1
|
Tang L, Ding K, Li M, Chao X, Sun T, Guo Y, Peng X, Jia W, Chen T, Xie G, Feng L. Differences in oral microbiota associated with type 2 diabetes mellitus between the Dai and Han populations. J Oral Microbiol 2025; 17. [DOI: 10.1080/20002297.2024.2442420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Affiliation(s)
- Lingtong Tang
- Department of Clinical Laboratory, The People’s Hospital of Gao County, Yibin, Sichuan, China
- Department of Clinical Laboratory, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Keke Ding
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengci Li
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowen Chao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Sun
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhuai Guo
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xufei Peng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Tianlu Chen
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoxiang Xie
- Human Metabolomics Institute Inc, Shenzhen, China
| | - Lei Feng
- Department of Clinical Laboratory, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Zhao S, Lin H, Li W, Xu X, Wu Q, Wang Z, Shi J, Chen Y, Ye L, Xi L, Chen L, Yuan M, Su J, Gao A, Jin J, Ying X, Wang X, Ye Y, Sun Y, Zhang Y, Deng X, Shen B, Gu W, Ning G, Wang W, Hong J, Wang J, Liu R. Post sleeve gastrectomy-enriched gut commensal Clostridia promotes secondary bile acid increase and weight loss. Gut Microbes 2025; 17:2462261. [PMID: 39915243 PMCID: PMC11810084 DOI: 10.1080/19490976.2025.2462261] [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: 05/22/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
The gut microbiome is altered after bariatric surgery and is associated with weight loss. However, the commensal bacteria involved and the underlying mechanism remain to be determined. We performed shotgun metagenomic sequencing in obese subjects before and longitudinally after sleeve gastrectomy (SG), and found a significant enrichment in microbial species in Clostridia and bile acid metabolizing genes after SG treatment. Bile acid profiling further revealed decreased primary bile acids (PBAs) and increased conjugated secondary bile acids (C-SBAs) after SG. Specifically, glycodeoxycholic acid (GDCA) and taurodeoxycholic acid (TDCA) were increased at different follow-ups after SG, and were associated with the increased abundance of Clostridia and body weight reduction. Fecal microbiome transplantation with post-SG feces increased SBA levels, and alleviated body weight gain in the recipient mice. Furthermore, both Clostridia-enriched spore-forming bacteria and GDCA supplementation increased the expression of genes responsible for lipolysis and fatty acid oxidation in adipose tissue and reduced adiposity via Takeda G-protein-coupled receptor 5 (TGR5) signaling. Our findings reveal post-SG gut microbiome and C-SBAs as contributory to SG-induced weight loss, in part via TGR5 signaling, and suggest SBA-producing gut microbes as a potential therapeutic target for obesity intervention.
Collapse
Affiliation(s)
- Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Qihan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Juan Shi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingxia Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuqing Xi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijia Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyang Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junlei Su
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiayang Ying
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiyong Shen
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Huang W, Zhu W, Lin Y, Chan FKL, Xu Z, Ng SC. Roseburia hominis improves host metabolism in diet-induced obesity. Gut Microbes 2025; 17:2467193. [PMID: 39976263 PMCID: PMC11845086 DOI: 10.1080/19490976.2025.2467193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Next-generation live biotherapeutics are promising to aid the treatment of obesity and metabolic diseases. Here, we reported a novel anti-obesity probiotic candidate, Roseburia hominis, that was depleted in stool samples of obese subjects compared with lean controls, and its abundance was negatively correlated with body mass index and serum triglycerides. Supplementation of R. hominis prevented body weight gain and disorders of glucose and lipid metabolism, prevented fatty liver, inhibited white adipose tissue expansion and brown adipose tissue whitening in mice fed with high-fat diet, and boosted the abundance of lean-related species. The effects of R. hominis could be partially attributed to the production of nicotinamide riboside and upregulation of the Sirtuin1/mTOR signaling pathway. These results indicated that R. hominis is a promising candidate for the development of next-generation live biotherapeutics for the prevention of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Wenli Huang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Geng P, Zhao N, Zhou Y, Harris RS, Ge Y. Faecalibacterium prausnitzii regulates carbohydrate metabolic functions of the gut microbiome in C57BL/6 mice. Gut Microbes 2025; 17:2455503. [PMID: 39841201 DOI: 10.1080/19490976.2025.2455503] [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: 07/05/2024] [Revised: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The probiotic impact of microbes on host metabolism and health depends on both host genetics and bacterial genomic variation. Faecalibacterium prausnitzii is the predominant human gut commensal emerging as a next-generation probiotic. Although this bacterium exhibits substantial intraspecies diversity, it is unclear whether genetically distinct F. prausnitzii strains might lead to functional differences in the gut microbiome. Here, we isolated and characterized a novel F. prausnitzii strain (UT1) that belongs to the most prevalent but underappreciated phylogenetic clade in the global human population. Genome analysis showed that this butyrate-producing isolate carries multiple putative mobile genetic elements, a clade-specific defense system, and a range of carbohydrate catabolic enzymes. Multiomic approaches were used to profile the impact of UT1 on the gut microbiome and associated metabolic activity of C57BL/6 mice at homeostasis. Both 16S rRNA and metagenomic sequencing demonstrated that oral administration of UT1 resulted in profound microbial compositional changes including a significant enrichment of Lactobacillus, Bifidobacterium, and Turicibacter. Functional profiling of the fecal metagenomes revealed a markedly higher abundance of carbohydrate-active enzymes (CAZymes) in UT1-gavaged mice. Accordingly, UT1-conditioned microbiota possessed the elevated capability of utilizing starch in vitro and exhibited a lower availability of microbiota-accessible carbohydrates in the gut. Further analysis uncovered a functional network wherein UT1 reduced the abundance of mucin-degrading CAZymes and microbes, which correlated with a concomitant reduction of fecal mucin glycans. Collectively, our results reveal a crucial role of UT1 in facilitating the carbohydrate metabolism of the gut microbiome and expand our understanding of the genetic and phenotypic diversity of F. prausnitzii.
