1
|
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
|
2
|
Wang P, Wang R, Zhao W, Zhao Y, Wang D, Zhao S, Ge Z, Ma Y, Zhao X. Gut microbiota-derived 4-hydroxyphenylacetic acid from resveratrol supplementation prevents obesity through SIRT1 signaling activation. Gut Microbes 2025; 17:2446391. [PMID: 39725607 DOI: 10.1080/19490976.2024.2446391] [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: 08/30/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Resveratrol (RSV), a natural polyphenol, has been suggested to influence glucose and lipid metabolism. However, the underlying molecular mechanism of its action remains largely unknown due to its multiple biological targets and low bioavailability. In this study, we demonstrate that RSV supplementation ameliorates high-fat-diet (HFD)-induced gut microbiota dysbiosis, enhancing the abundance of anti-obesity bacterial strains such as Akkermansia, Bacteroides and Blautia. The critical role of gut microbiota in RSV-mediated anti-obesity effects was confirmed through antibiotic-induced microbiome depletion and fecal microbiota transplantation (FMT), which showed that RSV treatment effectively mitigates body weight, histopathological damage, glucose dysregulation and systematic inflammation associated with HFD. Metabolomics analysis revealed that RSV supplementation significantly increases the levels of the gut microbial flavonoid catabolite 4-hydroxyphenylacetic acid (4-HPA). Notably, 4-HPA was sufficient to reverse obesity and glucose intolerance in HFD-fed mice. Mechanistically,4-HPA treatment markedly regulates SIRT1 signaling pathways and induces the expression of beige fat and thermogenesis-specific markers in white adipose tissue (WAT). These beneficial effects of 4-HPA are partially abolished by EX527, a known SIRT1 inhibitor. Collectively, our findings indicate that RSV improve obesity through a gut microbiota-derived 4-HPA-SIRT1 axis, highlighting gut microbiota metabolites as a promising target for obesity prevention.
Collapse
Affiliation(s)
- Pan Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ruiqi Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Wenting Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Yuanyuan Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Dan Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Shuang Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zhiwen Ge
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Yue Ma
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Xiaoyan Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
3
|
Liang M, Deng Z, Wu W, Dong Q, Fan J. Study on the correlation between intestinal flora and cytokines in children with Henoch-Schönlein purpura. Cytokine 2025; 191:156959. [PMID: 40373421 DOI: 10.1016/j.cyto.2025.156959] [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: 12/29/2024] [Revised: 04/20/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The pathogenesis of Henoch-Schönlein purpura (HSP) is complex. It is currently believed that the development of HSP involves abnormalities in humoral immunity and cellular immunity. The intestinal microbiota has a powerful regulatory effect on the human immune system and has been shown to serve a significant role in the pathogenesis of various immune-mediated disorders. This study examines changes in intestinal flora and cytokines(IFN-γ, IL-4, IL-10, and IL-17) in children with HSP and explores their correlation, offering fresh insights for the prevention and treatment of HSP. METHODS Blood and stool specimens were collected from 25 healthy children (control group) and 27 children with HSP (observation group). Enzyme-linked immunosorbent assay(ELISA) was used to detect the levels of cytokines IFN-γ, IL-4, IL-17, and IL-10 in the serum of all the study participants, and the 16S rRNA gene sequencing combined with high-throughput sequencing technology was used to analyze the intestinal flora of the study subjects. Finally, the correlation between serum cytokines and gut microbiota was analyzed in the children with HSP. RESULT 1)The serum levels of IL-4 and IL-17 in the observation group were higher than those in the control group, while the levels of IFN-γ and IL-10 were lower than those in the control group. 2) At the level of phylum, the abundance of Fusobacteria and Verrucomicrobia was higher than that in the control group, while the abundance of Firmicutes was lower than that of the control group, and the differences were statistically significant (P < 0.05); At the level of genus, the abundance of Prevotella and Akkermansia were higher than the control group, while the abundance of Bifidobacterium, Blautia, and Clostridium XlVa was lower than that in the control group, and the differences were all statistically significant (P < 0.05); At the species level, the abundance of Akkermansia muciniphila, Prevotella copri, and Subdoligranulum variabile was higher than that of the control group, while the abundance of Bifidobacterium pseudolongum, Brautella Weiss, and Bacteroides fragilis was lower than that in the control group, and the differences were statistically significant (P < 0.05). 3) The abundance of Blautia and Blautia wexlerae in the observation group was positively associated with the IL-10 level (r = 0.522, r = 0.578, P < 0.01). CONCLUSION Disturbances in intestinal flora and changes in serum cytokines IFN-γ, IL-4, IL-10, and IL-17 were present in children with HSP. The abundance of Blautia and Blautia wexlerae in the gut microbiota of children with HSP was positively correlated with serum IL-10 levels.
Collapse
Affiliation(s)
- Mingxin Liang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China; Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Zhaoxu Deng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Weiyi Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China; Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Qinqin Dong
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China.; Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Juan Fan
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China..
| |
Collapse
|
4
|
Huang Y, Xu W, Dong W, Chen G, Sun Y, Zeng X. Anti-diabetic effect of dicaffeoylquinic acids is associated with the modulation of gut microbiota and bile acid metabolism. J Adv Res 2025; 72:17-35. [PMID: 38969095 DOI: 10.1016/j.jare.2024.06.027] [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: 03/30/2024] [Revised: 06/06/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
INTRODUCTION The human gut microbiome plays a pivotal role in health and disease, notably through its interaction with bile acids (BAs). BAs, synthesized in the liver, undergo transformation by the gut microbiota upon excretion into the intestine, thus influencing host metabolism. However, the potential mechanisms of dicaffeoylquinic acids (DiCQAs) from Ilex kudingcha how to modulate lipid metabolism and inflammation via gut microbiota remain unclear. OBJECTIVES AND METHODS The objectives of the present study were to investigate the regulating effects of DiCQAs on diabetes and the potential mechanisms of action. Two mice models were utilized to investigate the anti-diabetic effects of DiCQAs. Additionally, analysis of gut microbiota structure and functions was conducted concurrently with the examination of DiCQAs' impact on gut microbiota carrying the bile salt hydrolase (BSH) gene, as well as on the enterohepatic circulation of BAs and related signaling pathways. RESULTS Our findings demonstrated that DiCQAs alleviated diabetic symptoms by modulating gut microbiota carrying the BSH gene. This modulation enhanced intestinal barrier integrity, increased enterohepatic circulation of conjugated BAs, and inhibited the farnesoid X receptor-fibroblast growth factor 15 (FGF15) signaling axis in the ileum. Consequently, the protein expression of hepatic FGFR4 fibroblast growth factor receptor 4 (FGFR4) decreased, accompanied by heightened BA synthesis, reduced hepatic BA stasis, and lowered levels of hepatic and plasma cholesterol. Furthermore, DiCQAs upregulated glucolipid metabolism-related proteins in the liver and muscle, including v-akt murine thymoma viral oncogene homolog (AKT)/glycogen synthase kinase 3-beta (GSK3β) and AMP-activated protein kinase (AMPK), thereby ameliorating hyperglycemia and mitigating inflammation through the down-regulation of the MAPK signaling pathway in the diabetic group. CONCLUSION Our study elucidated the anti-diabetic effects and mechanism of DiCQAs from I. kudingcha, highlighting the potential of targeting gut microbiota, particularly Acetatifactor sp011959105 and Acetatifactor muris carrying the BSH gene, as a therapeutic strategy to attenuate FXR-FGF15 signaling and ameliorate diabetes.
Collapse
Affiliation(s)
- Yujie Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, China
| | - Weiqi Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Wei Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Guijie Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yi Sun
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
5
|
Luo F, Yang J, Song Z, Zhao Y, Wang P, Liu K, Mou X, Liu W, Li W. Renshen Zhuye decoction ameliorates high-fat diet-induced obesity and insulin resistance by modulating gut microbiota and metabolic homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156655. [PMID: 40120542 DOI: 10.1016/j.phymed.2025.156655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Obesity, characterized by excessive adipose tissue accumulation, has become a global health challenge with rapidly increasing prevalence. It contributes significantly to metabolic disorders including insulin resistance (IR). Renshen-zhuye decoction (RZD), a traditional Chinese medicine formula historically used for diabetes, shows potential for improving metabolic parameters, but its effects and mechanisms in obesity and insulin resistance remain unclear. PURPOSE This study aimed to evaluate the therapeutic benefits of RZD on obesity and insulin resistance, and to elucidate the underlying mechanisms through which it improves glucose and lipid metabolism. METHODS The role of RZD was evaluated in a high-fat diet (HFD) mouse model. The formula was characterized using UPLC-MS. Comprehensive analyses including histopathological staining, immunofluorescence, biochemical assays, 16S rRNA gene sequencing of gut microbiota, and non-targeted metabolomic analysis were performed. To validate the role of gut microbiota, we employed antibiotic treatment (ABX) to deplete intestinal flora and conducted fecal microbiota transplantation (FMT) experiments. RESULTS RZD treatment dose-dependently alleviated HFD-induced dyslipidemia and insulin resistance, improving glucose tolerance, insulin sensitivity, and energy expenditure. Gut microbiota analysis revealed that RZD significantly modulated the composition of intestinal flora and their metabolic profiles. Additionally, RZD reduced intestinal and systemic inflammation by enhancing intestinal barrier integrity, particularly through increased expression of tight junction proteins such as Occludin. Importantly, the beneficial effects of RZD on weight management and glucose homeostasis were antagonized by antibiotic intervention, while FMT experiments confirmed that these improvements were mediated through gut microbiota modulation. CONCLUSION This study provides new insights into RZD's modulatory effects on gut microbiota and subsequent improvements in obesity-related metabolic parameters. RZD alleviates HFD-induced obesity and insulin resistance in mice by modulating gut microbiota composition and function, which subsequently improves intestinal barrier integrity, reduces inflammation, and enhances metabolic homeostasis.
Collapse
Affiliation(s)
- Fei Luo
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jie Yang
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiping Song
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Yuan Zhao
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Panpan Wang
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou 310000, PR China
| | - Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China
| | - Xin Mou
- Department of Endocrinology, Zhejiang Integrated Traditional Chinese and Western Medicine Hospital, Hangzhou, 310000, PR China.
| | - Wenhong Liu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| | - Wei Li
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, PR China.
| |
Collapse
|
6
|
Lepáček M, Boďo P, Prnová MŠ, Bučková M, Pangallo D, Pavlović J. Impact of novel aldose reductase inhibitor drug on gut microbiota composition and metabolic health in ZDF 'lean' rats. Chem Biol Interact 2025; 413:111490. [PMID: 40139546 DOI: 10.1016/j.cbi.2025.111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
A novel multi-target drug, cemtirestat, inhibiting aldose reductase (ALR2) has been developed to prevent secondary diabetic complications and act as an antioxidant against hyperglycemia-related processes. This study examines cemtirestat's impact on gut microbiome composition, drug metabolism, and therapeutic efficacy in male Zucker diabetic fatty (ZDF) "Lean" rats. Rats were divided into the control group (C) and the treated group (T), which received 7.7 mg/kg/day cemtirestat for two months, with weekly monitoring of food, fluid intake, and weight gain. Stool, urine, and plasma samples were analyzed biochemically, and fecal DNA was sequenced using Oxford Nanopore Technology. Treated rats exhibited less weight gain, likely due to cemtirestat's antioxidant effects. Biochemical analyses revealed no significant changes in glucose, liver enzymes, or cholesterol. Although there was a slight increase in alanine aminotransferase (ALT), our study found that levels of other liver enzymes such as aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bilirubin remained within normal limits, suggesting the observed increase in ALT was not indicative of drug-induced liver injury. LefSe microbiome analysis revealed an enrichment of beneficial bacteria like Blautia and Faecalibacterium in treated rats. Microbial community structure did not distinctly separate treated from control groups, but differences emerged over time. DeSeq2 analysis identified varying genera abundances over weeks, with treated samples enriched in beneficial bacteria by Week 8. Correlation analysis linked plasma insulin levels positively with Prevotella and negatively with Clostridium and Lactobacillus. Cemtirestat's impact on weight and microbiota suggests the potential to improve gut health. Further research is required to uncover cemtirestat's mechanism in diabetes management, drug metabolism, and therapeutic efficacy.
Collapse
Affiliation(s)
- Marek Lepáček
- Center of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04, Bratislava, Slovakia
| | - Pavol Boďo
- Center of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04, Bratislava, Slovakia
| | - Marta Šoltésová Prnová
- Center of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 841 04, Bratislava, Slovakia
| | - Mária Bučková
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 841 04, Bratislava, Slovakia
| | - Domenico Pangallo
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 841 04, Bratislava, Slovakia
| | - Jelena Pavlović
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská cesta 21, 841 04, Bratislava, Slovakia.
| |
Collapse
|
7
|
Dörr AK, Imangaliyev S, Karadeniz U, Schmidt T, Meyer F, Kraiselburd I. Distinguishing critical microbial community shifts from normal temporal variability in human and environmental ecosystems. Sci Rep 2025; 15:16934. [PMID: 40374711 DOI: 10.1038/s41598-025-01781-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025] Open
Abstract
Differentiating significant microbial community changes from normal fluctuations is vital for understanding microbial dynamics in human and environmental ecosystems. This knowledge could enable early warning systems to monitor critical changes affecting human or environmental health. We applied 16S rRNA gene sequencing and time-series analysis to model bacterial abundance trajectories in human gut and wastewater microbiomes. We evaluated various model architectures using datasets from two human studies and five wastewater settings. Long short-term memory (LSTM) models consistently outperformed other models in predicting bacterial abundances and detecting outliers, as measured by multiple metrics. Prediction intervals for each genus allowed us to identify significant changes and signaling shifts in community states. This study proposes a machine learning model capable of monitoring microbial communities and providing insights into their responses to internal and external factors in medical and environmental settings.
