1
|
Minty M, Germain A, Sun J, Kaglan G, Servant F, Lelouvier B, Misselis E, Neagoe RM, Rossella M, Cardellini M, Burcelin R, Federici M, Fernandez-Real JM, Blasco-Baque V. Identifying the location-dependent adipose tissue bacterial DNA signatures in obese patients that predict body weight loss. Gut Microbes 2025; 17:2439105. [PMID: 39714075 DOI: 10.1080/19490976.2024.2439105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Recent sets of evidence have described profiles of 16S rDNA sequences in host tissues, notably in fat pads that are significantly overrepresented and can serve as signatures of metabolic disease. However, these recent and original observations need to be further detailed and functionally defined. Here, using state-of-the-art targeted DNA sequencing and discriminant predictive approaches, we describe, from the longitudinal FLORINASH cohort of patients who underwent bariatric surgery, visceral, and subcutaneous fat pad-specific bacterial 16SrRNA signatures. The corresponding Porphyromonadaceae, Campylobacteraceae, Prevotellaceae, Actimomycetaceae, Veillonellaceae, Anaerivoracaceae, Fusobacteriaceae, and the Clostridium family XI 16SrRNA DNA segment profiles are signatures of the subcutaneous adipose depot while Pseudomonadaceae and Micrococcacecae, 16SrRNA DNA sequence profiles characterize the visceral adipose depot. In addition, we have further identified that a specific pre-bariatric surgery adipose tissue bacterial DNA signature predicts the efficacy of body weight loss in obese patients 5-10 years after the surgery. 16SrRNA signatures discriminate (ROC ~ 1) the patients who did not maintain bodyweight loss and those who did. Second, from the 16SrRNA sequences we infer potential pathways suggestive of catabolic biochemical activities that could be signatures of subcutaneous adipose depots that predict body weight loss.
Collapse
Affiliation(s)
- Matthieu Minty
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Alberic Germain
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Jiuwen Sun
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Gracia Kaglan
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | | | | | - Emiri Misselis
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Radu Mircea Neagoe
- Science and Technology "George Emil Palade" Tîrgu Mures, Second Department of Surgery, Emergency Mureş County Hospital, University of Medicine Pharmacy, Târgu Mureș, Romania
| | - Menghini Rossella
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Marina Cardellini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - José Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, University Hospital of Girona 'Dr Josep Trueta'
- Institut d'Investigacio Biomedica de Girona IdibGi, CIBER Fisiopatologia de la Obesidad y Nutricion, Girona, Spain
| | - Vincent Blasco-Baque
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| |
Collapse
|
2
|
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
|
3
|
Kou G, Yao S, Ullah A, Fang S, Guo E, Bo Y. Polystyrene microplastics impair brown and beige adipocyte function via the gut microbiota-adipose tissue crosstalk in high-fat diet mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138225. [PMID: 40220396 DOI: 10.1016/j.jhazmat.2025.138225] [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: 12/11/2024] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Microplastics (MPs) are pervasive in the environment and food. The potential health hazards of this emerging pollutant have raised significant concerns in recent years. However, the underlying mechanism by which MPs have any impact on brown and beige adipocytes in the context of obesity is yet to be investigated. METHODS The C57BL/6 J mice were randomly assigned to the HFD and HFD+MPs group for 12 weeks of exposure to explore the differences in brown and beige adipocyte function. The gut microbiota analysis, fecal microbiota transplantation and metabolomic profiling were carried out to further determine its potential mechanism. RESULTS The present work demonstrated that high-fat diet mice accumulate lipids and have reduced energy expenditure after three months of oral administration of MPs. In addition to escalating intestinal dysbiosis, exposing HFD mice to MPs induces thermogenic dysfunction in inguinal white adipose tissue and brown adipose tissue. Following the fecal microbiota transplantation, the accumulation of lipids and dysfunction in energy expenditure within the microbiota of recipient mice further elucidated the inhibitory effect of MPs. CONCLUSIONS Our results suggest that MPs induced the thermogenic dysfunction of BAT and iWAT by affecting gut microbiota composition. The present study highlights the mechanisms by which MPs produce thermogenic dysfunction in BAT and iWAT and disruption in the gastrointestinal microbiota.
Collapse
Affiliation(s)
- Guangning Kou
- Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou University, Zhengzhou 450001, China; Department of Nutrition and Food Hygiene, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shuai Yao
- Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou University, Zhengzhou 450001, China
| | - Amin Ullah
- Department of Nutrition and Food Hygiene, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Shuhao Fang
- Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou University, Zhengzhou 450001, China
| | - Erni Guo
- Centre for Nutritional Ecology and Centre for Sport Nutrition and Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Yacong Bo
- Department of Nutrition and Food Hygiene, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
4
|
Zhang Y, Ni P, Chen H, Tang L, Song H, Wen H, Miao Y, Li W, Li X. Vitamin D 3 ameliorates hyperglycemia by modulating gut microbiota and metabolites in prediabetic KKay mice. Food Res Int 2025; 211:116369. [PMID: 40356099 DOI: 10.1016/j.foodres.2025.116369] [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: 11/05/2024] [Revised: 03/16/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Prediabetes represents a pivotal stage in the development and pathogenesis of diabetes, during which notable alterations in the gut microbiota can be observed. Vitamin D (VD) showed anti-diabetic properties, but it is unknown whether the improvement of VD on hyperglycemia is associated with gut microbiota. Thus, our objective was to investigate and verify the effects of VD3 on glucose metabolism in prediabetes, as well as to elucidate the underlying mechanisms. In this study, different concentrations of VD3 were intraperitoneally administered to prediabetic mice induced by high fat diet for 16 weeks. Biochemical analyses, oral glucose tolerance test, 16S rRNA and untargeted metabolomics were used, the mechanism was explored. Then, fecal suspensions collected from the above donors were transplanted into KKay mice for 6 weeks, and the relevant indicators were measured. The results showed that VD3 intervention alleviated glucose metabolism in KKay mice. It increased the protein expression of colon tight junction proteins, alleviated metabolic endotoxemia and inflammation, so that reduced tumor necrosis factor alpha (TNFα) induced toll-like receptor 4/nuclear factor kappa-B (TLR4/NFκB) and improvement of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) insulin signaling pathway. VD3 affected the structure of gut microbiota and metabolites, and functional prediction analysis suggested that VD3 may affect carbohydrate. Besides, the effect of VD3 could be delivered by fecal microbiota transplantation (FMT). Consequently, VD3 ameliorate glucose metabolism by modulating gut microbiota and metabolites in KKay mice, and this ability could be transferred by FMT.
Collapse
Affiliation(s)
- Yujing Zhang
- Department of Food Hygiene and Nutrition, School of Medicine, Henan University of Chinese Medicine, Zhengzhou 450000, Henan, China; Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Peng Ni
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hao Chen
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Lulu Tang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hanlu Song
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Hongwei Wen
- Department of Public Health, Zhengzhou Shuqing Medical College, Zhengzhou 450000, Henan, China
| | - Yufan Miao
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Wenjie Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China.
| | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450000, Henan, China.
| |
Collapse
|
5
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
6
|
Huang C, Cao W, Zhou S, Deng Y. Biogenesis mechanisms, regulatory strategies, and applications of bacterial extracellular vesicles. Crit Rev Biotechnol 2025:1-17. [PMID: 40368580 DOI: 10.1080/07388551.2025.2496300] [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: 10/11/2024] [Revised: 11/29/2024] [Accepted: 02/08/2025] [Indexed: 05/16/2025]
Abstract
Bacterial extracellular vesicles (EVs) are produced by both Gram-negative and Gram-positive bacteria. These EVs are composed of lipid bilayers and various components derived from parent bacteria, including proteins, lipids, and nucleic acids. Previous studies have indicated the significant role of bacterial EVs in interactions between bacteria and between bacteria and hosts. Moreover, bacterial EVs are emerging as promising delivery vectors capable of transporting drug molecules over long distances to tissues. Therefore, understanding the biogenesis of bacterial EVs and how to regulate their production holds great importance for expanding their applications. In this review, we provide an overview of bacterial EVs, especially focusing on the distinct mechanisms of EVs biogenesis and the regulation of EVs production in both Gram-negative and Gram-positive bacteria. Additionally, we discuss various methods for cargos loading into bacteria EVs, as well as their diverse applications in vaccines, cancer therapy, and drug delivery. We anticipate that this review will advance the field of bacterial EVs, contributing to both the enhancement of existing applications and the emergence of novel applications.
Collapse
Affiliation(s)
- Chao Huang
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| | - Wenyan Cao
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| | - Shenghu Zhou
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| | - Yu Deng
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Huang M, Huang M, Liu L, Yang F, He C, Sun YC, Jiao YR, Tang X, Hou J, Chen KX, He WZ, Wei J, Chen HL, Li X, Zeng C, Lei GH, Li CJ. Gut Microbiota Modulates Obesity-Associated Skeletal Deterioration Through Macrophage Aging and Grancalcin Secretion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2502634. [PMID: 40349163 DOI: 10.1002/advs.202502634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/08/2025] [Indexed: 05/14/2025]
Abstract
Obesity is associated with skeletal deterioration and increased fracture risk, but the underlying mechanism is unclear. Herein, it is shown that obese gut microbiota promotes skeletal deterioration by inducing bone marrow macrophages (BMMs) senescence and grancalcin (GCA) secretion. Obese mice and those receiving obese fecal microbiota transplants exhibit increased senescent macrophages and elevated GCA expression in the bone marrow. In a study of 40 participants, it is found that obese patients are associated with higher serum GCA levels. It is further revealed that obese gut-microbiota derived lipopolysaccharides (LPS) stimulate GCA expression in senescent BMMs via activating Toll-like receptor 4 pathway. Mice with depletion of the Gca gene are resistant to the negative effects of obesity and LPS on bone. Moreover, neutralizing antibody against GCA mitigates skeletal deterioration in obese mice and LPS-induced chronic inflammation mouse model. The data suggest that the interaction between gut microbiota and the immune system contributes to obesity-associated skeletal deterioration, and targeting senescent macrophages and GCA shows potential of protecting skeletal health in obese population.
