1
|
Clasen F, Yildirim S, Arıkan M, Garcia-Guevara F, Hanoğlu L, Yılmaz NH, Şen A, Celik HK, Neslihan AA, Demir TK, Temel Z, Mardinoglu A, Moyes DL, Uhlen M, Shoaie S. Microbiome signatures of virulence in the oral-gut-brain axis influence Parkinson's disease and cognitive decline pathophysiology. Gut Microbes 2025; 17:2506843. [PMID: 40420833 PMCID: PMC12118390 DOI: 10.1080/19490976.2025.2506843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/25/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
The human microbiome is increasingly recognized for its crucial role in the development and progression of neurodegenerative diseases. While the gut-brain axis has been extensively studied, the contribution of the oral microbiome and gut-oral tropism in neurodegeneration has been largely overlooked. Cognitive impairment (CI) is common in neurodegenerative diseases and develops on a spectrum. In Parkinson's Disease (PD) patients, CI is one of the most common non-motor symptoms but its mechanistic development across the spectrum remains unclear, complicating early diagnosis of at-risk individuals. Here, we generated 228 shotgun metagenomics samples of the gut and oral microbiomes across PD patients with mild cognitive impairment (PD-MCI) or dementia (PDD), and a healthy cohort, to study the role of gut and oral microbiomes on CI in PD. In addition to revealing compositional and functional signatures, the role of pathobionts, and dysregulated metabolic pathways of the oral and gut microbiome in PD-MCI and PDD, we also revealed the importance of oral-gut translocation in increasing abundance of virulence factors in PD and CI. The oral-gut virulence was further integrated with saliva metaproteomics and demonstrated their potential role in dysfunction of host immunity and brain endothelial cells. Our findings highlight the significance of the oral-gut-brain axis and underscore its potential for discovering novel biomarkers for PD and CI.
Collapse
Affiliation(s)
- Frederick Clasen
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Suleyman Yildirim
- Department of Medical Microbiology, Istanbul Medipol University International School of Medicine, Istanbul, Türkiye
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Tűrkiye
| | - Muzaffer Arıkan
- Department of Medical Microbiology, Istanbul Medipol University International School of Medicine, Istanbul, Türkiye
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Tűrkiye
| | - Fernando Garcia-Guevara
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Lűtfű Hanoğlu
- Neuroscience Graduate Program and Department of Neurology, Istanbul Medipol University School of Medicine, Istanbul, Tűrkiye
| | - Nesrin H. Yılmaz
- Department of Neurology, Istanbul Medipol University School of Medicine, Istanbul, Tűrkiye
| | - Aysu Şen
- Department of Neurology, Bakırkoy Research and Training Hospital for Psychiatric and Neurological Diseases, Istanbul, Tűrkiye
| | - Handan Kaya Celik
- Department of Neurology, Kocaeli University Faculty of Medicine, Kocaeli, Türkiye
| | | | - Tuǧçe Kahraman Demir
- Department of Electroneurophysiology, Vocational School, Biruni University, Istanbul, Tűrkiye
| | - Zeynep Temel
- Department of Psychology, Faculty of Humanities and Social Sciences, Fatih Sultan Mehmet Vakif University, Istanbul, Tűrkiye
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - David L. Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
2
|
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
|
3
|
Qiao S, Wang T, Sun J, Han J, Dai H, Du M, Yang L, Guo CJ, Liu C, Liu SJ, Liu H. Cross-feeding-based rational design of a probiotic combination of Bacterides xylanisolvens and Clostridium butyricum therapy for metabolic diseases. Gut Microbes 2025; 17:2489765. [PMID: 40190016 PMCID: PMC11980479 DOI: 10.1080/19490976.2025.2489765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/13/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
The human gut microbiota has gained interest as an environmental factor that contributes to health or disease. The development of next-generation live biotherapeutic products (LBPs) is a promising strategy to modulate the gut microbiota and improve human health. In this study, we identified a novel cross-feeding interaction between Bacteroides xylanisolvens and Clostridium butyricum and developed their combination into a novel LBP for treating metabolic syndrome. Using in-silico analysis and in vitro experiments, we demonstrated that B. xylanisolvens supported the growth and butyrate production of C. butyricum by supplying folate, while C. butyricum reciprocated by providing pABA for folate biosynthesis. Animal gavage experiments showed that the two-strain combination LBP exhibited superior therapeutic efficacy against metabolic disorders in high-fat diet-induced obese (DIO) mice compared to either single-strain treatment. Further omics-based analyses revealed that the single-strain treatments exhibited distinct taxonomic preferences in modulating the gut microbiota, whereas the combination LBP achieved more balanced modulation to preserve taxonomic diversity to a greater extent, thereby enhancing the stability and resilience of the gut microbiome. Moreover, the two-strain combinations more effectively restored gut microbial functions by reducing disease-associated pathways and opportunistic pathogen abundance. This work demonstrates the development of new LBP therapy for metabolic diseases from cross-feeding microbial pairs which exerted better self-stability and robust efficacy in complex intestinal environments compared to conventional single-strain LBPs.
Collapse
Affiliation(s)
- Shanshan Qiao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Mengxuan Du
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Lan Yang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chun-Jun Guo
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
4
|
Olivares C, Ruppé E, Ferreira S, Corbel T, Andremont A, de Gunzburg J, Guedj J, Burdet C. A modelling framework to characterize the impact of antibiotics on the gut microbiota diversity. Gut Microbes 2025; 17:2442523. [PMID: 39711113 DOI: 10.1080/19490976.2024.2442523] [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: 09/13/2024] [Revised: 11/08/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
Metagenomic sequencing deepened our knowledge about the role of the intestinal microbiota in human health, and several studies with various methodologies explored its dynamics during antibiotic treatments. We compared the impact of four widely used antibiotics on the gut bacterial diversity. We used plasma and fecal samples collected during and after treatment from healthy volunteers assigned to a 5-day treatment either by ceftriaxone (1 g every 24 h through IV route), ceftazidime/avibactam (2 g/500 mg every 8 h through IV route), piperacillin/tazobactam (1 g/500 mg every 8 h through IV route) or moxifloxacin (400 mg every 24 h through oral route). Antibiotic concentrations were measured in plasma and feces, and bacterial diversity was assessed by the Shannon index from 16S rRNA gene profiling. The relationship between the evolutions of antibiotic fecal exposure and bacterial diversity was modeled using non-linear mixed effects models. We compared the impact of antibiotics on gut microbiota diversity by simulation, using various reconstructed pharmacodynamic indices. Piperacillin/tazobactam was characterized by the highest impact in terms of intensity of perturbation (maximal [IQR] loss of diversity of 27.3% [1.9; 40.0]), while moxifloxacin had the longest duration of perturbation, with a time to return to 95% of baseline value after the last administration of 13.2 d [8.3; 19.1]. Overall, moxifloxacin exhibited the highest global impact, followed by piperacillin/tazobactam, ceftazidime/avibactam and ceftriaxone. Their AUC between day 0 and day 42 of the change of diversity indices from day 0 were, respectively, -13.2 Shannon unit.day [-20.4; -7.9], -10.9 Shannon unit.day [-20.4; -0.6] and -10.1 Shannon unit.day [-18.3; -4.6]. We conclude that antibiotics alter the intestinal diversity to varying degrees, both within and between antibiotics families. Such studies are needed to help antibiotic stewardship in using the antibiotics with the lowest impact on the intestinal microbiota.
Collapse
Affiliation(s)
| | - Etienne Ruppé
- Université Paris Cité, IAME, INSERM, Paris, France
- APHP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, France
| | | | | | | | | | | | | |
Collapse
|
5
|
Aboulalazm FA, Kazen AB, deLeon O, Müller S, Saravia FL, Lozada-Fernandez V, Hadiono MA, Keyes RF, Smith BC, Kellogg SL, Grobe JL, Kindel TL, Kirby JR. Reutericyclin, a specialized metabolite of Limosilactobacillus reuteri, mitigates risperidone-induced weight gain in mice. Gut Microbes 2025; 17:2477819. [PMID: 40190120 PMCID: PMC11980487 DOI: 10.1080/19490976.2025.2477819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including Limosilactobacillus reuteri as a candidate for further study. Oral supplementation with L. reuteri protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and L. reuteri restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by L. reuteri as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.
Collapse
Affiliation(s)
- Fatima A. Aboulalazm
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexis B. Kazen
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Orlando deLeon
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susanne Müller
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fatima L. Saravia
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Matthew A. Hadiono
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert F. Keyes
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian C. Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephanie L. Kellogg
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tammy L. Kindel
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John R. Kirby
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
6
|
Jardon KM, Umanets A, Gijbels A, Trouwborst I, Hul GB, Siebelink E, Vliex LM, Bastings JJ, Argamasilla R, Chenal E, Venema K, Afman LA, Goossens GH, Blaak EE. Distinct gut microbiota and metabolome features of tissue-specific insulin resistance in overweight and obesity. Gut Microbes 2025; 17:2501185. [PMID: 40336254 PMCID: PMC12064058 DOI: 10.1080/19490976.2025.2501185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/24/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Insulin resistance (IR) is an early marker of cardiometabolic deterioration which may develop heterogeneously in key metabolic organs, including the liver (LIR) and skeletal muscle (MIR). This tissue-specific IR is characterized by distinct metabolic signatures, but the role of the gut microbiota in its etiology remains unclear. Here, we profiled the gut microbiota, its metabolites and the plasma metabolome in individuals with either a LIR or MIR phenotype (n = 233). We observed distinct microbial community structures LIR and MIR, and higher short-chain fatty acid (SCFA) producing bacteria, fecal SCFAs and branched-chain fatty acids and a higher postprandial plasma glucagon-like-peptide-1 response in LIR. In addition, we found variations in metabolome profiles and phenotype-specific associations between microbial taxa and functional metabolite groups. Overall, our study highlights association between gut microbiota and its metabolites composition with IR heterogeneity that can be targeted in precision-based strategies to improve cardiometabolic health. Clinicaltrials.gov registration: NCT03708419.
Collapse
Affiliation(s)
- Kelly M. Jardon
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Alexander Umanets
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Anouk Gijbels
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Inez Trouwborst
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gabby B. Hul
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lars M.M. Vliex
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jacco J.A.J. Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Lydia A. Afman
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
7
|
Huang W, Zhu W, Lin Y, Chan FKL, Xu Z, Ng SC. Roseburia hominis improves host metabolism in diet-induced obesity. Gut Microbes 2025; 17:2467193. [PMID: 39976263 PMCID: PMC11845086 DOI: 10.1080/19490976.2025.2467193] [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: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Next-generation live biotherapeutics are promising to aid the treatment of obesity and metabolic diseases. Here, we reported a novel anti-obesity probiotic candidate, Roseburia hominis, that was depleted in stool samples of obese subjects compared with lean controls, and its abundance was negatively correlated with body mass index and serum triglycerides. Supplementation of R. hominis prevented body weight gain and disorders of glucose and lipid metabolism, prevented fatty liver, inhibited white adipose tissue expansion and brown adipose tissue whitening in mice fed with high-fat diet, and boosted the abundance of lean-related species. The effects of R. hominis could be partially attributed to the production of nicotinamide riboside and upregulation of the Sirtuin1/mTOR signaling pathway. These results indicated that R. hominis is a promising candidate for the development of next-generation live biotherapeutics for the prevention of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Wenli Huang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Hornero-Ramirez H, Morisette A, Marcotte B, Penhoat A, Lecomte B, Panthu B, Lessard Lord J, Thirion F, Van-Den-Berghe L, Blond E, Simon C, Caussy C, Feugier N, Doré J, Sanoner P, Meynier A, Desjardins Y, Pilon G, Marette A, Cani PD, Laville M, Vinoy S, Michalski MC, Nazare JA. Multifunctional dietary approach reduces intestinal inflammation in relation with changes in gut microbiota composition in subjects at cardiometabolic risk: the SINFONI project. Gut Microbes 2025; 17:2438823. [PMID: 39710576 DOI: 10.1080/19490976.2024.2438823] [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: 07/03/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
The development of cardiometabolic (CM) diseases is associated with chronic low-grade inflammation, partly linked to alterations of the gut microbiota (GM) and reduced intestinal integrity. The SINFONI project investigates a multifunctional (MF) nutritional strategy's impact combining different bioactive compounds on inflammation, GM modulation and CM profile. In this randomized crossover-controlled study, 30 subjects at CM-risk consumed MF cereal-products, enriched with polyphenols, fibers, slowly-digestible starch, omega-3 fatty acids or Control cereal-products (without bioactive compounds) for 2 months. Metabolic endotoxemia (lipopolysaccharide (LPS), lipopolysaccharide-binding protein over soluble cluster of differentiation-14 (LBP/sCD14), systemic inflammation and cardiovascular risk markers, intestinal inflammation, CM profile and response to a one-week fructose supplementation, were assessed at fasting and post mixed-meal. GM composition and metabolomic analysis were conducted. Mixed linear models were employed, integrating time (pre/post), treatment (MF/control), and sequence/period. Compared to control, MF intervention reduced intestinal inflammation (fecal calprotectin, p = 0.007) and endotoxemia (fasting LPS, p < 0.05), without alteration of systemic inflammation. MF decreased serum branched-chain amino acids compared to control (p < 0.05) and increased B.ovatus, B.uniformis, A.butyriciproducens and unclassified Christensenellaceae.CAG-74 (p < 0.05). CM markers were unchanged. A 2-month dietary intervention combining multiple bioactive compounds improved intestinal inflammation and induced GM modulation. Such strategy appears as an effective strategy to target low-grade inflammation through multi-target approach.