Collapse
Affiliation(s)
- Ann-Kathrin Dörr
- Department of Medicine, Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Computer Science, University of Duisburg-Essen, Essen, Germany
| | - Sultan Imangaliyev
- Department of Medicine, Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Utku Karadeniz
- Department of Computer Science, University of Duisburg-Essen, Essen, Germany
| | - Tina Schmidt
- Emschergenossenschaft/Lippeverband, Kronprinzenstraße 24, 45128, Essen, Germany
| | - Folker Meyer
- Department of Medicine, Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Computer Science, University of Duisburg-Essen, Essen, Germany
| | - Ivana Kraiselburd
- Department of Medicine, Institute for Artificial Intelligence in Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
8
|
Yoshii K, Node E, Furuta M, Tojima Y, Matsunaga A, Adachi J, Takaai N, Morita M, Hosomi K, Kunisawa J. Establishment of enterotype-specific antibodies for various diagnostic systems. Sci Rep 2025; 15:16814. [PMID: 40368953 PMCID: PMC12078515 DOI: 10.1038/s41598-025-01144-6] [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: 11/26/2024] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
This study demonstrates that monoclonal antibodies can be developed to targeting specific gut bacteria prevalent in the Japanese population and the potential for creating a novel diagnostic system using these antibodies. In this study, we established specific antibodies against representative bacteria from the genera Bacteroides, Faecalibacterium, and Prevotella and showed that they could be detected using ELISA, flow cytometry, and western blot analysis. Furthermore, a technique to quantify target bacteria was developed by combining these antibodies in a sandwich ELISA, enabling the quantification of bacteria in human fecal samples. This technology serves as a foundational method for rapidly and easily measuring gut bacteria and is expected to evolve into a powerful tool for analyzing the impact of gut bacteria on health, as well as for personalized health management based on individual gut environments.
Collapse
Affiliation(s)
- Ken Yoshii
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Eri Node
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Mari Furuta
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Yoko Tojima
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Ayu Matsunaga
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
- Faculty of Agriculture, Department of Applied Biological Science, Takasaki University of Health and Welfare, 54 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, NIBN, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Narimi Takaai
- Laboratory of Proteomics for Drug Discovery, NIBN, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Makiko Morita
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
- Graduate School of Veterinary Science, Osaka Metropolitan University, 1-58 Rinku-oraikita, Izumisano, Osaka, 598-8531, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan.
- Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Science, Osaka University, 1-1 Machikaneyamacho, Toyonaka, Osaka, 560-0043, Japan.
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
- Research Organization for Nano and Life Innovation, Waseda University, 2-2 Wakamatsu, Shinjuku-ku, Tokyo, 162-8480, Japan.
| |
Collapse
|
9
|
Rampelli S, Pomstra D, Barone M, Fabbrini M, Turroni S, Candela M, Henry AG. Consumption of only wild foods induces large scale, partially persistent alterations to the gut microbiome. Sci Rep 2025; 15:16593. [PMID: 40360545 PMCID: PMC12075472 DOI: 10.1038/s41598-025-00319-5] [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: 11/29/2024] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The gut microbiome (GM) is implicated in human health and varies among lifestyles. So-called "traditional" diets have been suggested to promote health-associated taxa. However, most studies focused only on diets including domesticated foods. Historically, humans consumed only wild foods, which might uniquely shape GM composition. We explored the impact of a wild-food-only diet on GM, particularly whether it increases the presence of health-associated and/or "old friend" taxa, and if the alterations to GM are persistent or transient. One participant collected daily fecal samples and recorded daily food consumption over an eight-week period, the middle four weeks of which he consumed only wild foods. Samples were profiled by 16S rRNA sequencing, and oligotyping and network analysis were conducted to assess microbial co-occurrence patterns. A wild-food-only diet considerably alters the composition of the GM, and the magnitude of the changes is larger than that observed in other diet interventions. No new taxa, including "old friends" appeared; instead, the proportions of already-present taxa shifted. Network analysis revealed distinct microbial co-abundance groups restructuring across dietary phases. There is a clear successional shift from the pre-, during- and post-wild-food-only diet. This analysis highlighted structural and functional shifts in microbial interactions, underscoring diet's role in shaping the gut ecosystem.
Collapse
Affiliation(s)
- Simone Rampelli
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy.
| | - Diederik Pomstra
- Department of Archaeological Sciences, Faculty of Archaeology, Leiden University, Leiden, The Netherlands
| | - Monica Barone
- Department of Medical and Surgical Sciences (DiMeC), Microbiomics Unit, Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Marco Fabbrini
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
- Department of Medical and Surgical Sciences (DiMeC), Microbiomics Unit, Alma Mater Studiorum - University of Bologna, 40138, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Marco Candela
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, 40126, Bologna, Italy
| | - Amanda G Henry
- Department of Archaeological Sciences, Faculty of Archaeology, Leiden University, Leiden, The Netherlands.
- Naturalis Biodiversity Center, Leiden, The Netherlands.
| |
Collapse
|
10
|
Song CH, Kim N, Choi Y, Kim S, Kim KS, Park MH, Lee SH, Lee DH. Beneficial effect of consuming milk containing only A2 beta-casein on gut microbiota: A single-center, randomized, double-blind, cross-over study. PLoS One 2025; 20:e0323016. [PMID: 40338897 PMCID: PMC12061139 DOI: 10.1371/journal.pone.0323016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/01/2025] [Indexed: 05/10/2025] Open
Abstract
Cow milk contains essential nutrients, with β-casein existing in A1 and A2 forms. Studies suggest that A2 milk (containing only A2 β-casein) may offer gastrointestinal (GI) benefits compared to A1/A2 milk (containing both forms). This study investigated the effects of A2 milk consumption on the gut microbiota of South Korean cohort experiencing GI discomfort after consuming A1/A2 milk. Thirty-five participants with GI discomfort after milk consumption were included. Stool DNA was analyzed using 16S rRNA gene sequencing before and after consuming either A1/A2 or A2 milk. Beta diversity analysis using the generalized UniFrac distance method revealed a significant shift in gut microbiota composition after A2 milk consumption (p = 0.04), but no significant change after consuming A1/A2 milk. Significant differences in gut microbiota composition were found between A1/A2 and A2 milk drinkers after milk consumption (p = 0.031). Alpha diversity indices remained unchanged. Notable increases in beneficial microbes, including Bifidobacterium and Blautia, were observed after A2 milk intake. Linear discriminant analysis Effect Size (LEfSe) analysis identified significant enrichment of Actinobacteria, particularly Bifidobacterium longum and Blautia wexlerae, in the A2 group. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) analysis highlighted enriched transport systems related to energy, peptides, sugars, and raffinose family oligosaccharides in the A2 group. Spearman correlation showed significant associations between Bifidobacterium, Blautia, and enhanced transport systems exclusively in the A2 group. Two weeks of A2 milk consumption led to significant alterations in gut microbiota, promoting beneficial microbes and related functions. A2 milk could be a suitable alternative for subjects who experience milk-intake-related GI discomfort.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seulgi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyung Su Kim
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Min Hee Park
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Sang Hee Lee
- R&D Center, Seoul Dairy Cooperative, Ansan, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine and Liver Research institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Wang Z, Tian L, Jiang Y, Ning L, Zhu X, Chen X, Xuan B, Zhou Y, Ding J, Ma Y, Zhao Y, Huang X, Hu M, Fang JY, Shen N, Cao Z, Chen H, Wang X, Hong J. Synergistic role of gut-microbial L-ornithine in enhancing ustekinumab efficacy for Crohn's disease. Cell Metab 2025; 37:1089-1102.e7. [PMID: 39978335 DOI: 10.1016/j.cmet.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025]
Abstract
The role of the intestinal microbiome in Crohn's disease (CD) treatment remains poorly understood. This study investigates microbe-host interactions in CD patients undergoing ustekinumab (UST) therapy. Fecal metagenome, metabolome, and host transcriptome data from 85 CD patients were analyzed using multi-omics integration and mediation analysis. Our findings reveal significant microbiome-metabolite-host interactions. Specifically, Faecalibacterium prausnitzii was linked to altered L-ornithine biosynthesis, resulting in higher L-ornithine levels in patients before UST therapy. In vivo and in vitro studies demonstrated that microbiome-derived L-ornithine enhances UST treatment sensitivity in CD by disrupting the host IL-23 receptor signaling and inhibiting Th17 cell stabilization through the IL-12RB1/TYK2/STAT3 axis. L-ornithine significantly enhances the therapeutic efficacy of UST in CD patients, as demonstrated in a prospective clinical trial. These findings suggest that targeting specific microbe-host metabolic pathways may improve the efficacy of inflammatory bowel disease (IBD) treatments.
Collapse
Affiliation(s)
- Zhenyu Wang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Lijun Ning
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiaoqiang Zhu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xuejie Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Baoqin Xuan
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yilu Zhou
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jinmei Ding
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yanru Ma
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Ying Zhao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiaowen Huang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Muni Hu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Nan Shen
- Department of Infectious Disease, Shanghai Children's Medical Center, National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Dongfang Rd. 1678, Shanghai 200127, China
| | - Zhijun Cao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China.
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| |
Collapse
|
12
|
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
|
13
|
Li X, Song G, Cao Q, Mo Q, Fu Y, Chen J, Lukuyu B, Wang S, Ye H, Zhu Y, Muhammad KS, Yang L, Pan J, Wang W. Growth curve fitting and analysis of intestinal flora changes of the Magang Goose (Anser domesticus) during 1 to 70 days post hatch. Poult Sci 2025; 104:105267. [PMID: 40367566 DOI: 10.1016/j.psj.2025.105267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
Nonlinear models are frequently utilized to study the growth and development of livestock and poultry, and to investigate the dynamic relationship with the intestinal microbiota changes. In this study, a total of 180 Magang geese (1-day-old) were selected and randomly divided into 6 replicates with 30 geese in each replicate. The growth performance, organ development, and intestinal flora composition of geese aged 7, 14, 21, 28, 42, 50, 60, and 70 d were observed. A total of 3 nonlinear growth models were applied to fit the development curves, aiming to explore the ontogenic development of Magang geese and the dynamic changes in the intestinal flora. Our results demonstrated that the Gompertz model serves as the most suitable model for simulating the growth pattern of Magang geese (R2=0.996). Using this model, the weight of the inflection point in Magang geese was 3.470 kg, the age of the inflection point was 25.460 d, and the maximum daily gain was 0.061 kg. The development curves of the liver, kidney, and pancreas conform to the Logistic model (R2=0.901, 0.978, 0.971), while the intestinal development also followed this model. The bacteria involved in energy metabolism (Subdoligranulum, Bacteroides, Romboutsia) and the bacteria inhibiting the colonization of harmful bacteria (Blautia) in cecum changed rapidly from 7 to 14 d, and microbial community composition stabilized after 21 d. In conclusion, our findings indicated that the ontogenic pattern of the Magang goose conformed to the Gompertz growth curve. The period from 7 to 42 d represents the rapid growth phase for Magang geese, during which organ development occurs, and cecal microbiota composition becomes increasingly stable.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Guorong Song
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Qingyun Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Qianyuan Mo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Yang Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Jianying Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Ben Lukuyu
- International Livestock Research Institute, Nairobi, 00100, Kenya
| | - Shunxiang Wang
- Guangdong Haid Group Co., Limited, Guangzhou, 511400, PR China
| | - Hui Ye
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Yongwen Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | | | - Lin Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China
| | - Jie Pan
- Zhuhai Tianjiao Technology Co., Limited, Zhuhai, 519000, PR China.
| | - Wence Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510640, PR China.
| |
Collapse
|
14
|
Zhang S, Zhao R, Wang R, Lu Y, Xu M, Lin X, Lan R, Zhang S, Tang H, Fan Q, Yang J, Liu L, Xu J. Weissella viridescens Attenuates Hepatic Injury, Oxidative Stress, and Inflammation in a Rat Model of High-Fat Diet-Induced MASLD. Nutrients 2025; 17:1585. [PMID: 40362894 PMCID: PMC12073722 DOI: 10.3390/nu17091585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disorder globally. Probiotic supplementation has shown promise in its prevention and treatment. Although Weissella viridescens, a lactic acid bacterium with immunomodulatory effects, has antibacterial and anti-inflammatory activities, there is a lack of direct evidence for its role in alleviating MASLD. This study aimed to investigate the protective effects of W. viridescens strain Wv2365, isolated from healthy human feces, in a high-fat diet (HFD)-induced rat model of MASLD. Methods: Rats were randomly assigned to a normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with W. viridescens Wv2365 (Wv2365) groups. All groups were fed their respective diets for 8 weeks. During this period, the NC and HFD groups received a daily oral gavage of PBS, while the Wv2365 group received a daily oral gavage of Wv2365. Results: Wv2365 supplementation significantly reduced HFD-induced body weight gain, improved NAFLD activity scores, alleviated hepatic injury, and restored lipid metabolism. A liver transcriptomic analysis revealed the downregulation of inflammation-related pathways, along with decreased serum levels of TNF-α, IL-1β, IL-6, MCP-1, and LPS. Wv2365 also activated the Nrf2/HO-1 antioxidant pathway, enhanced hepatic antioxidant enzyme activities and reduced malondialdehyde levels. A gut microbiota analysis showed the enrichment of beneficial genera, including Butyricicoccus, Akkermansia, and Blautia. Serum metabolomic profiling revealed increased levels of metabolites including indole-3-propionic acid, indoleacrylic acid, and glycolithocholic acid. Conclusions: Wv2365 attenuates hepatic injury, oxidative stress, and inflammation in a rat model of high-fat-diet-induced MASLD, supporting its potential as a probiotic candidate for the modulation of MASLD.