Collapse
Affiliation(s)
- Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of General Medicine, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Fang Yang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu-Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xiang Tang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jie Wei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hui-Ling Chen
- Department of Endocrine Subspecialty of Gerontology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xia Li
- Department of General Medicine, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, China
| | - Chao Zeng
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guang-Hua Lei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- FuRong Laboratory, Changsha, 410008, China
- Laboratory Animal Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
8
|
Tsou TC, Yeh SC, Tsai FY, Chen PY. Palmitic acid and lipopolysaccharide induce macrophage TNFα secretion, suppressing browning regulators and mitochondrial respiration in adipocytes. Toxicol Appl Pharmacol 2025; 500:117389. [PMID: 40348028 DOI: 10.1016/j.taap.2025.117389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Obesity and its associated pro-inflammatory activity contribute significantly to metabolic dysfunction. In contrast, browning of white adipose tissue (WAT) generally improves metabolic health. Our prior research suggested that macrophage-derived pro-inflammatory cytokines suppress key regulators of browning-adrenergic receptor β3 (Adrb3) and peroxisome proliferator-activated receptor γ (Pparg)-as well as energy metabolism mediators-insulin receptor substrate 1 (Irs1) and hormone-sensitive lipase (Lipe)-in diet-induced obese mice. To explore this mechanism, we developed an in vitro model using RAW264.7 macrophages and 3T3-L1 adipocytes exposed to palmitic acid (PA) and/or lipopolysaccharide (LPS). PA (200 μM) and LPS (1.0 μg/ml) synergistically promoted M1 polarization of macrophages and secretion of pro-inflammatory cytokines, with tumor necrosis factor-α (TNFα), C-C motif chemokine ligand 2 (CCL2), CCL5, and interleukin-6 (IL-6) being predominant. Conditioned media from both control and PA-treated macrophages, when exposed to LPS ≥0.01 μg/ml, significantly downregulated Adrb3, Pparg, Irs1, and Lipe in adipocytes. At physiologically relevant LPS levels (≤0.001 μg/ml), PA-treated macrophage media exerted greater suppression of these genes than controls. Among the cytokines, TNFα emerged as the primary mediator, significantly reducing expression of the four key regulators. Furthermore, adipocytes treated with TNFα exhibited significant reductions in both uncoupling protein 1 (Ucp1) expression and mitochondrial respiration. These findings demonstrate that exposure to obesity-associated factors (PA and LPS) induces macrophage-derived TNFα, which suppresses browning and mitochondrial function in adipocytes. This mechanism may inform new therapeutic strategies targeting TNFα to alleviate obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Tsui-Chun Tsou
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan.
| | - Szu-Ching Yeh
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Feng-Yuan Tsai
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Pei-Yu Chen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| |
Collapse
|
9
|
Wu S, Tong C, Liu J. Obesogenic effects of six classes of emerging contaminants. J Environ Sci (China) 2025; 151:252-272. [PMID: 39481937 DOI: 10.1016/j.jes.2024.03.032] [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: 11/29/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 11/03/2024]
Abstract
There is growing concern about the concept that exposure to environmental chemicals may be contributing to the obesity epidemic. However, there is no consensus on the obesogenic effects of emerging contaminants from a toxicological and environmental perspective. The potential human exposure and experimental evidence for obesogenic effects of emerging contaminants need to be systematically discussed. The main objective of this review is to provide recommendations for further subsequent policy development following a critical analysis of the literature for humans and experimental animals exposed to emerging contaminants. This article reviews human exposure to emerging contaminants (with a focus on antimicrobials, preservatives, water and oil repellents, flame retardants, antibiotics and bisphenols) and the impact of emerging contaminants on obesity. These emerging contaminants have been widely detected in human biological samples. Epidemiological studies provide evidence linking exposure to emerging contaminants to the risks of obesity in humans. Studies based on animal models and adipose cells show the obesogenic effects of emerging contaminants and identify modes of action by which contaminants may induce changes in body fat accumulation and lipid metabolic homeostasis. Some knowledge gaps in this area and future directions for further investigation are discussed.
Collapse
Affiliation(s)
- Siying Wu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaoyu Tong
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
10
|
Murugesan R, Kumar J, Leela KV, Meenakshi S, Srivijayan A, Thiruselvam S, Satheesan A, Chaithanya V. The role of gut microbiota and bacterial translocation in the pathogenesis and management of type 2 diabetes mellitus: Mechanisms, impacts, and dietary therapeutic strategies. Physiol Behav 2025; 293:114838. [PMID: 39922411 DOI: 10.1016/j.physbeh.2025.114838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE OF REVIEW The influence of gut microbiota on Type 2 Diabetes Mellitus (T2DM) is an emerging area of research. This review investigates the relationship between gut microbiota dysbiosis, bacterial translocation, and T2DM. It aims to elucidate how microbial imbalances contribute to the progression of T2DM through bacterial translocation and to evaluate dietary and therapeutic strategies to manage these effects. RECENT FINDINGS Recent studies highlight that dysbiosis in T2DM patients often leads to increased systemic inflammation, impaired glucose metabolism, and disrupted gut barrier integrity. These disruptions promote elevated levels of harmful bacterial components, such as lipopolysaccharides, in the bloodstream. This, in turn, is linked to worsening insulin resistance and metabolic dysfunction. Advances in molecular methods and biomarkers have provided deeper insights into bacterial translocation and its impact on diabetes. Dietary interventions, including nutraceutical agents, high-fiber and low-glycemic index diets, as well as the use of probiotics and prebiotics, have shown promise in restoring gut health and mitigating bacterial translocation. CONCLUSION Maintaining a balanced gut microbiota and intestinal barrier integrity is crucial for managing T2DM. Therapeutic strategies, including dietary modifications and nutraceuticals, have demonstrated potential in reducing bacterial translocation and systemic inflammation. Continued research is needed to refine these approaches and explore novel treatment modalities for improving metabolic health in T2DM patients.
Collapse
Affiliation(s)
- Ria Murugesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Janardanan Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Kakithakara Vajravelu Leela
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Sachdev Meenakshi
- Department of Dietary, Tamil Nadu Government Multi Super Speciality Hospital, Chennai 600002, Tamil Nadu, India
| | - Appandraj Srivijayan
- Department of Internal Medicine, Melmaruvathur Adhiparasakthi Institute of Medical Sciences and Research, Melmaruvathur 603319, Tamil Nadu, India
| | - Shubhashree Thiruselvam
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Venkata Chaithanya
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| |
Collapse
|
11
|
Li Y, Feng T, Zhao Y, Zhang X, Chen H, Xia P, Yang D, Liang Z. Medicinal and edible homologous poly/oligo-saccharides: Structural features, effect on intestinal flora and preventing and treating type 2 diabetes, and their applications: A review. Int J Biol Macromol 2025; 305:141031. [PMID: 39965679 DOI: 10.1016/j.ijbiomac.2025.141031] [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: 09/24/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is the third most common chronic metabolic disorder worldwide and seriously dangerous. Novel therapeutics are sought due to the paucity of safe and effective metabolic disorder-related diabetes medicines. Intestinal flora impacts glucose and lipid balance, making it a unique T2DM therapeutic target. Due to gut fermentation, poly/oligo-saccharides are highly beneficial prebiotic carbohydrates for intestinal health. Moreover, supplementation with naturally occurring medicinal and edible homologous traditional Chinese medicines (MEHTCM) poly/oligo-saccharides has significant antidiabetic effects with few side effects. Now, a comprehensive review of research developments of MEHTCM poly/oligo-saccharides was presented to explore their prospects. We outlined the structural characteristics, structure classification, and structure-activity relationships. Notably, structure-activity relationships illustrated that molecular weight, monosaccharide composition, and glycosidic bond type could influence the hypoglycemic activity and prebiotic effect of MEHTCM poly/oligo-saccharides. Additionally, the review systematically summarized the effect and potential mechanism of MEHTCM poly/oligo-saccharide on T2DM, focusing on gut microbiota. The potential applications in formulations for special medical purposes, common food, health care product, agriculture and other fields have also been summarized. This review emphasizes MEHTCM poly/oligo-saccharides' potential as prebiotics for T2DM treatment. This information provides new insights and a theoretical foundation for MEHTCM poly/oligo-saccharide nutritional and medicinal research.
Collapse
Affiliation(s)
- Yuan Li
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Tinghui Feng
- College of Life Sciences, Northwest A & F University, Xi'an 710000, China
| | - Yaxin Zhao
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xiaodan Zhang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Haimin Chen
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Pengguo Xia
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Dongfeng Yang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zongsuo Liang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
12
|
Piłot M, Dzięgielewska-Gęsiak S, Walkiewicz KW, Bednarczyk M, Waniczek D, Muc-Wierzgoń M. Gut Microbiota and Metabolic Dysregulation in Elderly Diabetic Patients: Is There a Gender-Specific Effect. J Clin Med 2025; 14:3103. [PMID: 40364140 PMCID: PMC12073094 DOI: 10.3390/jcm14093103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: The aim of this study was to qualitatively and quantitatively assess the bacterial domain of the gut microbiome in elderly patients with type 2 diabetes (T2D), with a focus on sex differences, glycemic control, and lipid disorders. Methods: This study included 60 older adults with T2D (38 women and 22 men) treated with metformin or a combination of metformin and insulin. The gut microbiota was profiled using 16S rRNA gene sequencing. Statistical analyses, including correlation analysis and multiple regression, were performed to identify the associations between microbial taxa, sex, and metabolic parameters. Results: No statistically significant differences in alpha or beta diversity were observed between the sexes. Multiple regression analysis indicated a positive relationship between Tenericutes and HbA1c in male participants (β = 2.22931, CI [0.75, 3.70], R = 0.67; R2 = 0.36; unadjusted p = 0.0052; adjusted p = 0.0496). In female participants, G0' (β = -2.24107, CI [-3.19, -1.30], R = 0.78; R2 = 0.58; unadjusted p = 0.00003; adjusted p = 0.0005) and HbA1c (β = -1.86670, CI [-2.61, -1.12], R = 0.78; R2 = 0.58; unadjusted p = 0.00001; adjusted p = 0.0003) correlated negatively with Verrucomicrobia as well G0' (β = -1.90427, CI [-2.95, -0.85], R = 0.46; R2 = 0.17; unadjusted p = 0.0008; adjusted p = 0.007) and HbA1c (β = -1.69561, CI [-2.52, -0.87], R = 0.46; R2 = 0.17; unadjusted p = 0.0002; adjusted p = 0.002) correlated negatively with OD1 bacteria, known as Parcubacteria. Conclusions: In this elderly population with type 2 diabetes, biological sex did not significantly affect the gut microbiota diversity. However, several exploratory associations between microbial taxa and metabolic parameters differed between men and women, suggesting that sex may influence specific aspects of microbiota-metabolism interactions. These preliminary findings underscore the importance of considering both age- and sex-related factors when investigating the gut microbiome in the context of type 2 diabetes.
Collapse
Affiliation(s)
- Magdalena Piłot
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Sylwia Dzięgielewska-Gęsiak
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Katarzyna Weronika Walkiewicz
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Martyna Bednarczyk
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Katowice, Poland;
| | - Małgorzata Muc-Wierzgoń
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| |
Collapse
|
13
|
Hernández-Martín M, Bocanegra A, Garcimartín A, Macho-González A, Redondo-Castillejo R, García-Fernández RA, Apaza-Ticona L, Bastida S, Benedí J, Sánchez-Muniz FJ, López-Oliva ME. Silicon-enriched functional meat enhances colonic barrier function by regulating tight junction protein expression, oxidative stress, and inflammation responses in a diabetic dyslipidemia model. Food Funct 2025. [PMID: 40302652 DOI: 10.1039/d4fo06277a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Western diets are linked to metabolic disorders such as Type 2 diabetes mellitus (T2DM) and diabetic dyslipidemia, which involve hyperglycemia, insulin resistance, high plasma cholesterol levels and altered lipoprotein profiles. The T2DM progression also involves glucolipotoxicity, wherein elevated glucose and fatty acid levels induce oxidative stress and inflammation. Excessive intake of saturated fats and/or cholesterol can trigger dysbiosis, which weakens the colonic barrier, increases its permeability, and promotes chronic low-grade inflammation, thereby accelerating the progression of T2DM. Silicon, an essential trace element, has demonstrated antidiabetic, hypolipidemic, antioxidant and anti-inflammatory properties, suggesting its potential as a nutritional adjuvant in therapeutic management of T2DM and the maintenance of gut health. In this study, 24 male Wistar rats were divided into three groups: (1) an early-stage T2DM group (ED) fed a control meat incorporated into a high saturated-fat diet; (2) a late-stage T2DM group (LD) fed a control meat incorporated into a high-saturated fat and high cholesterol diet combined with streptozotocin and nicotinamide injection; and (3) a late-stage T2DM group fed a silicon enriched meat (LD-Si). Microbiota composition, lipoperoxidation and concentrations of fat, cholesterol, oxysterols and short-chain fatty acids and silicon were assayed in feces. The colonic tissue morphology, barrier integrity, antioxidant capacity and inflammatory markers were measured to evaluate the impact of silicon on colonic health and intestinal barrier function. Silicon enriched meat (Si-RM) consumption increased faecal fat and cholesterol excretion and reduced toxic luminal environments by modulating oxysterols. Si-RM consumption also enhanced colonic barrier integrity, increasing tight junction proteins and goblet cells, and exhibited antioxidant effects via the pNrf2 pathway and superoxide dismutase activity. Furthermore, silicon reduced the pro-inflammatory cytokines TNFα and IL-6, likely through inhibition of the TLR4/NFκB pathway. The results suggest that silicon's ability to enhance intestinal barrier integrity, reduce oxidative stress, and prevent inflammation could slow down T2DM progression, making it a promising nutritional adjuvant for managing the disease.