Collapse
Affiliation(s)
- Hugo Hornero-Ramirez
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Arianne Morisette
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Bruno Marcotte
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Armelle Penhoat
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Béryle Lecomte
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Baptiste Panthu
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | | | | | - Laurie Van-Den-Berghe
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
| | - Emilie Blond
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
- Biochemistry Department, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Chantal Simon
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Cyrielle Caussy
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
- Endocrinology, Diabetes and Nutrition Department, Lyon South Hospital, Civil Hospices of Lyon, Pierre-Bénite, France
| | - Nathalie Feugier
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
| | - Joël Doré
- INRAE, MGP, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Sanoner
- iSymrise-Diana Food SAS, R&D, Naturals Food & Beverage, Rennes, France
| | - Alexandra Meynier
- Nutrition Research, Paris-Saclay Tech Center, Mondelez International R&D, Saclay, France
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute, Université Laval, Québec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute, Université Laval, Québec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, (LDRI) Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Louvain Drug Research Institute; Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Sophie Vinoy
- Nutrition Research, Paris-Saclay Tech Center, Mondelez International R&D, Saclay, France
| | - Marie-Caroline Michalski
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| |
Collapse
|
9
|
Saadh MJ, Allela OQB, Kareem RA, Sanghvi G, Ballal S, Naidu KS, Bareja L, Chahar M, Gupta S, Sameer HN, Yaseen A, Athab ZH, Adil M. Exploring preventive and treatment strategies for oral cancer: Modulation of signaling pathways and microbiota by probiotics. Gene 2025; 952:149380. [PMID: 40089085 DOI: 10.1016/j.gene.2025.149380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/11/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
The evidence suggests that the microbiome plays a crucial role in cancer development. The oral cavity has many microorganisms that can influence oral cancer progression. Understanding the mechanisms and signaling pathways of the oral, gum, and teeth microbiome in tumor progression can lead to new treatment strategies. Probiotics, which are friendly microorganisms, have shown potential as anti-cancer agents. These positive characteristics of probiotic strains make them suitable for cancer prevention or treatment. The oral-gut microbiome axis supports health and homeostasis, and imbalances in the oral microbiome can disrupt immune signaling pathways, epithelial barriers, cell cycles, apoptosis, genomic stability, angiogenesis, and metabolic processes. Changes in the oral microbiome in oral cancer may suggest using probiotics-based treatments for their direct or indirect positive roles in cancer development, progression, and metastasis, specifically oral squamous cell carcinoma (OSCC). Here, reported relationships between probiotics, oral microbiota, and oral cancer are summarized.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003 Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401 Punjab, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Sofia Gupta
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
10
|
Gao Y, Liu Y, Li Z, Lu L, Guo Y, Su D, Zhang H. Multi-strain probiotics attenuate carbohydrate-lipid metabolic dysregulation in type 2 diabetic rats via gut-liver axis modulation. mSystems 2025:e0036925. [PMID: 40492727 DOI: 10.1128/msystems.00369-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 04/19/2025] [Indexed: 06/12/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a pervasive chronic metabolic disorder characterized by dysregulation of carbohydrate, protein, and lipid metabolism. The objective of this study was to elucidate the impact of multi-strain probiotic supplementation on the metabolism of carbohydrates and lipids and the interplay between the gut and liver in a T2DM rat model. A total of 32 rats were randomly assigned to four experimental groups: a control group, a T2DM model group, a low-dose probiotics group, and a high-dose probiotics group. The impact of probiotic intervention on glycemic and lipid profiles was evaluated, with a specific emphasis on the high-dose cohort. The treatment with multi-strain probiotics, consisting of three Lacticaseibacillus species and one Bifidobacterium species, resulted in a significant improvement in blood glucose and lipid profiles in T2DM rats, with the highest dosage demonstrating the most pronounced effects. Probiotic administration modulated gut microbiota composition and diversity, enriching potentially beneficial bacterial species and altering gut metabolic modules and carbohydrate-active enzyme profiles. Multi-omics analyses indicated that alterations in fecal short-chain fatty acids and serum bile acids may serve as pivotal mediators for crosstalk between hepatic and gut transcriptomic pathways. This study offers novel insights into the role of probiotics in managing T2DM via the gut-liver axis, emphasizing the potential of probiotic therapy in modulating key metabolic pathways.IMPORTANCEType 2 diabetes mellitus (T2DM) is a chronic metabolic disease characterized by hyperglycemia, caused by defects in insulin secretion, insulin action, or both. For individuals diagnosed with T2DM, managing diabetes-related complications is often the most challenging aspect. Exogenous probiotics have the potential to serve as a promising therapeutic strategy to improve diabetes-related symptoms. We conducted a 64-day animal experiment to investigate the effects of probiotics on T2DM-related metabolic disorders and dyslipidemia by feeding four mixed probiotics to T2DM rats. The results showed that probiotics exerted beneficial effects on glucose- and lipid-related homeostasis indices in diabetic rats to some extent and modulated the gut microbiota to manage T2DM via the gut-liver axis.
Collapse
Affiliation(s)
- Yecheng Gao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Perfect (Guangdong) Co., Ltd., Zhongshan, Guangdong, China
| | - Yanfang Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zelong Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Liyi Lu
- Perfect (Guangdong) Co., Ltd., Zhongshan, Guangdong, China
| | - Yajuan Guo
- Perfect (Guangdong) Co., Ltd., Zhongshan, Guangdong, China
| | - Dun Su
- Perfect (Guangdong) Co., Ltd., Zhongshan, Guangdong, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| |
Collapse
|
11
|
Huang YX, Tang Q, Fu SH, Zhong HJ, Liu Z, Zhong QY, Wang YH, Luo YD, Li XY, Chen S, Zhu HP. Comparison of Efavirenz and Dolutegravir on Gut Microbiome and Gut Barrier Functions. ACS OMEGA 2025; 10:23099-23110. [PMID: 40521525 PMCID: PMC12163826 DOI: 10.1021/acsomega.5c01210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 06/18/2025]
Abstract
Dolutegravir (DTG) is increasingly replacing efavirenz (EFV) as the recommended first-line antiretroviral therapy (cART) component due to its superior tolerance. However, both EFV-based and DTG-based cART regimens are associated with metabolic and neuropsychiatric disorders through mechanisms that remain poorly understood. In this study, the abundance of Lactobacillus was significantly reduced, along with elevated soluble CD14 levels, in HIV-1-infected individuals receiving EFV-based cART compared to those on DTG-based cART. Additionally, EFV treatment in mice resulted in the destruction of intestinal crypts, loss of goblet cells, decreased expression of intestinal ZO-1, and more severe damage to gut barrier integrity compared to the DTG and normal control groups. Furthermore, we identified a significant reduction in Lactobacillus in EFV-treated mice. Our results suggest that EFV and DTG may directly lead to gut dysbiosis and compromised gut barrier integrity, which could be targeted to prevent the side effects associated with EFV and DTG.
Collapse
Affiliation(s)
- Yong-Xiang Huang
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, No. 12 Jichang Road, Guangzhou510405, China
| | - Qin Tang
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, No. 12 Jichang Road, Guangzhou510405, China
- Jiangxi
Provincial People’s Hospital & The First Affiliated Hospital
of Nanchang Medical College, No. 152 Ai-guo Road, Nanchang330006, China
| | - Shi-Hua Fu
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| | - Hui-Jun Zhong
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| | - Zhe Liu
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, No. 12 Jichang Road, Guangzhou510405, China
| | - Qing-Yang Zhong
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| | - Yan-Hao Wang
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| | - Yin-di Luo
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| | - Xue-Ying Li
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, No. 12 Jichang Road, Guangzhou510405, China
| | - Song Chen
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, No. 12 Jichang Road, Guangzhou510405, China
| | - Hai-Peng Zhu
- Dongguan
Ninth People’s hospital, No. 88 Sha-di-tang Road, Dongguan523001, China
| |
Collapse
|
12
|
Hijová E, Bertková I, Štofilová J. Incorporating Postbiotics into Intervention for Managing Obesity. Int J Mol Sci 2025; 26:5362. [PMID: 40508171 PMCID: PMC12154039 DOI: 10.3390/ijms26115362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/20/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
Obesity is reaching global epidemic proportions worldwide, posing a significant burden on individual health and society. Altered gut microbiota is considered a key factor in the pathogenesis of many diseases, producing metabolites that contribute to the health-beneficial properties of postbiotics. Postbiotics, bioactive microbial components derived from probiotics, are emerging as a valuable strategy in modern medicine and a promising alternative for managing obesity without the need for live bacteria. This work provides a comprehensive overview of the potential health benefits of postbiotics, particularly in relation to obesity, which represents an important health challenge. Despite the encouraging insights into the health benefits of postbiotics, we highlight the need for further research to clarify the mechanisms and the specific roles of different postbiotic components. Integrating postbiotics into health interventions has the potential to enhance preventive care and significantly improve health outcomes in at-risk populations.
Collapse
Affiliation(s)
- Emília Hijová
- Center of Clinical and Preclinical Research-MediPark, Faculty of Medicine, Pavol Jozef Šafárik University, SNP 1, 040 11 Košice, Slovakia; (I.B.); (J.Š.)
| | | | | |
Collapse
|
13
|
Caille A, Connan C, Lyon Belgy N, Borezée E, Cherbuy C, Meunier N, Meslier V. Positive nutritional selection of adults with healthy lifestyle and high daily fiber consumption for the isolation of beneficial intestinal bacteria: The iTARGET cohort study protocol. MethodsX 2025; 14:103268. [PMID: 40224142 PMCID: PMC11987684 DOI: 10.1016/j.mex.2025.103268] [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: 12/06/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Recent advances in the study of the gut microbiota has pointed to its under-utilized source of potentially beneficial bacteria, known as next generation probiotics, offering a promising avenue to restore or compensate impaired gut microbiota toward a healthy state. Aside from the difficulties to achieve in-lab adequate culture conditions, the use of beneficial bacterial isolates is also limited by their bioavailability in the donor itself. In the iTARGET study, we positively selected donors based on their diet enriched in fiber, that has been shown to increase the prevalence of bacterial species associated with health. The iTARGET study is a monocenter, prospective, observational study of adults with healthy lifestyle and high daily fiber consumption. We aim to recruit individuals in two phases, the first one for all individuals that will permit the identification of carriers for bacteria of interest and the second phase for a subset of individuals to allow for culture and isolation of previously identified potentially beneficial bacteria. Our primary outcome is the isolation and culture of at least one potentially beneficial isolate. The secondary outcomes comprised the high throughput metagenomic profiles of the intestinal microbiota and the characterization of the cultured isolates. The study was approved by the French Research Ethics Committees (Comité de Protection des Personnes Sud-Est I) under the National reference ID 2023-A01677-38. Study findings and results will be published in peer-reviewed Open Access journals. (Trial registration number on ClinicalTrials.gov: NCT06166810).
Collapse
Affiliation(s)
- Aurélie Caille
- Centre Hospitalier Universitaire de Clermont-Ferrand, CRNH Auvergne, Clermont-Ferrand, France
| | - Chloé Connan
- Université Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Noelle Lyon Belgy
- Centre Hospitalier Universitaire de Clermont-Ferrand, CRNH Auvergne, Clermont-Ferrand, France
| | - Elise Borezée
- Université Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Claire Cherbuy
- Université Paris-Saclay, INRAE, Micalis, 78350 Jouy-en-Josas, France
| | - Nathalie Meunier
- Centre Hospitalier Universitaire de Clermont-Ferrand, CRNH Auvergne, Clermont-Ferrand, France
| | - Victoria Meslier
- Université Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| |
Collapse
|
14
|
Yoshimura Y, Wakabayashi H, Nagano F, Matsumoto A, Shimazu S, Shiraishi A, Kido Y, Bise T, Hamada T, Yoneda K, Maeda K. Gut microbiome diversity is associated with muscle mass, strength and quality in post-stroke patients. Clin Nutr ESPEN 2025; 67:25-33. [PMID: 40049396 DOI: 10.1016/j.clnesp.2025.02.027] [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: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The gut microbiome has emerged as a potential influencer of muscle health; however, its role in hospitalized patients remains unclear. This study aimed to investigate the association between gut microbiome diversity and skeletal muscle mass, strength, and quality in hospitalized post-stroke patients. METHODS We conducted a cross-sectional study of post-stroke patients admitted to a rehabilitation facility. Gut microbiome diversity was assessed using 16S ribosomal ribonucleic acid (rRNA) gene sequencing, calculating Operational Taxonomic Unit (OTU) Richness, Faith's Phylogenetic Diversity (PD), and Shannon index. Muscle health was evaluated using skeletal muscle index (SMI) for muscle mass, handgrip strength (HGS) for muscle strength, and bioimpedance analysis-derived phase angle (PhA) for muscle quality. Multiple linear regression analyses were performed, adjusting for potential confounders. RESULTS A total of 156 patients (mean age 78.4 years; 55.7 % male) were analyzed. OTU Richness showed significant positive associations with SMI (β = 0.197, p = 0.025), HGS (β = 0.180, p = 0.005), and PhA (β = 0.178, p = 0.022). The Shannon index was also positively associated with SMI (β = 0.120, p = 0.041), HGS (β = 0.140, p = 0.028), and PhA (β = 0.164, p = 0.032). Faith's PD did not demonstrate significant associations with muscle health parameters. CONCLUSIONS Higher gut microbiome diversity, assessed by OTU Richness and Shannon index, is associated with better muscle mass, strength, and quality in post-stroke patients. These findings suggest a potential role for gut microbiota in muscle health during stroke rehabilitation.
