MP. Dynamic human gut microbiome and immune shifts during an immersive psychosocial intervention program. Brain Behav Immun 2025; 125:428-443. [PMID: 39701328 PMCID: PMC11903166 DOI: 10.1016/j.bbi.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/24/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Although depression is a leading cause of disability worldwide, the pathophysiological mechanisms underlying this disorder-particularly those involving the gut microbiome-are poorly understood. METHOD To investigate, we conducted a community-based observational study to explore complex associations between changes in the gut microbiome, cytokine levels, and depression symptoms in 51 participants (Mage = 49.56, SD = 13.31) receiving an immersive psychosocial intervention. A total of 142 multi-omics samples were collected from participants before, during, and three months after the nine-day inquiry-based stress reduction program. RESULTS Results revealed that depression was associated with both an increased presence of putatively pathogenic bacteria and reduced microbial beta-diversity. Following the intervention, we observed reductions in neuroinflammatory cytokines and improvements in several mental health indicators. Interestingly, participants with a Prevotella-dominant microbiome showed milder symptoms when depressed, along with a more resilient microbiome and more favorable inflammatory cytokine profile, including reduced levels of CXCL-1. CONCLUSIONS These findings reveal a potentially protective link between the Prevotella-dominant microbiome and depression, as evidenced by a reduced pro-inflammatory environment and fewer depressive symptoms. These insights, coupled with observed improvements in neuroinflammatory markers and mental health from the intervention, may highlight potential avenues for microbiome-targeted therapies for managing depression.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, CA, USA
| | - Ariel B Ganz
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA
| | - Andre Rayner
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Tess Yan Cheng
- Department of Genetics, Stanford University School of Medicine, CA, USA; Department of Microbiology, College of Arts and Sciences, University of Washington, WA, USA
| | - Haley Oba
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Benjamin Rolnik
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA
| | - Samuel Lancaster
- Department of Genetics, Stanford University School of Medicine, CA, USA
| | - Xinrui Lu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Sichuan, China
| | - Yizhou Li
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Sichuan, China
| | - Jethro S Johnson
- Oxford Centre for Microbiome Studies, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Rebecca Hoyd
- The Ohio State University Comprehensive Cancer Center, OH, USA
| | | | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, CA, USA; Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, CA, USA; Stanford Healthcare Innovation Lab, Stanford University, CA, USA.
| |
Collapse
|
43
|
Tao S, Fan J, Li J, Wu Z, Yao Y, Wang Z, Wu Y, Liu X, Xiao Y, Wei H. Extracellular vesicles derived from Lactobacillus johnsonii promote gut barrier homeostasis by enhancing M2 macrophage polarization. J Adv Res 2025; 69:545-563. [PMID: 38508446 PMCID: PMC11954842 DOI: 10.1016/j.jare.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/19/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024] Open
Abstract
INTRODUCTION Diarrheic disease is a common intestinal health problem worldwide, causing great suffering to humans and animals. Precise manipulation strategies based on probiotics to combat diarrheic diseases have not been fully developed. OBJECTIVES The aim of this study was to investigate the molecular mechanisms by which probiotics manipulate macrophage against diarrheic disease. METHODS Metagenome reveals gut microbiome profiles of healthy and diarrheic piglets. Fecal microbial transplantation (FMT) was employed to explore the causal relationship between gut microbes and diarrhea. The protective role of probiotics and their derived extracellular vesicles (EVs) was investigated in ETEC K88-infected mice. Macrophage depletion was performed to assess the role of macrophages in EVs against diarrhea. Execution of in vitro cell co-culture and transcriptome analyses elucidated the molecular mechanisms by which EVs modulate the macrophage and intestinal epithelial barrier. RESULTS Escherichia coli was enriched in weaned diarrheic piglets, while Lactobacillus johnsonii (L. john) showed a negative correlation with Escherichia coli. The transmission of diarrheic illness symptoms was achieved by transferring fecal microbiota, but not metabolites, from diarrheic pigs to germ-free (GF) mice. L. john's intervention prevented the transmission of disease phenotypes from diarrheic piglets to GF mice. L. john also reduces the gut inflammation induced by ETEC K88. The EVs secreted by L. john demonstrated enhanced efficacy in mitigating the adverse impacts induced by ETEC K88 through the modulation of macrophage phenotype. In vitro experiments have revealed that EVs activate M2 macrophages in a manner that shuts down ERK, thereby inhibiting NLRP3 activation in intestinal epithelial cells. CONCLUSION Our results reveal that intestinal microbiota drives the onset of diarrheic disease and that probiotic-derived EVs ameliorate diarrheic disease symptoms by modulating macrophage phenotypes. These findings can enhance the advancement of innovative therapeutic approaches for diarrheic conditions based on probiotic-derived EVs.