Collapse
Affiliation(s)
- Shuwei Zhang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiqing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruoshi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yao Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mingchao Xu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang 050010, China
| | - Xiaoying Lin
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Suping Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Huijing Tang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qianhua Fan
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Liyun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Jianguo Xu
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| |
Collapse
|
15
|
Nohesara S, Mostafavi Abdolmaleky H, Pirani A, Pettinato G, Thiagalingam S. The Obesity-Epigenetics-Microbiome Axis: Strategies for Therapeutic Intervention. Nutrients 2025; 17:1564. [PMID: 40362873 PMCID: PMC12073275 DOI: 10.3390/nu17091564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Obesity (OB) has become a serious health issue owing to its ever-increasing prevalence over the past few decades due to its contribution to severe metabolic and inflammatory disorders such as cardiovascular disease, type 2 diabetes, and cancer. The unbalanced energy metabolism in OB is associated with substantial epigenetic changes mediated by the gut microbiome (GM) structure and composition alterations. Remarkably, experimental evidence also indicates that OB-induced epigenetic modifications in adipocytes can lead to cellular "memory" alterations, predisposing individuals to weight regain after caloric restriction and subsequently inducing inflammatory pathways in the liver. Various environmental factors, especially diet, play key roles in the progression or prevention of OB and OB-related disorders by modulating the GM structure and composition and affecting epigenetic mechanisms. Here, we will first focus on the key role of epigenetic aberrations in the development of OB. Then, we discuss the association between abnormal alterations in the composition of the microbiome and OB and the interplays between the microbiome and the epigenome in the development of OB. Finally, we review promising strategies, including prebiotics, probiotics, a methyl-rich diet, polyphenols, and herbal foods for the prevention and/or treatment of OB via modulating the GM and their metabolites influencing the epigenome.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Ahmad Pirani
- Mental Health Research Center, Psychosocial Health Research Institute, Iran University of Medical Sciences, Tehran 14535, Iran;
| | - Giuseppe Pettinato
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
16
|
Ma N, Li R, Zhang GF, Gao RH, Zhang DJ. Fermentation-enriched quinoa β-glucan ameliorates disturbed gut microbiota and metabolism in type 2 diabetes mellitus mice. Int J Biol Macromol 2025; 306:141666. [PMID: 40032090 DOI: 10.1016/j.ijbiomac.2025.141666] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Quinoa β-glucan (QBG) has shown potential benefits in treating type 2 diabetes mellitus (T2DM); however, comprehensive evaluations of its effects remain limited. This study investigates the impact of QBG-derived from hot water extraction (Q-) and microbial fermentation enrichment (Q+)-on serum glucose levels, lipid profiles, appetite-regulating hormones, fecal short-chain fatty acids (SCFAs), and gut microbiota composition and function in streptozotocin/high-fat diet (STZ/HFD)-induced T2DM mice. The results indicate that QBG treatment significantly reduced fasting blood glucose, insulin levels, triglycerides (TG) and total cholesterol (TC), while concurrently increasing high-density lipoprotein cholesterol (HDLC) levels. Additionally, liver and pancreatic function improved, as evidenced by decreased levels of malondialdehyde (MDA), aspartate transaminase (AST), and alanine transaminase (ALT). SCFA levels were significantly higher in QBG-treated groups compared to MC group. QBG treatment also reduced the abundance of Firmicutes and Patescibacteria, along with the Firmicutes/Bacteroidota ratio, while increasing levels of Bacteroidota and Actinobacteria. These findings suggest that QBG can regulate the dysbiosis of SCFAs production in T2DM mice and may indirectly modulate the secretion of appetite-regulating hormones by influencing gut microbiota composition. Furthermore, PICRUSt analysis revealed that QBG treatment, particularly Q + _H, could enhance disrupted metabolism and improve gut microbiota functions, helping restore normal physiological function.
Collapse
Affiliation(s)
- Nan Ma
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Heilongjiang Bayi Agricultural University Mudanjiang Institute of Food and Biotechnology, Mudanjiang 157000, China; Daqing Center of Inspection and Testing for Rural Affairs Agricultural Products and Processed Products, Ministry of Agriculture and Rural Affairs, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Rong Li
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Gui-Fang Zhang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ruo-Han Gao
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Dong-Jie Zhang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Daqing Center of Inspection and Testing for Rural Affairs Agricultural Products and Processed Products, Ministry of Agriculture and Rural Affairs, Heilongjiang Bayi Agricultural University, Daqing 163319, China; National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, China.
| |
Collapse
|
17
|
Khan I, Khan I, Xie P, Xiaohui Y, Lei S, Song T, Li Z, Xie X. Insights into the blood, gut, and oral microbiomes in Chinese patients with myocardial infarction: a case-control study. BMC Microbiol 2025; 25:226. [PMID: 40253360 PMCID: PMC12008866 DOI: 10.1186/s12866-025-03878-9] [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: 10/29/2024] [Accepted: 03/10/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that changes in the blood microbes might be associated with cardiovascular disease, especially myocardial infarction (MI). However, some researchers are questioning whether a true "blood microbiome" actually exists. They hypothesized that these microbes may translocate into the bloodstream from the gut or oral cavities. To test this hypothesis, we analyzed the microbial composition, diversity, and potential role in disease progression by comparing blood, gut, and oral microbiota profiles in a cohort of MI patients and healthy controls. METHODS In this study, 144 samples, including blood, fecal, and saliva, were collected from twenty-four myocardial infarction patients and twenty-four healthy controls. These samples were analyzed using 16 S rRNA sequencing to characterize the microbial profiles across the three distinct microbial compartments. Differential analyses were conducted to find key differential microbiota for MI. Spearman's rank correlation analysis was used to study the association between microbiota and clinical indicators. RESULTS Our findings revealed striking microbial shifts across blood, gut, and oral compartments in MI patients compared to healthy controls. In the blood, we observed significant enrichment of the phyla Armatimonadota and Caldatribacteriota, alongside the genera Bacillus, Pedobacter, and Odoribacter. The gut microbiota of MI patients showed a notable increase in the phyla Proteobacteria, Verrucomicrobiota, Cyanobacteria, Synergistota, and Crenarchaeota, as well as the genera Eubacterium_coprostanoligenes_group, Rothia, Akkermansia, Lachnospiraceae_ NK4A136_ group, and Eubacterium_ruminantium_group. Meanwhile, the oral microbiota of MI patients was uniquely enriched with the phylum Elusimicrobiota and the genera Streptococcus, Rothia, and Granulicatella. These distinct microbial signatures highlight compartment-specific alterations that may play a role in the pathophysiology of MI. Additionally, LEfSe analysis identified 64 distinct taxa that differed across the three compartments. Of these, eight taxa were unique to blood, eighteen to the gut, and thirty-eight to the oral microbiota, all of which demonstrated significant associations with clinical markers of MI. Functional pathways were predicted and analyzed via KEGG annotation, but no statistically significant differences were found between MI patients and healthy controls in any of the microbiome compartments. CONCLUSION This study demonstrates significant alterations in the blood, gut, and oral microbiome profiles of MI patients, identifying specific bacterial taxa strongly associated with key markers of myocardial infarction. The unique microbial patterns detected in the blood provide compelling evidence for the existence of a stable core blood microbiome, highlighting its importance as a key contributor to cardiovascular health and disease progression.
Collapse
Affiliation(s)
- Ikram Khan
- Department of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Imran Khan
- Department of Microecology, School of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Ping Xie
- Department of Cardiology, Gansu Province People's Hospital, Lanzhou, Gansu, China
| | - Yu Xiaohui
- Department of Gastroenterology, The 940 Hospital Joint Logistic Support Force of PLA, Lanzhou, Gansu, China
| | - Shengnan Lei
- School of Stomatology, Key Laboratory of Oral Disease, Northwest Minzu University, Lanzhou, Gansu, China
| | - Tianzhu Song
- School of Stomatology, Key Laboratory of Oral Disease, Northwest Minzu University, Lanzhou, Gansu, China
| | - Zhiqiang Li
- School of Stomatology, Key Laboratory of Oral Disease, Northwest Minzu University, Lanzhou, Gansu, China.
| | - Xiaodong Xie
- Department of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
18
|
Qi Y, Du S, Li W, Qiu X, Zhou F, Bai L, Zhang B, Mi Z, Qian W, Li L, Zhao X, Li Y. Sanye tablet regulates gut microbiota and bile acid metabolism to attenuate hepatic steatosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119514. [PMID: 39971018 DOI: 10.1016/j.jep.2025.119514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sanye Tablet (SYT), a patent traditional Chinese prescription, is commonly used in treating type 2 diabetes mellitus and hyperlipidemia. Both clinical and animal studies suggest that SYT effectively regulates lipid metabolism. However, its mode of action on hepatic steatosis has yet to be fully elucidated. AIM OF STUDY This study investigates the lipid-regulating effects and underlying mechanism of SYT in high-fat diet (HFD)-induced hepatic steatosis mice. MATERIAL AND METHODS The inhibitory effects of SYT on developing hepatic steatosis were investigated in HFD-fed C57BL/6N mice. Biochemical markers, including total cholesterol (TC) and triglycerides (TG), were measured using specific kits. Hepatic histological alterations were determined by Hematoxylin and Eosin (H&E) and Oil Red O staining. Hepatic, fecal, and systemic bile acids (BAs) profiles were detected by UPLC-MS. mRNA and protein levels of BAs synthesis-related enzymes and critical nodes of farnesoid X receptor (FXR)/fibroblast growth factor 15 (FGF15)/fibroblast growth factor receptor 4 (FGFR4) signaling were detected. Fecal microbial composition was analyzed by 16S rRNA gene sequencing and the antimicrobial activity of SYT was further evaluated in vitro. RESULTS SYT alleviated HFD-induced hepatic steatosis by decreasing TG and TC levels, relieving hepatocyte ballooning, and promoting hepatic BAs synthesis. Moreover, SYT significantly increased the levels of taurine-conjugated BAs in the liver and feces, which in turn inhibited the FXR/FGF15/FGFR4 signaling. Consequently, the hepatic BAs synthesis-related enzyme expression was promoted to reduce lipid accumulation. Notably, SYT remodeled the gut microbiota composition of HFD-fed mice, especially inhibiting the growth of bile salt hydrolase (BSH)-producing bacteria, such as Lactobacillus murinus, Lactobacillus johnsonii, and Enterococcus faecalis. CONCLUSION The findings illustrated that SYT prevented hepatic steatosis by improving hepatic lipid accumulation, which is reflected in modulating the gut-liver axis. SYT corrects BAs profile, restores perturbed FXR/FGF15/FGFR4 signaling and promotes hepatic BAs synthesis, which is associated with modulation on certain BSH-producing bacteria.
Collapse
Affiliation(s)
- Yulin Qi
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Siqi Du
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenwen Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xianzhe Qiu
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fengjie Zhou
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liding Bai
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Boli Zhang
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhuoxin Mi
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Weiqiang Qian
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xin Zhao
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yuhong Li
- Key Laboratory of Traditional Chinese Medical Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
19
|
Jiang Y, Zhou T, Zhang S, Leng J, Li L, Zhao W. β-Glucan-based superabsorbent hydrogel ameliorates obesity-associated metabolic disorders via delaying gastric emptying, improving intestinal barrier function, and modulating gut microbiota. Int J Biol Macromol 2025; 304:140846. [PMID: 39933677 DOI: 10.1016/j.ijbiomac.2025.140846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The global obesity epidemic and its associated metabolic syndrome highlight the urgent need for new weight-loss therapies that provide high efficacy and patient compliance. Herein, we propose a novel, noninvasive approach using an orally administered β-glucan-based superabsorbent hydrogel (βC-MA hydrogel) to improve obesity-associated metabolic disorders. Results demonstrated that βC-MA hydrogel functioned as a dynamic exoskeleton within the gastrointestinal tract, slowing gastric emptying and reducing the digestion and absorption of ingested food. Furthermore, βC-MA hydrogel alleviated hepatic lipid accumulation and prevented hepatic steatosis and fibrosis by regulating the expression levels of key genes involved in lipid metabolism, including Cd36, SREBP 1c, FAS, ACC1, Cpt1a, and HSL, thereby limiting the progression of nonalcoholic fatty liver disease. In addition, βC-MA hydrogel reduced intestinal inflammation by lowering tumor necrosis factor-α and interleukin-6 levels while enhancing gut barrier function through increased expression of claudin-1, ZO-1, and MUC2. Finally, βC-MA hydrogel, enriched with obesity-negative probiotics such as Akkermansia, norank_f__Muribaculaceae, and Faecalibaculum, promoted the production of short-chain fatty acids. Consequently, βC-MA hydrogel significantly reduced body weight and fat accumulation and improved blood glucose and lipid levels, with efficacy comparable to semaglutide therapy and superior to β-glucan and sodium carboxymethylcellulose interventions. Overall, these findings suggest that βC-MA hydrogel could serve as a promising next-generation ingestible medical device for alleviating diet-induced obesity and related metabolic disorders by modulating food digestion and absorption, improving intestinal inflammation and barrier function, and regulating gut microbiota composition.
Collapse
Affiliation(s)
- Yiming Jiang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Tingyi Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Shiqi Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Juncai Leng
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Li Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wei Zhao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
20
|
Schütz B, Krause FF, Taudte RV, Zaiss MM, Luu M, Visekruna A. Modulation of Host Immunity by Microbiome-Derived Indole-3-Propionic Acid and Other Bacterial Metabolites. Eur J Immunol 2025; 55:e202451594. [PMID: 40170399 PMCID: PMC11962249 DOI: 10.1002/eji.202451594] [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: 11/07/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/03/2025]
Abstract
In recent years, we have witnessed a rapidly growing interest in the intricate communications between intestinal microorganisms and the host immune system. Research on the human microbiome is evolving from merely descriptive and correlative studies to a deeper mechanistic understanding of the bidirectional interactions between gut microbiota and the mucosal immune system. Despite numerous challenges, it has become increasingly evident that an imbalance in gut microbiota composition, known as dysbiosis, is associated with the development and progression of various metabolic, immune, cancer, and neurodegenerative disorders. A growing body of evidence highlights the importance of small molecules produced by intestinal commensal bacteria, collectively referred to as gut microbial metabolites. These metabolites serve as crucial diffusible messengers, translating the microbial language to host cells. This review aims to explore the complex and not yet fully understood molecular mechanisms through which microbiota-derived metabolites influence the activity of the immune cells and shape immune reactions in the gut and other organs. Specifically, we will discuss recent research that reveals the close relationship between microbial indole-3-propionic acid (IPA) and mucosal immunity. Furthermore, we will emphasize the beneficial effects of IPA on intestinal inflammation and discuss its potential clinical implications.