Collapse
Affiliation(s)
- Yoshihiro Yoshimura
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Hidetaka Wakabayashi
- Department of Rehabilitation Medicine, Tokyo Women's Medical University Hospital, Japan.
| | - Fumihiko Nagano
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Ayaka Matsumoto
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Sayuri Shimazu
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Ai Shiraishi
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Yoshifumi Kido
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Takahiro Bise
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Takenori Hamada
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Kouki Yoneda
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Keisuke Maeda
- Nutrition Therapy Support Center, Aichi Medical University Hospital, Japan; Department of Geriatric Medicine, Hospital, National Center for Geriatrics and Gerontology, Japan.
| |
Collapse
|
15
|
Xia J, Wang Y, Li X, Liu L, Zhang P, Dai W, Luo P, Wang G, Li Y. The mechanism of perilla oil in regulating lipid metabolism. Food Chem 2025; 476:143318. [PMID: 39977980 DOI: 10.1016/j.foodchem.2025.143318] [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: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
Emerging science supports the role of lipid metabolism disorders in the occurrence and development of chronic diseases. Dietary intervention has been shown to be an effective strategy for regulating lipid metabolism. Recent studies showed that perilla is rich in various effective ingredients, including fatty acids, flavonoids, and phenolic acids. These ingredients exhibit a myriad of benefits, notably enhancing intestinal health and helping to manage metabolic diseases. Perilla oil stands out as a promising agent for regulating lipid metabolism, underscoring its potential for various health applications. This review introduces the active ingredients in perilla and provides a systematic overview of the mechanism by which perilla oil regulates lipid metabolism to expand its application value. Further research should focus on exploring the dose effect and absorption efficiency of perilla oil in clinical applications.
Collapse
Affiliation(s)
- Jiawei Xia
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Yi Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Xin Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Li Liu
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Pin Zhang
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Wendong Dai
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Peng Luo
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Guoze Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| | - Yanhong Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
16
|
Byrd DA, Gomez M, Hogue S, Wan Y, Ortega-Villa A, Warner A, Dagnall C, Jones K, Hicks B, Albert P, Murphy G, Sinha R, Vogtmann E. Effects of a high-fiber, high-fruit and high-vegetable, low-fat dietary intervention on the rectal tissue microbiome. J Natl Cancer Inst 2025; 117:1237-1244. [PMID: 39980340 PMCID: PMC12145913 DOI: 10.1093/jnci/djaf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/11/2025] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that bacteria residing in colorectal tissue are plausibly associated with colorectal cancer. Prior studies investigated the effects of dietary interventions on the fecal microbiome, but few assessed colorectal tissue microbiome endpoints. We investigated the effects of a high-fiber, high-fruit, high-vegetable, and low-fat dietary intervention on the rectal tissue microbiome in the Polyp Prevention Trial (PPT). METHODS PPT is a 4-year randomized clinical trial with intervention goals of consuming (1) at least 18 g of fiber per 1000 kcal/day; (2) at least 3.5 servings of fruits and vegetables per 1000 kcal/day; and (3) no more than 20% of kcal/day from fat. Using 16S ribosomal RNA gene sequencing, we characterized bacteria in rectal biopsies collected at baseline and the end of years 1 and 4 (n = 233 in intervention arm and n = 222 in control arm). We estimated effects of the intervention on alpha and beta diversity and relative abundance of a priori-selected bacteria using repeated-measures linear mixed-effects models. RESULTS The intervention did not statistically significantly modify rectal tissue alpha diversity. Compared with the control arm, relative abundance of a priori-selected Porphyromonas (absolute intervention effects [standard errors] at T1 vs T0 = -0.24 [0.07] and T4 vs T0 = -0.12 [0.07]; P = .004) and Prevotella (absolute intervention effects at T1 vs T0 = -0.40 [0.14] and at T4 vs T0 = -0.32 [0.15]; P = .01) were more strongly decreased in the intervention arm. CONCLUSION The PPT intervention did not influence rectal tissue microbiome diversity or the relative abundance of most bacteria, except for 2 oral-originating bacteria that were previously associated with colorectal cancer presence.
Collapse
Affiliation(s)
- Doratha A Byrd
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Maria Gomez
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Stephanie Hogue
- Non-therapeutic Research Office, Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Yunhu Wan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
| | - Ana Ortega-Villa
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, United States
| | - Andrew Warner
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD 21701, United States
| | - Casey Dagnall
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD 21701, United States
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD 21701, United States
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD 21701, United States
| | - Paul Albert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
| | - Gwen Murphy
- Cancer Screening and Prevention Research Group, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, United Kingdom
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
| | - Emily Vogtmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, United States
| |
Collapse
|
17
|
Deng M, Tang F, Zhu Z. Altered cognitive function in obese patients: relationship to gut flora. Mol Cell Biochem 2025; 480:3553-3567. [PMID: 39937394 PMCID: PMC12095350 DOI: 10.1007/s11010-024-05201-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/24/2024] [Indexed: 02/13/2025]
Abstract
Obesity is a risk factor for non-communicable diseases such as cardiovascular disease and diabetes, which are leading causes of death and disability. Today, China has the largest number of overweight and obese people, imposing a heavy burden on China's healthcare system. Obesity adversely affects the central nervous system (CNS), especially cognitive functions such as executive power, working memory, learning, and so on. The gradual increase in adult obesity rates has been accompanied by a increase in childhood obesity rates. In the past two decades, the obesity rate among children under 5 years of age has increased from 32 to 42 million. If childhood obesity is not intervened in the early years, it will continue into adulthood and remain there for life. Among the potential causative factors, early lifestyle may influence the composition of the gut flora in childhood obesity, such as the rate and intake of high-energy foods, low levels of physical activity, may persist into adulthood, thus, early lifestyle interventions may improve the composition of the gut flora in obese children. Adipose Axis plays an important role in the development of obesity. Adipose tissue is characterized by increased expression of nucleoside diphosphate-linked molecule X-type motif 2 (NUDT2), amphiphilic protein AMPH genes, which encode proteins that all play important roles in the CNS. NUDT2 is associated with intellectual disability. Furthermore, amphiphysin (AMPH) is involved in glutamatergic signaling, ganglionic synapse development, and maturation, which is associated with mild cognitive impairment (MCI) and Alzheimer's disease (AD). All of the above studies show that obesity is closely related to cognitive decline in patients. Animal experiments have confirmed that obesity causes changes in cognitive function. For example, high-fat diets rich in long- and medium-chain saturated fatty acids may adversely affect cognitive function in obese mice. This process may be attributed to the Short-Chain Fatty Acid (SCFA)-rich high-fat diet (HFD) activating enterocyte TLR signaling, especially TLR-2 and TLR-4, altering the downstream MyD88-4 signaling, thereby impacting the downstream MyD88-NF-κB signaling cascade and up-regulating the levels of pro-inflammatory factors and lipopolysaccharide (LPS). These changes result in the loss of integrity of the intestinal mucosa and cause an imbalance in the internal environment. Obesity may lead to the disruption of the intestinal flora and damage the intestinal barrier function, causing intestinal flora dysbiosis. In recent years, a growing number of studies have investigated the relationship between obesity and the intestinal flora. For example, high-fat and high-sugar diets have been found to lead to the thinning of the mucus layer of the colon, a decrease in the number of tight junction proteins, and an increase in intestinal permeability in mice. Such changes alter the composition of intestinal microorganisms, allow endotoxins into the blood circulation, and induce neuroinflammation and brain damage. Therefore, obesity affects cognitive function and is even hereditary. This paper reviews the obesity-induced cognitive dysfunction, the underlying mechanisms, the research progress of intestinal flora dysregulation in obese patients, the relationship between intestinal flora and cognitive function changes, and the research progress on intestinal flora dysregulation in obese patients. We want to regulate the internal environment of obese patients from the perspective of intestinal flora, improving the cognitive function of obese patients, and prevent obesity-induced changes in related neurological functions.
Collapse
Affiliation(s)
- Mengyuan Deng
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China
| | - Fushan Tang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, 563006, China
| | | |
Collapse
|
18
|
Koller AM, Săsăran MO, Mărginean CO. The Role of Gut Microbiota in Pediatric Obesity and Metabolic Disorders: Insights from a Comprehensive Review. Nutrients 2025; 17:1883. [PMID: 40507152 PMCID: PMC12158192 DOI: 10.3390/nu17111883] [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: 05/07/2025] [Revised: 05/28/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Pediatric obesity represents a multifactorial condition in which gut microbiota dysbiosis, low-grade systemic inflammation, and metabolic dysfunction are intricately connected. Objectives: This systematic review sought to evaluate and integrate current findings regarding the interactions between gut microbial composition, dietary influences, inflammatory status, and metabolic outcomes in obese pediatric populations. Methods: A comprehensive search of PubMed, Scopus, and Web of Science databases was conducted for studies published from January 2010 onward. Eligible studies comprised randomized controlled trials, and cohort, cross-sectional, and longitudinal designs involving individuals aged ≤18 years. Study quality was appraised using the NIH Study Quality Assessment Tool. Results: Sixteen studies fulfilled the inclusion criteria. Dysbiosis was consistently observed among obese children, characterized by alterations in microbial diversity and abundance associated with increased inflammation and adverse metabolic profiles. Dietary interventions, notably symbiotic supplementation and adherence to Mediterranean diet patterns, were associated with favorable modulation of gut microbiota and inflammatory parameters. The majority of studies demonstrated high methodological quality, although minor observational limitations were noted. Conclusions: Gut microbiota dysregulation plays a central role in the development of metabolic and inflammatory complications associated with pediatric obesity. Although dietary and microbiota-modifying strategies show therapeutic promise, their effectiveness must be substantiated through robust, long-term studies.
Collapse
Affiliation(s)
- Ana Maria Koller
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania;
| | - Maria Oana Săsăran
- Department of Pediatrics 3, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania;
| |
Collapse
|
19
|
Jin L, Zhang P, Sun K, Wang H. Gut microbiota dynamics and their impact on body condition in nestlings of the yellow-rumped flycatchers, Ficedula zanthopygia. Front Microbiol 2025; 16:1595357. [PMID: 40520375 PMCID: PMC12165319 DOI: 10.3389/fmicb.2025.1595357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 05/06/2025] [Indexed: 06/18/2025] Open
Abstract
Investigating the gut microbiome during host development is essential for understanding its influence on host health and fitness. While host body condition is a crucial fitness-related trait and a strong predictor of viability in numerous animal species, its relationship with gut bacteria remains underexplored, particularly in non-model organisms. This study examines the gut microbiome of the altricial wild bird species, yellow-rumped flycatchers (Ficedula zanthopygia), by analyzing nestling feces through 16S rRNA sequencing at four developmental stages: Day 3, Day 6, Day 9 and Day 12 post-hatching. We explored the temporal dynamics of the gut microbiome and its correlation with body condition, a key indicator of fitness. Our results demonstrate signinficant shifts in microbial community composition and diversity throughout development. Notably, Day 3 nestlings displayed lower alpha diversity compared to later stages, while microbial diversity stabilized from Days 6 to 12. Both the age of the nestlings and the environmental conditions of the nest box significantly shaped the gut microbial community structure. A contemporaneous relationship was observed, where the scaled-mass index (SMI) at Day 6 positively correlating with microbial diversity at that time. Additionally, a time-lagged effect emerged, linking SMI at Day 9 to microbial diversity at Day 6. These findings highlight the vital role of the gut microbiome in the development of nestlings, particularly emphasizing Day 6 as a critical period due to its stable microbial diversity and association with SMI. This study underscores the influence of gut bacteria on host fitness in developing birds.
Collapse
Affiliation(s)
- Longru Jin
- Jilin Engineering Laboratory for Avian Ecology and Conservation Genetics, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Northeast Normal University, Changchun, China
- Key Laboratory of Vegetation Ecology, Ministry of Education, Changchun, China
| | - Pai Zhang
- Jilin Engineering Laboratory for Avian Ecology and Conservation Genetics, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Northeast Normal University, Changchun, China
- Key Laboratory of Vegetation Ecology, Ministry of Education, Changchun, China
| | - Keping Sun
- Jilin Engineering Laboratory for Avian Ecology and Conservation Genetics, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Northeast Normal University, Changchun, China
- Key Laboratory of Vegetation Ecology, Ministry of Education, Changchun, China
| | - Haitao Wang
- Jilin Engineering Laboratory for Avian Ecology and Conservation Genetics, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Northeast Normal University, Changchun, China
- Key Laboratory of Vegetation Ecology, Ministry of Education, Changchun, China
| |
Collapse
|
20
|
Miura N, Tabata T, Ishihama Y, Okuda S. Phylogenetic tree-based amino acid sequence generation for proteomics data analysis of unknown species. Comput Struct Biotechnol J 2025; 27:2313-2322. [PMID: 40520598 PMCID: PMC12167054 DOI: 10.1016/j.csbj.2025.05.041] [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: 01/21/2025] [Revised: 05/24/2025] [Accepted: 05/24/2025] [Indexed: 06/18/2025] Open
Abstract
In bottom-up proteomics, selecting an appropriate protein amino acid sequence database is vital for reliable peptide identification. However, this approach excludes species with unsequenced genomes, limiting the comprehensiveness. This is a major challenge in current microbiota proteomics, a rapidly developing field, which involves simultaneously assigning proteins to species in a sample and analyzing them using databases of protein amino acid sequences with known genomes. We aimed to develop a method to extend the database species diversity by generating protein amino acid sequences of unknown species using phylogenetic relationships among known species. To evaluate this approach, we generated the Helicobacter pylori F16 strain sequence based on the phylogenetic relationships of 29 closely related strains (excluding F16). Consequently, the percentages of peptides that matched the peptides obtained from the reference F16 strain increased by 5 %, based on sequence generation. Proteomics data analyses were performed on the F16 strain using the generated sequence database to validate peptide identification. Peptide spectral match decreased when the database was expanded using sequence generation owing to a decrease in sensitivity primarily caused by an increase in decoy hits. The decrease in identification sensitivity caused by large-scale databases could be improved by introducing a novel score, Ion Cover Score, based on spectral matching. The sequence generation method used in the present study and the introduction of scores based on spectral matching could accelerate proteomics development.