Collapse
Affiliation(s)
- Peiling Geng
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ni Zhao
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yong Ge
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
5
|
Gong X, Peng Q, Jiang R, Yang N, Xing C, Wang R. Mn-oxidizing microalgae and woodchip-denitrifying bioreactor system for recovering manganese and removing nitrogen from electrolytic manganese metal industrial tailwater. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137383. [PMID: 39889605 DOI: 10.1016/j.jhazmat.2025.137383] [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: 10/30/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Excess manganese (Mn) and NH4+-N emissions from electrolytic manganese metal industrial tailwater may harm the environment. However, previous studies have not combined Mn-oxidizing microalgae to reclaim Mn with woodchip substrates for nitrogen removal from tailwater. Here, a two-stage bioreactor system was constructed to recover Mn by microalgal-mediated bio-oxidation in an algae reactor (AR) and remove nitrogen by denitrification in a woodchip reactor (WR). The results showed that up to 100 % of Mn2+ in the tailwater was removed after a 3-day incubation period. The maximum amount of biogenerated Mn oxide nanoparticles reached 13.34 mg/L with Mn4+ as the main Mn valence. Mn recovery reached 65.69 % through precipitate collection, and the NH4+-N removal efficiency reached 97 % in the AR. Mn oxidation by algae might promote oxidative removal of NH4+-N. NO3--N and total nitrogen removal efficiencies in the WR reached 82-90 % and 65-87 %, respectively, which was attributed to denitrification. The predominance of the denitrification gene narG in the WR may have driven the efficient nitrate removal. Flavobacterium, Acidovora, Massilia, Arcticibacter, and Acinetobacter were the most abundant genera in the WR and represented dominant denitrifying bacteria in the woodchip microbiome, indicating their important contribution to denitrification. Overall, the combined application of Mn-oxidizing algae and woodchip-denitrifying bioreactors may represent an efficient treatment technology for electrolytic manganese wastewater remediation.
Collapse
Affiliation(s)
- Xinyue Gong
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China
| | - Qin Peng
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China
| | - Ruixin Jiang
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China
| | - Na Yang
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China
| | - Cijun Xing
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China
| | - Rui Wang
- Key Laboratory of Land Resources Evaluation and Monitoring in Southwest, Sichuan Normal University, Ministry of Education, China; College of Life Science, Sichuan Normal University, Chengdu 610041, China.
| |
Collapse
|
6
|
Murugesan R, Kumar J, Leela KV, Meenakshi S, Srivijayan A, Thiruselvam S, Satheesan A, Chaithanya V. The role of gut microbiota and bacterial translocation in the pathogenesis and management of type 2 diabetes mellitus: Mechanisms, impacts, and dietary therapeutic strategies. Physiol Behav 2025; 293:114838. [PMID: 39922411 DOI: 10.1016/j.physbeh.2025.114838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025]
Abstract
PURPOSE OF REVIEW The influence of gut microbiota on Type 2 Diabetes Mellitus (T2DM) is an emerging area of research. This review investigates the relationship between gut microbiota dysbiosis, bacterial translocation, and T2DM. It aims to elucidate how microbial imbalances contribute to the progression of T2DM through bacterial translocation and to evaluate dietary and therapeutic strategies to manage these effects. RECENT FINDINGS Recent studies highlight that dysbiosis in T2DM patients often leads to increased systemic inflammation, impaired glucose metabolism, and disrupted gut barrier integrity. These disruptions promote elevated levels of harmful bacterial components, such as lipopolysaccharides, in the bloodstream. This, in turn, is linked to worsening insulin resistance and metabolic dysfunction. Advances in molecular methods and biomarkers have provided deeper insights into bacterial translocation and its impact on diabetes. Dietary interventions, including nutraceutical agents, high-fiber and low-glycemic index diets, as well as the use of probiotics and prebiotics, have shown promise in restoring gut health and mitigating bacterial translocation. CONCLUSION Maintaining a balanced gut microbiota and intestinal barrier integrity is crucial for managing T2DM. Therapeutic strategies, including dietary modifications and nutraceuticals, have demonstrated potential in reducing bacterial translocation and systemic inflammation. Continued research is needed to refine these approaches and explore novel treatment modalities for improving metabolic health in T2DM patients.