Collapse
Affiliation(s)
- Marina Hernández-Martín
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain.
| | - Aránzazu Bocanegra
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Alba Garcimartín
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Adrián Macho-González
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Rocío Redondo-Castillejo
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Rosa A García-Fernández
- Animal Medicine and Surgery Department, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Luis Apaza-Ticona
- Animal Medicine and Surgery Department, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain
- Organic Chemistry Unit, Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Spain
| | - Sara Bastida
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Juana Benedí
- Pharmacology, Pharmacognosy and Botany Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Francisco J Sánchez-Muniz
- Nutrition and Food Science Department, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - M Elvira López-Oliva
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
14
|
Toledo M, Martínez-Martínez S, Van Hul M, Laudo B, Eyre E, Pelicaen R, Puel A, Altirriba J, Gómez-Valadés AG, Inderhees J, Moreno-Indias I, Pozo M, Chivite I, Milà-Guasch M, Haddad-Tóvolli R, Obri A, Fos-Domènech J, Tahiri I, Llana SR, Ramírez S, Monelli E, Schwaninger M, Cani PD, Nogueiras R, Claret M. Rapid modulation of gut microbiota composition by hypothalamic circuits in mice. Nat Metab 2025:10.1038/s42255-025-01280-3. [PMID: 40263603 DOI: 10.1038/s42255-025-01280-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
In recent years, the gut microbiota and derived metabolites have emerged as relevant players in modulating several brain functions, including energy balance control1-3. This form of distant communication mirrors that of metabolic hormones (for example, leptin, ghrelin), which convey information about the organism's energy status by exerting effects on diverse brain regions, including the master homeostatic centre, the hypothalamus4. However, whether the hypothalamus is also able to influence gut microbiota composition remains enigmatic. Here we present a study designed to unravel this challenging question. To this aim, we used chemogenetics5 (to selectively activate or inhibit hypothalamic pro-opiomelanocortin or agouti-related peptide neurons) or centrally administered leptin or ghrelin to male mice. Subsequently, we conducted microbiota composition analysis throughout the gut using 16S rRNA gene sequencing. Our results showed that these brain interventions significantly changed the gut microbiota in an anatomical and short-term (2-4 h) fashion. Transcriptomic analysis indicated that these changes were associated with the reconfiguration of neuronal and synaptic pathways in the duodenum concomitant with increased sympathetic tone. Interestingly, diet-induced obesity attenuated the brain-mediated changes triggered by leptin in gut microbiota communities and sympathetic activation. Our findings reveal a previously unanticipated brain-gut axis that acutely attunes microbiota composition on fast timescales, with potential implications for meal-to-meal adjustments and systemic energy balance control.
Collapse
Affiliation(s)
- Míriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Martínez-Martínez
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Berta Laudo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rudy Pelicaen
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Anthony Puel
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julica Inderhees
- Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iasim Tahiri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergio R Llana
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erika Monelli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Markus Schwaninger
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
| | - Rubén Nogueiras
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Galicia Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
15
|
Yang H, Lei C, Li D, Ma L, Zhang N, Lang Y, Wu L, Wang M, Tian H, Li C. An integrated fecal microbiome and metabolomics in type 2 diabetes mellitus rats reveal mechanism of action of Moringa oleifera Lamarck seeds polysaccharides to alleviate diabetes. Int J Biol Macromol 2025; 310:143437. [PMID: 40274155 DOI: 10.1016/j.ijbiomac.2025.143437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Moringa oleifera Lamarck seeds (MOS) have been traditionally used in folk medicine and documented for their potential to alleviate type 2 diabetes symptoms, but the potential mechanisms are still unknown. The purpose of this article is to investigate the effects of MSAP (alkali-extracted polysaccharide from MOS) on diabetic rats by assessing its impact on the gut microbiome, diabetes-related biochemical markers, and fecal metabolomics. The results demonstrated that the fasting blood glucose, glucose tolerance, insulin resistance, insulin level and lipopolysaccharides (LPS) level in the rats treated with MSAP were all improved. Specifically, MSAP was found to modulate the composition and diversity of the gut microbiota, increasing the ratio of Firmicutes/Bacteroidetes, which enhanced the quantity of probiotic Lactobacillus and butyrate-producing bacteria, such as Roseburia, thereby reinforcing the intestinal epithelial barrier. Furthermore, fecal metabolomics indicates that MSAP actively regulates pathways closely associated with diabetes, including sphingolipid metabolism, amino acid synthesis and catabolism, retrograde endogenous cannabinoid signaling, and the modulation of TRP channels by inflammatory mediators. By integrating microbiome and metabolomics data, this study elucidated the mechanisms through which MSAP alleviates diabetes. In conclusion, the findings suggest that polysaccharides from MOS hold potential as a medicinal and edible homologous food for diabetes management.
Collapse
Affiliation(s)
- Hongru Yang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Chongbin Lei
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Dongyao Li
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Lei Ma
- College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Na Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; College of Biochemistry and Environmental Engineering, Baoding University, Baoding, Hebei 071000, China
| | - Yumiao Lang
- College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Liping Wu
- College of Nursing, Hebei University, Baoding, Hebei 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co. Ltd., Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China
| | - Hongtao Tian
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; National Engineering Research Center for Agriculture in Northern Mountainous Areas, Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China.
| | - Chen Li
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China.
| |
Collapse
|
16
|
Thonusin C, Suparan K, Kunasol C, Lungruammit N, Nawara W, Arunsak B, Kerdphoo S, Kongkaew A, Songtrai S, Pintana H, Maneechote C, Pratchayasakul W, Kaewsuwan S, Chattipakorn N, Chattipakorn SC. Interruptins Extracted from Cyclosorus terminans Protect Gut Pathologies Induced by High-Fat Diet in Rats. Nutrients 2025; 17:1387. [PMID: 40284250 PMCID: PMC12030309 DOI: 10.3390/nu17081387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The fern "Cyclosorus terminans" (C. terminans) or "Maiden Fern" contains interruptin A and interruptin B. This plant could attenuate obesity, insulin resistance, and fatty liver in rats fed a high-fat/calorie diet. However, the benefits of C. terminans to the gut remain unknown. We investigated the protective effect of C. terminans extract against gut dysfunction in rats exposed to a high-fat/calorie diet. METHODS Male Wistar rats were assigned to receive either (1) a normal diet treated with vehicle, (2) a high-fat/calorie diet treated with vehicle, (3) a high-fat/calorie diet treated with 100 mg per kg per day (mg·kg-1·day-1) of C. terminans extract, or (4) a high-fat/calorie diet treated with 200 mg·kg-1·day-1 of C. terminans extract. The rats were euthanized after 12 weeks of treatment to enable feces and colon tissue collection. RESULTS Both 100 and 200 mg·kg-1·day-1 of C. terminans extract reduced body weight (-10.49%; p = 0.030 and -10.54%; p = 0.037, respectively) and ameliorated gut inflammation, gut barrier disruption, changes in short-chain fatty acid levels, and gut dysbiosis caused by high-fat/calorie diet. CONCLUSIONS C. terminans extract attenuated an increase in body weight and exerted prophylactic effects against gut pathologies induced by high-fat/calorie diet.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokphong Suparan
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanon Kunasol
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sujinda Songtrai
- Faculty of Medical Technology, Rangsit University, Pathumthani 12000, Thailand;
| | - Hiranya Pintana
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sireewan Kaewsuwan
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkhla University, Songkhla 90110, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (C.T.); (W.P.); (N.C.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (C.K.); (W.N.); (B.A.); (S.K.); (H.P.); (C.M.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
17
|
Lu J, Huang Y, Zhang Y, Xie J, Guo Q, Yang H, Yang Y, Chen J, Su L. Quercetin ameliorates obesity and inflammation via microbial metabolite indole-3-propionic acid in high fat diet-induced obese mice. Front Nutr 2025; 12:1574792. [PMID: 40308638 PMCID: PMC12040668 DOI: 10.3389/fnut.2025.1574792] [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: 02/11/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Obesity is a chronic metabolic disease, mainly caused by excessive/abnormal fat accumulation, as well as accompanied by endotoxemia and chronic inflammation. Quercetin, a natural flavonoid, may alleviate obesity by regulating gut microbiota and metabolites, but its exact mechanism for improving obesity is unknown. Objectives The aim of this study was to investigate the effects of quercetin on high-fat diet (HFD)-induced obesity in mice. In particular, we focused on the regulatory effects of quercetin on gut microbiota and the tryptophan metabolite indole-3-propionic acid (IPA). Methods The C57BL/6J mice were subjected to a 20-week HFD feeding regimen with concurrent daily oral administration of quercetin or IPA. The body weight, fat accumulation, gut barrier function, and chronic inflammation were determined. Gut microbiota composition was analyzed by 16S rRNA sequencing and IPA levels were measured in serum and feces. In vitro experiments, Caco-2 cells were used to evaluate the effects of IPA and fecal dilutions from quercetin-treated mice on tight junction protein expression and aryl hydrocarbon receptor (AhR) activation. Results Our results revealed that quercetin supplementation significantly mitigated obesity and chronic inflammation, and improved the disrupted gut barrier function through the actvation of AhR/interleukin 22 (IL-22) pathway. 16S rRNA sequencing revealed that quercetin treatment increased the abundance of Lactobacillus. Quercetin intervention increased the levels of IPA in the serum and feces of mice. IPA supplementation alleviated obesity and chronic inflammation, and enhanced intestinal barrier function through AhR activation. The findings were further corroborated by Caco-2 cell experiment, which indicated that the modulation of the dysregulated gut microbiota to change microbial metabolite IPA coordinated the improvement effect of quercetin on gut barrier disruption. Conclusion Quercetin supplementation alleviates obesity by restoring high-fat diet induced gut microbiota disorder, which elevates IPA level to activate AhR/IL-22 pathway, thereby enhancing intestinal barrier integrity and suppressing chronic inflammation.
Collapse
Affiliation(s)
- Jiaxin Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanting Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yujing Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiayu Xie
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qingjun Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huifan Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunyan Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Chen
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lijie Su
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Ionescu VA, Diaconu CC, Gheorghe G, Mihai MM, Diaconu CC, Bostan M, Bleotu C. Gut Microbiota and Colorectal Cancer: A Balance Between Risk and Protection. Int J Mol Sci 2025; 26:3733. [PMID: 40332367 PMCID: PMC12028331 DOI: 10.3390/ijms26083733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiome, a complex community of microorganisms residing in the intestinal tract, plays a dual role in colorectal cancer (CRC) development, acting both as a contributing risk factor and as a protective element. This review explores the mechanisms by which gut microbiota contribute to CRC, emphasizing inflammation, oxidative stress, immune evasion, and the production of genotoxins and microbial metabolites. Fusobacterium nucleatum, Escherichia coli (pks+), and Bacteroides fragilis promote tumorigenesis by inducing chronic inflammation, generating reactive oxygen species, and producing virulence factors that damage host DNA. These microorganisms can also evade the antitumor immune response by suppressing cytotoxic T cell activity and increasing regulatory T cell populations. Additionally, microbial-derived metabolites such as secondary bile acids and trimethylamine-N-oxide (TMAO) have been linked to carcinogenic processes. Conversely, protective microbiota, including Lactobacillus, Bifidobacterium, and Faecalibacterium prausnitzii, contribute to intestinal homeostasis by producing short-chain fatty acids (SCFAs) like butyrate, which exhibit anti-inflammatory and anti-carcinogenic properties. These beneficial microbes enhance gut barrier integrity, modulate immune responses, and inhibit tumor cell proliferation. Understanding the dynamic interplay between pathogenic and protective microbiota is essential for developing microbiome-based interventions, such as probiotics, prebiotics, and fecal microbiota transplantation, to prevent or treat CRC. Future research should focus on identifying microbial biomarkers for early CRC detection and exploring personalized microbiome-targeted therapies. A deeper understanding of host-microbiota interactions may lead to innovative strategies for CRC management and improved patient outcomes.