Collapse
Affiliation(s)
- Nobuaki Miura
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| | - Tsuyoshi Tabata
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
- Medical AI Center, Niigata University School of Medicine, 2-5274 Gakkocho-dori, Chuo-ku, Niigata 951-8514, Japan
| |
Collapse
|
21
|
Anvarbatcha R, Kunnathodi F, Arafat AA, Azmi S, Mustafa M, Ahmad I, Alotaibi HF. Harnessing Probiotics: Exploring the Role of the Gut Microbiome in Combating Obesity. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10605-3. [PMID: 40434504 DOI: 10.1007/s12602-025-10605-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Obesity has become a global health crisis driven by genetic, environmental, and lifestyle factors, often linked to gut microbiome imbalances. Probiotics, particularly Lactobacillus and Bifidobacterium strains, have shown promise in clinical trials by promoting weight loss, improving lipid profiles, and addressing gut dysbiosis associated with obesity. This review surveys the literature on the microbiome and obesity, emphasizing the clinical relevance of probiotics in treatment strategies. Our comprehensive PubMed search highlights the mechanisms through which probiotics influence metabolic health, including their effects on inflammation and appetite regulation. We also explore promising future research directions and the potential for integrating probiotics into clinical practice. While results are encouraging, the evidence is limited by strain variability, small sample sizes, short trial durations, and individual differences in microbiota composition. More extensive, long-term studies with standardized methods are crucial to confirm the effectiveness of probiotics as viable anti-obesity treatments.
Collapse
Affiliation(s)
- Riyasdeen Anvarbatcha
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Faisal Kunnathodi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Amr A Arafat
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
- Departments of Adult Cardiac Surgery Department, Kingdom of Saudi Arabia, Prince Sultan Cardiac Center, Riyadh, Kingdom of Saudi Arabia
| | - Sarfuddin Azmi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Mustafa
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Ishtiaque Ahmad
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia
| | - Haifa F Alotaibi
- Health Research Center, Kingdom of Saudi Arabia, Ministry of Defense Health Services, Riyadh, Kingdom of Saudi Arabia.
- Department of Family Medicine, Kingdom of Saudi Arabia, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
22
|
Yin C, Fan P, Mou X, Zhao W. Clostridium butyricum Ameliorates Atherosclerosis by Regulating Host Linoleic Acid Metabolism. Microorganisms 2025; 13:1220. [PMID: 40572107 PMCID: PMC12194821 DOI: 10.3390/microorganisms13061220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 06/29/2025] Open
Abstract
Dysbiosis of the gut microbiota is strongly implicated in atherosclerosis (AS), thus prompting microbial modulation to be explored as a therapeutic strategy. However, limited evidence exists for probiotic interventions capable of alleviating AS. Here, we focused on Clostridium butyricum (C. butyricum; CB), a probiotic known for its production of short-chain fatty acids (SCFAs). We found that administration of C. butyricum to high-fat diet (HFD)-fed Apoe deficient (Apoe-/-) mice reduced plaque area by improving blood lipid profiles, decreasing macrophage infiltration in the aortic roots, and lowering the levels of circulating pro-inflammatory monocytes and macrophages. By non-targeted serum metabolomics analysis, C. butyricum treatment significantly reduced the levels of both linoleic acid and its downstream metabolites. Collectively, these findings establish C. butyricum-mediated amelioration of AS through modulation of linoleic acid metabolism.
Collapse
Affiliation(s)
| | | | - Xiangyu Mou
- Shenzhen Key Laboratory for Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (C.Y.); (P.F.)
| | - Wenjing Zhao
- Shenzhen Key Laboratory for Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (C.Y.); (P.F.)
| |
Collapse
|
23
|
Trapanese V, Dagostino A, Natale MR, Giofrè F, Vatalaro C, Melina M, Cosentino F, Sergi S, Imoletti F, Spagnuolo R, Arturi F. Bidirectional Interactions Between the Gut Microbiota and Incretin-Based Therapies. Life (Basel) 2025; 15:843. [PMID: 40566497 PMCID: PMC12193786 DOI: 10.3390/life15060843] [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: 04/10/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 06/28/2025] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and metabolic syndrome have been largely correlated to a reduction in bacterial load and diversity, resulting in a condition known as intestinal dysbiosis. The recent emergence of novel antidiabetic medications has been demonstrated to exert a favourable influence on the composition of the intestinal microbiota. Incretin-based therapy exerts a multifaceted influence on the composition of the gut microbiota, leading to alterations in bacterial flora. Of particular significance is the capacity of numerous metabolites produced by the gut microbiota to modulate the activity and hormonal secretion of enteroendocrine cells. This review examines the effects of dipeptidyl peptidase 4 (DPP-4) inhibitors, glucagon-like peptide 1 (GLP-1) receptor agonists and GLP-1/gastric inhibitory polypeptide (GIP) receptor dual agonists on the composition of the gut microbiota in both mice and human subjects. The nature of this interaction is complex and bidirectional. The present study demonstrates the involvement of the incretinic axis in modulating the microbial composition, with the objective of providing novel preventative strategies and potential personalised therapeutic targets for obesity and T2DM.
Collapse
Affiliation(s)
- Vincenzo Trapanese
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Annamaria Dagostino
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Maria Resilde Natale
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Federica Giofrè
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Clara Vatalaro
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Melania Melina
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Francesca Cosentino
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Silvia Sergi
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
| | - Felice Imoletti
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (F.I.); (R.S.)
| | - Rocco Spagnuolo
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (F.I.); (R.S.)
| | - Franco Arturi
- Internal Medicine Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (A.D.); (M.R.N.); (F.G.); (C.V.); (M.M.); (F.C.); (S.S.); (F.A.)
- Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
24
|
Sun J, Brooks EC, Houshyar Y, Connor SJ, Paven G, Grimm MC, Hold GL. Unravelling the Relationship Between Obesity and Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf098. [PMID: 40397482 DOI: 10.1093/ibd/izaf098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Indexed: 05/22/2025]
Abstract
Mirroring the global obesity epidemic, obesity rates in inflammatory bowel disease (IBD) patients is rising. Several epidemiological studies propose that 15%-40% of adult patients with IBD are obese, and an additional 25%-40% fall into the overweight category. This article examines the pathophysiologic relationship between obesity and IBD concerning the role of visceral adipose tissue, microbiota shifts, dietary patterns, and hunger hormone changes. Additionally, increasing evidence is demonstrating the negative impact that obesity is having on disease course and quality of life in patients with IBD. Obesity has been demonstrated to be associated with an attenuated response to immunomodulators and biological agents, as well as higher rates of peri-operative surgical complications. A better understanding of the relationship between obesity and IBD can be applied to clinical decision-making in personalizing treatment plans, promoting weight loss in patients with obesity, and identifying areas of future research.
Collapse
Affiliation(s)
- Jessica Sun
- Microbiome Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Prince of Wales Hospital, South Eastern Sydney Local Health District, Sydney, NSW, Australia
| | - Ella C Brooks
- Microbiome Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Yashar Houshyar
- Microbiome Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- St. George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| | - Susan J Connor
- Department of Gastroenterology and Hepatology, Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Gokulan Paven
- St. George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| | - Michael C Grimm
- Microbiome Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- St. George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| | - Georgina L Hold
- Microbiome Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- St. George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
25
|
Hillman EBM, Baumgartner M, Carson D, Amos GCA, Wazir I, Khan HA, Khan MA, Rijpkema S, Walters JRF, Wellington EMH, Arasaradnam R, Lewis SJ. Changing Gastrointestinal Transit Time Alters Microbiome Composition and Bile Acid Metabolism: A Cross-Over Study in Healthy Volunteers. Neurogastroenterol Motil 2025:e70075. [PMID: 40394972 DOI: 10.1111/nmo.70075] [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: 08/22/2024] [Revised: 03/21/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND The specific influence of whole gut transit time (WGTT) on microbiome dynamics and bile acid metabolism remains unclear, despite links between changes in WGTT and certain gastrointestinal disorders. Our investigation aimed to determine the impact of WGTT changes on the composition of the fecal microbiome and bile acid profile. METHODS Healthy volunteers (n = 18) received loperamide, to decrease bowel movement frequency, and senna, a laxative, each over a 6-day period, in a randomized sequence, with a minimum 16-day interval between each treatment. Stool samples were analyzed for microbiome by shotgun sequencing and bile acid composition determined with high-performance liquid chromatography coupled to tandem mass spectrometry. Sera were examined for markers of bile acid synthesis. KEY RESULTS Senna or loperamide decreased or increased WGTT, respectively. Treatment altered stool characteristics, bowel movement frequency, and stool weight. The senna-treated group had increased primary and secondary fecal bile acids; serum levels of fibroblast growth factor 19 were significantly reduced. Increasing WGTT with loperamide led to an increase in bile salt hydrolase genes, along with elevated bacterial species richness (p = 0.04). Thirty-six species exhibiting significant differences were identified, several of which have notable implications for gut health. WGTT displayed negative correlations with total primary (particularly chenodeoxycholic acid) and secondary bile acids (ursodeoxycholic acid and glycochenodeoxycholic acid). Treatment-induced changes in microbiome composition and bile acid metabolism reverted back to baseline within 16 days. CONCLUSION Whole gut transit time changes significantly affect fecal microbiome composition and function, as well as bile acid composition and synthesis in healthy subjects. This consideration is likely to have long-term implications.
Collapse
Affiliation(s)
- Evette B M Hillman
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - Maximilian Baumgartner
- Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Danielle Carson
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Gregory C A Amos
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Imad Wazir
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Haider A Khan
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Malik A Khan
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
| | - Sjoerd Rijpkema
- Diagnostics, Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Julian R F Walters
- Division of Digestive Diseases, Imperial College London, London, UK
- Imperial College Healthcare Trust, London, UK
| | | | - Ramesh Arasaradnam
- Department of Gastroenterology, University Hospitals Coventry & Warwickshire, Coventry, UK
- Warwick Medical School, The University of Warwick, Coventry, UK
| | - Stephen J Lewis
- Department of Gastroenterology, University Hospital Plymouth, Plymouth, UK
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| |
Collapse
|
26
|
Yang X, Zhang Y, Xu Y, Xu Y, Zhang M, Guan Q, Hu W, Tun HM, Xia Y. Microbial Disturbances Caused by Pesticide Exposure and Their Predictive Implications for Gestational Diabetes Mellitus. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:9449-9460. [PMID: 40327666 DOI: 10.1021/acs.est.5c01076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Previous studies have suggested that pesticide exposure and gut microbiome alterations are associated with gestational diabetes mellitus (GDM) risk. Understanding the complex interactive effect of these factors on GDM is essential. In a cohort of 852 pregnant women, we assessed pesticide levels in serum and analyzed the gut microbiota using 16S rRNA and shotgun metagenomic sequencing. We explored the interactions between pesticides and gut microbiota, assessed their roles in GDM development, and proposed a predictive model based on identified biomarkers. We identified an environmental risk score (ERS), denoting the pesticide mixture level significantly associated with GDM, with the gut microbiota, particularly involving the Dorea branch, playing a crucial mediating role. In addition, we found an interactive effect of pesticide exposure and gut microbiota on GDM risk. Notably, low Prevotella enrichment combined with high ERS arisen from pesticide levels led to a 10.36-fold increased GDM risk. The identified pesticide and gut microbial biomarkers achieved high predictive accuracy for GDM (AUC: 0.833, 95% CI: 0.748-0.918). Collectively, maternal pesticide exposure may induce disrupted microbiome-dependent glycemic alteration, necessitating future assessment of clinical implications. Potential GDM markers can serve as targets for therapeutic intervention caused by pesticides, leading to prevention.
Collapse
Affiliation(s)
- Xu Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Yuqing Zhang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yifan Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yadan Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mingzhi Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Quanquan Guan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Weiyue Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Hein Min Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health and Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
27
|
Veseli I, Chen YT, Schechter MS, Vanni C, Fogarty EC, Watson AR, Jabri B, Blekhman R, Willis AD, Yu MK, Fernàndez-Guerra A, Füssel J, Eren AM. Microbes with higher metabolic independence are enriched in human gut microbiomes under stress. eLife 2025; 12:RP89862. [PMID: 40377187 PMCID: PMC12084026 DOI: 10.7554/elife.89862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
A wide variety of human diseases are associated with loss of microbial diversity in the human gut, inspiring a great interest in the diagnostic or therapeutic potential of the microbiota. However, the ecological forces that drive diversity reduction in disease states remain unclear, rendering it difficult to ascertain the role of the microbiota in disease emergence or severity. One hypothesis to explain this phenomenon is that microbial diversity is diminished as disease states select for microbial populations that are more fit to survive environmental stress caused by inflammation or other host factors. Here, we tested this hypothesis on a large scale, by developing a software framework to quantify the enrichment of microbial metabolisms in complex metagenomes as a function of microbial diversity. We applied this framework to over 400 gut metagenomes from individuals who are healthy or diagnosed with inflammatory bowel disease (IBD). We found that high metabolic independence (HMI) is a distinguishing characteristic of microbial communities associated with individuals diagnosed with IBD. A classifier we trained using the normalized copy numbers of 33 HMI-associated metabolic modules not only distinguished states of health vs IBD, but also tracked the recovery of the gut microbiome following antibiotic treatment, suggesting that HMI is a hallmark of microbial communities in stressed gut environments.