Collapse
Affiliation(s)
- Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhifeng Wu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yong Yao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China.
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
44
|
Liang JH, Pu YQ, Yang XZ, Chen JQ, Wu ZW, Liu ML, Jiang N, Huang S, Zhang YS, Hu LX, Jin ZG, Ge WX, Pu XY, Huang SY, Chen YJ. Mediating effect of physical activity on the relationship between high dietary live microbe intake and obesity among U.S adolescents, finding from NHANES 1999-2018. Nutr Metab Cardiovasc Dis 2025; 35:103786. [PMID: 39674724 DOI: 10.1016/j.numecd.2024.103786] [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: 03/13/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS The impact of dietary live microbe intake on adolescent obesity is still not fully understood. This study aims to investigate the potential relationship between dietary live microbe intake and obesity among U.S adolescents, and to explore the mediating role of physical activity (PA). METHODS AND RESULTS Data from NHANES (1999-2018) were analyzed, and dietary live microbe intake was categorized into low, medium, and high groups using a developed framework. Survey-weighted logistic regression and mediation analysis models were used to examine the association between live microbe intake and adolescent obesity, as well as the potential mediating effect of PA. Our study included 8443 participants aged 6-18, representing the noninstitutionalized U.S population of 184.5 million. We found that participants with a high dietary intake of live microbes had lower odds of developing obesity compared to those with the lowest exposure to live microbes (AOR = 0.900, 95 % CI: 0.812, 0.997). Additionally, our mediation analysis revealed a significant indirect effect of live microbes on obesity risk through PA (P-value <0.001), with 39.4 % (95 % CI: 24.5 %, 86.5 %) of the effect mediated by PA. CONCLUSION Our study highlights the association between consuming a higher amount of live microbes in the diet and a decreased risk of obesity among U.S adolescents. It also suggests that PA may act as a mediator in this relationship. Therefore, it is crucial to emphasize the incorporation of both dietary interventions and PA in the development of prevention and therapy policies for managing adolescent obesity.
Collapse
Affiliation(s)
- Jing-Hong Liang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Ying-Qi Pu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xiu-Zhi Yang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Jia-Qi Chen
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Zhuo-Wen Wu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Mei-Ling Liu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Nan Jiang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Shan Huang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Yu-Shan Zhang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Li-Xin Hu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Zheng-Ge Jin
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Wen-Xin Ge
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Xue-Ya Pu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Shao-Yi Huang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China
| | - Ya-Jun Chen
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
45
|
Romaní‐Pérez M, Líebana‐García R, Flor‐Duro A, Bonillo‐Jiménez D, Bullich‐Vilarrubias C, Olivares M, Sanz Y. Obesity and the gut microbiota: implications of neuroendocrine and immune signaling. FEBS J 2025; 292:1397-1420. [PMID: 39159270 PMCID: PMC11927058 DOI: 10.1111/febs.17249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Obesity is a major health challenge due to its high prevalence and associated comorbidities. The excessive intake of a diet rich in fat and sugars leads to a persistent imbalance between energy intake and energy expenditure, which increases adiposity. Here, we provide an update on relevant diet-microbe-host interactions contributing to or protecting from obesity. In particular, we focus on how unhealthy diets shape the gut microbiota and thus impact crucial intestinal neuroendocrine and immune system functions. We describe how these interactions promote dysfunction in gut-to-brain neuroendocrine pathways involved in food intake control and postprandial metabolism and elevate the intestinal proinflammatory tone, promoting obesity and metabolic complications. In addition, we provide examples of how this knowledge may inspire microbiome-based interventions, such as fecal microbiota transplants, probiotics, and biotherapeutics, to effectively combat obesity-related disorders. We also discuss the current limitations and gaps in knowledge of gut microbiota research in obesity.