Collapse
Affiliation(s)
- Vlad Alexandru Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
- Academy of Romanian Scientists, 050085 Bucharest, Romania;
| | - Gina Gheorghe
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Mara-Madalina Mihai
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Department of Oncologic Dermathology, “Elias” University Emergency Hospital, 010024 Bucharest, Romania
| | - Carmen Cristina Diaconu
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
| | - Marinela Bostan
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
- Department of Immunology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| | - Coralia Bleotu
- Academy of Romanian Scientists, 050085 Bucharest, Romania;
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060023 Bucharest, Romania
| |
Collapse
|
19
|
Shin C, Lee HJ, Kim YJ, Choi S, Park SK. Dietary exposure assessment of 9 food emulsifiers to Korean population and their health effects. Food Chem 2025; 471:142807. [PMID: 39793355 DOI: 10.1016/j.foodchem.2025.142807] [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: 12/28/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
In this study, dietary exposures to 9 food emulsifiers, including 4 polysorbates and 5 esters of fatty acids, were assessed in Korean population. For the exposure assessment, three scenarios of the consumption, including mean and P95 in whole population and mean in consumed population, were applied. As a result, the EDIs of 9 emulsifiers were overall low compared to the ADIs. The total EDI to polysorbates was 1.72 % of the ADI in whole population. Exposures to polysorbate 20, 60, 65, and 80 were 0.02, 1.14, 0.39, and 0.29 % of the ADIs in whole population, respectively. Exposures to 5 esters of fatty acids were ranged from 0.57 to 6.13 % of the ADIs in whole population. In conclusion, it is considered that there are no safety concerns regarding the current consumption of the food emulsifiers. However, considering the change of food consumption, use levels, and health-based guidance values, periodic re-evaluation is necessary.
Collapse
Affiliation(s)
- Choonshik Shin
- Food Additives Standard Division, Ministry of Food and Drug Safety, Osong, Cheongju 28159, Republic of Korea.
| | - Hyun Ji Lee
- Food Additives Standard Division, Ministry of Food and Drug Safety, Osong, Cheongju 28159, Republic of Korea
| | - Young-Jun Kim
- Department of Food Science and Technology, Seoul National University of Science & Technology, Seoul 01811, Republic of Korea
| | - Shinai Choi
- KnA Consulting Co., Ltd, Suji, Yongin 16864, Republic of Korea
| | - Sung-Kwan Park
- Food Additives Standard Division, Ministry of Food and Drug Safety, Osong, Cheongju 28159, Republic of Korea
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Liu Z, Chen M, Zheng W, Zhan X, Sui W, Huang H, Jiang Q, Zhao W. Effect of gut symbiotic bacteria Akkermansia muciniphila on aging-related obesity. Biochem Biophys Res Commun 2025; 756:151606. [PMID: 40081236 DOI: 10.1016/j.bbrc.2025.151606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Recent studies have shown that Akkermansia muciniphila may play a role in regulating lipid metabolism and immune response in diet-induced obese mice. However, in contrast to diet-induced obesity, aging-related obesity is characterized by a gradual increase in body fat proportion over time. This type of obesity is thought to be caused by a combination of factors, including slow metabolism, unhealthy lifestyle choices, and chronic inflammation. Unlike diet-induced obesity, which can occur relatively quickly, aging-related obesity is a long-term and slow process. In this study, we administered Akkermansia muciniphila to aged mice and collected fecal samples to analyze the targeted metabolism of short chain fatty acids (SCFAs). The mice were then euthanized and their abdominal fat was weighed. hematoxylin-eosin (H&E) staining was performed to examine tissue samples. quantitative polymerase chain reaction (qPCR) was used to detect the expression of IL-6 and TNF-α. Flow cytometry was used to examine the proportion of lymphocytes. Enzyme-linked immunosorbent assays (ELISAs) kits were used to measure the levels of inflammatory factors and aging-related indicators. The results indicate that following intragastric administration, the body weight of the aged mice decreased, along with a decrease in abdominal fat and a reduction in the size of fat cells. Additionally, there was a decrease in the mRNA level of inflammatory factors, a decrease in the total number of immune cells in abdominal fat, and a decrease in the proportion of CD8+ CD4-cells. In addition, our findings showed that serum levels of IL-6, TNF-α, and lipopolysaccharide (LPS) were reduced, and catalase (CAT) and thyroid-stimulating hormone (TSH) levels were comparable to those of young mice. The findings revealed that Akkermansia muciniphila has the potential to enhance immune regulation in aged mice, alleviate persistent inflammation, and decrease obesity in this aged mice.
Collapse
Affiliation(s)
- Zebiao Liu
- Pathology, Huizhou First Hospital, Huizhou, 516001, China
| | - Mingquan Chen
- Pathology, Huizhou First Hospital, Huizhou, 516001, China
| | - Wanhua Zheng
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, School of Life Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Xuebing Zhan
- Pathology, Huizhou First Hospital, Huizhou, 516001, China
| | - Wenyan Sui
- Pathology, Huizhou First Hospital, Huizhou, 516001, China
| | - Haijie Huang
- Pathology, Huizhou First Hospital, Huizhou, 516001, China
| | - Qingjiu Jiang
- First Affiliated Hospital of Jiamusi University, Jiamusi, 154000, China
| | - Wenli Zhao
- Pathology, Huizhou First Hospital, Huizhou, 516001, China.
| |
Collapse
|
23
|
Crain E, Minaya DM, de La Serre CB. Microbiota-induced inflammation mediates the impacts of a Western diet on hippocampal-dependent memory. Nutr Res 2025; 138:89-106. [PMID: 40339190 DOI: 10.1016/j.nutres.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/10/2025]
Abstract
Obesity is associated with impaired hippocampal-dependent memory, but the mechanisms driving this pathology are not fully understood. Western diets (WD) contribute to obesity, and previous reviews have described a role for WD in impaired hippocampal-dependent memory. However, there is need for a more detailed description of the pathways by which WD may impair memory. The short vs long-term effect of specific dietary components on brain structure and functions as well as the precise mechanism and molecular pathways involved are still not fully understood. This review focuses on the mechanisms and effects of gut microbiota-driven neuroinflammation. WD leads to changes and imbalance in bacterial taxa abundances that are deleterious to the host health (gut dysbiosis) and studies in rodent models show these changes are sufficient to impair hippocampal-dependent memory. Here, we discuss a variety of proposed mechanisms linking microbiota composition to hippocampal function, with a focus on neuroinflammation. Gut microbiota impacts gastrointestinal barrier function, leading to increased circulating proinflammatory bacterial products, increased blood-brain barrier permeability, and neuroinflammation.
Collapse
Affiliation(s)
- Eden Crain
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Dulce M Minaya
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
24
|
Iqbal M, Yu Q, Tang J, Xiang J. Unraveling the gut microbiota's role in obesity: key metabolites, microbial species, and therapeutic insights. J Bacteriol 2025:e0047924. [PMID: 40183584 DOI: 10.1128/jb.00479-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Obesity, characterized by excessive fat accumulation, stems from an imbalance between energy intake and expenditure, with the gut microbiota playing a crucial role. This review highlights how gut microbiota influences metabolic pathways, inflammation, and adipose tissue regulation in obesity. Specific bacteria and metabolites, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), modulate gut permeability, inflammation, and energy harvest, impacting obesity development. Certain gut bacteria, including Clostridium XIVb, Dorea spp., Enterobacter cloacae, and Collinsella aerofaciens, promote obesity by increasing energy harvest, gut permeability, and inflammatory response through LPS translocation into the bloodstream. Conversely, beneficial bacteria like Akkermansia muciniphila, Lactobacillus spp., and Bifidobacterium spp. enhance gut barrier integrity, regulate SCFA production, and modulate fasting-induced adipose factor, which collectively support metabolic health by reducing fat storage and inflammation. Metabolites such as SCFAs (acetate, propionate, and butyrate) interact with G-protein coupled receptors to regulate lipid metabolism and promote the browning of white adipose tissue (WAT), thus enhancing thermogenesis and energy expenditure. However, LPS contributes to insulin resistance and fat accumulation, highlighting the dual roles of these microbial metabolites in both supporting and disrupting metabolic function. Therapeutic interventions targeting gut microbiota, such as promoting WAT browning and activating brown adipose tissue (BAT), hold promise for obesity management. However, personalized approaches are necessary due to individual microbiome variability. Further research is essential to translate these insights into microbiota-based clinical therapies.
Collapse
Affiliation(s)
- Majid Iqbal
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Yu
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingqun Tang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Changsha, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- NHC Key Laboratory of Carcinogenesis and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
26
|
Ramezan M, Arzhang P, Shin AC. Milk-derived bioactive peptides in insulin resistance and type 2 diabetes. J Nutr Biochem 2025; 138:109849. [PMID: 39870329 DOI: 10.1016/j.jnutbio.2025.109849] [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: 08/20/2024] [Revised: 01/18/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
Diabetes is a global health issue affecting over 6% of the world and 11% of the US population. It is closely linked to insulin resistance, a pivotal factor in Type 2 diabetes development. This review explores a promising avenue for addressing insulin resistance through the lens of Milk-Derived Bioactive Peptides (MBAPs). Taken from casein or whey fractions of various milks, MBAPs exhibit diverse health-promoting properties. Specific interactions between these peptides and enzymes involved in glucose digestion and metabolism have been examined, leading to the identification of some key peptides exerting the effects. This review emphasizes the positive impact of MBAPs on glycemic control through various mechanisms. Different cell lines have been used to investigate MBAPs' effects on insulin signaling, inflammation, and oxidative stress. Preclinical in vivo studies have also shown that MBAPs lower glucose, stimulate insulin, and reduce inflammation. Human trials further substantiate these findings and suggest the potential utility of milk protein hydrolysates containing MBAPs in individuals with insulin resistance or T2D to improve insulin action and glucose homeostasis.