Collapse
Affiliation(s)
- Iva Veseli
- Biophysical Sciences Program, The University of ChicagoChicagoUnited States
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Yiqun T Chen
- Data Science Institute and Department of Biomedical Data Science, Stanford UniversityStanfordUnited States
| | - Matthew S Schechter
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Chiara Vanni
- MARUM Center for Marine Environmental Sciences, University of BremenBremenGermany
| | - Emily C Fogarty
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Andrea R Watson
- Department of Medicine, The University of ChicagoChicagoUnited States
- Committee on Microbiology, The University of ChicagoChicagoUnited States
| | - Bana Jabri
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Ran Blekhman
- Department of Medicine, The University of ChicagoChicagoUnited States
| | - Amy D Willis
- Department of Biostatistics, University of WashingtonSeattleUnited States
| | - Michael K Yu
- Toyota Technological Institute at ChicagoChicagoUnited States
| | - Antonio Fernàndez-Guerra
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of CopenhagenCopenhagenDenmark
| | - Jessika Füssel
- Department of Medicine, The University of ChicagoChicagoUnited States
- Institute for Chemistry and Biology of the Marine Environment, University of OldenburgOldenburgGermany
| | - A Murat Eren
- Department of Medicine, The University of ChicagoChicagoUnited States
- Institute for Chemistry and Biology of the Marine Environment, University of OldenburgOldenburgGermany
- Marine ‘Omics Bridging Group, Max Planck Institute for Marine MicrobiologyBremenGermany
- Helmholtz Institute for Functional Marine BiodiversityOldenburgGermany
- Alfred Wegener Institute for Polar and Marine ResearchBremerhavenGermany
| |
Collapse
|
28
|
Guo Y, Sun S, Wang Y, Chen S, Kou Z, Yuan P, Han W, Yu X. Microbial dysbiosis in obstructive sleep apnea: a systematic review and meta-analysis. Front Microbiol 2025; 16:1572637. [PMID: 40444003 PMCID: PMC12119640 DOI: 10.3389/fmicb.2025.1572637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/01/2025] [Indexed: 06/02/2025] Open
Abstract
Background The association between the microbiota and obstructive sleep apnea (OSA) remains understudied. In this study, we conducted a comprehensive systematic review and meta-analysis of studies investigating the diversity and relative abundance of microbiota in the gut, respiratory tracts and oral cavity of patients with OSA, aiming to provide an in-depth characterization of the microbial communities associated with OSA. Methods A comprehensive literature search across PubMed, the Cochrane Library, Web of Science, and Embase databases were conducted to include studies published prior to Dec 2024 that compared the gut, respiratory and oral microbiota between individuals with and without OSA. The findings regarding alpha-diversity, beta-diversity, and relative abundance of microbiota extracted from the included studies were summarized. This meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, and the study protocol was registered with PROSPERO (CRD42024525114). Results We identified a total of 753 articles, out of which 27 studies were ultimately included in the systematic review, involving 1,381 patients with OSA and 692 non-OSA populations, including 1,215 OSA patients and 537 non-OSA populations in adults and 166 OSA patients and 155 non-OSA populations in children. The results of alpha diversity revealed a reduction in the Chao1 index (SMD = -0.40, 95% CI = -0.76 to -0.05), Observed species (SMD = -0.50, 95% CI = -0.89 to -0.12) and Shannon index (SMD = -0.27, 95% CI = -0.47 to -0.08) of the gut microbiota in patients with OSA. Beta diversity analysis indicated significant differences in the gut, respiratory and oral microbial community structure between individuals with OSA and those without in more than half of the included studies. Furthermore, in comparison to the non-OSA individuals, the gut environment of patients with OSA exhibited an increased relative abundance of phylum Firmicutes, along with elevated levels of genera Lachnospira; conversely, there was a decreased relative abundance of phylum Bacteroidetes and genus Ruminococcus and Faecalibacterium. Similarly, within the oral environment of OSA patients, there was an elevated relative abundance of phylum Actinobacteria and genera Neisseria, Rothia, and Actinomyces. Conclusion Patients with OSA exhibit reduced diversity, changes in bacterial abundance, and altered structure in the microbiota, especially in the gut microbiota. The results of this study provide basic evidence for further exploration of microbiome diagnostic markers and potential intervention strategies for OSA.
Collapse
Affiliation(s)
- Yang Guo
- School of Medical Laboratory, Shandong Second Medical University, Weifang, China
| | - Shuqi Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yaoyao Wang
- Department of Medicine, Qingdao University, Qingdao, China
| | - Shiyang Chen
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Ziwei Kou
- Department of Medicine, Qingdao University, Qingdao, China
| | - Peng Yuan
- Qingdao Key Laboratory of Common Diseases, Department of Respiratory and Critical Medicine, Department of Emergency, Department of General Practice, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Wei Han
- Qingdao Key Laboratory of Common Diseases, Department of Respiratory and Critical Medicine, Department of Emergency, Department of General Practice, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Xinjuan Yu
- Clinical Research Center, Qingdao Key Laboratory of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
29
|
Connan C, Fromentin S, Benallaoua M, Alvarez AS, Pons N, Quinquis B, Morabito C, Nazare JA, Borezée-Durant E, Le French Gut Consortium, Haimet F, Ehrlich SD, Valeille K, Cavezza A, Blottière H, Veiga P, Almeida M, Doré J, Benamouzig R. Associations Among Diet, Health, Lifestyle, and Gut Microbiota Composition in the General French Population: Protocol for the Le French Gut - Le Microbiote Français Study. JMIR Res Protoc 2025; 14:e64894. [PMID: 40358997 PMCID: PMC12117270 DOI: 10.2196/64894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/28/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Over the past 2 decades, the gut microbiota has emerged as a key player in human health, being involved in many different clinical contexts. Yet, many aspects of the relationship with its host are poorly documented. One obstacle is the substantial variability in wet-laboratory procedures and data processing implemented during gut microbiota studies, which poses a challenge of comparability and potential meta-analysis. OBJECTIVE The study protocol described here aimed to better understand the relationship between health, dietary habits, and the observed heterogeneity of gut microbiota composition in the general population. "Le French Gut - Le microbiote français" aimed to collect, sequence, and analyze 100,000 fecal samples from French residents using a high-quality shotgun metagenomic pipeline, complemented with comprehensive health, lifestyle, and dietary metadata. METHODS "Le French Gut - Le microbiote français" is a prospective, noninterventional French national study involving individuals, the creation of a biological collection (feces), and the exploitation of data from questionnaires and the National Health Data System (Système National des Données de Santé). This national study is open to all metropolitan French adult residents, excluding those who have undergone a colectomy or digestive stoma, or who have had a colonoscopy or taken antibiotics in the last 3 months. This is a home-based trial in which volunteers complete a questionnaire with insights about their health and habits, and in which stool samples are self-collected. Data analysis is structured into 6 work packages, each focusing on a specific aspect of the gut microbiome, including its composition and associations with lifestyle, quality of life, and health. RESULTS This paper outlines the study protocol, with recruitment having started in September 2022 and expected to continue until the end of December 2025. As of January 2025, a total of 20,000 participants have been enrolled. The first scientific publications based on the data analysis are expected by mid-2025. CONCLUSIONS "Le French Gut" aims to provide a reference database and new ecosystem tools for understanding the relationship between the gut microbiota, its host, and diet. We expect to be able to find new signatures or targets and promote the design of innovative preventive strategies, personalized nutrition, and precision medicine. TRIAL REGISTRATION ClinicalTrials.gov NCT05758961; https://clinicaltrials.gov/study/NCT05758961. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/64894.
Collapse
Affiliation(s)
- Chloe Connan
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | | | - Mourad Benallaoua
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Bobigny, France
| | | | - Nicolas Pons
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Benoît Quinquis
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Christian Morabito
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Julie-Anne Nazare
- Univ-Lyon, CarMeN Laboratory, Inserm, Inrae, Université Claude Bernard Lyon-1, Lyon, France
| | - Elise Borezée-Durant
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Florence Haimet
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- INRAE Mica division, Jouy-en-Josas, France
| | | | - Karine Valeille
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Alexandre Cavezza
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Hervé Blottière
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Nantes Université, INRAE, UMR 1280, PhAN, Nantes, France
| | - Patrick Veiga
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Mathieu Almeida
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Robert Benamouzig
- Department of Gastroenterology, Avicenne Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Bobigny, France
| |
Collapse
|
30
|
Dai Y, Vgontzas AN, Chen L, Zheng D, Chen B, Wu J, Shao R, Li Y. A multi-omics study of the association between insomnia with objective short sleep duration phenotype and high blood pressure. Sleep 2025; 48:zsaf030. [PMID: 39888642 DOI: 10.1093/sleep/zsaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/26/2025] [Indexed: 02/01/2025] Open
Abstract
STUDY OBJECTIVES Insomnia with objective short sleep duration is associated with increased hypertension risk. We aimed to explore the mechanism underlying the association between objective short sleep duration and hypertension in patients with chronic insomnia disorder (CID) by multi-omics. METHODS CID was defined according to International Classification of Sleep Disorders-3, and objective short sleep duration was based on the median value of total sleep time of the overall subjects during an overnight polysomnography. We used the mean values of measured nighttime and morning systolic (SBP) and diastolic blood pressure (DBP) for analysis. Serum metabolomics and fecal 16S rDNA amplicon sequencing were used to explore characteristic metabolites and analyze gut microbiota distribution, respectively. RESULTS One hundred and three patients with CID and 70 normal sleepers were included. We found 52 objective short sleep duration insomnia phenotype (ISSD)-related serum metabolites. Among the 52 ISSD-related serum metabolites, indoxyl sulfate was positively correlated with BP after adjusting for confounding factors (SBP: β = 0.250, p = .028; DBP: β = 0.256, p = .030) in ISSD. In addition, the level of serum indoxyl sulfate was significantly correlated with the genera Prevotella 9 (r = .378, p = .027), CAG-56 (r = -.359, p = .037), Ruminiclostridium 9 (r = -.340, p = .049), and Ruminococcus 2 (r = -.356, p = .039) in ISSD. CONCLUSIONS Our study suggests that the ISSD phenotype is associated with significant changes in serum metabolic profile, including high levels of indoxyl sulfate. The latter molecule correlates both with BP and gut microbiota in patients with the ISSD phenotype, suggesting that indoxyl sulfate may be the molecular path resulting in increased hypertension risk in this phenotype.
Collapse
Affiliation(s)
- Yanyuan Dai
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Alexandros N Vgontzas
- Sleep Research and Treatment Center, Department of Psychiatry and Behavioral Health, Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Le Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Dandan Zheng
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Baixin Chen
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Jun Wu
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Ruifan Shao
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| | - Yun Li
- Department of Sleep Medicine, Shantou University Mental Health Center, Shantou, Guangdong, China
- Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong, China
| |
Collapse
|
31
|
Kehrmann J, Dostmohammadi A, Stumpf AL, Best L, Consten L, Sievert H, Maischack F, Sammet S, Albayrak-Rena S, Doerr AK, Bohlen K, von Velsen O, Kraiselburd I, Karsten CB, Farahpour F, Meyer F, Esser S, Buer J. Gut microbiota differences linked to weight gain and ART in people living with HIV are enterotype specific and minor compared to the large differences linked to sexual behavior. Front Cell Infect Microbiol 2025; 15:1568352. [PMID: 40406514 PMCID: PMC12095285 DOI: 10.3389/fcimb.2025.1568352] [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: 01/29/2025] [Accepted: 04/03/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Specific antiretroviral therapy (ART) regimens are associated with weight gain in people living with HIV (PLWH). Gut microbiota is involved in weight gain in humans and animals. Human gut microbiota can be classified into enterotypes with distinct microbial and functional profiles. Methods In a cohort of 118 PLWH, we analyzed the gut microbiome in relation to weight gain and ART regimen using 16S rRNA gene sequencing, taking enterotype classification into account. Results The enterotype was strongly associated with sexual orientation. Of the 67 individuals forming a Prevotella-dominated enterotype cluster in principal coordinates analysis, 93% were men who had sex with men (MSM), while 31% of individuals in the Bacteroides-dominated enterotype cluster were MSM and 69% were non-MSM. Forty-nine genera differed significantly between the MSM and non-MSM individuals. When stratified by dominant genus, only six taxa were associated with weight gain. Of these, five were restricted to Bacteroides-dominated individuals. Among them, the class Actinobacteria and genus Bifidobacterium differed between individuals gaining more than 5% weight and less than 5% weight 1 year after ART switch. Additionally, three taxa were significantly different between 15% of individuals with the highest weight gain (≥6.3%) and the highest weight loss (≤3.19%) 1 year after ART switch, including the phyla Firmicutes, Verrucomicrobia, and Synergistetes. Distinct functional properties in Bacteroides, but not Prevotella-dominated enterotype individuals, linked to weight gain were observed, particularly for glycan and lipid metabolism. Additionally, ART regimen-associated differences were observed for the phylum Actinobacteria, although this was limited to Prevotella-dominated enterotype individuals. Discussion Differences in the composition and functional characteristics of the gut microbiome associated with weight gain and ART regimens were enterotype-specific and relatively small compared with differences linked to sexual orientation. Due to the substantial differences in gut microbiome structure among many MSM, categorization into enterotypes is useful for identifying differences in microbiome composition associated with variables such as weight gain or ART, which may be limited to a single enterotype. This may further advance the identification of microbes that contribute to weight gain or alter the gut microbiome composition in the context of the enterotype.