Collapse
Affiliation(s)
- Marina Romaní‐Pérez
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Rebeca Líebana‐García
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Alejandra Flor‐Duro
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Daniel Bonillo‐Jiménez
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Clara Bullich‐Vilarrubias
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Marta Olivares
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Yolanda Sanz
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| |
Collapse
|
46
|
Syal A, Martell M, Sikdar R, Dietz M, Ziegert Z, Jahansouz C, Elias MH, Staley C. Quorum quenching enzymes disrupt bacterial communication in a sex- and dose-dependent manner. Animal Model Exp Med 2025; 8:473-482. [PMID: 39948046 PMCID: PMC11904096 DOI: 10.1002/ame2.12520] [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: 07/10/2024] [Accepted: 11/11/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Over the past 50 years, the incidence of obesity has gradually increased, necessitating investigation into the multifactorial contributors to this disease, including the gut microbiota. Bacteria within the human gut microbiome communicate using a density-dependent process known as quorum sensing (QS), in which autoinducer (AI) molecules (e.g., N-acyl-homoserine lactones [AHLs]) are produced to enable bacterial interactions and regulate gene expression. METHODS We aimed to disrupt QS using quorum quenching (QQ) lactonases GcL and SsoPox, which cleave AHL signaling molecules in a taxa-specific manner based on differing enzyme affinities for different substrates. We hypothesized that QQ hinders signals from obesity-associated pathobionts, thereby slowing or preventing obesity. RESULTS In a murine model of diet-induced obesity, we observed GcL and SsoPox treatments have separate sex-dependent and dose-dependent effects on intestinal community composition and diversity. Notably, male mice given 2 mg/mL SsoPox exhibited significant changes in the relative abundances of gram-negative taxa, including Porphyromonadaceae, Akkermansiaceae, Muribaculaceae, and Bacteroidales (Kruskal-Wallis p < 0.001). Additionally, we used covariance matrix network analysis to model bacterial taxa co-occurrence due to QQ enzyme administration. There were more associations among taxa in control mice, particularly among gram-negative bacteria, whereas mice receiving SsoPox had the fewest associations. CONCLUSIONS Overall, our study establishes proof of concept that QQ is a targetable strategy for microbial control in vivo. Further characterization and dosage optimization of QQ enzymes are necessary to harness their therapeutic capability for the treatment of chronic microbial-associated diseases.