Collapse
Affiliation(s)
- Burkhard Schütz
- Institute of Anatomy and Cell BiologyPhilipps‐University MarburgMarburgGermany
| | - Felix F. Krause
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| | - R. Verena Taudte
- Core Facility for MetabolomicsDepartment of MedicinePhilipps‐University MarburgMarburgGermany
| | - Mario M. Zaiss
- Department of Internal Medicine 3Rheumatology and ImmunologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
- Deutsches Zentrum Immuntherapie (DZI)Friedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU) and Universitätsklinikum ErlangenErlangenGermany
| | - Maik Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik IIUniversitätsklinikum WürzburgWürzburgGermany
| | - Alexander Visekruna
- Institute for Medical Microbiology and HygienePhilipps‐University MarburgMarburgGermany
| |
Collapse
|
21
|
Duan L, Zhang X, Wang D, Xin J, Jiang Y, Sun N, Chen B, Luo J, He Y, Pan K, Zeng Y, Jing B, Ni X, Liu H. Effect of Probiotic Product Containing Heyndrickxia coagulans TBC169 on Hyperuricemia in Rats. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10519-0. [PMID: 40126815 DOI: 10.1007/s12602-025-10519-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Hyperuricemia (HUA) is a metabolic disease characterized by elevated serum uric acid, which is closely related to the gut microbiota. Probiotics have great potential in improving HUA. The purpose of this study was to evaluate the effect and mechanism of probiotic product (SQK) containing Heyndrickxia coagulans TBC169 on HUA rats. Forty SD rats (6 weeks old, 200 ± 20 g) were randomly divided into four groups (Ctrl group, HUA group, SQK1 group, and SQK2 group) of 10 rats each. Rats were given potassium oxonate (100 mg potassium oxonate/100 g BW/day) for 12 weeks to establish HUA model and simultaneously administered with sterile saline (HUA group) or different dose of SQK (SQK1 group, 20.48 mg SQK/100 g BW/day; SQK2 group, 40.95 mg SQK/100 g BW/day) throughout the 12 weeks. The results showed that SQK could degrade uric acid precursors and inhibit the xanthine oxidase (XOD) activity in vitro. Oral supplementation of SQK can reverse the increase of serum uric acid, the increase of the liver and serum XOD activity, and the decrease of ABCG2 expression in the ileum induced by HUA. In addition, SQK could restore the changes in α and β diversity of the ileal microbiota and prevent the increase in pathogenic Helicobacter and Staphylococcus caused by HUA. 16S rRNA sequencing and correlation analysis showed that the chondroitin sulfate (CS) degradation pathway of the gut microbiota played a key role in the prevention of HUA in the SQK group. These findings suggest that SQK may improve HUA by reducing uric acid synthesis and increasing uric acid excretion and provide a basis for its development into a probiotic product to improve HUA.
Collapse
Affiliation(s)
- Lixiao Duan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xingting Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dandan Wang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jinge Xin
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Jiang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Benhao Chen
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jiuyang Luo
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuhao He
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.
| | - Hongfa Liu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Krueger ME, Boles JS, Simon ZD, Alvarez SD, McFarland NR, Okun MS, Zimmermann EM, Forsmark CE, Tansey MG. Comparative analysis of Parkinson's and inflammatory bowel disease gut microbiomes reveals shared butyrate-producing bacteria depletion. NPJ Parkinsons Dis 2025; 11:50. [PMID: 40108151 PMCID: PMC11923181 DOI: 10.1038/s41531-025-00894-4] [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: 05/25/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Epidemiological studies reveal that inflammatory bowel disease (IBD) is associated with an increased risk of Parkinson's disease (PD). Gut dysbiosis has been documented in both PD and IBD, however it is currently unknown whether gut dysbiosis underlies the epidemiological association between both diseases. To identify shared and distinct features of the PD and IBD microbiome, we recruited 54 PD, 26 IBD, and 16 healthy control individuals and performed the first joint analysis of gut metagenomes. Larger, publicly available PD and IBD metagenomic datasets were also analyzed to validate and extend our findings. Depletions in short-chain fatty acid (SCFA)-producing bacteria, including Roseburia intestinalis, Faecalibacterium prausnitzii, Anaerostipes hadrus, and Eubacterium rectale, as well depletion in SCFA-synthesis pathways were detected across PD and IBD datasets, suggesting that depletion of these microbes in IBD may influence the risk for PD development.
Collapse
Affiliation(s)
- Maeve E Krueger
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jake Sondag Boles
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zachary D Simon
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stephan D Alvarez
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Nikolaus R McFarland
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Michael S Okun
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher E Forsmark
- Department of Medicine, Division of Gastroenterology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
23
|
Nie LJ, Cheng Z, He YX, Yan QH, Sun YH, Yang XY, Tian J, Zhu PF, Yu JY, Zhou HP, Zhou XQ. Role of duodenal mucosal resurfacing in controlling diabetes in rats. World J Diabetes 2025; 16:102277. [PMID: 40093272 PMCID: PMC11885968 DOI: 10.4239/wjd.v16.i3.102277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 01/03/2025] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND The duodenum plays a significant role in metabolic regulation, and thickened mucous membranes are associated with insulin resistance. Duodenal mucosal resurfacing (DMR), a new-style endoscopic procedure using hydrothermal energy to ablate this thickened layer, shows promise for enhancing glucose and lipid metabolism in type 2 diabetes (T2D) patients. However, the mechanisms driving these improvements remain largely unexplored. AIM To investigate the mechanisms by which DMR improves metabolic disorders using a rat model. METHODS Rats with T2D underwent a revised DMR procedure via a gastric incision using a specialized catheter to abrade the duodenal mucosa. The duodenum was evaluated using histology, immunofluorescence, and western blotting. Serum assays measured glucose, lipid profiles, lipopolysaccharide, and intestinal hormones, while the gut microbiota and metabolomics profiles were analyzed through 16S rRNA gene sequencing and ultra performance liquid chromatography-mass spectrum/mass spectrum, severally. RESULTS DMR significantly improved glucose and lipid metabolic disorders in T2D rats. It increased the serum levels of cholecystokinin, gastric inhibitory peptide, and glucagon-like peptide 1, and reduced the length and depth of duodenal villi and crypts. DMR also enhanced the intestinal barrier integrity and reduced lipopolysaccharide translocation. Additionally, DMR modified the gut microbiome and metabolome, particularly affecting the Blautia genus. Correlation analysis revealed significant links between the gut microbiota, metabolites, and T2D phenotypes. CONCLUSION This study illustrates that DMR addresses metabolic dysfunctions in T2D through multifaceted mechanisms, highlighting the potential role of the Blautia genus on T2D pathogenesis and DMR's therapeutic impact.
Collapse
Affiliation(s)
- Li-Juan Nie
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhe Cheng
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yi-Xian He
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Qian-Hua Yan
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yao-Huan Sun
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Xin-Yi Yang
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Jie Tian
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Peng-Fei Zhu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Jiang-Yi Yu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Hui-Ping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Xi-Qiao Zhou
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
24
|
Caesar R. The impact of novel probiotics isolated from the human gut on the gut microbiota and health. Diabetes Obes Metab 2025; 27 Suppl 1:3-14. [PMID: 39726216 PMCID: PMC11894790 DOI: 10.1111/dom.16129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The gut microbiota plays a pivotal role in influencing the metabolism and immune responses of the body. A balanced microbial composition promotes metabolic health through various mechanisms, including the production of beneficial metabolites, which help regulate inflammation and support immune functions. In contrast, imbalance in the gut microbiota, known as dysbiosis, can disrupt metabolic processes and increase the risk of developing diseases, such as obesity, type 2 diabetes, and inflammatory disorders. The composition of the gut microbiota is dynamic and can be influenced by environmental factors such as diet, medication, and the consumption of live bacteria. Since the early 1900s, bacteria isolated from food and have been used as probiotics. However, the human gut also offers an enormous reservoir of bacterial strains, and recent advances in microbiota research have led to the discovery of strains with probiotic potentials. These strains, derived from a broad spectrum of microbial taxa, differ in their ecological properties and how they interact with their hosts. For most probiotics bacterial structural components and metabolites, such as short-chain fatty acids, contribute to the maintenance of metabolic and immunological homeostasis by regulating inflammation and reinforcing gut barrier integrity. Metabolites produced by probiotic strains can also be used for bacterial cross-feeding to promote a balanced microbiota. Despite the challenges related to safety, stability, and strain-specific properties, several newly identified strains offer great potential for personalized probiotic interventions, allowing for targeted health strategies.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
25
|
Du Z, Liu X, Xie Z, Wang Q, Lv Z, Li L, Wang H, Xue D, Zhang Y. The relationship between a high-fat diet, gut microbiome, and systemic chronic inflammation: insights from integrated multiomics analysis. Am J Clin Nutr 2025; 121:643-653. [PMID: 39746397 DOI: 10.1016/j.ajcnut.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/29/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND The detrimental effects of a high-fat diet (HFD) extend beyond metabolic consequences and include systemic chronic inflammation (SCI), immune dysregulation, and gut health disruption. OBJECTIVES In this study, we used Mendelian randomization (MR) to investigate the relationship between HFD, gut microbiota, and SCI. METHODS Genetic variants associated with dietary fat were utilized to explore causal relationships. Genome-wide association study data for the analyses of the gut microbiota, inflammatory cytokines, immune cell characteristics, and serum metabolites were obtained from European individuals. Mediation analysis was used to reveal potential mediating factors. The GMrepo database was used to analyze the bacterial composition in different groups. Transcriptomic and single-cell sequencing analyses explored inflammation and barrier function in colonic tissue. RESULTS HFD consumption was linked to changes in the abundance of 3 bacterial families and 11 bacterial genera. Combined with the GMrepo database, the increased abundance of the genus Lachnospiraceae_FCS020group and the decreased abundance of genus Bacteroides and genus Barnesiella are consistent with the MR results. Transcriptomic and single-cell sequencing analyses revealed intestinal inflammation and mucosal barrier dysfunction in HFD-fed mice. MR revealed a link between HFD consumption and increased levels of interleukin (IL)-18 [odds ratio (OR): 3.64, 95%CI: 1.24, 10.69, P = 0.02], MIG (OR = 3.14, 95%CI: 1.17, 8.47, P = 0.02), IL-13 [OR = 3.21, 95% confidence interval (CI): 1.08, -9.52, P = 0.04], and IL-2RA (OR = 2.93, 95%CI: 1.01, 8.53, P = 0.049). Twenty-nine immune cell signatures, including altered monocyte and T-cell subsets, were affected by HFD consumption. Twenty-six serum metabolites that are linked to HFD consumption, particularly lipid and amino acid metabolites, were identified. The positive gut microbiota exhibit extensive associations with inflammatory cytokines. In particular, Lachnospiraceae_FCS020 group (OR: 1.93, 95% CI: 1.11, 3.37, P = 0.02) may play a mediating role in HFD-induced increases in IL-2RA concentrations. CONCLUSIONS Microbial dysbiosis appears to be an important mechanism for HFD-induced SCI. The Lachnospiraceae_FCS020 group may act as a key genus in HFD-mediated elevation of IL-2RA.
Collapse
Affiliation(s)
- Zhiwei Du
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhihong Xie
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Zhenyi Lv
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lianghao Li
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heming Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yingmei Zhang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
26
|
Liu D, Wei M, Fang Y, Yuan T, Sun Y, Xie H, Yan W, Yuan B, Zhuang B, Jin Y. Oral colon-retentive inulin gels protect against radiation-induced hematopoietic and gastrointestinal injury by improving gut homeostasis. Int J Biol Macromol 2025; 292:139199. [PMID: 39730057 DOI: 10.1016/j.ijbiomac.2024.139199] [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: 08/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Ionizing radiation-induced injury often occurs in nuclear accidents or large-dose radiotherapy, leading to acute radiation syndromes characterized by hematopoietic and gastrointestinal injuries even to death. However, current radioprotective drugs are only used in hospitals with unavoidable side effects. Here, we heated the aqueous solution of inulin, a polysaccharide dietary fiber, forming colon-retentive gel as a radiation protector in radiotherapy. Mouse models were established after 60Co γ-ray irradiation of the total body or abdomen. Inulin gels were orally administered to the mice every day from 3 days pre-radiation to 3 days post-radiation. The hematopoietic system was well protected with good blood cell recovery and cell proliferation in the femur and spleen. Oral inulin gels increased the relative abundances of key commensal microorganisms including f_Lachnospiraceae, Akkermansia, Blautia, and short-chain fatty acid metabolites. The secretion of the anti-inflammation cytokines IL-22 and IL-10 in the intestinal cells also increased. Similarly, the expression of the tight junction proteins claudin-1 and occludin in the gut mucosa was affected. In an orthotopic murine colorectal cancer model, oral inulin gels followed by 10-Gy abdomen radiation improved the radiotherapy efficiency with low attenuated radiation injury. Taken the data together, these results suggest that oral inulin gels are a bioactive material against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yubao Fang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yingbao Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hua Xie
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Institute of NBC Defense, Beijing 102205, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
27
|
Romaní‐Pérez M, Líebana‐García R, Flor‐Duro A, Bonillo‐Jiménez D, Bullich‐Vilarrubias C, Olivares M, Sanz Y. Obesity and the gut microbiota: implications of neuroendocrine and immune signaling. FEBS J 2025; 292:1397-1420. [PMID: 39159270 PMCID: PMC11927058 DOI: 10.1111/febs.17249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Obesity is a major health challenge due to its high prevalence and associated comorbidities. The excessive intake of a diet rich in fat and sugars leads to a persistent imbalance between energy intake and energy expenditure, which increases adiposity. Here, we provide an update on relevant diet-microbe-host interactions contributing to or protecting from obesity. In particular, we focus on how unhealthy diets shape the gut microbiota and thus impact crucial intestinal neuroendocrine and immune system functions. We describe how these interactions promote dysfunction in gut-to-brain neuroendocrine pathways involved in food intake control and postprandial metabolism and elevate the intestinal proinflammatory tone, promoting obesity and metabolic complications. In addition, we provide examples of how this knowledge may inspire microbiome-based interventions, such as fecal microbiota transplants, probiotics, and biotherapeutics, to effectively combat obesity-related disorders. We also discuss the current limitations and gaps in knowledge of gut microbiota research in obesity.