Collapse
Affiliation(s)
- Marjan Ramezan
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Health & Human Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Pishva Arzhang
- Qods Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Andrew C Shin
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Health & Human Sciences, Texas Tech University, Lubbock, Texas, USA.
| |
Collapse
|
27
|
Bona MD, Mota ACCC, Nascimento DSM, Magalhães LMVC, Lima SCVC, Lyra CO, Morais AHA, Marchioni DM, Lima AAM, Maciel BLL. Intestinal absorption area is correlated with cardiovascular risk factors associated with obesity and metabolic syndrome. Nutr Metab Cardiovasc Dis 2025; 35:103803. [PMID: 39939256 DOI: 10.1016/j.numecd.2024.103803] [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: 06/05/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND AND AIM This study aimed to evaluate if the intestinal permeability is associated with overweight/obesity with or without metabolic syndrome (MetS) and correlate intestinal permeability parameters with cardiovascular risk factors. METHODS AND RESULTS This was a cross-sectional study that individuals were divided in three groups: 1) controls (n = 34), 2) overweight/obesity (n = 29), and 3) overweight/obesity + MetS (n = 29). Anthropometric and blood biochemical parameters were used to estimate cardiovascular risk factors. Intestinal permeability was evaluated using the lactulose/mannitol test in urine samples analyzed by High Performance Liquid Chromatography with Pulsed Amperometry Detection. Correlations between intestinal permeability and anthropometric and biochemical parameters were evaluated using Spearman's correlations (r2). Logistic regression models were performed to elucidate variables associated with intestinal permeability parameters. The percentage of urinary excretion of lactulose, mannitol, and the lactulose/mannitol ratio was similar between the studied groups. The percentage of urinary mannitol excretion was positively associated with diastolic blood pressure (r2 = 0.24, p = 0.23), fasting glucose (r2 = 0.26, p = 0.013), fasting insulin (r2 = 0.25, p = 0.015) and HOMA-IR (r2 = 0.26, p = 0.012). The logistic regression showed fasting insulin was associated with a higher mannitol urinary excretion (AOR = 1.08, 95 % CI = 1.02-1.14). CONCLUSION Intestinal permeability was not disrupted in overweight/obesity and MetS, however the results suggest that the increased cardiovascular risk factors were associated with a higher intestinal absorption area. Further studies should investigate other intestinal parameters related to overweight/obesity and MetS in humans.
Collapse
Affiliation(s)
- Mariana D Bona
- Institute of Biomedicine, Department of Medicine, Federal University of Ceará, Fortaleza, 60430-270, Brazil; Graduate Medical Science Program, Department of Medicine, Federal University of Ceará, Fortaleza, 60430-270, Brazil
| | - Ana C C C Mota
- Graduate Health Science Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil
| | - Daniele S M Nascimento
- Graduate Health Science Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil
| | - Lyvia M V C Magalhães
- Institute of Biomedicine, Department of Medicine, Federal University of Ceará, Fortaleza, 60430-270, Brazil
| | - Severina C V C Lima
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Nutrition Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Health Science Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil
| | - Clélia O Lyra
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Nutrition Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Health Science Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil
| | - Ana H A Morais
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Nutrition Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil
| | - Dirce M Marchioni
- University of São Paulo, School of Public Health, Department of Nutrition, SP, Brazil
| | - Aldo A M Lima
- Institute of Biomedicine, Department of Medicine, Federal University of Ceará, Fortaleza, 60430-270, Brazil; Graduate Medical Science Program, Department of Medicine, Federal University of Ceará, Fortaleza, 60430-270, Brazil
| | - Bruna L L Maciel
- Department of Nutrition, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Nutrition Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil; Graduate Health Science Program, Center for Health Science, Federal University of Rio Grande do Norte, Natal, 59078-970, Brazil.
| |
Collapse
|
28
|
Babakhani K, Kucinskas AL, Ye X, Giles ED, Sun Y. Aging immunity: unraveling the complex nexus of diet, gut microbiome, and immune function. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00061. [PMID: 40352822 PMCID: PMC12063687 DOI: 10.1097/in9.0000000000000061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is associated with immune senescence and gut dysbiosis, both of which are heavily influenced by the diet. In this review, we summarize current knowledge regarding the impact of diets high in fiber, protein, or fat, as well as different dietary components (tryptophan, omega-3 fatty acids, and galacto-oligosaccharides) on the immune system and the gut microbiome in aging. Additionally, this review discusses how aging alters tryptophan metabolism, contributing to changes in immune function and the gut microbiome. Understanding the relationship between diet, the gut microbiome, and immune function in the context of aging is critical to formulate sound dietary recommendations for older individuals, and these personalized nutritional practices will ultimately improve the health and longevity of the elderly.
Collapse
Affiliation(s)
| | - Amanda L. Kucinskas
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xiangcang Ye
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Erin D. Giles
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| |
Collapse
|
29
|
Shah K, Khan AS, Kunwar D, Jacob SR, Akbar A, Singh A, Ahmed MMH. Influence of gut microbiota on the pediatric endocrine system and associated disorders. Ann Med Surg (Lond) 2025; 87:2149-2162. [PMID: 40212169 PMCID: PMC11981368 DOI: 10.1097/ms9.0000000000003099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/15/2025] [Indexed: 04/13/2025] Open
Abstract
The microbiota, a complex assembly of microorganisms residing in various body systems, including the gastrointestinal tract, plays a crucial role in influencing various physiological processes in the human body. The dynamic nature of gut microbiota is especially pronounced in children and is influenced by factors like breastfeeding and antibiotic use. Dysbiosis, characterized by alterations in microbiota composition or function, is associated with several pediatric endocrine disorders, such as precocious puberty, polycystic ovarian syndrome, and diabetes mellitus. This review focuses on the intricate relationship between gut microbiota and the pediatric endocrine system. The aim of this narrative review is to critically examine the existing literature to elucidate the impact of gut microbiota on the pediatric endocrine system and associated disorders. Additionally, potential interventions, such as probiotics and current gaps in knowledge, will be discussed. Despite emerging treatments like probiotics, further research is needed to understand and validate their effectiveness in treating pediatric endocrine disorders associated with dysbiosis.
Collapse
Affiliation(s)
- Krutik Shah
- Byramjee Jeejeebhoy (BJ) Medical College and Civil Hospital, Ahmedabad, India
| | - Alina Sami Khan
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Digbijay Kunwar
- Department of Internal Medicine, Bagahi Primary Healthcare Center, Birgunj, Nepal
| | | | - Anum Akbar
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ajeet Singh
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
30
|
Jayasinghe T, Jenkins J, Medara N, Choowong P, Dharmarathne G, Kong F, Cho H, Kim SH, Zhang Y, Franco-Duarte R, Eberhard J, Spahr A. Dietary Fibre Modulates Body Composition, Blood Glucose, Inflammation, Microbiome, and Metabolome in a Murine Model of Periodontitis. Nutrients 2025; 17:1146. [PMID: 40218904 PMCID: PMC11990244 DOI: 10.3390/nu17071146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Dietary fibre plays a crucial role in metabolic regulation, inflammation, and microbiome composition. However, its impact on systemic and oral health, particularly in periodontitis, remains unclear. This study investigated the effects of high- and low-fibre diets on body composition, glycaemic control, inflammation, microbiome, and metabolome in a murine model of experimental periodontitis. Methods: Thirty-six male C57BL/6 mice were randomised to a high-fibre (40% fibre) or low-fibre (5% fibre) diet for eight weeks. Body weight, fat mass, lean mass, fasting blood glucose, serum inflammatory markers, alveolar bone loss, and root length were assessed. Oral and faecal microbiome composition was analysed using 16S rRNA sequencing. Metabolomic and short-chain fatty acid (SCFA) profiling was conducted using liquid chromatography-mass spectrometry (LC-MS). Results: Mice on the high-fibre diet exhibited significantly lower body weight (p < 0.0001), fat mass (p = 0.0007), and lean mass (p < 0.0001) compared to the low-fibre group. Fasting blood glucose levels were significantly lower in the high-fibre group (p = 0.0013). TNF-α and IFN-γ levels were significantly elevated in the low-fibre group (p < 0.0001), suggesting a heightened pro-inflammatory state. While alveolar bone loss and root length did not differ significantly, microbiome analysis revealed distinct bacterial compositions (PERMANOVA, p < 0.05), with fibre-fermenting taxa enriched in high-fibre-fed mice. Metabolomic analysis identified 19 significantly altered metabolites, indicating dietary adaptations. Conclusions: A high-fibre diet improves glycaemic control, reduces systemic inflammation, and alters microbial and metabolic profiles in experimental periodontitis. These findings highlight dietary fibre's role in modulating metabolic and inflammatory pathways relevant to periodontal and systemic diseases.
Collapse
Affiliation(s)
- Thilini Jayasinghe
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Josie Jenkins
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Nidhi Medara
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Phannaphat Choowong
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia;
| | - Fay Kong
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Hanna Cho
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Se Hun Kim
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Yuchen Zhang
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Ricardo Franco-Duarte
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal;
| | - Joerg Eberhard
- The Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (J.J.); (P.C.); (J.E.)
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| | - Axel Spahr
- School of Dentistry, Faculty of Medicine and Health, University of Sydney, Surry Hills, NSW 2006, Australia; (N.M.); (F.K.); (H.C.); (S.H.K.); (Y.Z.); (A.S.)
| |
Collapse
|
31
|
Pang L, Liu Y, Yuan C, Ju Y, Wu J, Cheng M, Jin S, Fan Y, Zhang H, Wang Y, Min D. Yi Mai Granule Improves High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Mice by Regulating Gut Microbiota and Metabolites. Int J Microbiol 2025; 2025:2273986. [PMID: 40166691 PMCID: PMC11955292 DOI: 10.1155/ijm/2273986] [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: 12/14/2023] [Revised: 12/24/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Yi Mai granule (YMG) is a traditional Chinese medicine (TCM) herbal decoction consisting of two TCM formulas: Gua-Lou-Ban-Xia decoction and Si-Jun-Zi decoction. YMG has shown clinical benefit in the treatment of nonalcoholic fatty liver disease (NAFLD), which may be due to its regulatory effects on lipid metabolism. Previous studies have highlighted the importance of the gut microbiota and its metabolites in the use of TCM. However, the effect of YMG on the gut microbiota in the treatment of NAFLD remains unclear. In this study, we established an NAFLD model in ApoE-/- mice and treated them with YMG. High-performance liquid chromatography was adopted to identify the chemical components of YMG. By mapping the candidate targets using network pharmacology, we found that the targets of the main components of YMG were significantly enriched in NAFLD-related pathways. Moreover, 16S rRNA gene sequencing revealed that YMG affected the constitution and metabolism of the gut microbiota in NAFLD model mice, including lipid and carbohydrate metabolism. Similarly, metabolites related to lipid and carbohydrate metabolism in mouse serum were significantly altered by YMG. The correlation heat map and network analyses showed that the gut microbiota and metabolites affected by YMG were closely related to the blood lipid content. Collectively, YMG may exert therapeutic effects by affecting the metabolism of gut microbiota, thus regulating lipid and carbohydrate metabolism. These findings offer novel insight into the pharmacological mechanism of YMG in the treatment of NAFLD and provide theoretical bases for its clinical applications.
Collapse
Affiliation(s)
- Linlin Pang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- Department of Cardiovascular Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yongming Liu
- Experimental Center of Traditional Chinese Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Changbin Yuan
- First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yetao Ju
- Experimental Center of Traditional Chinese Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Junpeng Wu
- First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Meijia Cheng
- Experimental Center of Traditional Chinese Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Sian Jin
- First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Ying Fan
- College of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Huiyong Zhang
- Department of Traditional Chinese Medicine, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Wang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Dongyu Min
- Experimental Center of Traditional Chinese Medicine, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| |
Collapse
|
32
|
Zheng L, Zhang J, Yang J, Wang Y, Zhang Y, Fang K, Wu J, Zheng M. Association of the use of nonfood prebiotics, probiotics, and synbiotics with total and cause-specific mortality: a prospective cohort study. Nutr J 2025; 24:45. [PMID: 40114150 PMCID: PMC11924732 DOI: 10.1186/s12937-025-01104-w] [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: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The use of nonfood prebiotics, probiotics, and synbiotics has approximately tripled in the last 20 years. It is necessary to examine the associations of these substances with all-cause and cause-specific mortality in a large prospective cohort. METHODS This study included 53,333 adults from the National Health and Nutrition Examination Survey 1999-2018. All participants answered questions on the use of dietary supplements and medications, including prebiotics, probiotics, and synbiotics. Death outcomes were determined by linkage to National Death Index records through 31 December 2019. Cox proportional hazards models were used to estimate hazard ratios (HR) and 95% confidence intervals (CI) for mortality from all causes, heart diseases, cancer, and other causes. RESULTS During a mean follow-up of 10.6 years, 9117 deaths were documented, including 2364 heart disease deaths, 1964 cancer deaths, and 4700 other causes deaths. Compared to nonusers, nonfood prebiotic, probiotic, and synbiotic users had a 59% (HR 0.41, 95% CI 0.30 to 0.56), 56% (HR 0.44, 95% CI 0.26 to 0.74), 49% (HR 0.51, 95% CI 0.31 to 0.83), and 64% (HR 0.36, 95% CI 0.23 to 0.59) for lower risk of all-cause, cancer, heart disease, and other causes mortality, respectively. Moreover, the inverse association of the use of prebiotics, probiotics, and synbiotics with mortality was stronger in female participants and participants without hypertension. CONCLUSION The use of nonfood prebiotics, probiotics, and synbiotics is significantly associated with lower all-cause mortality, as well as deaths from heart disease, cancer, and other causes.