Collapse
Affiliation(s)
- Aneesh Syal
- Division of Basic and Translational Research, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Maria Martell
- Division of Basic and Translational Research, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Rakesh Sikdar
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Matthew Dietz
- Division of Basic and Translational Research, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Zachary Ziegert
- Division of Basic and Translational Research, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Cyrus Jahansouz
- Division of Colon and Rectal Surgery, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Mikael H. Elias
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Christopher Staley
- Division of Basic and Translational Research, Department of SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- BioTechnology InstituteUniversity of MinnesotaSt. PaulMinnesotaUSA
| |
Collapse
|
47
|
Jia L, Wang R, Huang Z, Sun N, Sun H, Wang H, Lu F, Liu Y. Phosphatidylcholine ameliorates lipid accumulation and liver injury in high-fat diet mice by modulating bile acid metabolism and gut microbiota. Int J Food Sci Nutr 2025; 76:165-178. [PMID: 39632393 DOI: 10.1080/09637486.2024.2437469] [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: 07/13/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Phosphatidylcholine (PC) has garnered considerable attention due to its involvement in a wide array of crucial biological functions. However, there is still much to active explore regarding the precise mechanisms that underlie PC's actions in the context of high-fat diet. In this study, we found that both PC intervention and treatment significantly mitigated lipid accumulation, liver damage, and body weight gaining triggered by the high-fat diet. Untargeted and targeted metabolomic analyses uncovered substantial effects of PC on bile acid metabolism, especially led to a substantial reduction in elevated levels of free bile acids. 16S rRNA gene sequencing revealed that PC modulated the gut microbiota structures and compositions in high-fat diet mice, particularly exhibiting a positive association with Pseudoflavonifractor abundance, and a negative correlation with Olsenella, Parasutterella, and Allobaculum abundance. Our study suggested that PC held promise as a potential candidate for alleviating lipid metabolism injury, liver disease or obesity.
Collapse
Affiliation(s)
- Longgang Jia
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Ruijia Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Zhiqi Huang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Nana Sun
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Hui Sun
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Hongbin Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| | - Yihan Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, P. R. China
| |
Collapse
|
48
|
Chao J, Coleman RA, Keating DJ, Martin AM. Gut Microbiome Regulation of Gut Hormone Secretion. Endocrinology 2025; 166:bqaf004. [PMID: 40037297 PMCID: PMC11879239 DOI: 10.1210/endocr/bqaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Indexed: 03/06/2025]
Abstract
The gut microbiome, comprising bacteria, viruses, fungi, and bacteriophages, is one of the largest microbial ecosystems in the human body and plays a crucial role in various physiological processes. This review explores the interaction between the gut microbiome and enteroendocrine cells (EECs), specialized hormone-secreting cells within the intestinal epithelium. EECs, which constitute less than 1% of intestinal epithelial cells, are key regulators of gut-brain communication, energy metabolism, gut motility, and satiety. Recent evidence shows that gut microbiota directly influence EEC function, maturation, and hormone secretion. For instance, commensal bacteria regulate the production of hormones like glucagon-like peptide 1 and peptide YY by modulating gene expression and vesicle cycling in EE cells. Additionally, metabolites such as short-chain fatty acids, derived from microbial fermentation, play a central role in regulating EEC signaling pathways that affect metabolism, gut motility, and immune responses. Furthermore, the interplay between gut microbiota, EECs, and metabolic diseases, such as obesity and diabetes, is examined, emphasizing the microbiome's dual role in promoting health and contributing to disease states. This intricate relationship between the gut microbiome and EECs offers new insights into potential therapeutic strategies for metabolic and gut disorders.