Collapse
Affiliation(s)
- Marina Romaní‐Pérez
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Rebeca Líebana‐García
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Alejandra Flor‐Duro
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Daniel Bonillo‐Jiménez
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Clara Bullich‐Vilarrubias
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Marta Olivares
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| | - Yolanda Sanz
- Institute of Agrochemistry and Food TechnologySpanish National Research Council (IATA‐CSIC)ValenciaSpain
| |
Collapse
|
28
|
Guo X, Su Y, Du Y, Zhang F, Yu W, Ren W, Li S, Kuang H, Wu L. Vinegar-processed Schisandra chinensis polysaccharide ameliorates type 2 diabetes via modulation serum metabolic profiles, gut microbiota, and fecal SCFAs. Int J Biol Macromol 2025; 294:139514. [PMID: 39761882 DOI: 10.1016/j.ijbiomac.2025.139514] [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: 07/03/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/21/2025]
Abstract
Numerous studies indicate that Schisandra chinensis (Turcz.) Baill (SC) has anti-type 2 diabetes mellitus (T2DM) effects, and its processed products are commonly used in clinical practice. However, limited reports exist on the mechanisms of polysaccharides from its vinegar products and their role in T2DM. We purified a novel polysaccharide from vinegar-processed Schisandra chinensis (VSC) and used intestinal microbiota 16S rRNA analysis and metabolomics to study changes in T2DM mice after vinegar-processed Schisandra chinensis polysaccharide (VSP) intervention, aiming to elucidate how VSP alleviates T2DM. VSP has shown significant therapeutic effects in T2DM mice, which can regulate the imbalance of glucose and lipid metabolism, alleviate pancreatic and liver damage, restore the integrity of the intestinal barrier, and inhibit the inflammatory response. Serum metabolomics and microbiological analysis showed that VSP could significantly regulate 104 endogenous metabolites and rectify gut microbiota disorders in T2DM mice. Additionally, VSP enhanced the levels of short-chain fatty acids (SCFAs) and the expression of GPR41/43 in the colon of T2DM mice. Correlation analysis revealed significant correlations among specific gut microbiota, serum metabolites, and fecal SCFAs. Overall, these findings will provide a basis for further VSP development.
Collapse
Affiliation(s)
- Xingyu Guo
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Yang Su
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Yongqiang Du
- Heilongjiang Province Healthcare Security Administration, Harbin 150036, China
| | - Fan Zhang
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Wenting Yu
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Wenchen Ren
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Shanshan Li
- Heilongjiang Province Health Management Service Evaluation Center, Harbin 150030, China
| | - Haixue Kuang
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Lun Wu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
29
|
Chang C, Gu Z, Du L, Guo J, Yang Y, Wu Z. Effects of L-β-Galactoglucan Supplementation on Growth Performance, Palatability, and Intestinal Microbiota in Adult Beagle Dogs. Metabolites 2025; 15:160. [PMID: 40137125 PMCID: PMC11944019 DOI: 10.3390/metabo15030160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Background: This study was conducted to investigate the effects of different levels of L-β-galactoglucan on growth performance, palatability, and health condition of dogs. Methods: A total of 32 healthy beagle dogs (2.0 ± 0.5 yr; 13.2 ± 2.1 kg) were randomly assigned into four treatment groups, with 8 dogs in each group. The dogs were fed basal diets supplemented with 0 (control), 0.25, 0.5, or 1% L-β-galactoglucan. Results: The results showed that the feed intake ratio of the dogs in the Low_Gal (0.25%) group was significantly higher (p < 0.05) as compared with the control (Con) group. The low-density lipoprotein cholesterol (LDL-C) levels of the Mid_Gal (0.5%) group showed a trend toward lower levels as compared with the control (Con) group (p = 0.069). Compared with the control (Con) group, the alpha diversity of the bacterial flora of the Shannon index of the Mid_Gal (0.5%) group was significantly higher (p < 0.05). The Simpson index was significantly reduced (p < 0.05), and a PCoA indicated a significant change in the gut microbiota structure among the four groups (p < 0.05). The relative abundance of Blautia and Peptoclostridium in the Low_Gal (0.25%) group was significantly higher as compared with the control (Con) group (p < 0.05). Conclusions: These results indicated that L-β-galactoglucan exhibited a positive effect on improving the palatability and gut microbiota of dogs.
Collapse
Affiliation(s)
- Chenghe Chang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (C.C.); (Z.G.); (Z.W.)
| | - Zifeng Gu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (C.C.); (Z.G.); (Z.W.)
| | - Lingling Du
- Chengdu Sydix Biotech Co., Ltd., Chengdu 610000, China;
| | - Jiantao Guo
- Beijing Shanchongshuifu Technology Development Co., Beijing 100084, China;
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (C.C.); (Z.G.); (Z.W.)
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (C.C.); (Z.G.); (Z.W.)
| |
Collapse
|
30
|
Zhou L, Chen SZ, Li YY, Xue RY, Duan X, Lin XY, Chen S, Zhou D, Li HB. Gut Dysbiosis Exacerbates Intestinal Absorption of Cadmium and Arsenic from Cocontaminated Rice in Mice Due to Impaired Intestinal Barrier Functions. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:3459-3471. [PMID: 39945512 DOI: 10.1021/acs.est.5c00817] [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: 02/26/2025]
Abstract
Globally, humans face gut microbiota dysbiosis; however, its impact on the bioavailability of cadmium (Cd) and arsenic (As) from rice consumption─a major source of human exposure to these metals─remains unclear. In this study, we compared Cd and As accumulation in the liver and kidneys of mice with disrupted gut microbiota (administered cefoperazone sodium), restored microbiota (administered probiotics and prebiotics following antibiotic exposure), and normal microbiota, all after consuming cocontaminated rice. Compared to normal mice, microbiota-disrupted mice exhibited 30.9-119% and 30.0-100% (p < 0.05) higher Cd and As levels in tissues after a 3 week exposure period. The increased Cd and As bioavailability was not due to changes in the duodenal expression of Cd-related transporters or As speciation biotransformation in the intestine. Instead, it was primarily attributed to a damaged mucus layer and depleted tight junctions associated with gut dysbiosis, which increased intestinal permeability. These mechanisms were confirmed by observing 34.3-74.3% and 25.0-75.0% (p < 0.05) lower Cd and As levels in the tissues of microbiota-restored mice with rebuilt intestinal barrier functions. This study enhances our understanding of the increased risk of dietary metal(loid) exposure in individuals with gut microbiota dysbiosis due to impaired intestinal barrier functions.
Collapse
Affiliation(s)
- Lei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Sheng-Zhi Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Yuan-Yuan Li
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rong-Yue Xue
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Xu Duan
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Xin-Ying Lin
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Shan Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Hong-Bo Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|
31
|
Yin Y, Nie W, Tang ZQ, Zhu SJ. Flavonoid-Rich Extracts from Chuju ( Asteraceae Chrysanthemum L.) Alleviate the Disturbance of Glycolipid Metabolism on Type 2 Diabetic Mice via Modulating the Gut Microbiota. Foods 2025; 14:765. [PMID: 40077469 PMCID: PMC11898795 DOI: 10.3390/foods14050765] [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: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) and its associated complications represent a significant public health issue affecting hundreds of millions of people globally; thus, measures to prevent T2DM are urgently needed. Chuju has been proven to possess antihyperglycemic activity. However, the bioactive ingredients in chuju that contribute to its antihyperglycemic activity, as well as the relationship between its antihyperglycemic activity and the gut microbiota, remain unclear. To understand the potential effects that it has on T2DM, the glycolipid metabolism and gut microbiota regulation of flavonoid-rich extracts from chuju (CJE) were investigated. The results showed that the top ten flavonoid compounds in CJE are Apigenin 6, 8-digalactoside, Apigenin 6-C-glucoside 8-C-arabinoside, Luteolin-4'-O-glucoside, Isoshaftoside, Scutellarin, Quercetin 3-O-malonylglucoside, Chrysoeriol 7-O-glucoside, Quercetin-3,4'-O-di-beta-glucoside, Luteolin 6-C-glucoside 8-C-arabinoside, and Homoorientin. Furthermore, CJE mitigated hyperglycemia and glycolipid metabolism by reducing the abundance of Faecalibaculum, Coriobacteriaceae, and Romboutsia and increasing the abundance of Alistipes. In addition, the results of Western blot analysis showed that CJE could enhance glycogen synthesis and glucose transport by up-regulating the phosphorylation of IRS1-PI3K-Akt and AMPK-GLUT4. Simultaneously, CJE could decrease gluconeogenesis by down-regulating the phosphorylation of FoxO1/GSK 3β. In conclusion, the findings of this study provide new evidence supporting the hypothesis that CJE can be used as part of a therapeutic approach for treating disturbances in glycolipid metabolism via regulating the gut microbiota and mediating the IRS1-PI3K-Akt-FoxO1/GSK 3β and AMPK-GLUT4 pathways.
Collapse
Affiliation(s)
- Yu Yin
- School of Life Sciences, Anhui University, Hefei 230601, China;
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| | - Wen Nie
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| | - Zheng-Quan Tang
- School of Life Sciences, Anhui University, Hefei 230601, China;
| | - Shuang-Jie Zhu
- School of Life Sciences, Anhui University, Hefei 230601, China;
- School of Biological and Food Engineering, Chuzhou University, Chuzhou 239001, China;
| |
Collapse
|
32
|
Tian Z, Zhang J, Fan Y, Sun X, Wang D, Liu X, Lu G, Wang H. Diabetic peripheral neuropathy detection of type 2 diabetes using machine learning from TCM features: a cross-sectional study. BMC Med Inform Decis Mak 2025; 25:90. [PMID: 39966886 PMCID: PMC11837659 DOI: 10.1186/s12911-025-02932-w] [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: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
AIMS Diabetic peripheral neuropathy (DPN) is the most common complication of diabetes mellitus. Early identification of individuals at high risk of DPN is essential for successful early intervention. Traditional Chinese medicine (TCM) tongue diagnosis, one of the four diagnostic methods, lacks specific algorithms for TCM symptoms and tongue features. This study aims to develop machine learning (ML) models based on TCM to predict the risk of diabetic peripheral neuropathy (DPN) in patients with type 2 diabetes mellitus (T2DM). METHODS A total of 4723 patients were included in the analysis (4430 with T2DM and 293 with DPN). TFDA-1 was used to obtain tongue images during a questionnaire survey. LASSO (least absolute shrinkage and selection operator) logistic regression model with fivefold cross-validation was used to select imaging features, which were then screened using best subset selection. The synthetic minority oversampling technique (SMOTE) algorithm was applied to address the class imbalance and eliminate possible bias. The area under the receiver operating characteristic curve (AUC) was used to evaluate the model's performance. Four ML algorithms, namely logistic regression (LR), random forest (RF), support vector classifier (SVC), and light gradient boosting machine (LGBM), were used to build predictive models for DPN. The importance of covariates in DPN was ranked using classifiers with better performance. RESULTS The RF model performed the best, with an accuracy of 0.767, precision of 0.718, recall of 0.874, F-1 score of 0.789, and AUC of 0.77. With a value of 0.879, the LGBM model appeared to be the best regarding recall Age, sweating, dark red tongue, insomnia, and smoking were the five most significant RF features. Age, yellow coating, loose teeth, smoking, and insomnia were the five most significant features of the LGBM model. CONCLUSIONS This cross-sectional study demonstrates that the RF and LGBM models can screen for high-risk DPN in T2DM patients using TCM symptoms and tongue features. The identified key TCM-related features, such as age, tongue coating, and other symptoms, may be advantageous in developing preventative measures for T2DM patients.
Collapse
Affiliation(s)
- Zhikui Tian
- School of Rehabilitation Medicine, Qilu Medical University, Shandong, 255300, China
| | - JiZhong Zhang
- School of Rehabilitation Medicine, Qilu Medical University, Shandong, 255300, China
| | - Yadong Fan
- Medical College of Yangzhou University, YangZhou, 225000, China
| | - Xuan Sun
- College of Traditional Chinese Medicine, Binzhou Medical University, Shandong, China
| | - Dongjun Wang
- College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, 063000, China
| | - XiaoFei Liu
- School of Rehabilitation Medicine, Qilu Medical University, Shandong, 255300, China
| | - GuoHui Lu
- School of Rehabilitation Medicine, Qilu Medical University, Shandong, 255300, China.
| | - Hongwu Wang
- School of Health Sciences and Engineering, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
33
|
Jia J, Guo J, Yan C, Gu Y, Xia X. Oyster powder supplementation enhances immune function in mice partly through modulating the gut microbiota and arginine metabolism. Food Funct 2025; 16:1254-1266. [PMID: 39868593 DOI: 10.1039/d4fo06068g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oysters are well-known for their health benefits such as immuno-modulatory functions. The intestinal microbiome serves as a key mediator between diet and immune regulation. This study aimed to investigate whether oyster consumption could alleviate cyclophosphamide (Cy)-induced immunosuppression by promoting intestinal homeostasis. In mice treated with Cy, a significant decrease in immune cells and cytokines was observed. In contrast, mice supplemented with oyster powder demonstrated elevated numbers of immune cells in the spleen and small intestine, as well as enhanced serum production of IL-1β, IL-2, TNF-α, and IFN-γ. Furthermore, oyster consumption improved the composition of the gut microbiota by promoting beneficial bacteria and inhibiting harmful ones. Metabolomics analysis revealed that oyster powder treatment significantly enhanced the arginine biosynthesis pathway, and further analysis found that the consumption of oysters led to increased arginine levels. Correlation analysis showed a significant positive correlation between L-arginine and immune-related markers. Collectively, these findings suggest that oyster consumption may enhance immunity by modulating the gut microbiota and boosting arginine biosynthesis pathways. Dietary oyster consumption could be an effective strategy to support immune health.