Collapse
Affiliation(s)
- Luyan Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Jiaqi Zhang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Yanbo Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Yina Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Kailu Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China
| | - Jie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China.
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, #79 Qingchun Road, 310003, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
33
|
Ge Y, Yang H, Fu Y, Zhou J, Cheng Z, Fan X, Yu Y. A Mendelian randomization study to reveal gut-disc axis: causal associations between gut microbiota with intervertebral disc diseases. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025:10.1007/s00586-025-08795-z. [PMID: 40105993 DOI: 10.1007/s00586-025-08795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/15/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE Emerging evidence suggests a link between gut microbiota and intervertebral disc diseases (IDDs); however, the causal relationships remain unclear. This study aimed to evaluate the causal effects of gut microbiota on the risk of cervical disc disorders (CDD), other intervertebral disc disorders (OIDD), pyogenic intervertebral disc infections, and discitis, shedding light on the potential "gut-disc axis". METHODS Genetic variation data for 202 gut microbiota taxa were obtained from the Dutch Microbiome Project, and disease outcome data were sourced from the FinnGen consortium. A Mendelian Randomization (MR) approach was employed to assess causal relationships, using genetic variants as instrumental variables. Sensitivity analyses, including tests for pleiotropy, heterogeneity, and reverse causation, ensured robust findings. RESULTS The study identified 20 gut microbial taxa with significant associations to IDDs. Notably, taxa within the Erysipelotrichaceae family showed consistent protective effects against OIDD after Bonferroni correction (P < 0.05). Associations between several species and specific diseases, such as Alistipes senegalensis with CDD and Ruminococcus lactaris with discitis, were also observed. Sensitivity analyses confirmed no evidence of confounding or reverse causation. CONCLUSION This study provides evidence of causal relationships between specific gut microbiota and IDDs, supporting the existence of a "gut-disc axis." The findings suggest that microbial dysbiosis may influence spinal health through systemic inflammation and immune regulation. These insights open new possibilities for microbiota-targeted interventions, such as probiotics or dietary modifications, to prevent or manage IDDs. However, further research is required to validate these therapeutic strategies.
Collapse
Affiliation(s)
- Yuanxin Ge
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Huifang Yang
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Yang Fu
- Department of Rehabilitation, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Jie Zhou
- Department of Rehabilitation, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zilin Cheng
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohong Fan
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Yang Yu
- Sichuan Provincial Hospital of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
34
|
He Z, Song C, Wang Z, Dong C, Jiang Q, Yu X, Shan G. Bioinformatics revealed biomarkers for diagnosis in kidney stones. Front Genet 2025; 16:1542840. [PMID: 40171220 PMCID: PMC11959007 DOI: 10.3389/fgene.2025.1542840] [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: 12/11/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
Background One of the most prevalent urinary illnesses is kidney stone formation, often known as nephrolithiasis. The precise processes of kidney stone remain poorly known after substantial investigation. In order to successfully prevent and cure stone formation and recurrence, additional research into the pathophysiology of stone formation is of paramount importance. Ferroptosis is linked to a variety of renal diseases and is a critical factor in the death of cells. However, little is known about how ferroptosis-related genes (FRGs) contribute to the development of kidney stones. Methods The Ferroptosis Database and the Gene Expression Omnibus (GEO) database provided us with information on kidney stones and FRGs, respectively (FerrDb). Results Eight DE-FRGs related to kidney stones were found in total, and they were all closely related to immune response and autophagy management. Following this, among the 8 DE-FRGs, LASSO and SVM-RFE algorithms chose FZD7, STK11, SUV39H1, and LCN2 as marker genes with suitable diagnostic capabilities. These marker genes may be involved in the control of the PPAR signaling pathway, mTOR signaling system, and fatty acid production of kidney stones, according to the functional enrichment analysis that followed. Additionally, 24 drugs that target two marker genes have been found. Despite this, the ceRNA networks have gained that the regulatory relationship between marker genes is rather complex. Additionally, the findings of the CIBERSORT investigation indicated that FZD7 and SUV39H1 may be linked to variations in the immune milieu of people who have kidney stones. Conclusion We developed a diagnostic tool and provided information on the development of kidney stones. In order to confirm its diagnostic applicability for kidney stones, more studies are needed before it may be used in clinical practice.
Collapse
Affiliation(s)
- Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Caitao Dong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinhong Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guang Shan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Fan K, Hua X, Wang S, Efferth T, Tan S, Wang Z. A promising fusion: Traditional Chinese medicine and probiotics in the quest to overcome osteoporosis. FASEB J 2025; 39:e70428. [PMID: 40047492 DOI: 10.1096/fj.202403209r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/23/2025] [Accepted: 02/19/2025] [Indexed: 05/13/2025]
Abstract
Botanical drugs and probiotic supplements present safer alternative options for the prevention and treatment of osteoporosis (OP). However, pathological disorders of the gut microbiota and the specific properties of probiotics and traditional Chinese medicine (TCM) significantly limit their therapeutic efficacy. Given the favorable synergistic and complementary effects between probiotics and herbal medicines, a creative combination of these approaches may address the issue of their current limited efficacy. A comprehensive analysis is necessary to provide a detailed review of their potential for combination, the mechanisms behind their synergy, scientific applications, and future developments. There exists a complex relationship between gut microbiota and OP, and the underlying regulatory mechanisms are multidimensional, involving the production of pro-inflammatory metabolites, immune system disruption, and the impairment of the intestinal mucosal barrier. Furthermore, we analyzed the complex mechanisms and potential connections between probiotics, TCM, and their combined applications. We highlighted the principle of complementary gain and the substantial therapeutic potential of their organic combination, which facilitates the release of active substances in TCM, increases the bioavailability of TCM, enhances probiotic delivery efficiency, and exerts synergistic effects. The combined use of probiotics and TCM offers a safe and effective strategy for managing OP and presents an innovative and promising direction for the future development of modern phytomedicine.
Collapse
Affiliation(s)
- Kangcheng Fan
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin Hua
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Shuwan Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Shengnan Tan
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
36
|
Hyder N, Raza ML. Stress and the gut microbiota-brain axis. PROGRESS IN BRAIN RESEARCH 2025; 291:175-203. [PMID: 40222779 DOI: 10.1016/bs.pbr.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
The gut microbiota-brain axis is a complex system that links the bacteria in our gut with our brain, it plays a part in what way we respond to stress. This chapter explores how stress affects the types of bacteria in the gut and shows the two-way connection between them. Stress can change the bacteria in our gut, which can cause various problems related to stress, like depression, anxiety, and irritable bowel syndrome (IBS). Figuring out how these interactions may help us develop new treatments that focus on the connection between gut bacteria and the brain. This chapter looks at how gut bacteria could help identify stress-related problems. It also discusses the difficulties and possibilities of using this research in medical practice. In the end, the chapter talks about what comes next in this quickly changing area. It highlights how important it is to include research about the gut-brain connection in overall public health plans.
Collapse
Affiliation(s)
- Noorulain Hyder
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Karachi, Pakistan; HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.
| | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| |
Collapse
|
37
|
Saad MJA, Santos A. The Microbiota and Evolution of Obesity. Endocr Rev 2025; 46:300-316. [PMID: 39673174 PMCID: PMC11894537 DOI: 10.1210/endrev/bnae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Obesity is a major global concern and is generally attributed to a combination of genetic and environmental factors. Several hypotheses have been proposed to explain the evolutionary origins of obesity epidemic, including thrifty and drifty genotypes, and changes in thermogenesis. Here, we put forward the hypothesis of metaflammation, which proposes that due to intense selection pressures exerted by environmental pathogens, specific genes that help develop a robust defense mechanism against infectious diseases have had evolutionary advantages and that this may contribute to obesity in modern times due to connections between the immune and energy storage systems. Indeed, incorporating the genetic variations of gut microbiota into the complex genetic framework of obesity makes it more polygenic than previously believed. Thus, uncovering the evolutionary origins of obesity requires a multifaceted approach that considers the complexity of human history, the unique genetic makeup of different populations, and the influence of gut microbiome on host genetics.
Collapse
Affiliation(s)
- Mario J A Saad
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| |
Collapse
|
38
|
Nguyen MT, Lian A, Guilford FT, Venketaraman V. A Literature Review of Glutathione Therapy in Ameliorating Hepatic Dysfunction in Non-Alcoholic Fatty Liver Disease. Biomedicines 2025; 13:644. [PMID: 40149620 PMCID: PMC11940638 DOI: 10.3390/biomedicines13030644] [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: 01/13/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global cause of liver dysfunction. This spectrum of hepatic disorders can progress to severe conditions, such as non-alcoholic steatohepatitis (NASH) and cirrhosis, due to oxidative stress and sustained cellular injury. With limited pharmacological options, glutathione (GSH), a key antioxidant, has shown promising potential in reducing oxidative stress, maintaining redox balance, and improving liver function. This literature review examines studies from 2014-2024 exploring GSH therapy in NAFLD patients. Eligible studies assessed GSH as the primary intervention for NAFLD in human subjects, reporting outcomes such as liver function or oxidative stress markers. Randomized clinical trials (RCTs) were eligible, while combination therapy studies were included if GSH's effect could be isolated. Exclusions applied to non-NAFLD studies, animal/in vitro models, and non-GSH antioxidant interventions. Analysis of three studies (totaling 109 participants) demonstrated consistent improvements in alanine transaminase (ALT) levels and reductions in oxidative stress markers like 8-hydroxy-2-deoxyguanosine (8-OHdG). However, small sample sizes and inconsistent protocols limit generalizability. Further large-scale RCTs are required to confirm GSH's efficacy, determine optimal dosing, and assess long-term effects. This literature review highlights GSH's potential as a novel NAFLD therapeutic strategy while emphasizing the need for further studies to refine its clinical application.