Collapse
Affiliation(s)
- Ria Murugesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Janardanan Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India.
| | - Kakithakara Vajravelu Leela
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Sachdev Meenakshi
- Department of Dietary, Tamil Nadu Government Multi Super Speciality Hospital, Chennai 600002, Tamil Nadu, India
| | - Appandraj Srivijayan
- Department of Internal Medicine, Melmaruvathur Adhiparasakthi Institute of Medical Sciences and Research, Melmaruvathur 603319, Tamil Nadu, India
| | - Shubhashree Thiruselvam
- Department of Obstetrics and Gynaecology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| | - Venkata Chaithanya
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalapattu 603203, Tamil Nadu, India
| |
Collapse
|
7
|
Wang X, Lin Y, Li S, Wang J, Li X, Zhang D, Duan D, Shao Z. Metagenomic analysis reveals the composition and sources of antibiotic resistance genes in coastal water ecosystems of the Yellow Sea and Yangtze River Delta. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 371:125923. [PMID: 40010597 DOI: 10.1016/j.envpol.2025.125923] [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: 11/26/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
The rapid development of coastal areas has raised concerns about marine environmental pollution. In this study, metagenomics was employed to investigate antibiotic resistance genes (ARGs), mobile genetic elements (MGEs), and bacterial communities in the Yellow Sea and Yangtze River Delta in China. Multidrug resistance genes were the most abundant ARGs in these regions. Transposons and insertion_element_IS91 were the dominant MGEs, closely related to the horizontal gene transfer of ARGs. Temperature, dissolved oxygen, pH, and depth were identified as important environmental factors influencing the distribution of ARGs in seawater. Oil, agriculture, animal husbandry, and wastewater treatment plants are likely the primary sources of ARGs. From the perspective of ARG control, bacterial communities contributed the most to the development of the resistome and may carry ARGs, spreading from the Yangtze River Delta to the Yellow Sea along ocean currents. A comparison with Tara Oceans datasets revealed that the dominant ARG types and bacterial genera in coastal waters were consistent with global characteristics, with variations in ARG subtypes. This study expands knowledge on the distribution patterns of ARGs at an offshore scale and provides a reference for the prevention and control of resistant gene pollution in the Yellow Sea and Yangtze River Delta.
Collapse
Affiliation(s)
- Xin Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yude Lin
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Shaoxuan Li
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Jiahui Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; College of Life Sciences, Shandong Province Key Laboratory of Applied Mycology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xiaohui Li
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, 315211, China
| | - Demeng Zhang
- Key Laboratory of Seaweed Fertilizers, Ministry of Agriculture and Rural Affairs, Qingdao Brightmoon Seaweed Group Co. Ltd., Qingdao, 266400, China
| | - Delin Duan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Zhanru Shao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
8
|
Liu W, Jin R, Ma F, Zhao P, Su Y, Wang J, Zhang Y, Wang R, Zhu J, Liu X. Effects of Dioscorea opposita polysaccharides on insulin resistance and gut microbiota in high-fat-diet induced type 2 diabetic rats. Int J Biol Macromol 2025; 304:141004. [PMID: 39952518 DOI: 10.1016/j.ijbiomac.2025.141004] [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: 03/26/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
This study investigates the effects Dioscorea opposita polysaccharides (DOP) on insulin resistance, lipid metabolism, oxidative stress, and intestine microbiota in high-fat-diet and streptozotocin induced type 2 diabetes (T2DM) rats. Low dose (DOP-L, 200 mg/kg BW), high dose (DOP-H, 400 mg/kg BW) and D. opposita powder (DO, 400 mg/kg BW) were oral-administrated to T2DM rats. After 6 weeks of treatment, supplementation of DOP-H and DO improved body weight and glucose/lipid metabolism-related indicators, including glucagon-like peptide 1, total cholesterol and high-density lipoprotein cholesterol. DOP-H and DO suppressed liver oxidative stress through increasing the level of superoxide dismutase, catalase, glutathione and reducing malondialdehyde. DOP attenuated the pathological change in liver, such as hepatic steatosis, and thus improved the liver function. Furthermore, the anti-diabetic effects of DOP was correlated with alterations of the gut microbiota, including an increase in Firmicutes and Bacteroidetes and a decrease in Actinobacteria and Proteobacteria, which promoted a healthier gut environment. Further analysis of short-chain fatty acids and metabolites provided evidences of DOP's regulatory effects on cecal contents in T2DM rats. Therefore, DOP-H present decent effects on T2DM, suggesting that DOP can ameliorate the insulin resistance and restore blood lipid level of T2DM rats with high-fat-diet by regulating intestinal microbiota.