Collapse
Affiliation(s)
- Jinhui Jia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Jian Guo
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Yunqi Gu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China
| |
Collapse
|
34
|
Tang SS, Zhao XF, An XD, Sun WJ, Kang XM, Sun YT, Jiang LL, Gao Q, Li ZH, Ji HY, Lian FM. Classification and identification of risk factors for type 2 diabetes. World J Diabetes 2025; 16:100371. [PMID: 39959280 PMCID: PMC11718467 DOI: 10.4239/wjd.v16.i2.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/24/2024] [Accepted: 11/26/2024] [Indexed: 12/30/2024] Open
Abstract
The risk factors for type 2 diabetes mellitus (T2DM) have been increasingly researched, but the lack of systematic identification and categorization makes it difficult for clinicians to quickly and accurately access and understand all the risk factors, which are categorized in this paper into five categories: Social determinants, lifestyle, checkable/testable risk factors, history of illness and medication, and other factors, which are discussed in a narrative review. Meanwhile, this paper points out the problems of the current research, helps to improve the systematic categorisation and practicality of T2DM risk factors, and provides a professional research basis for clinical practice and industry decision-making.
Collapse
Affiliation(s)
- Shan-Shan Tang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Xue-Fei Zhao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Dong An
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Wen-Jie Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xiao-Min Kang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Yu-Ting Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Lin-Lin Jiang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Qing Gao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Ze-Hua Li
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Hang-Yu Ji
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Feng-Mei Lian
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| |
Collapse
|
35
|
Zhou X, Zhang Y, Wei L, Yang Y, Wang B, Liu C, Bai J, Wang C. In vitro fermentation characteristics of fucoidan and its regulatory effects on human gut microbiota and metabolites. Food Chem 2025; 465:141998. [PMID: 39549519 DOI: 10.1016/j.foodchem.2024.141998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Dietary polysaccharides affect the intestinal microorganisms and their metabolites in the host. Clarifying the relationship among polysaccharides, intestinal microflora, and their metabolites is helpful to formulate dietary nutrition intervention strategies. Thus, we explored the regulatory effects of fucoidan on the human gut microbiota and its metabolites. After 48 h of fermentation, fucoidan significantly reduced the pH value in the broth, accompanied by an increase in total short-chain fatty acids, acetic acid, and propanoic acid contents. Fucoidan significantly reduced the relative abundance of Escherichia_shigella and Blebsiella and increased the relative abundance of Bifidobacterium and Lactobacillus. Concurrently, fucoidan altered the composition of intestinal microbial metabolites. These results indicate that fucoidan can regulate the metabolism of the intestinal flora and host, which may contribute to the intestinal health of the host.
Collapse
Affiliation(s)
- Xu Zhou
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yuyan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Li Wei
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yuhan Yang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Botao Wang
- Bloomage Biotechnology CO, LTD, Jinan, Shandong 250000, China
| | - Cuiping Liu
- Department of Radiology, Yuxi Children's Hospital, Yuxi, Yunnan 653100, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400700, China.
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
36
|
Yao N, Kinouchi K, Katoh M, Ashtiani KC, Abdelkarim S, Morimoto H, Torimitsu T, Kozuma T, Iwahara A, Kosugi S, Komuro J, Kato K, Tonomura S, Nakamura T, Itoh A, Yamaguchi S, Yoshino J, Irie J, Hashimoto H, Yuasa S, Satoh A, Mikami Y, Uchida S, Ueki T, Nomura S, Baldi P, Hayashi K, Itoh H. Maternal circadian rhythms during pregnancy dictate metabolic plasticity in offspring. Cell Metab 2025; 37:395-412.e6. [PMID: 39814018 PMCID: PMC11872692 DOI: 10.1016/j.cmet.2024.12.002] [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: 08/07/2023] [Revised: 04/29/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Tissue-level oscillation is achieved by tissue-intrinsic clocks along with network-dependent signals originating from distal organs and organismal behavior. Yet, it remains unexplored whether maternal circadian rhythms during pregnancy influence fetal rhythms and impact long-term susceptibility to dietary challenges in offspring. Here, we demonstrate that circadian disruption during pregnancy decreased placental and neonatal weight yet retained transcriptional and structural maturation. Intriguingly, diet-induced obesity was exacerbated in parallel with arrhythmic feeding behavior, hypothalamic leptin resistance, and hepatic circadian reprogramming in offspring of chronodisrupted mothers. In utero circadian desynchrony altered the phase-relationship between the mother and fetus and impacted placental efficiency. Temporal feeding restriction in offspring failed to fully prevent obesity, whereas the circadian alignment of caloric restriction with the onset of the active phase virtually ameliorated the phenotype. Thus, maternal circadian rhythms during pregnancy confer adaptive properties to metabolic functions in offspring and provide insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Na Yao
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kenichiro Kinouchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Sherif Abdelkarim
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Hiroyuki Morimoto
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takuto Torimitsu
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahide Kozuma
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihide Iwahara
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Kosugi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Health Center, Keio University, Yokohama, Japan
| | - Jin Komuro
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Kyosuke Kato
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shun Tonomura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Nakamura
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Arata Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shintaro Yamaguchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Jun Yoshino
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Japan; The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Japan
| | - Junichiro Irie
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hisayuki Hashimoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan; Department of Cardiovascular Medicine, Academic Field, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akiko Satoh
- Department of Integrative Physiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Integrative Physiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shusaku Uchida
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takatoshi Ueki
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Pierre Baldi
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697, USA
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Center for Preventive Medicine, Keio University, Tokyo, Japan.
| |
Collapse
|
37
|
Zou W, Ma D, Sun F, Chen Z, Chen Y, Li X, Chen M, Lin M, Shi H, Wu B, Chen L, Liang Z, Liu J. Maternal OM-85 administration alleviates offspring allergic airway inflammation by downregulating IL-33/ILC2 axis. Pediatr Allergy Immunol 2025; 36:e70044. [PMID: 39927900 DOI: 10.1111/pai.70044] [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: 07/09/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Type 2 innate lymphoid cells (ILC2s) are essential for maintaining immune regulation and promoting tissue homeostasis in allergic asthma. How the development of gut microbiota on neonatal ILC2s influences allergic airway inflammation remains unclear. Here we focus on offspring ILC2 development in the context of alterations in maternal gut microbiota. METHODS C57BL/6 maternal mice were gavaged with OM-85 during pregnancy and/or lactation, ILC2-driven allergic airway inflammation in the OVA-sensitized adult offspring was observed. ILC2 development in offspring early life were investigated using recombinant (r)IL-33, rIL-25 and Bromodeoxyuridine in the vivo experiments. Further ILC2 promoting factors- IL-33 and IL-25 production in offspring early life were analysed. Finally, we examined the changes in gut microbiota and its metabolites in both dams and pups, and explored the effects of short-chain fatty acids (SCFAs) on IL-33 expression and secretion. RESULTS Maternal OM-85 administration restrained ILC2-driven allergic airway inflammation in the OVA-sensitized adult offspring. During ILC2 development in offspring early life, maternal OM-85 administration suppressed IL-33 and IL-25 production to inhibit ILC2 expansion and ILC2 responsiveness to alarmins, and infantile ILC2s could persist into adulthood. Maternal OM-85 administration increased SCFAs in breast milk and SCFA-producing gut probiotics (predominant Bacteroides and Blautia) in offspring, especially during pregnancy and lactation. SCFAs down-regulated IL-33 expression and reduced IL-33 secretion by inhibited gasdermin D (GSDMD) formation. CONCLUSION Maternal OM-85 administration restrains ILC2-driven allergic airway inflammation in adult offspring by increasing offspring intestinal SCFAs to modulate ILC2 development at an early stage, demonstrating that the transgenerational effects of maternal OM-85 exposure on offspring innate immunity.
Collapse
Affiliation(s)
- Wei Zou
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Donghai Ma
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Fengfei Sun
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zehu Chen
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Ying Chen
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xuegang Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Meizhu Chen
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Minmin Lin
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Honglei Shi
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Baihe Wu
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Lei Chen
- Oncology Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zibin Liang
- Department of Thoracic Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Jing Liu
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
- Department of Allergy, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| |
Collapse
|
38
|
Pan C, Hou Y, Hou Y, Wang R, Qian M, Bai X, Liang M, Wang J, Liu J, Wei Q, Pan Z, Wang T, Hu C, Xiang K, Yang C, Wang C, Chen H, Zhang J. Integrated analysis reveals that miR-548ab promotes the development of obesity and T2DM. J Genet Genomics 2025; 52:231-244. [PMID: 39608671 DOI: 10.1016/j.jgg.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Dysregulation of microRNA (miRNA) expression following the development of obesity is closely linked to the onset of type 2 diabetes mellitus (T2DM). Identifying differentially expressed miRNAs and their roles in regulating glucose metabolism will provide a theoretical foundation for the molecular mechanisms underlying obesity-induced T2DM. Here, we perform a genome-wide association study involving 5 glycolipid metabolism traits in 1783 Kazakh and 1198 Uyghur individuals to identify miRNAs associated with fasting plasma glucose (FPG) levels. A miR-548ab mimic and inhibitor are administered to hepatocytes and adipocytes, as well as obese and diabetic mice, to determine miR-548ab-related downstream signalling pathways. The effects of miR-548ab on glucose metabolism are validated using the glucose tolerance test and insulin tolerance test. Collectively, these results indicate that miR-548ab is significantly associated with FPG levels and obesity-related T2DM in both Kazakh and Uyghur populations. The miR-548ab-GULP1/SLC25A21-GLUT4 network exerts regulatory effects on glucose metabolism, obesity, and T2DM, positioning it as a candidate risk factor, potential diagnostic marker, and therapeutic target for obesity-induced T2DM. Additionally, through evolutionary analysis, the authentic variants or haplotypes of GULP1 and SLC25A21 are categorized according to their genetic susceptibility to T2DM. The miR-548ab inhibitor shows beneficial effects in obese and diabetic mice.
Collapse
Affiliation(s)
- Chongge Pan
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Yali Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanting Hou
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Ruizhen Wang
- China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meiyu Qian
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Xue Bai
- China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Maodi Liang
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Jingzhou Wang
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Jie Liu
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Qianqian Wei
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Ziyan Pan
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Ting Wang
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Chenyu Hu
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China
| | - Kun Xiang
- The First People's Hospital of Yunnan Province, Kunming, Yunnan 650223, China
| | - Chun Yang
- China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Cuizhe Wang
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China.
| | - Hua Chen
- China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Jun Zhang
- Medical College of Shihezi University, Shihezi, Xinjiang 832000, China; Laboratory of Xinjiang Endemic and Ethic Diseases of Shihezi University, Shihezi, Xinjiang 832000, China.
| |
Collapse
|
39
|
Prattico C, Gonzalez E, Dridi L, Jazestani S, Low KE, Abbott DW, Maurice CF, Castagner B. Identification of novel fructo-oligosaccharide bacterial consumers by pulse metatranscriptomics in a human stool sample. mSphere 2025; 10:e0066824. [PMID: 39699190 PMCID: PMC11774028 DOI: 10.1128/msphere.00668-24] [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: 08/06/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Dietary fibers influence the composition of the human gut microbiota and directly contribute to its downstream effects on host health. As more research supports the use of glycans as prebiotics for therapeutic applications, the need to identify the gut bacteria that metabolize glycans of interest increases. Fructo-oligosaccharide (FOS) is a common diet-derived glycan that is fermented by the gut microbiota and has been used as a prebiotic. Despite being well studied, we do not yet have a complete picture of all FOS-consuming gut bacterial taxa. To identify new bacterial consumers, we used a short exposure of microbial communities in a stool sample to FOS or galactomannan as the sole carbon source to induce glycan metabolism genes. We then performed metatranscriptomics, paired with whole metagenomic sequencing, and 16S amplicon sequencing. The short incubation was sufficient to cause induction of genes involved in carbohydrate metabolism, like carbohydrate-active enzymes (CAZymes), including glycoside hydrolase family 32 genes, which hydrolyze fructan polysaccharides like FOS and inulin. Interestingly, FOS metabolism transcripts were notably overexpressed in Blautia species not previously reported to be fructan consumers. We therefore validated the ability of different Blautia species to ferment fructans by monitoring their growth and fermentation in defined media. This pulse metatranscriptomics approach is a useful method to find novel consumers of prebiotics and increase our understanding of prebiotic metabolism by CAZymes in the gut microbiota. IMPORTANCE Complex carbohydrates are key contributors to the composition of the human gut microbiota and play an essential role in the microbiota's effects on host health. Understanding which bacteria consume complex carbohydrates, or glycans, provides a mechanistic link between dietary prebiotics and their beneficial health effects, an essential step for their therapeutic application. Here, we used a pulse metatranscriptomics pipeline to identify bacterial consumers based on glycan metabolism induction in a human stool sample. We identified novel consumers of fructo-oligosaccharide among Blautia species, expanding our understanding of this well-known glycan. Our approach can be applied to identify consumers of understudied glycans and expand our prebiotic repertoire. It can also be used to study prebiotic glycans directly in stool samples in distinct patient populations to help delineate the prebiotic mechanism.