Collapse
Affiliation(s)
- Michelle Thuy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| |
Collapse
|
39
|
Wang W, Wang Y, Sanidad KZ, Wang Y, Zhang J, Yang W, Sun Q, Bayram I, Song R, Yang H, Johnson D, Sherman HL, Kim D, Minter LM, Wong JJL, Zeng MY, Decker EA, Zhang G. Oxidized Polyunsaturated Fatty Acid Promotes Colitis and Colitis-Associated Tumorigenesis in Mice. J Crohns Colitis 2025; 19:jjae148. [PMID: 39279209 DOI: 10.1093/ecco-jcc/jjae148] [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: 04/29/2024] [Revised: 07/23/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND AND AIMS Human studies suggest that a high intake of polyunsaturated fatty acid (PUFA) is associated with an increased risk of inflammatory bowel disease (IBD). PUFA is highly prone to oxidation. To date, it is unclear whether unoxidized or oxidized PUFA is involved in the development of IBD. Here, we aim to compare the effects of unoxidized PUFA vs oxidized PUFA on the development of IBD and associated colorectal cancer. METHODS We evaluated the effects of unoxidized and oxidized PUFA on dextran sodium sulfate (DSS)-induced and IL-10 knockout-induced colitis, and azoxymethane/DSS-induced colon tumorigenesis in mice. Additionally, we studied the roles of gut microbiota and Toll-like receptor 4 (TLR4) signaling involved. RESULTS Administration of a diet containing oxidized PUFA, at human consumption-relevant levels, increases the severity of colitis and exacerbates the development of colitis-associated colon tumorigenesis in mice. Conversely, a diet rich in unoxidized PUFA does not promote colitis. Furthermore, oxidized PUFA worsens colitis-associated intestinal barrier dysfunction and leads to increased bacterial translocation, and it fails to promote colitis in TLR4 knockout mice. Finally, oxidized PUFA alters the diversity and composition of gut microbiota, and it fails to promote colitis in mice lacking the microbiota. CONCLUSIONS These results support that oxidized PUFA promotes the development of colitis and associated tumorigenesis in mouse models via TLR4- and gut microbiota-dependent mechanisms. Our findings highlight the potential need to update regulation policies and industrial standards for oxidized PUFA levels in food.
Collapse
Affiliation(s)
- Weicang Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Yuxin Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - Katherine Z Sanidad
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Yige Wang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Jianan Zhang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Wenqi Yang
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Quancai Sun
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Ipek Bayram
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Haixia Yang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - David Johnson
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Heather L Sherman
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Daeyoung Kim
- Department of Mathematics & Statistics, University of Massachusetts, Amherst, MA, USA
| | - Lisa M Minter
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, USA
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Justin J-L Wong
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Eric A Decker
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Guodong Zhang
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| |
Collapse
|
40
|
Huang H, Liu S, Peng Z, Wang B, Zhan S, Huang S, Li W, Liu D, Yang X, Zhu Y, Xiao W. Comparative effects of different sugar substitutes: Mogroside V, stevioside, sucralose, and erythritol on intestinal health in a type 2 diabetes mellitus mouse. Food Funct 2025; 16:2108-2123. [PMID: 39969196 DOI: 10.1039/d4fo04446k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Intestinal health disorders significantly contribute to the development of type 2 diabetes mellitus (T2DM). Sugar substitutes such as mogroside V (MOG), stevioside (ST), sucralose (TGS), and erythritol (ERT), are increasingly used in T2DM management as alternatives to sucrose (SUC). However, their effects on intestinal health in T2DM have not been fully compared. In the present study, we established a T2DM mouse model using a high-fat diet and streptozotocin injection. These mice were treated with equal doses of SUC, MOG, ST, TGS, or ERT for 4 weeks to evaluate the effects of these sugar substitutes on intestinal health in T2DM. T2DM mice exhibited increased intestinal permeability, reduced goblet cell numbers, elevated pro-inflammatory cytokine levels, and alterations in both gut microbiota and metabolite composition. After 4 weeks of treatment, MOG showed the most significant benefits. MOG activates the PI3K/AKT pathway, enhancing the expression of tight junction proteins, which improves intestinal barrier function and reduces permeability. This is accompanied by NF-κB inhibition, leading to reduced pro-inflammatory cytokine production and increased mucus secretion. These changes help maintain healthy gut microbiota and metabolites, preventing pathogenic bacteria from entering the bloodstream. ST downregulates NF-κB to alleviate intestinal inflammation and improves gut microbiota and metabolic homeostasis in T2DM. ERT has less beneficial effects. TGS and SUC reduce intestinal inflammation and have a better effect on the duodenum. However, TGS has a negative effect on the colon microbiota and metabolites, whereas SUC has a negative effect on the colon microbiota alone. MOG improved intestinal health in T2DM by modulating the PI3K/AKT and NF-κB pathways, whereas ST primarily modulated NF-κB to alleviate intestinal inflammation. Both treatments were effective, with MOG showing the best performance. Therefore, MOG can be considered a viable alternative to SUC for T2DM management.
Collapse
Affiliation(s)
- Huaxue Huang
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sha Liu
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Zhi Peng
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Bin Wang
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Shuang Zhan
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sirui Huang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Wei Li
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Dai Liu
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Xiulian Yang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Yizhun Zhu
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
| | - Wenjun Xiao
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
41
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
42
|
Chen D, Bi X, Feng Q, Sun Y. Supplementation with Lentil ( Lens culinaris) Hull Soluble Dietary Fiber Ameliorates Sodium Dextran Sulfate-Induced Colitis and Behavioral Deficits via the Gut-Brain Axis. Foods 2025; 14:870. [PMID: 40077572 PMCID: PMC11898428 DOI: 10.3390/foods14050870] [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: 02/08/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
In this study, the impact of lentil hull soluble dietary fibers (SDFs) on colitis and behavioral deficits in mice was assessed. Structural characterizations of SDFs confirmed that cellulase-modified soluble dietary fiber exhibited better physicochemical properties: more porous microstructure; similar polysaccharide structure; more stable particle size distribution; higher crystallinity; better adsorption capacity; and lower viscosity. Additionally, we explored its potential cognitive benefits via the gut-brain axis by behavioral tests, histopathology, 16S rRNA sequencing, gas chromatography and metabolomics analysis. The results showed that SDFs significantly improved inflammatory symptoms in colon and brain and cognitive behaviors. LSDF had better efficacy than HSDF. LSDF intervention decreased the harmful bacteria abundance (Bacteroides, Flexispira and Escherichia, etc.) and increased beneficial bacteria abundance (Aggregatibacter and Helicobacter, etc.). LSDF also affected brain metabolites through the sphingolipid metabolism. Spearman correlation analysis showed that there was a positive correlation between harmful bacteria with inflammatory factors (LPS, IL-1β, IL-6, and TNF-α, etc.) and sphingolipid metabolites, while beneficial bacteria were positively correlated with brain-derived neurotrophic factor (BDNF), IL-10, and cognitive behavior. This study highlights the value of SDFs in future diet-based therapeutic strategies targeting gut-brain interactions.
Collapse
Affiliation(s)
- Dongying Chen
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Xin Bi
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Qian Feng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| |
Collapse
|
43
|
Santos GL, Dias Costa EF, Dalla Costa AP, Zanesco AM, Simoes MR, Rogério F, Demolin DMR, Navarro CDC, Velloso LA, Francisco A, Castilho RF. Influence of Mitochondrial NAD(P) + Transhydrogenase (NNT) on Hypothalamic Inflammation and Metabolic Dysfunction Induced by a High-Fat Diet in Mice. Horm Metab Res 2025; 57:199-207. [PMID: 39481390 DOI: 10.1055/a-2420-6549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
The mitochondrial protein NAD(P)+ transhydrogenase (NNT) has been implicated in the metabolic derangements observed in obesity. Mice with the C57BL/6J genetic background bear a spontaneous mutation in the Nnt gene and are known to exhibit increased susceptibility to diet-induced metabolic disorders. Most of the studies on NNT in the context of diet-induced obesity have compared C57BL/6J mice with other mouse strains, where differences in genetic background can serve as confounding factors. Moreover, these studies have predominantly employed a high-fat diet (HFD) consisting of approximately 60% of calories from fat, which may not accurately mimic real-world fat-rich diets. In this study, we sought to examine the role of NNT in diet-induced hypothalamic inflammation and metabolic syndrome by using a congenic mice model lacking NNT, along with a HFD providing approximately 45% of calories from fat. Our findings indicate that mice lacking NNT were more protected from HFD-induced weight gain but presented a worse performance on glucose tolerance test, albeit not in insulin tolerance test. Interestingly, the brown adipose tissue of HFD-fed Nnt +/+ mice presented a greater mass and a higher whole-tissue ex-vivo oxygen consumption rate. Also, HFD increased the expression of the inflammatory markers Il1β, Tlr4 and Iba1 in the hypothalamus of Nnt -/- mice. In conclusion, our study highlights the importance of NNT in the context of diet-induced obesity and metabolic syndrome, indicating its contribution to mitigate hypothalamic inflammation and suggesting its role in the brown adipose tissue increased mass.
Collapse
Affiliation(s)
| | | | | | - Ariane Maria Zanesco
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Marcela Reymond Simoes
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Fábio Rogério
- Department of Pathology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniele Masselli Rodrigues Demolin
- Multidisciplinary Center for Biological Investigation on Laboratory Animals Science, State University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Annelise Francisco
- Department of Experimental Medical Science, Lunds Universitet, Lund, Sweden
- Department of Pathology, State University of Campinas (UNICAMP), Campinas, Brazil
| | | |
Collapse
|
44
|
Hashemi D, Fard MV, Mohammadhasani K, Barati M, Nattagh‐Eshtivani E. Carotenoids Improve Obesity and Fatty Liver Disease via Gut Microbiota: A Narrative Review. Food Sci Nutr 2025; 13:e70092. [PMID: 40071130 PMCID: PMC11893484 DOI: 10.1002/fsn3.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Carotenoids are natural micronutrients found in plants and microorganisms, but not synthesized by animals. Carotenoids show various biological activities, including antioxidant properties, regulation of cell growth, and modulation of gene expression and immune responses. The rising global incidence of fatty liver disease (FLD) and obesity highlights the importance of carotenoids in chronic progressive conditions. Gut microbiota (GM) dysbiosis is associated with the development and progression of obesity and FLD due to the effects of metabolites such as lipopolysaccharide (LPS), bile acids (BAs), and short-chain fatty acids (SCFAs). Furthermore, GM may affect intestinal barrier integrity. This review evaluates the potential impact of carotenoids on GM and intestinal barrier function, and their subsequent effects on obesity and FLD. We searched through a wide range of databases, such as Web of Science, Scopus, EMBASE, and PubMed, to collect data for our non-systematic review of English literature. Carotenoids such as lycopene, zeaxanthin, fucoxanthin, capsanthin, astaxanthin, and lutein can regulate GM composition and improve obesity and FLD by affecting energy expenditure, food intake, lipid profile, liver fat deposition, liver enzymes, inflammatory markers, glucose homeostasis, and bile acids. These carotenoids improve obesity and FLD through GM metabolites such as SCFAs and LPS. Our findings show that dietary supplementation of lycopene, zeaxanthin, fucoxanthin, capsanthin, astaxanthin, and lutein can positively affect obesity and FLD by regulating GM and intestinal barrier integrity.