Collapse
Affiliation(s)
- Jan Kehrmann
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alireza Dostmohammadi
- Bioinformatics and Computational Biophysics, Faculty of Biology and Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
| | - Anna-Lena Stumpf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lara Best
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leah Consten
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hannah Sievert
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Felix Maischack
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefanie Sammet
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Albayrak-Rena
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ann-Kathrin Doerr
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Katharina Bohlen
- Department of Gastroenterology, Hepatology and Transplantational Medicine, University Hospital Essen, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Otgonzul von Velsen
- Institute of Medical Informatics, Biometrics, and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Clinical Trials, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ivana Kraiselburd
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christina B. Karsten
- Institute for the Research on HIV and AIDS-associated Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Farnoush Farahpour
- Bioinformatics and Computational Biophysics, Faculty of Biology and Centre for Medical Biotechnology (ZMB), University of Duisburg-Essen, Essen, Germany
- Institute of Cell Biology (Tumor Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Folker Meyer
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefan Esser
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
32
|
Origüela V, Lopez-Zaplana A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025; 13:1084. [PMID: 40431257 PMCID: PMC12113704 DOI: 10.3390/microorganisms13051084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
The importance of the microbiome, particularly the gut microbiota and its implications for health, is well established. However, an increasing number of studies further strengthen the link between an imbalanced gut microbiota and a greater predisposition to different diseases. The gut microbiota constitutes a fundamental ecosystem for maintaining human health. Its alteration, known as dysbiosis, is associated with a wide range of conditions, including intestinal, metabolic, immunological, or neurological pathologies, among others. In recent years, there has been a substantial increase in knowledge about probiotics-bacterial species that enhance health or address various diseases-with numerous studies reporting their benefits in preventing or improving these conditions. This review aims to analyze the most common pathologies resulting from an imbalance in the gut microbiota, as well as detail the most important and known gut probiotics, their functions, and mechanisms of action in relation to these conditions.
Collapse
Affiliation(s)
- Valentina Origüela
- Department of Physiology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain;
| | | |
Collapse
|
33
|
Wei C, Xu X, Zhang J, Wang X, Han T, Zhang Y, Pan S, Ming Z, Li R, Lou F, Cheng Y, Xu H, Sun X, Geng G, Pan Y, Liu Q, Qi H, Yan X, Dang K, Zhou J, Sun C, Li Y. Timing of unsaturated fat intake improves insulin sensitivity via the gut microbiota-bile acid axis: a randomized controlled trial. Nat Commun 2025; 16:4211. [PMID: 40328731 PMCID: PMC12056104 DOI: 10.1038/s41467-025-58937-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
The timing of dietary total fat intake influences glucose homeostasis, however, the impact of unsaturated fat (USFA) intake has yet to be explored. This 12-week, double-blind, randomized, controlled, 2 × 2 factorial-designed feeding trial investigated the effects of timing (lunch or dinner) and types of dietary USFA (high monounsaturated fat or polyunsaturated fat diet) intake on glucose metabolism in seventy prediabetes participants (mean age, 57 years). Sixty participants with completed fecal samples were included in the final analysis (n = 15 for each group). Postprandial serum glucose was first primary outcome, postprandial insulin levels and insulin sensitivity indices were co-primary outcomes Secondary outcomes were continuous glucose levels, serum fatty acid profile, gut microbiome (metagenomic sequencing) and fecal metabolites. Results showed no significant differences in postprandial glucose between groups. However, USFA intake at lunch (vs. dinner) improved insulin sensitivity and reduced postprandial insulin and serum free saturated fatty acid (Ptiming < 0.05, Ptype > 0.05, Pinteraction > 0.05), which was associated with alterations in gut microbiome and bile acid metabolism, regardless of USFA type. In summary, these results suggest that advancing timing of USFA intake improves insulin sensitivity through the gut microbiome and bile acid metabolism. Trial registration: ChiCTR2100045645.
Collapse
Affiliation(s)
- Chunbo Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoqing Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jia Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuanyang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yingfeng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhu Ming
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ran Li
- Department of Clinical Nutrition, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fengge Lou
- Public Health Research Office, School of Public Health, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yu Cheng
- Public Health Research Office, School of Public Health, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Huan Xu
- Department of Clinical Nutrition, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xingyuan Sun
- Department of Neurology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Guannan Geng
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yujun Pan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qianmin Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Haitao Qi
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuemin Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Keke Dang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaofeng Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Ying Li
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
34
|
Castañeda S, Tomiak J, Andersen LO, Acosta CP, Vasquez-A LR, Stensvold CR, Ramírez JD. Impact of Blastocystis carriage and colonization intensity on gut microbiota composition in a non-westernized rural population from Colombia. PLoS Negl Trop Dis 2025; 19:e0013111. [PMID: 40354411 PMCID: PMC12097710 DOI: 10.1371/journal.pntd.0013111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 05/22/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The role of Blastocystis, a common intestinal parasitic protist of humans and other animals, in human health and disease remains elusive. Recent studies suggest a connection between Blastocystis colonization, healthier lifestyles, and high-diversity gut microbiota. Nevertheless, studies concerning the relationship between Blastocystis colonization, its intensity, and gut microbiota composition -involving both bacterial and eukaryotic communities- remain limited. METHODS This study examines the impact of Blastocystis carriage and colonization intensity on gut microbiota composition in a rural community in Colombia. A total of 88 human samples were collected from the rural population of Las Guacas village, located in the Cauca department in southwest Colombia. We utilized 16S and 18S rDNA sequencing to analyze both bacterial and eukaryotic microbiota, comparing Blastocystis-positive and -negative individuals, as well as groups with varying Blastocystis colonization intensity (low, medium, high), to identify distinct microbiota profiles and differentially abundant taxa linked to each condition. RESULTS The analysis revealed significant differences between Blastocystis-positive and -negative individuals. In terms of bacterial composition and structure, Blastocystis-positive individuals exhibited distinct microbiota profiles, as shown by beta diversity analysis. Taxa associated with colonization included Bacteroides, Prevotella, Oscillibacter, Faecalibacterium, and Alistipes. Higher Blastocystis colonization intensity was associated with an increased abundance of taxa such as Alistipes and Lachnospira, while lower intensities correlated with beneficial bacteria such as Akkermansia. Regarding eukaryotic composition, beta diversity analysis revealed distinct profiles associated with Blastocystis colonization. Differentially abundant taxa, including Entamoeba coli, were more prevalent in Blastocystis-positive individuals, while Blastocystis-negative individuals exhibited a higher abundance of opportunistic fungi, such as Candida albicans. Machine learning models, including random forest classifiers, supported these findings, identifying Faecalibacterium and Bacteroides as predictors of Blastocystis colonization. CONCLUSIONS These findings suggest that Blastocystis may modulate gut microbiota, contributing to microbial balance providing new insights into the ecological implications of Blastocystis in rural populations.
Collapse
Affiliation(s)
- Sergio Castañeda
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Jeff Tomiak
- Laboratory of Parasitology, Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
- University of Stavanger, Department of Chemistry, Bioscience, and Environmental Engineering, Stavanger, Norway
| | - Lee O’Brien Andersen
- Laboratory of Parasitology, Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Claudia Patricia Acosta
- Grupo de investigación en Genética Humana, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Universidad del Cauca,
| | - Luis Reinel Vasquez-A
- Centro de Estudios en Microbiología y Parasitología, Facultad de Ciencias de la Salud, Universidad del Cauca, Popayán, Colombia
| | - Christen Rune Stensvold
- Laboratory of Parasitology, Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York city, New York, United States of America
| |
Collapse
|
35
|
Liu L, Lei S, Lin X, Bodjrenou DM, Zhang Y, Zheng B, Zeng H. Synergistic regulation of colon microflora and metabolic environment by resistant starch and sodium lactate in hyperlipidemic rats. Int J Biol Macromol 2025; 307:141933. [PMID: 40074132 DOI: 10.1016/j.ijbiomac.2025.141933] [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: 12/04/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Type 3 resistant starch (RS3) regulates diet-related metabolic diseases by promoting intestinal short-chain fatty acids (SCFAs) and lactate production, and facilitating microbial lactate-to-butyrate fermentation. However, its precise in vivo mechanism remains unclear. Therefore, we studied the effects of type 3 lotus seed resistant starch (LRS3) and sodium lactate (SL) on colonic microbiota composition, metabolism, and lipid parameters. This study aimed to elucidate the mechanism by which LRS3 and SL modulate colonic microbiota and metabolism to mitigate hyperlipidemia in rats induced by a high-fat diet. Results showed LRS3 increased colonic microbial diversity, shifting the composition towards that of healthy rats. LRS3 intake reduced lactic acid-producing bacteria such as Allobaculum, Collinsella, and Blautia in the colon while promoting SCFAs-producing Ruminococcaceae. SL alone stimulated Lachnospiraceae growth. When both were administered, there was a significant increase in Treponema and Ruminococcaceae. The co-intervention of LRS3 and SL significantly affected lipid metabolism-related metabolites, up-regulating palmitic acid while down-regulating androsterone and phosphatidylcholine (PC) substances PC (14:0/20:4(8Z,11Z,14Z,17Z)), influencing unsaturated fatty acid biosynthesis pathways and inhibiting steroid hormone biosynthesis. Finally, via the microbial-metabolism-lipid correlation network, we identified that LRS3 and SL increased SCFAs production through Treponema and Ruminococcaceae metabolism, influencing organic acid and lipid composition in the colon. This indirectly reduced blood lipid levels in hyperlipidemic rats by modulating intestinal microecology.
Collapse
Affiliation(s)
- Lu Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Suzhen Lei
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoli Lin
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - David Mahoudjro Bodjrenou
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yi Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Baodong Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hongliang Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
36
|
Yamamura R, Okubo R, Ukawa S, Nakamura K, Okada E, Nakagawa T, Imae A, Kimura T, Tamakoshi A. Increased fecal glycocholic acid levels correlate with obesity in conjunction with the depletion of archaea: The Dosanco Health Study. J Nutr Biochem 2025; 139:109846. [PMID: 39863085 DOI: 10.1016/j.jnutbio.2025.109846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Recent studies have focused on the relationship between obesity and gut microbiota. This study aims to identify fecal components and gut bacterial species associated with different BMI categories. In this study, 538 participants aged ≥18 years were categorized into underweight, normal, and obese groups based on BMI (cutoffs: 18.5 and 25.0 kg/m²). We compared 30 fecal components among these groups and calculated correlation coefficients between each component and BMI. Participants were then divided into quartiles based on fecal component levels correlated with BMI, and the prevalence ratio (PR) of obesity was calculated, adjusted for confounding factors. We also analyzed the composition and diversity of gut microbiota and bacterial gene expression among the quartiles for each fecal component. Fecal glycocholic acid (GCA) showed a significant positive correlation with BMI. The PR for obesity in the highest quartile of fecal GCA was 3.30 (95% CI, 1.21-9.54), indicating a significantly higher risk of obesity compared to the lowest quartile. Gut microbiota analysis revealed significant differences in the abundance of Ruminococcaceae Incertae Sedis, Faecalibacterium, and Methanobrevibacter, with Methanobrevibacter being absent in the higher quartiles of fecal GCA. Additionally, gene expression for enzymes involved in the deconjugation of conjugated bile acids, including GCA, was downregulated in the highest quartile. Increased fecal GCA levels are positively correlated with obesity, alongside a depletion of archaea.
Collapse
Affiliation(s)
- Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| | - Ryo Okubo
- Department of Neuropsychiatry, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shigekazu Ukawa
- Osaka Metropolitan University Graduate School of Human Life and Ecology, Sumiyoshi, Osaka, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Koshi Nakamura
- Department of Public Health and Epidemiology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Emiko Okada
- The Health Care Science Institute, Minato-ku, Tokyo, Japan; Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Akihiro Imae
- The Hokkaido Centre for Family Medicine, Sapporo, Japan
| | - Takashi Kimura
- Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Akiko Tamakoshi
- Department of Public Health, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
37
|
Charalambous H, Brown C, Vogazianos P, Katsaounou K, Nikolaou E, Stylianou I, Papageorgiou E, Vraxnos D, Aristodimou A, Chi J, Costeas P, Shammas C, Apidianakis Y, Antoniades A. Dysbiosis in the Gut Microbiome of Pembrolizumab-Treated Non-Small Lung Cancer Patients Compared to Healthy Controls Characterized Through Opportunistic Sampling. Thorac Cancer 2025; 16:e70075. [PMID: 40356191 PMCID: PMC12069221 DOI: 10.1111/1759-7714.70075] [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: 11/22/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The gut microbiome influences the host immune system, cancer development and progression, as well as the response to immunotherapy during cancer treatment. Here, we analyse the composition of the gut bacteriome in metastatic Non-Small Cell Lung Cancer (NSCLC) patients receiving Pembrolizumab immunotherapy within a prospective maintenance trial through opportunistic sampling during treatment. METHODS The gut microbiome profiles of NSCLC patients were obtained from stool samples collected during Pembrolizumab treatment and analysed with 16S rRNA metagenomics sequencing. Patient profiles were compared to a group of healthy individuals of matching ethnic group, age, sex, BMI and comorbidities. RESULTS A significant decrease in the treated patients was observed in two prominent bacterial families of the phylum Firmicutes, Lachnospiraceae and Ruminoccocaceae, which comprised 31.6% and 21.8% of the bacteriome in the healthy group but only 10.9% and 14.2% in the treated patient group, respectively. Species within the Lachnospiraceae and Ruminococcaceae families are known to break down undigested carbohydrates generating short chain fatty acids (SCFA), such as butyrate, acetate and propionate as their major fermentation end-products, which have been implicated in modifying host immune responses. In addition, a significant increase of the Bacteroidacaeae family (Bacteroidetes phylum) was observed from 10.7% in the healthy group to 23.3% in the treated patient group. Moreover, and in agreement with previous studies, a decrease in the Firmicutes to Bacteroidetes ratio in the metastatic NSCLC Pembrolizumab-treated patients was observed. CONCLUSION The observed differences indicate dysbiosis and a compromised intestinal health status in the metastatic NSCLC Pembrolizumab-treated patients. This data could inform future studies of immunotherapy treatment responses and modulation of the gut microbiome to minimise dysbiosis prior or concurrent to treatment. TRIAL REGISTRATION SWIPE Trial (NCT02705820).