Collapse
Affiliation(s)
- Catherine Prattico
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics, McGill Genome Centre, McGill University, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Lharbi Dridi
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Shiva Jazestani
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Kristin E. Low
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - D. Wade Abbott
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Corinne F. Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
- McGill Centre for Microbiome Research, McGill University, Montréal, Québec, Canada
| | - Bastien Castagner
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
- McGill Centre for Microbiome Research, McGill University, Montréal, Québec, Canada
| |
Collapse
|
40
|
Wang Y, Xue Y, Xu H, Zhu Q, Qin K, He Z, Huang A, Mu M, Tao X. Pediococcus acidilactici Y01 reduces HFD-induced obesity via altering gut microbiota and metabolomic profiles and modulating adipose tissue macrophage M1/M2 polarization. Food Funct 2025; 16:554-569. [PMID: 39699275 DOI: 10.1039/d4fo04301d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Obesity-related metabolic syndrome is intimately associated with infiltrated adipose tissue macrophages (ATMs), gut microbiota, and metabolic disorders. Pediococcus acidilactici holds the potential to mitigate obesity; however, there exist strain-specific functionalities and diverse mechanisms, which deserve extensive exploration. This study aims to explore the potential of P. acidilactici Y01, isolated from traditional sour whey, in alleviating HFD-induced metabolic syndrome in mice and elucidating its underlying mechanism. The results showed that P. acidilactici Y01 could inhibit the increase of body weight gain, the deposition of fat, lipid disorders and chronic low-grade inflammation, improve glucose tolerance and insulin resistance, and could reduce adipose tissue inflammation by decreasing M1-type ATMs and increasing M2-type ATMs. Meanwhile, P. acidilactici Y01 significantly increased the abundance of potentially beneficial intestinal bacteria, such as Akkermansia, Alistipes, Bifidobacterium, Lachnospiraceae_NK4A136_group, Lactobacillus, norank_f__Muribaculaceae, and Parabacteroides, and partially restored the levels of metabolites, such as phosphatidylcholines, glycerophosphocholines, sphingolipids and unsaturated fatty acids. The fecal microbiota transplantation experiment demonstrated that P. acidilactici Y01 ameliorated obesity-related metabolic syndrome by modulating the polarization of M1/M2 ATMs mediated by gut microbiota. Overall, as a dietary supplement, P. acidilactici Y01 has good potential in the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Yujing Wang
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Yu Xue
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan 232001, China
| | - Huan Xu
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Qian Zhu
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
| | - Kaili Qin
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Zhonglei He
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| | - Aixiang Huang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Min Mu
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Xinrong Tao
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| |
Collapse
|
41
|
Lu K, Shan S, Zeng Y, Yang G. Neohesperidin Mitigates High-Fat-Diet-Induced Colitis In Vivo by Modulating Gut Microbiota and Enhancing SCFAs Synthesis. Int J Mol Sci 2025; 26:534. [PMID: 39859249 PMCID: PMC11764976 DOI: 10.3390/ijms26020534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Previous research has consistently shown that high-fat diet (HFD) consumption can lead to the development of colonic inflammation. Neohesperidin (NHP), a naturally occurring flavanone glycoside in citrus fruits, has anti-inflammatory properties. However, the efficacy and mechanism of NHP in countering prolonged HFD-induced inflammation remains unclear. In this study, rats on HFD were intragastrically administered (i.g.) with NHP for 12 consecutive weeks. Results indicate that this natural compound is effective in reducing colorectal inflammation at doses of 40-80 mg/kg body weight (BW) by i.g. administration, with significant decreases in inflammation markers such as TNF-α and IL-1β levels. It also improved intestinal mucosal tissue integrity and reduced HFD-stimulated colorectal inflammation via the JAK2/STAT3 pathway. Furthermore, intestinal microbiota sequencing results show that NHP intervention significantly downregulated the Firmicutes/Bacteroidetes ratio. This ratio is closely related to the preventive role in the context of glycolipid metabolism disorder. Compared with fecal cultures of rats from the HFD group, after 48 h in vitro fermentation, those from the NHP group had distinct microbiota composition and notably higher concentrations of SCFAs. Collectively, these observations suggest that 80 mg/kg BW NHP possesses biological activities in downregulating HFD-induced colorectal inflammation by regulating intestinal flora and promoting SCFAs formation.
Collapse
Affiliation(s)
- Kun Lu
- National Engineering Laboratory for Rice and By-Products Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha 410004, China; (K.L.); (S.S.)
| | - Sijie Shan
- National Engineering Laboratory for Rice and By-Products Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha 410004, China; (K.L.); (S.S.)
| | - Yanling Zeng
- Key Laboratory of Cultivation and Protection for Non-Wood Forest Trees, Ministry of Education, Central South University of Forestry and Technology, Changsha 410004, China;
| | - Guliang Yang
- National Engineering Laboratory for Rice and By-Products Processing, Food Science and Engineering College, Central South University of Forestry and Technology, Changsha 410004, China; (K.L.); (S.S.)
| |
Collapse
|
42
|
Correa Lopes B, Turck J, Tolbert MK, Giaretta PR, Suchodolski JS, Pilla R. Prolonged storage reduces viability of Peptacetobacter (Clostridium) hiranonis and core intestinal bacteria in fecal microbiota transplantation preparations for dogs. Front Microbiol 2025; 15:1502452. [PMID: 39839105 PMCID: PMC11747423 DOI: 10.3389/fmicb.2024.1502452] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Fecal microbiota transplantation (FMT) has been described useful as an adjunct treatment for chronic enteropathy in dogs. Different protocols can be used to prepare and store FMT preparations, however, the effect of these methods on microbial viability is unknown. We aimed (1) to assess the viability of several core intestinal bacterial species by qPCR and (2) to assess Peptacetobacter (Clostridium) hiranonis viability through culture to further characterize bacterial viability in different protocols for FMT preparations. Methods Bacterial abundances were assessed in feces from six healthy dogs by qPCR after propidium monoazide (PMA-qPCR) treatment for selective quantitation of viable bacteria. Conservation methods tested included lyophilization (stored at 4°C and at -20°C) and freezing with glycerol-saline solution (12.5%) and without any cryoprotectant (stored at -20°C). Additionally, the abundance of P. hiranonis was quantified using bacterial culture. Results Using PMA-qPCR, the viability of Faecalibacterium, Escherichia coli, Streptococcus, Blautia, Fusobacterium, and P. hiranonis was reduced in lyophilized fecal samples kept at 4°C and -20°C up to 6 months (p < 0.05). In frozen feces without cryoprotectant, only Streptococcus and E. coli were not significantly reduced for up to 3 months (p > 0.05). Lastly, no differences were observed in the viability of those species in glycerol-preserved samples up to 6 months (p > 0.05). When using culture to evaluate the viability of P. hiranonis, we observed that P. hiranonis abundance was lower in lyophilized samples kept at 4°C than -20°C; and P. hiranonis abundance was higher in glycerol-preserved samples for up to 6 months than in samples preserved without glycerol for up to 3 months. Moreover, the highest abundance of P. hiranonis was observed in glycerol-preserved feces. After 3 months, P. hiranonis was undetectable by culture in 83% (5/6) of the frozen samples without glycerol. Discussion While the lyophilization procedure initially reduced P. hiranonis abundance, P. hiranonis viability was stable thereafter for up to 6 months at -20°C. The higher bacterial viability detected in fecal samples preserved with glycerol confirms the use of this cryoprotectant as a reliable method to keep bacteria alive in the presence of fecal matrix for FMT purposes.
Collapse
Affiliation(s)
- Bruna Correa Lopes
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jonathan Turck
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - M. Katherine Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Paula R. Giaretta
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
- Department of Veterinary Pathology, Hygiene and Public Health, University of Milan, Milan, Italy
| |
Collapse
|
43
|
Gao Y, Huang B, Qin Y, Qiao B, Ren M, Cao L, Zhang Y, Han M. Dietary supplementation with proanthocyanidins and rutin alleviates the symptoms of type 2 diabetes mice and regulates gut microbiota. Front Microbiol 2025; 15:1513935. [PMID: 39834378 PMCID: PMC11743507 DOI: 10.3389/fmicb.2024.1513935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Background Obesity and high fasting blood glucose (FBG) resulting from high-fat diets (HFDs) have emerged as significant public health concerns, garnering increasing attention. Recently, gut microbiota has been linked with metabolic diseases such as type 2 diabetes (T2DM), and its mediating role in dietary supplements has been confirmed. Seeking various dietary supplements to lose body weight (BW) and decrease FBG and explaining the underlying mechanism have become the research hotspots in T2DM studies. Methods In this study, rutin and proanthocyanidins (PA) were selected as dietary supplements (200 mg/kg × day, oral gavage, 6 weeks) in T2DM mice induced with HFD to assess their efficacy in weight loss, FBG reduction, gut microbiota alterations, and the associated underlying mechanisms. Results Our findings indicate that rutin was more effective than PA in relieving inflammation and fat hypertrophy, although both significantly reduced BW and FBG within 2 weeks after the intervention. Analysis of 16S rRNA amplicons revealed substantial alterations in the gut microbial community composition of mice administered with PA and rutin compared to HFD-fed mice. Importantly, several core microbes, particularly a series of probiotics, such as Akkermansia, Lactococcus, Odoribacter, Faecalibaculum, and Roseburia were identified, which were significantly correlated with the changes in BW and FBG. Conclusion Overall, our study highlights that rutin and PA can reduce BW, FBG, and inflammation by modulating the gut microbiota composition, providing novel perspectives for managing and treating weight and FBG concerns in obesity and T2DM patients through dietary supplements in clinical treatment.
Collapse
Affiliation(s)
- Yue Gao
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
- School of Life Sciences, Hefei Normal University, Hefei, Anhui, China
- Microbial Medicinal Resources Development Research Team, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| | - Binbin Huang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yunyi Qin
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Bing Qiao
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Mengfei Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Liqing Cao
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yan Zhang
- School of Life Sciences, Hefei Normal University, Hefei, Anhui, China
| | - Maozhen Han
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
- Microbial Medicinal Resources Development Research Team, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| |
Collapse
|
44
|
Oba PM, Swanson OR, Kang Y, Mioto JC, Menton JF, Vinay E, Millette M, Kelly MR, Swanson KS. Effects of Bacillus subtilis ATCC PTA-122264 on apparent total tract macronutrient digestibility and fecal characteristics, metabolites, and microbiota of healthy adult dogs. J Anim Sci 2025; 103:skaf038. [PMID: 39923129 PMCID: PMC11926536 DOI: 10.1093/jas/skaf038] [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: 11/04/2024] [Accepted: 02/06/2025] [Indexed: 02/10/2025] Open
Abstract
Gastrointestinal and stool quality issues are common in companion animals. In addition to dietary fibers and prebiotics, the consumption of live microorganisms may be used to support the gastrointestinal health of pets. Spore-forming Bacillus species are gaining interest due to their viability during processing, storage, and within the gastrointestinal tract. The objective of the current study was to determine the effects of B. subtilis ATCC PTA-122264 supplementation on dietary apparent total tract macronutrient digestibility and the fecal characteristics, metabolites, and microbiota of healthy adult dogs. Twelve healthy adult beagle dogs (6 ± 1.14 yr; 8.71 ± 0.91 kg body weight) were used in a replicated 3 × 3 Latin square design. Dogs were fed to maintain body weight and allotted to 1 of the 3 treatments each experimental period (n = 12/treatment): Control [kibble diet + placebo (1.25 g of maltodextrin)], Low [kibble diet + 1 × 109 colony-forming units (CFU)/d of B. subtilis], and High (kibble diet + 5 × 109 CFU/d of B. subtilis). Each experimental period was composed of a 22-d adaptation phase, 5-d fecal collection phase, and 1 d for blood collection. Fecal microbiota data were evaluated using QIIME2. All other data were analyzed using the Mixed Models procedure of SAS, with P < 0.05 being considered significant. B. subtilis supplementation tended to decrease (P < 0.10) apparent total tract dry matter, organic matter, and energy digestibilities but did not influence food or energy intake, fecal output, and apparent total tract protein or fat digestibilities. Most serum metabolites, hematology, fecal characteristics, and fecal bacterial alpha and beta diversity indices were not affected. Fecal dysbiosis index tended to be affected and fecal Streptococcus, Escherichia coli, and Blautia abundances were lower (P < 0.05) in dogs allotted to the Low treatment. These data suggest that daily supplementation of up to 5 × 109 CFU/d of B. subtilis ATCC PTA-122264 is safe and does not affect markers of general health and fecal characteristics of healthy dogs, warranting further exploration.