Collapse
Affiliation(s)
- Dorna Hashemi
- Department of NutritionSarvestan Branch, Islamic Azad UniversitySarvestanIran
| | - Mohammad Vahedi Fard
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research CenterGonabad University of Medical SciencesGonabadIran
| | - Kimia Mohammadhasani
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research CenterGonabad University of Medical SciencesGonabadIran
| | - Mehdi Barati
- Department of Pathobiology and Laboratory SciencesNorth Khorasan University of Medical SciencesBojnurdIran
| | - Elyas Nattagh‐Eshtivani
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research CenterGonabad University of Medical SciencesGonabadIran
| |
Collapse
|
45
|
Sellami E, Evangelista-Silva PH, Jordão Teixeira C, Diop K, Mitchell P, Forato Anhê F. High fructose rewires gut glucose sensing via glucagon-like peptide 2 to impair metabolic regulation in mice. Mol Metab 2025; 93:102101. [PMID: 39855562 PMCID: PMC11830333 DOI: 10.1016/j.molmet.2025.102101] [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: 10/30/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Increased fructose consumption contributes to type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD), but the mechanisms are ill-defined. Gut nutrient sensing involves enterohormones like Glucagon-like peptide (Glp)2, which regulates the absorptive capacity of luminal nutrients. While glucose is the primary dietary energy source absorbed in the gut, it is unknown whether excess fructose alters gut glucose sensing to impair blood glucose regulation and liver homeostasis. METHODS Mice were fed diets where carbohydrates were either entirely glucose (70 %Kcal) or glucose partially replaced with fructose (8.5 %Kcal). Glp2 receptor (Glp2r) was inhibited with Glp2 (3-33) injections. Glucose tolerance, insulin sensitivity, and gut glucose absorption were concomitantly assessed, and enteric sugar transporters and absorptive surface were quantified by RT-qPCR and histological analysis, respectively. RESULTS High fructose feeding led to impairment of blood glucose disposal, ectopic fat accumulation in the liver, and hepatic (but not muscle or adipose tissue) insulin resistance independent of changes in fat mass. This was accompanied by increased gut glucose absorption, which preceded glucose intolerance and liver steatosis. Fructose upregulated glucose transporters and enlarged the gut surface, but these effects were prevented by Glp2r inhibition. Blocking Glp2r prevented fructose-induced impairments in glucose disposal and hepatic lipid handling. CONCLUSION Excess fructose impairs blood glucose and liver homeostasis by rewiring gut glucose sensing and exacerbating gut glucose absorption. Our findings are positioned to inform novel early diagnostic tools and treatments tailored to counter high fructose-induced metabolic derangements predisposing to T2D and MASLD.
Collapse
Affiliation(s)
- Eya Sellami
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Medicine, Faculty of Medicine, Université Laval - 1050, Av. de la Médecine, Québec, QC, G1V 0A6, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada
| | - Paulo Henrique Evangelista-Silva
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo - 1524, Av. Prof. Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Caio Jordão Teixeira
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo - 1524, Av. Prof. Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Khoudia Diop
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada
| | - Patricia Mitchell
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada
| | - Fernando Forato Anhê
- Québec Heart and Lung Institute Research Center, Université Laval - 2725, Ch. Sainte-Foy, Québec, QC, G1V 4G5, Canada; Department of Medicine, Faculty of Medicine, Université Laval - 1050, Av. de la Médecine, Québec, QC, G1V 0A6, Canada; Institute of Nutrition and Functional Foods, Université Laval - 2440 Bd. Hochelaga, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
46
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [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: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
47
|
García G, Carlin M, Cano RDJ. Holobiome Harmony: Linking Environmental Sustainability, Agriculture, and Human Health for a Thriving Planet and One Health. Microorganisms 2025; 13:514. [PMID: 40142407 PMCID: PMC11945859 DOI: 10.3390/microorganisms13030514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The holobiome is an interconnected network of microbial ecosystems spanning soil, plants, animals, humans, and the environment. Microbial interactions drive nutrient cycling, pathogen suppression, and climate regulation. Soil microbiomes facilitate carbon sequestration and enhance soil fertility, while marine microbiomes contribute to carbon capture and climate stability. However, industrial agriculture, extensive herbicide use, antibiotic overuse, and climate change threaten microbial diversity, leading to ecosystem and health disruptions. Probiotic interventions help to restore microbial balance. In human health, probiotics support gut microbiota diversity, reduce inflammation, and regulate metabolism. In agriculture, soil probiotics enhance microbial diversity, improve nutrient cycling, and degrade contaminants, increasing crop yields and soil health. Case studies show that microbial inoculants effectively remediate degraded soils and enhance nutrient uptake. Artificial intelligence is transforming microbiome research by enabling predictive modeling, precision probiotic design, and microbial consortia optimization. Interdisciplinary collaboration and supportive policies are essential for restoring microbial equilibria, ensuring ecosystem resilience, and promoting long-term sustainability. The integration of artificial intelligence, clinical research, and sustainable practices is crucial for advancing holobiome science. The holobiome framework underscores the need for interdisciplinary collaboration to address global challenges, bridging environmental sustainability, agriculture, and public health for a resilient future.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Hospital Hermanos Ameijeiras, La Habana 10400, Cuba;
| | | | - Raul de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
- Chauvell, LLC, San Luis Obispo, CA 93401, USA
| |
Collapse
|
48
|
Bianchi F, Roccabianca P, Vianello E, Gentile G, La Sala L, Bandera F, Tacchini L, Zoia R, Corsi Romanelli MM, Dozio E. Inhibition of DPP-4 Attenuates Endotoxemia-Induced NLRC4 Inflammasome and Inflammation in Visceral Adipose Tissue of Mice Fed a High-Fat Diet. Biomolecules 2025; 15:333. [PMID: 40149869 PMCID: PMC11940500 DOI: 10.3390/biom15030333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammasomes are protein complexes that trigger pro-inflammatory responses and promote many diseases, including adipose tissue dysfunction. Linagliptin (L), a DPP-4 inhibitor used for type 2 diabetes therapy, has putative anti-inflammatory effects. This work explores L effects on inflammasome regulation, inflammation, and adipose tissue dysfunction in obese mice. Male C57BL/6N mice were fed a normal chow (NC) diet, high-fat (HF) diet, or HF diet with L (HFL) for 15 weeks. Gene expression and histological examinations were performed on visceral (VAT) and subcutaneous (SAT) adipose tissue samples. Biomarkers were quantified on sera. Murine macrophages were utilized for in vitro analyses. L decreased HF-induced endotoxemia and circulating inflammatory indicators. Despite having no effect on body weight, L reduced VAT inflammation by decreasing endotoxemia-induced NLRC4 inflammasome, inflammation severity, and fat cell hypertrophy. Although SAT response differed from VAT, inflammation was slightly reduced in this tissue too. In vitro, L modulated inflammation by directly reducing the pro-inflammatory macrophage phenotype. In obesity, increased NLRC4 inflammasome expression links endotoxemia and VAT inflammation. L protected against endotoxemia, maybe by affecting gut permeability and VAT responses. The decreased polarization of macrophages toward a pro-inflammatory phenotype and the reduction in adipocyte hypertrophy are involved in the response to L.
Collapse
Affiliation(s)
- Francesca Bianchi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Paola Roccabianca
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy;
| | - Elena Vianello
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Guendalina Gentile
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
| | - Lucia La Sala
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Francesco Bandera
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- IRCCS MultiMedica, 20138 Milan, Italy
| | - Lorenza Tacchini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Riccardo Zoia
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
| | - Massimiliano M. Corsi Romanelli
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Dipartimento di Patologia Clinica e Sperimentale, IRCCS Istituto Auxologico, 20149 Milan, Italy
| | - Elena Dozio
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy; (F.B.); (E.V.); (G.G.); (L.L.S.); (F.B.); (L.T.); (R.Z.); (M.M.C.R.)
- Laboratorio Sperimentale Ricerche Biomarcatori di Danno d’Organo, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
49
|
Peña-Durán E, García-Galindo JJ, López-Murillo LD, Huerta-Huerta A, Balleza-Alejandri LR, Beltrán-Ramírez A, Anaya-Ambriz EJ, Suárez-Rico DO. Microbiota and Inflammatory Markers: A Review of Their Interplay, Clinical Implications, and Metabolic Disorders. Int J Mol Sci 2025; 26:1773. [PMID: 40004236 PMCID: PMC11854938 DOI: 10.3390/ijms26041773] [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: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The human microbiota, a complex ecosystem of microorganisms, plays a pivotal role in regulating host immunity and metabolism. This review investigates the interplay between microbiota and inflammatory markers, emphasizing their impact on metabolic and autoimmune disorders. Key inflammatory biomarkers, such as C-reactive protein (CRP), interleukin-6 (IL-6), lipopolysaccharides (LPS), zonulin (ZO-1), and netrin-1 (Ntn1), are discussed in the context of intestinal barrier integrity and chronic inflammation. Dysbiosis, characterized by alterations in microbial composition and function, directly modulates the levels and activity of these biomarkers, exacerbating inflammatory responses and compromising epithelial barriers. The disruption of microbiota is further correlated with increased intestinal permeability and chronic inflammation, serving as a precursor to conditions like type 2 diabetes (T2D), obesity, and non-alcoholic fatty liver disease. Additionally, this review examines therapeutic strategies, including probiotics and prebiotics, designed to restore microbial balance, mitigate inflammation, and enhance metabolic homeostasis. Emerging evidence positions microbiota-targeted interventions as critical components in the advancement of precision medicine, offering promising avenues for diagnosing and treating inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Emiliano Peña-Durán
- Licenciatura en Médico Cirujano y Partero, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jesús Jonathan García-Galindo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Luis Daniel López-Murillo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Alfredo Huerta-Huerta
- Hospital Medica de la Ciudad, Santa Catalina, Calle. Pablo Valdez 719, La Perla, Guadalajara 44360, Mexico
| | - Luis Ricardo Balleza-Alejandri
- Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alberto Beltrán-Ramírez
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Elsa Janneth Anaya-Ambriz
- Departamento de Ciencias de la Salud, Centro Universitario de los Valles, Universidad de Guadalajara, Ameca 46708, Mexico
| | - Daniel Osmar Suárez-Rico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Guadalajara 44430, Mexico
| |
Collapse
|
50
|
Wu T, Dai Z, Luo Y, Yu Q, Zhang Y, Bao X, Li R, Zhang Y, Hao J, Shen Q, Xue Y. Refined highland barley ameliorates obesity-associated insulin resistance in high-fat diet-fed mice by targeting the gut microbiota and liver transcriptomics. Eur J Nutr 2025; 64:96. [PMID: 39964534 DOI: 10.1007/s00394-025-03614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE It is generally believed that refined grains lack nutritional value compared to whole grains. The objective of this study was to investigate whether refined highland barley (RHB) holds the potential to combat obesity-associated insulin resistance. METHODS Thirty-two male 6-week-old C57BL/6J mice were randomly divided into four groups fed with a normal chow diet, a high-fat diet (HFD), a 30% RHB supplemented HFD diet, and a 30% whole-grain highland barley (WGHB) supplemented HFD diet. We examined the anti-obesity and anti-insulin resistance effects of RHB and compared them with WGHB in mice. RESULTS RHB intervention effectively improved obesity and insulin resistance, enhanced the intestinal mucosal barrier, and reduced inflammation. Moreover, it promoted the abundance of beneficial gut bacteria such as Akkermansia, Bifidobacterium, Lachnospiraceae_NK4A136_group, Lachnospiraceae_UCG-001, Alloprevotella, and increased the production of short-chain fatty acids (SCFAs) in faeces. Additionally, RHB intervention modulated liver gene transcription, downregulating inflammatory genes like IRF3/7, STAT1/2, NLRP3, and TLR2. CONCLUSIONS RHB could effectively alleviate obesity-related insulin resistance by targeting gut microbiota and liver transcriptomics, and its beneficial impacts are comparable to those of WGHB.
Collapse
Affiliation(s)
- Tong Wu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Zijian Dai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yingting Luo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Qinye Yu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yiyun Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Xin Bao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Rong Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yuhong Zhang
- Tibet Academy of Agriculture and Animal Sciences, Lhasa, 850002, China
| | - Jing Hao
- Qinghai Tianyoude Technology Investment Management Group Co., Ltd, Qinghai, 810500, China
- Qinghai Engineering Technology Research Institute for Comprehensive Utilization of Highland Barley Resources, Qinghai, 810016, China
| | - Qun Shen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China
| | - Yong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
- National Center of Technology Innovation in Food Industry, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|