Collapse
MESH Headings
- Humans
- Gastrointestinal Microbiome/drug effects
- Dysbiosis/chemically induced
- Dysbiosis/microbiology
- Dysbiosis/pathology
- Male
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/microbiology
- Female
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/microbiology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Aged
- Prospective Studies
- Case-Control Studies
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
| | | | - Paris Vogazianos
- Stremble Ventures LtdLimassolCyprus
- European University CyprusEngomiCyprus
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang N, Wu M, Gu W, Dai C, Shao Z, Subbalakshmi KP. MSFT-transformer: a multistage fusion tabular transformer for disease prediction using metagenomic data. Brief Bioinform 2025; 26:bbaf217. [PMID: 40370098 PMCID: PMC12078939 DOI: 10.1093/bib/bbaf217] [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: 01/26/2025] [Revised: 04/05/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
More and more recent studies highlight the crucial role of the human microbiome in maintaining health, while modern advancements in metagenomic sequencing technologies have been accumulating data that are associated with human diseases. Although metagenomic data offer rich, multifaceted information, including taxonomic and functional abundance profiles, their full potential remains underutilized, as most approaches rely only on one type of information to discover and understand their related correlations with respect to disease occurrences. To address this limitation, we propose a multistage fusion tabular transformer architecture (MSFT-Transformer), aiming to effectively integrate various types of high-dimensional tabular information extracted from metagenomic data. Its multistage fusion strategy consists of three modules: a fusion-aware feature extraction module in the early stage to improve the extracted information from inputs, an alignment-enhanced fusion module in the mid stage to enforce the retainment of desired information in cross-modal learning, and an integrated feature decision layer in the late stage to incorporate desired cross-modal information. We conduct extensive experiments to evaluate the performance of MSFT-Transformer over state-of-the-art models on five standard datasets. Our results indicate that MSFT-Transformer provides stable performance gains with reduced computational costs. An ablation study illustrates the contributions of all three models compared with a reference multistage fusion transformer without these novel strategies. The result analysis implies the significant potential of the proposed model in future disease prediction with metagenomic data.
Collapse
Affiliation(s)
- Ning Wang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Minghui Wu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Wenchao Gu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Chenglong Dai
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | | | - K P Subbalakshmi
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, Castle Point Terrace, Hoboken, NJ 07030, United States
| |
Collapse
|
39
|
He X, Chen H, Chen F, Su W, Wang Y, Hu D, Hu J, Zhou X. Characterization of Fecal Microbial Communities in Patients With Type 2 Diabetes Mellitus Combined With Helicobacter pylori Infection. Helicobacter 2025; 30:e70041. [PMID: 40338991 DOI: 10.1111/hel.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection has the capacity to alter the gut microbiota composition. There is a significant correlation between H. pylori infection and type 2 diabetes mellitus (T2DM). Further research is necessary to explore whether gut microbiota plays a role in the relationship between H. pylori and T2DM. METHOD Fecal samples were obtained from 44 patients with T2DM, including 20 who tested positive for H. pylori and 24 who tested negative. Intestinal microbiota composition was analyzed via 16S rRNA V3-V4 amplicon sequencing. Differences in microbial distribution and significant microbial biomarkers were identified between H. pylori positive and negative groups. A Spearman correlation analysis assessed the relationship between intestinal microbiota and glycemic parameters. Additionally, PICRUSt2 was used to predict intestinal bacterial functions. RESULTS Results indicate that in H. pylori (+) T2DM patients, HbA1c levels were significantly higher (8.9% vs. 8.1%, p = 0.021), while both the C-peptide peak (3.70 vs. 5.98 ng/mL, p = 0.040) and fasting C-peptide levels (1.42 vs. 2.31 ng/mL, p = 0.008) were significantly lower compared to H. pylori (-) T2DM groups. A total of 11 colonic phyla and 100 genera were identified in all fecal samples. In groups positive for H. pylori, there was a significant enrichment of the phylum Proteobacteria, while the genera Lactobacillus, Butyricimonas, and Akkermansia were significantly reduced (all p < 0.05). Correlation analysis showed that the abundance of the genera Butyricimonas (p = 0.01) and Akkermansia (p = 0.048) were negatively correlated with fasting plasma glucose. KEGG pathway analysis indicated a significant enrichment of methylmalonyl-CoA mutase and succinyl-CoA in H. pylori-infected T2DM patients. CONCLUSIONS This study suggests that T2DM patients with H. pylori infection exhibit more impaired pancreatic islet function potentially due to H. pylori-induced alterations in the gut microbiota.
Collapse
Affiliation(s)
- Xiaoyan He
- Department of Gastroenterology, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang, China
| | - Han Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengdan Chen
- Department of Endocrinology, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang, China
| | - Wei Su
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Die Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianwen Hu
- Department of Gastroenterology, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang, China
| | - Xiaoying Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Shen Q, Yang Z, Hu C, Liu Y, Zhao L, Li C, Ma Y, Bian H. Non-starch polysaccharides and health: gut-target organ axis influencing obesity. Food Sci Biotechnol 2025; 34:1771-1788. [PMID: 40196321 PMCID: PMC11972281 DOI: 10.1007/s10068-024-01745-3] [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: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 04/09/2025] Open
Abstract
Obesity is recognized as a global epidemic that can result in changes in the human body and metabolism. Accumulating evidence indicates that gut microbiota (GM) can affect the development of obesity. The GM not only plays a crucial role in digesting and absorbing nutrients, but also in maintaining the overall health of the host. Dietary supplements such as non-starch polysaccharides are mainly fermented by the GM in the colon. Recent findings suggest that shaping the GM through the prebiotic function of non-starch polysaccharides may be a viable strategy against obesity. In this paper, the effects of non-starch polysaccharides on host health, together with their prebiotic function influencing the GM to control obesity via the gut-target organ axis, are reviewed. Potential perspectives of non-starch polysaccharides exhibiting anti-obesity effects via the gut-target organ axis are proposed for future research. Graphical abstract
Collapse
Affiliation(s)
- Qingshan Shen
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Zhuan Yang
- School of Life Science and Agricultural Engineering, Nanyang Normal University, Wolong Road 1638, Nanyang, 473061 China
| | - Chengzhi Hu
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071000 China
| | - Yilin Liu
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Lei Zhao
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Cuicui Li
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Yanli Ma
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| | - Hua Bian
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Changjiang Road 80, Nanyang, 473004 Henan China
| |
Collapse
|
41
|
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] [MESH Headings] [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
|
42
|
Niu Y, Xiao L, Feng L. Association between dietary index for gut microbiota and metabolic syndrome risk: a cross-sectional analysis of NHANES 2007-2018. Sci Rep 2025; 15:15153. [PMID: 40307409 PMCID: PMC12044051 DOI: 10.1038/s41598-025-99396-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
Metabolic syndrome (MetS) poses a significant global health challenge, closely associated with cardiovascular diseases, diabetes, and other conditions. With the global prevalence of MetS steadily rising, the potential role of gut microbiota in its development has garnered increasing attention. Against this backdrop, the present study aims to explore the association between the dietary index for gut microbiota (DI-GM) score and MetS. This cross-sectional study utilized data from the 2007-2018 U.S. National Health and Nutrition Examination Survey (NHANES), including 339,242 adults aged ≥ 18 years. The DI-GM score, constructed based on 14 food or nutrient components, served as the exposure variable. MetS was defined according to the Adult Treatment Panel III (ATP III) criteria, including abdominal obesity (waist circumference ≥ 102 cm in men and ≥ 88 cm in women), elevated triglycerides (≥ 150 mg/dL), reduced HDL cholesterol (< 40 mg/dL in men and < 50 mg/dL in women), elevated blood pressure (≥ 130/85 mmHg), and elevated fasting glucose (≥ 100 mg/dL). Multivariable logistic regression analyses were performed to adjust for demographic characteristics, lifestyle factors, and other potential confounders. Higher DI-GM scores were significantly associated with a reduced risk of MetS. After adjusting for all confounders, individuals in the highest quartile (Q4) of DI-GM scores had a 16% lower risk of MetS compared to those in the lowest quartile (Q1) (OR: 0.84; 95%CI: 0.70-1.01). Mediation analyses revealed that systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) mediated 4.63% and 3.83% of the association between DI-GM and MetS, respectively. There is an inverse association between DI-GM scores and the risk of MetS, potentially mediated in part by inflammatory markers. These findings provide new evidence supporting dietary interventions aimed at improving gut microbiota to prevent MetS.
Collapse
Affiliation(s)
- Yueyue Niu
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Xiao
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ling Feng
- the Cadre Health Care Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
43
|
Andreu-Sánchez S, Blanco-Míguez A, Wang D, Golzato D, Manghi P, Heidrich V, Fackelmann G, Zhernakova DV, Kurilshikov A, Valles-Colomer M, Weersma RK, Zhernakova A, Fu J, Segata N. Global genetic diversity of human gut microbiome species is related to geographic location and host health. Cell 2025:S0092-8674(25)00416-7. [PMID: 40311618 DOI: 10.1016/j.cell.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/23/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
The human gut harbors thousands of microbial species, each exhibiting significant inter-individual genetic variability. Although many studies have associated microbial relative abundances with human-health-related phenotypes, the substantial intraspecies genetic variability of gut microbes has not yet been comprehensively considered, limiting the potential of linking such genetic traits with host conditions. Here, we analyzed 32,152 metagenomes from 94 microbiome studies across the globe to investigate the human microbiome intraspecies genetic diversity. We reconstructed 583 species-specific phylogenies and linked them to geographic information and species' horizontal transmissibility. We identified 484 microbial-strain-level associations with 241 host phenotypes, encompassing human anthropometric factors, biochemical measurements, diseases, and lifestyle. We observed a higher prevalence of a Ruminococcus gnavus clade in nonagenarians correlated with distinct plasma bile acid profiles and a melanoma and prostate-cancer-associated Collinsella clade. Our large-scale intraspecies genetic analysis highlights the relevance of strain diversity as it relates to human health.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Daoming Wang
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Golzato
- Department of CIBIO, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of CIBIO, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of CIBIO, University of Trento, Trento, Italy
| | | | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Mireia Valles-Colomer
- Department of CIBIO, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | - Nicola Segata
- Department of CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
44
|
Bhowmick K, Yang X, Mohammad T, Xiang X, Molmenti CL, Mishra B, Dasarathy S, Krainer AR, Hassan MI, Crandall KA, Mishra L. Microbial metabolite ammonia disrupts TGF-β signaling to promote colon cancer. J Biol Chem 2025:108559. [PMID: 40311681 DOI: 10.1016/j.jbc.2025.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Colorectal cancer (CRC) is rising alarmingly in younger populations, potentially arising from factors such as obesity, pro-inflammatory gut microbiome and the accumulation of toxic metabolites. However, how metabolites such as ammonia impact key signaling pathways to promote CRC remains unclear. Our study investigates a critical link between gut microbiome alterations, ammonia, and their toxic effects on the TGF-β signaling pathway, driving CRC progression. We observe altered microbial populations in an obesity-induced mouse model of cancer, where ammonia promotes caspase-3-mediated cleavage of the SMAD3 adaptor βII-spectrin (SPTBN1). Cleaved SPTBN1 fragments form adducts with ammonia that induce pro-inflammatory cytokine expression and disrupt TGF-β signaling. Extending from AlphaFold docking simulations, we identified that ammonia interacts with N-terminal SPTBN1 potentially through residues D81, Y556, S663, Y666, N986, and D1177 to form hydrogen bonds that disrupt downstream SMAD3 signaling, altering TGF-β signaling to a protumorigenic phenotype. Blocking SPTBN1, through an SPTBN1-specific siRNA blocks ammonia toxicity and restores normal SMAD3/TGF-β signaling by reducing the abundance of SPTBN1 cleaved fragments in SW480 and Caco-2 (CRC) cell lines. In addition, our research establishes crosstalk between TGF-β signaling and a microbial sensor, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), which is significantly overexpressed in CRC patients. We identified CEACAM1-SPTBN1 interactions at specific residues (E517 and Y520) within the immunoreceptor tyrosine-based inhibitory motif (ITIM) of CEACAM1 cytoplasmic domain, identifying distinguishing a potential axis that is harnessed by the altered microbiome. Our study identifies mechanistic insights into how microbial metabolites target TGF-β as a major signaling pathway to promote CRC.