Collapse
Affiliation(s)
- Jessica Chao
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Rosemary A Coleman
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Damien J Keating
- Gut Sensory Systems Group, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Alyce M Martin
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| |
Collapse
|
49
|
Bai J, Tian Y, Lu Y, Chen Y, Yu M, Gao X. Differential effects of high-fat diet on salivary and gut microbiota. Front Cell Infect Microbiol 2025; 15:1547555. [PMID: 40066066 PMCID: PMC11891374 DOI: 10.3389/fcimb.2025.1547555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Objectives Microorganisms contribute to the pathogenesis of obesity, while more studies focus on gut microbiome. However, the relationship between oral microbiota and obesity has yet to be elucidated. This study was designed to investigate the similarities and differences in the effects of a high-fat diet on salivary and gut microbiota through mouse experiments, exploring the hypothesis that oral microbial mechanisms may contribute to obesity. Methods An obese mouse model was established in male C57BL/6J mice by feeding a high-fat diet, confirmed by body weight records and blood glucose tests. This study evaluated the physiological effects of the high-fat diet on mice. 16S rRNA sequencing technology was used to analyze changes in salivary and gut microbiota, and gas chromatography-mass spectrometry was employed to evaluate 17 short-chain and medium-chain fatty acids quantitatively. Results The microbiota distribution in salivary was different between the high-fat diet (HFD) and normal chow diet (NCD) groups. At the genus level of salivary microbiota, Streptococcus and Escherichia were highly abundant in the HFD group. Rodentibacter and Turicibacter were more abundant in the NCD group. Regarding the gut microbiome, the diversity changes of gut microbiota are more significant than those of salivary microbiota. The HFD group had a significantly higher abundance of Kineothrix, Cryptobacteroides, and a lower abundance of CAG-485. Nine genera had consistent alterations in salivary and gut microbiota, among which Akkermansia, Lactobacillus, and Intestinimonas were significantly correlated with physiological indicators, and Muribaculum was significantly correlated with increased decanoic acid levels in the HFD group. The dysregulated nine genera were associated with significant upregulation of certain metabolic pathways of the HFD group, including the pentose phosphate, bacterial invasion of epithelial cells, and steroid biosynthesis pathways. Conclusions There are differences and similarities in the effects of HFD on salivary and gut microbiota. Certain genera of the oral-gut axis altered consistently by HFD may affect obesity through mechanisms involving metabolic pathways and inflammation.
Collapse
Affiliation(s)
- Jingxuan Bai
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| | - Yixue Tian
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| | - Yujia Lu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| | - Yuke Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| | - Min Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| | - Xuemei Gao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Oral Therapy of Sleep Apnea, Peking University Hospital of Stomatology, Beijing, China
| |
Collapse
|
50
|
Rondanelli M, Borromeo S, Cavioni A, Gasparri C, Gattone I, Genovese E, Lazzarotti A, Minonne L, Moroni A, Patelli Z, Razza C, Sivieri C, Valentini EM, Barrile GC. Therapeutic Strategies to Modulate Gut Microbial Health: Approaches for Chronic Metabolic Disorder Management. Metabolites 2025; 15:127. [PMID: 39997751 PMCID: PMC11857149 DOI: 10.3390/metabo15020127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Numerous recent studies have suggested that the composition of the intestinal microbiota can trigger metabolic disorders, such as diabetes, prediabetes, obesity, metabolic syndrome, sarcopenia, dyslipidemia, hyperhomocysteinemia, and non-alcoholic fatty liver disease. Since then, considerable effort has been made to understand the link between the composition of intestinal microbiota and metabolic disorders, as well as the role of probiotics in the modulation of the intestinal microbiota. The aim of this review was to summarize the reviews and individual articles on the state of the art regarding ideal therapy with probiotics and prebiotics in order to obtain the reversion of dysbiosis (alteration in microbiota) to eubiosis during metabolic diseases, such as diabetes, prediabetes, obesity, hyperhomocysteinemia, dyslipidemia, sarcopenia, and non-alcoholic fatty liver diseases. This review includes 245 eligible studies. In conclusion, a condition of dysbiosis, or in general, alteration of the intestinal microbiota, could be implicated in the development of metabolic disorders through different mechanisms, mainly linked to the release of pro-inflammatory factors. Several studies have already demonstrated the potential of using probiotics and prebiotics in the treatment of this condition, detecting significant improvements in the specific symptoms of metabolic diseases. These findings reinforce the hypothesis that a condition of dysbiosis can lead to a generalized inflammatory picture with negative consequences on different organs and systems. Moreover, this review confirms that the beneficial effects of probiotics on metabolic diseases are promising, but more research is needed to determine the optimal probiotic strains, doses, and administration forms for specific metabolic conditions.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Sara Borromeo
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Cavioni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Ilaria Gattone
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Elisa Genovese
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Lazzarotti
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Leonardo Minonne
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessia Moroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Zaira Patelli
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Razza
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Sivieri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Eugenio Marzio Valentini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Gaetan Claude Barrile
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| |
Collapse
|