Collapse
Affiliation(s)
- Patrícia M Oba
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Olivia R Swanson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Yifei Kang
- Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Julio C Mioto
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | | | | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801,USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801,USA
| |
Collapse
|
45
|
Qian SX, Bao YF, Li XY, Dong Y, Zhang XL, Wu ZY. Multi-omics Analysis Reveals Key Gut Microbiota and Metabolites Closely Associated with Huntington's Disease. Mol Neurobiol 2025; 62:351-365. [PMID: 38850348 DOI: 10.1007/s12035-024-04271-9] [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: 02/09/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Dysbiosis of the gut microbiota is closely associated with neurodegenerative diseases, including Huntington's disease (HD). Gut microbiome-derived metabolites are key factors in host-microbiome interactions. This study aimed to investigate the crucial gut microbiome and metabolites in HD and their correlations. Fecal and serum samples from 11 to 26 patients with HD, respectively, and 16 and 23 healthy controls, respectively, were collected. The fecal samples were used for shotgun metagenomics while the serum samples for metabolomics analysis. Integrated analysis of the metagenomics and metabolomics data was also conducted. Firmicutes, Bacteroidota, Proteobacteria, Uroviricota, Actinobacteria, and Verrucomicrobia were the dominant phyla. At the genus level, the presence of Bacteroides, Faecalibacterium, Parabacteroides, Alistipes, Dialister, and Christensenella was higher in HD patients, while the abundance of Lachnospira, Roseburia, Clostridium, Ruminococcus, Blautia, Butyricicoccus, Agathobaculum, Phocaeicola, Coprococcus, and Fusicatenibacter decreased. A total of 244 differential metabolites were identified and found to be enriched in the glycerophospholipid, nucleotide, biotin, galactose, and alpha-linolenic acid metabolic pathways. The AUC value from the integrated analysis (1) was higher than that from the analysis of the gut microbiota (0.8632). No significant differences were found in the ACE, Simpson, Shannon, Sobs, and Chao indexes between HD patients and controls. Our study determined crucial functional gut microbiota and potential biomarkers associated with HD pathogenesis, providing new insights into the role of the gut microbiota-brain axis in HD occurrence and development.
Collapse
Affiliation(s)
- Shu-Xia Qian
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
- Nanhu Brain-Computer Interface Institute, Hangzhou, China
- Department of Neurology, the Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, China
| | - Yu-Feng Bao
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Xiao-Yan Li
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China
| | - Xiao-Ling Zhang
- Department of Neurology, the Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, China.
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, Department of Neurology in the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, China.
- Nanhu Brain-Computer Interface Institute, Hangzhou, China.
| |
Collapse
|
46
|
Han X, Huangfu B, Xu T, Huang K, He X. Zearalenone exacerbates lipid metabolism disorders by promoting liver lipid droplet formation and disrupting gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117664. [PMID: 39808876 DOI: 10.1016/j.ecoenv.2024.117664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
Zearalenone (ZEA), produced by Fusarium, is a fungal toxin commonly found in maize, wheat, and other cereals. ZEA has the ability to bind to estrogen receptors of humans and animals and is an environmental endocrine disruptor that may interfere with glucose homeostasis and lipid metabolism. In this study, we first investigated the effects of chronic exposure to low doses of ZEA with a high-fat-diet (HFD) in obese C57BL/6 J mice. In the absence of significant toxicity and without affecting glucose tolerance, 50 and 100 μg/kg b. w. ZEA was found to significantly exacerbate lipid synthesis, accumulation and alter the overall transcriptional profile of the liver in mice synergistically with HFD. Validation in combination with AML-12 cells revealed that ZEA promoted lipid synthesis and increased hepatic lipid droplet accumulation via the HNF1β/PPARγ and SREBP1c-HSD17B13/PLINs signal pathways. Further, by analyzing the changes in the intestinal flora of mice and their relationship with lipid metabolism genes, it was found that ZEA decreased the relative abundance of Lactobacillus and increased the relative abundance of Ileococcus, E. faecalis, and Ricardia. These changes were significantly correlated with the expression of Pparg and Srebf1, etc. ZEA may contribute to the abnormality of lipid metabolism by influencing the intestinal microbiota. This study highlights the synergistic effects of long-term low-dose ZEA and excess lipids, providing a theoretical basis for elucidating the mechanism of chronic toxicity of ZEA and its negative impact on metabolic diseases.
Collapse
Affiliation(s)
- Xiao Han
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bingxin Huangfu
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Tongxiao Xu
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety),Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China.
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety),Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China.
| |
Collapse
|
47
|
Liu Y, Wang F, Zhou Z, Liu B, Wu Z, Pan X. Profiling and comprehensive analysis of microbiome and ARGs of nurses and nursing workers in China: a cross-sectional study. Sci Rep 2024; 14:31301. [PMID: 39732868 PMCID: PMC11682234 DOI: 10.1038/s41598-024-82659-2] [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: 07/31/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Hospital-acquired infection (HAI) and antimicrobial resistance (AMR) represent major challenges in healthcare system. Despite numerous studies have assessed environmental and patient samples, very few studies have explored the microbiome and resistome profiles of medical staff including nursing workers. This cross-sectional study was performed in a tertiary hospital in China and involved 25 nurses (NSs), 25 nursing workers (NWs), and 55 non-medical control (NC). Stool samples from all participants and hand samples (i.e., the microbiome sample from hand skin, which were collected by swabbing both hands with a sponge-swab soaked with neutralized buffer and centrifuging the liquid buffer) from NSs and NWs were collected for metagenomic analysis. Metagenomic analysis revealed that medical staff exhibited lower abundances of beneficial species such as Blautia, and Bifidobacterium in the gut microbiome. However, an important potential pathogen, Staphylococcus haemolyticus, was enriched in the hands of NWs, suggesting a considerable prevalence of pathogenesis and multi-drug resistance. Accordantly, ARG analysis revealed worse hand hygiene among NWs than among NSs, characterized by a higher diversity of ARGs and a higher abundance of ARGs conferring multi-drug resistance including mdtF, acrB, AcrF and evgS. This study provides a comprehensive overview of the microbial and ARG profiles in the gut and hands of NSs and NWs. The higher abundance of potential pathogens and diverse multi-drug resistant ARGs in NWs hands indicates insufficient hand hygiene and a higher risk of HAI in this subgroup. This study is the first to highlight the critical need to improve hand hygiene among NWs, thus mitigating the risks of AMR and HAI.
Collapse
Affiliation(s)
- Ye Liu
- Department of Nursing, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79, Qingchun Rd, Hangzhou, 310003, China
| | - Fang Wang
- Department of infectious diseases, Beilun District People's Hospital, Ningbo, 315800, Zhejiang, China
| | - Ziyuan Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Bowen Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhongwen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaxia Pan
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
48
|
Yang W, Jiang M, Chen B, Jiang K, Ma N, Li Y, Wang M, Bao M, Wang C, Yang X. Study the effect of Lactobacillus plantarum ATCC 14917 for caries prevention and anti-obesity. Front Nutr 2024; 11:1511660. [PMID: 39777074 PMCID: PMC11703752 DOI: 10.3389/fnut.2024.1511660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction A complicated scenario where "multiple disease threats coexist and multiple health influencing factors are intertwined" is demonstrated by the fact that dental caries, obesity myopia and scoliosis have emerged as global public health issues. The problem of diseases co-existing in living things can be resolved by using probiotics. Lactobacillus plantarum, has gained attention recently due to its probiotic properties, useful traits, and potential medical applications. Objective Examining the anti-obesity and anti-caries effects of L. plantarum ATCC 14917 on dental caries and obese rat models caused by a high-fat and high-sugar diet is the aim of this study. Method In vitro, we assessed the L. plantarum strain's probiotic properties, such as its antibacterial activity and ability to build biofilms, to determine its ability to inhibit Streptococcus mutans. Prior to the in vivo experiment, the subsist test for L. plantarum ATCC 14917 was carried out by mimicking its capacity to lower blood sugar and blood lipid levels as well as its tolerance to gastrointestinal disorders. In order to assess the health promotion effect of L. plantarum in vivo. Three-week-old rats were fed a high-sugar, high-fat diet for 8 weeks. They were split into three groups: the control group (Control), the caries and obesity group (CA _OB) and the caries and obesity treated with L. plantarum ATCC14917 group (LP). L. plantarum ATCC 14917 was applied during the experiment, and the associated indices were then thoroughly assessed. These included the use of Mirco-CT to calculate the enamel volume, the staining of liver and fat cell sections, serological analysis, and 16S rRNA sequencing of feces. Results It was proved that the L. plantarum could inhibit the proliferation of S. mutans and remove dental plaque biofilm in time, which showed the remarkable effects of anti-caries in vitro. The demineralization rate of enamel decreased by 44.10% due to the inhibition of acid production by pathogenic bacteria. Moreover, In intestinal and stomach juice simulations, L. plantarum has a high survival rate. The characteristics of bacterial activity in a wide range of pH could degrade triglycerides and glucose in vitro smoothly. The LP group demonstrated it by reducing animal weight, serum biochemical indices, and HE-stained adipocytes as compared to the CA_OB group. 16S rRNA sequencing data showed that a high-fat and high-sugar diet induced the imbalance of intestinal flora, which showed an increase in microbial abundance, including unclassified_o__Clostridia_UCG-014, unclassified_f__Oscillospiraceae, Turicibacter, unclassified_f__Lachnospiraceae, Clostridium_sensu_stricto_1. After the intervention of L. plantarum, the number of Lactobacillus, Limosilactobacillus, unclassified_f__Muribaculaceae, Blautia, Faecalibaculum increased significantly. Conclusion Therefore, L. plantarum ATCC 14917 performed the potential of reducing tooth decay and controlling weight gain by a single strain. Support the management of dental caries and obesity, and establish a foundation for future functional food research and development.
Collapse
Affiliation(s)
- Wei Yang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Mingxin Jiang
- Department of Micro-endodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Bairu Chen
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Kongzhao Jiang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Nan Ma
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yimin Li
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meng Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meihua Bao
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chengyue Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiaopeng Yang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
49
|
Cui Z, Wang S, Niu J, Ma J, Yang H. Bifidobacterium species serve as key gut microbiome regulators after intervention in gestational diabetes mellitus. BMC Microbiol 2024; 24:520. [PMID: 39643877 PMCID: PMC11622461 DOI: 10.1186/s12866-024-03680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024] Open
Abstract
Gut microbiome dysbiosis is associated with gestational diabetes mellitus (GDM), and its modulation represents a promising approach for enhancing glycemic control. In this study, we aimed to discover specific alterations in the gut microbiome through lifestyle management. We performed metagenome sequencing on fecal samples and measured short-chain fatty acid (SCFA) in plasma samples from 27 well-controlled GDM pregnancies before and after glycemic control. At the same time, 38 normal glucose tolerance (NGT) samples served as controls. Additionally, we employed two-sample Mendelian Randomization (MR) to validate our findings against Genome-Wide Association Study (GWAS) database. Our dynamic analysis revealed Bifidobacterium genus increased in GDM patients after intervention. The MR analysis confirmed that the family of Bifidobacteriaceae (OR 0.929, 95% CI, 0.886-0.975; P = 0.003) was the only negatively associated family with GDM. Further analysis indicated the increased abundance of Bifidobacterium species were negatively correlated with glycemic traits (Spearman rho mean - 0.32 ± 0.34) but positively correlated with plasma SCFA levels (Spearman rho mean 0.24 ± 0.19). Functional analysis revealed that the quorum-sensing pathway had the strongest effect on the ability of Bifidobacterium to promote glucose homeostasis (Spearman rho = -0.34), suggesting its role in regulating intestinal microbiota. Finally, the multivariable MR analysis demonstrated that two pathways, COLANSYN PWY and PWY 7323, responsible for cell surface compound synthesis in gram-negative bacteria, mediated 14.83% (P = 0.017) and 16.64% (P = 0.049) of the protective effects of Bifidobacteriaceae against GDM, respectively. In summary, Bifidobacterium is an effective gut microbiota regulator for GDM-related glucose homeostasis.
Collapse
Affiliation(s)
- Zifeng Cui
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jianhua Niu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jingmei Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China.
| |
Collapse
|
50
|
Wang C, Qiu M, Wang S, Luo J, Huang L, Deng Q, Fang Z, Sun L, Gooneratne R. Gut-Microbiota-Derived Butyric Acid Overload Contributes to Ileal Mucosal Barrier Damage in Late Phase of Chronic Unpredictable Mild Stress Mice. Int J Mol Sci 2024; 25:12998. [PMID: 39684708 DOI: 10.3390/ijms252312998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Intestinal mucosal barrier damage is regarded as the critical factor through which chronic unpredictable mild stress (CUMS) leads to a variety of physical and mental health problems. However, the exact mechanism by which CUMS induces intestinal mucosal barrier damage is unclear. In this study, 14, 28, and 42 d CUMS model mice were established. The indicators related to ileal mucosal barrier damage (IMBD), the composition of the ileal microbiota and its amino acid (AA) and short-chain fatty acid (SCFA) metabolic functions, and free amino acid (FAA) and SCFA levels in the ileal lumen were measured before and after each stress period. The correlations between them are analyzed to investigate how CUMS induces intestinal mucosal barrier damage in male C57BL/6 mice. With the progression of CUMS, butyric acid (BA) levels decreased (14 and 28 d) and then increased (42 d), and IMBD progressively increased. In the late CUMS stage (42 d), the degree of IMBD is most severe and positively correlated with significantly increased BA levels (p < 0.05) in the ileal lumen and negatively correlated with significantly decreased FAAs, such as aspartic, glutamic, alanine, and glycine levels (p < 0.05). In the ileal lumen, the abundance of BA-producing bacteria (Muribaculaceae, Ruminococcus, and Butyricicoccus) and the gene abundance of specific AA degradation and BA production pathways and their related enzymes are significantly increased (p < 0.05). In addition, there is a significant decrease (p < 0.05) in the abundance of core bacteria (Prevotella, Lactobacillus, Turicibacter, Blautia, and Barnesiella) that rely on these specific AAs for growth and/or are sensitive to BA. These changes, in turn, promote further colonization of BA-producing bacteria, exacerbating the over-accumulation of BA in the ileal lumen. These results were validated by ileal microbiota in vitro culture experiments. In summary, in the late CUMS stages, IMBD is related to an excessive accumulation of BA caused by dysbiosis of the ileal microbiota and its overactive AA degradation.
Collapse
Affiliation(s)
- Chen Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Mei Qiu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Shuo Wang
- College of Agriculture and Biotechnology, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinjin Luo
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ling Huang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qi Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhijia Fang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Lijun Sun
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, P.O. Box 85084, Lincoln 7647, New Zealand
| |
Collapse
|