Collapse
Affiliation(s)
- Krishanu Bhowmick
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA.
| | - Xiaochun Yang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Xiyan Xiang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Christine L Molmenti
- Department of Occupational Medicine, Epidemiology and Prevention, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Feinstein Institutes for Medical Research, Institute of Cancer Research, Manhasset, NY, USA; Department of Surgery, Northwell Health, Manhasset, NY, USA
| | - Bibhuti Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Department of Neurology, Hempstead, NY, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Keith A Crandall
- Computational Biology Institute and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington DC, USA
| | - Lopa Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA; Department of Surgery, George Washington University, Washington DC, USA.
| |
Collapse
|
45
|
Xu Q, Lv Y, Yuan X, Huang G, Guo Z, Tan J, Qiu S, Wang X, Wei C. Simulated Gastrointestinal Digestion and In Vitro Fecal Fermentation of Purified Pyracantha fortuneana (Maxim.) Li Fruit Pectin. Foods 2025; 14:1529. [PMID: 40361612 PMCID: PMC12071275 DOI: 10.3390/foods14091529] [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: 03/20/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pyracantha fortuneana, an underutilized wild plant, has been found to have a high nutritional value. This study used simulated digestion and fecal fermentation models to investigate the digestive properties of the purified acidic pectin polysaccharide of Pyracantha fortuneana and its impact on the gut microbiota and metabolites. Pyracantha fortuneana polysaccharide (PFP) is mainly composed of rhamnose (Rha), galacturonic acid (GalA), glucose (Glc), galactose (Gal), and arabinose (Ara), with a molecular weight (Mw) of 851.25 kDa. Following simulated digestion, the Mw of PFP remained consistent. The reduced sugar content showed minimal change, suggesting that PFP exhibits resistance to gastrointestinal digestion and can effectively reach the colon. Following fecal fermentation, the molecular weight, monosaccharide, and carbohydrate contents of PFP decreased, while the short-chain fatty acid content increased. This suggests that PFP is susceptible to degradation by microorganisms and can be metabolized into acetic acid and n-butyric acid, contributing to the regulation of intestinal health. Meanwhile, PFP promotes the reproduction of beneficial bacteria such as Bacteroides, Dialister, and Dysgonomonas, inhibits the growth of harmful bacteria like Proteus, and generates metabolites such as thiamine, leonuriside A, oxoadipic acid, S-hydroxymethylglutathione, and isonicotinic acid, which exert beneficial effects on human health. These results indicate that PFP has great potential in regulating the gut microbiota and generating beneficial metabolites to promote intestinal functional health and can be used as a prebiotic to prevent diseases by improving intestinal health.
Collapse
Affiliation(s)
- Qingrui Xu
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Yiyi Lv
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Xiaohui Yuan
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Guichun Huang
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Institute of Agro-Bioengineering, College of Life Sciences, Guizhou University, Guiyang 550025, China
| | - Zhongxia Guo
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Jiana Tan
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Shuyi Qiu
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Xiaodan Wang
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Chaoyang Wei
- Key Laboratory of Fermentation Engineering and Biological Pharmacy of Guizhou Province, School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Institute of Agro-Bioengineering, College of Life Sciences, Guizhou University, Guiyang 550025, China
| |
Collapse
|
46
|
Xu Y, Li J, Cui M, Li X, Zhai H, Wu D, Chu X. Therapeutic effects of medicinal and food-based traditional herbal couples on type 2 diabetes mellitus based on pharmacodynamics and pharmacokinetics. Front Pharmacol 2025; 16:1560271. [PMID: 40351422 PMCID: PMC12062019 DOI: 10.3389/fphar.2025.1560271] [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: 01/14/2025] [Accepted: 03/14/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Cinnamomi Ramulus (CR) is the dried bark of Cinnamomum cassia Presl, Lauraceae. Puerariae Lobatae Radix (PLR) is the dried root of the Pueraria lobata (Wild.) Ohwi, Leguminosae. This Chinese herb couple come from the classic formula "Gui Zhi Ge Gen Tang," which is included in the TCM classic "Treatise on Febrile Diseases." Our previous studies have found that CR related herbal compound and PLR related herbal compound are useful in improving type 2 diabetes mellitus (T2DM), which is expected to be an antidiabetic candidate with fewer side effects. However the mechanism of action of CR-PLR on T2DM has not yet been fully elucidated. Methods The decoction of CR-PLR was prepared by aqueous extraction method and the composition of it was analyzed using UPLC-Q-TOF-MS and HPLC. The T2DM model was established by intraperitoneal injection of streptozotocin, and the groups of drug administration were metformin, CR, PLR and CR-PLR groups, with continuous gastric gavage for 6 weeks, and the serological indexes were detected by ELISA. The abundance of rats' gut flora was detected by 16s rDNA sequencing, and changes in the content of short-chain fatty acids (SCFAs) in feces of rats were detected by GC-MS; and the expression of G protein-coupled receptor43 (GPR43) and glucagon-like peptide-1 (GLP-1) proteins in colonic tissues of rats were detected by Western Blot. The pharmacokinetic behavior of CR-PLR was investigated in both normal and T2DM model rats. Caco-2/HT29 co-culture cell model was established in vitro, transepithelial electrical resistance (TEER) and ALP activity of epithelial cells were measured to evaluate cell model integrity and cell polarization, Alcian blue staining was used to verify the presence of mucus production, and CCK-8 was used to screen drug safe concentration. The bidirectional transport of puerarin was studied to investigate the transport mechanism of puerarin and the effect of leuric acid on puerarin transport. Results and discussion The results indicated that CR-PLR can stimulate intestinal flora, increase the content of SCFAs, activate intestinal GPR43 protein, and promote the secretion of GLP-1 in intestinal L cells, which plays a therapeutic role in the treatment of T2DM. Additionally, cytology and pharmacokinetics experiments have proved that cinnamic acid (CA) can enhance the absorption and transport of puerarin (PUR) by inhibiting the efflux effects mediated by P-gp and MRP efflux transporters. The present study exhibites the scientific and reasonable menaning of this novel Chinese herb couple treating T2DM from the perspecives of pharmacodynamics and pharmacokinetics.
Collapse
Affiliation(s)
- Yuhang Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengyao Cui
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xiaoliang Li
- Anhui Joyfar Pharmaceutical Research Institute Co., Ltd., Hefei, China
| | - Hongyan Zhai
- Anhui Province Institute for Food and Drug Control, Hefei, China
| | - Deling Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Bozhou University, Bozhou, China
- Anhui Provincial Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui, China
| |
Collapse
|
47
|
Petitclerc I, Perron J, Dugas C, Mayer T, Raymond F, Di Marzo V, Veilleux A, Robitaille J. Association between gestational diabetes mellitus, maternal health and diet, and gut microbiota in mother-infant dyads. BMC Pregnancy Childbirth 2025; 25:486. [PMID: 40275186 PMCID: PMC12023395 DOI: 10.1186/s12884-025-07584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) increasingly affects women and predisposes both mothers and their infants to short- and long-term health consequences. Emerging research links GDM to maternal gut microbiota dysbiosis. However, the impact of GDM on the infant gut microbiota remains unclear. This cross-sectional study aims to explore potential associations between GDM and the gut microbiota in mothers and their infants, as well as correlations between maternal diet, cardiometabolic profile, and gut microbiota composition. METHODS Gut microbiota taxonomic composition was characterized by 16S rRNA gene sequencing on fecal samples collected at 2 months postpartum from 28 mothers, including 17 with (GDM+) and 11 without (GDM-) GDM, as well as 30 infants, 17 GDM + and 13 GDM-. Variations in overall composition and specific taxa between GDM + and GDM- were assessed. Correlations between maternal cardiometabolic profile, dietary intakes, and taxa were performed. RESULTS GDM was associated with the overall composition of gut microbiota between GDM + and GDM- in the maternal group, but not in infants. No statistically significant difference in alpha diversity between groups was found in either mothers or infants. However, 14 taxa showed significantly different abundance between GDM + and GDM- mothers, and 4 taxa differed in infants. Specific taxa at the family rank were correlated with maternal dietary and cardiometabolic variables in both mothers and infants. CONCLUSIONS GDM exposition was associated with gut microbiota composition in both mothers and infants at two months postpartum. This study enhances our understanding of how maternal health could be linked with the gut microbiota of mothers and their infants. TRIAL REGISTRATION NCT02872402 (2016-08-04, https://clinicaltrials.gov/study/NCT02872402?term=NCT02872402&rank=1 ) and NCT04263675 (2020-02-07, https://clinicaltrials.gov/study/NCT04263675?term=NCT04263675&rank=1 ).
Collapse
Affiliation(s)
- Isabelle Petitclerc
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Perron
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Camille Dugas
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Thomas Mayer
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Frédéric Raymond
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Vincenzo Di Marzo
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Quebec City, QC, G1V 4G5, Canada
| | - Alain Veilleux
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Robitaille
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada.
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada.
| |
Collapse
|
48
|
Boicean A, Ichim C, Sasu SM, Todor SB. Key Insights into Gut Alterations in Metabolic Syndrome. J Clin Med 2025; 14:2678. [PMID: 40283508 PMCID: PMC12028006 DOI: 10.3390/jcm14082678] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Over time, extensive research has underscored the pivotal role of gut microbiota in the onset and progression of various diseases, with a particular focus on fecal microbiota transplantation (FMT) as a potential therapeutic approach. The practice of transferring fecal matter from a healthy donor to a patient provides valuable insights into how alterations in gut microbiota can impact disease development and how rectifying dysbiosis may offer therapeutic benefits. Re-establishing a balanced symbiotic relationship in the gastrointestinal tract has shown positive results in managing both intestinal and systemic conditions. Currently, one of the most pressing global health issues is metabolic syndrome-a cluster of conditions that includes insulin resistance, lipid imbalances, central obesity and hypertension. In this context, FMT has emerged as a promising strategy for addressing key components of metabolic syndrome, such as improving insulin sensitivity, body weight and lipid profiles. However, further well-structured studies are needed to refine treatment protocols and establish the long-term safety and efficacy of this intervention.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | - Sabina-Maria Sasu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.)
| | | |
Collapse
|
49
|
Poulsen CE, Vinding R, Rasmussen MA, Shah S, Trivedi U, Rodriguez CL, Widdowson ML, Jiang J, Poulsen CS, Eliasen A, Chawes B, Bønnelykke K, Hansen CHF, Sørensen SJ, Thorsen J, Stokholm J. No association between the early-life gut microbiota and childhood body mass index and body composition. MED 2025; 6:100538. [PMID: 39536756 DOI: 10.1016/j.medj.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The gut microbiota has been implicated in adult obesity, but the causality is still unclear. It has been hypothesized that an obesity-prone gut microbiota can be established in infancy, but only few studies have examined the early-life gut microbiota in relation to obesity in childhood, and no consistent associations have been reported. Here, we examine the association between the early-life gut microbiota and body mass index (BMI) development and body composition throughout childhood. METHODS Gut microbiota from stool were collected from 700 children in the Copenhagen Prospective Studies on Asthma in Childhood2010 (COPSAC2010) cohort at ages of 1 week, 1month, 1 year, 4 years, and 6 years and analyzed by 16S rRNA gene sequencing. Outcomes included BMI World Health Organization (WHO) Z scores (zBMI), overweight (zBMI > 1.04) and obesity (zBMI > 1.64) (0-10 years), and adiposity rebound and body composition from dual-energy X-ray absorptiometry at 6 years. FINDINGS The early-life gut microbiota diversity, overall composition, and individual taxon abundances in unsupervised and supervised models were not consistently associated with either current or later BMI Z scores, overweight, obesity, adiposity rebound, or body composition in childhood. CONCLUSIONS In a deeply characterized longitudinal birth cohort, we did not observe any consistent associations between the early-life gut microbiota and BMI or risk of obesity in later childhood. While this does not conclusively rule out a relationship, it suggests that if such associations exist, they may be more complex and potentially influenced by factors emerging later in life, including lifestyle changes. FUNDING COPSAC is funded by private and public research funds (all listed on www.copsac.com).
Collapse
Affiliation(s)
- Christina Egeø Poulsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Rebecca Vinding
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Morten A Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | - Shiraz Shah
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Urvish Trivedi
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Leal Rodriguez
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Michael L Widdowson
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jie Jiang
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Casper S Poulsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Eliasen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Camilla H F Hansen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Søren J Sørensen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| |
Collapse
|
50
|
Chang Z, Zhu Y, Wang P, Du L, Wu M, Wang X, Kong C, Huang D, Xie R, Ji G, Wang C, Cheng L, Yan X, Wei Q, Qin H. Multi-omic analyses of the development of obesity-related depression linked to the gut microbe Anaerotruncus colihominis and its metabolite glutamate. Sci Bull (Beijing) 2025:S2095-9273(25)00359-7. [PMID: 40274437 DOI: 10.1016/j.scib.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025]
Abstract
Emerging evidence implicates gut microbiota in the pathogenesis of obesity-related depression (OD); however, the underlying molecular mechanisms remain inadequately explored. This study compared the microbial and transcriptional profiles between patients with OD and healthy individuals. The results revealed an enrichment of Anaerotruncus colihominis (A. colihominis) and glutamate metabolism-related genes in the OD group. Fecal microbiota transplantation (FMT) from patients with OD induced weight gain, compromised barrier function, and intensified depression-like behaviors in high-fat diet (HFD) mice. Microbial analysis in the mice feces corroborated the clinical findings. Single-cell RNA sequencing highlighted the pivotal role of the Efnb2-Ephb2 interaction in cell communication among colon epithelial and hippocampal neuron subtypes in OD mice. Notably, A. colihominis correlated with glutamate levels in the OD mice and patients. It produced glutamate through a glutamic acid metabolism-related DNA sequence, verified in an engineered Escherichia coli MG1655 strain. Both A. colihominis and glutamate reduced barrier proteins in colon epithelial cells and modulated cognitive proteins in neurons. Finally, A. colihominis treatment induced the Efnb2-Ephb2 interaction, exacerbating depression-like behaviors in germ-free HFD mice. Collectively, these findings reveal that A. colihominis and glutamate are potential intervention targets for OD treatment.
Collapse
Affiliation(s)
- Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Yefei Zhu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225012, China
| | - Ping Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Lei Du
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Minkang Wu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xingchun Wang
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Cheng Kong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Dengfeng Huang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Ruting Xie
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Chao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China.
| | - Xuebing Yan
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining First People's Hospital, Affiliated Hospital of Xuzhou Medical University, Yangzhou University, Suining 221200, China.
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.
| | - Huanlong Qin
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.
| |
Collapse
|