Collapse
Affiliation(s)
- Wei Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China; Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rumeng Jin
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Fanyi Ma
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| | - Peng Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yuting Su
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jiani Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Yun Zhang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Ruijiao Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jinhua Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Xiuhua Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| |
Collapse
|
9
|
Nageswaran V, Carreras A, Reinshagen L, Beck KR, Steinfeldt J, Henricsson M, Ramezani Rad P, Peters L, Strässler ET, Lim J, Verhaar B, Döring Y, Weber C, König M, Steinhagen-Thiessen E, Demuth I, Kränkel N, Leistner DM, Potente M, Nieuwdorp M, Knaus P, Kuebler WM, Ferrell M, Nemet I, Hazen SL, Landmesser U, Bäckhed F, Haghikia A. Gut Microbial Metabolite Imidazole Propionate Impairs Endothelial Cell Function and Promotes the Development of Atherosclerosis. Arterioscler Thromb Vasc Biol 2025. [PMID: 40143816 DOI: 10.1161/atvbaha.124.322346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND The microbially produced amino acid-derived metabolite imidazole propionate (ImP) contributes to the pathogenesis of type 2 diabetes. However, the effects of ImP on endothelial cell (EC) physiology and its role in atherosclerotic coronary artery disease are unknown. Using both human and animal model studies, we investigated the potential contributory role of ImP in the development of atherosclerosis. METHODS Plasma levels of ImP were measured in patients undergoing elective cardiac angiography (n=831) by ultra-high performance liquid chromatography coupled to tandem mass spectrometry. Odds ratios and corresponding 95% CIs for coronary artery disease were calculated based on the ImP quartiles using both univariable and multivariable logistic regression models. The effects of ImP on functional properties of ECs were assessed using human aortic ECs. In a mouse model of carotid artery injury, the impact of ImP on vascular regeneration was examined. Additionally, atheroprone Apoe-/- mice fed a high-fat diet were treated with and without ImP (800 µg), and aortic atherosclerotic lesion area was evaluated after 12 weeks. Next-generation sequencing, Western blot analysis, small interfering RNA-based gene knockdown, and tamoxifen-inducible Cre-loxP experiments were performed to investigate ImP-mediated molecular mechanisms. RESULTS Plasma ImP levels in subjects undergoing cardiac evaluation were associated with increased risk of prevalent coronary artery disease. We found that ImP dose dependently impaired migratory and angiogenic properties of human ECs and promoted an increased inflammatory response. Long-term exposure to ImP compromised the repair potential of the endothelium after an arterial insult. In atheroprone Apoe-/- mice, ImP increased atherosclerotic lesion size. Mechanistically, ImP attenuated insulin receptor signaling by suppressing the PI3K (phosphoinositide 3-kinase)/AKT pathway leading to sustained activation of the FOXO1 (forkhead box protein O1) transcription factor. Genetic inactivation of endothelial FOXO1 signaling in ImP-treated mice enhanced the angiogenic activity and preserved the vascular repair capacity of ECs after carotid injury. CONCLUSIONS Our findings reveal a hitherto unknown role of the microbially produced histidine-derived metabolite ImP in endothelial dysfunction and atherosclerosis, suggesting that ImP metabolism is a potential therapeutic target in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Vanasa Nageswaran
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Alba Carreras
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Leander Reinshagen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
| | - Katharina R Beck
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Jakob Steinfeldt
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
| | - Pegah Ramezani Rad
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Lisa Peters
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- Institute of Biology, Freie Universität Berlin, Germany (L.P.)
| | - Elisabeth T Strässler
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
| | - Joseph Lim
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Barbara Verhaar
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.V., M.N.)
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital (Y.D.)
- Department for BioMedical Research, University of Bern (Y.D.), Switzerland
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Germany (Y.D., C.W.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany (Y.D., C.W.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands (C.W.)
- Munich Cluster for Systems Neurology, Germany (C.W.)
| | - Maximilian König
- Department of Internal Medicine D-Geriatrics, University Medicine Greifswald, Germany (M.K.)
| | - Elisabeth Steinhagen-Thiessen
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (E.S.-T., I.D.)
- Charité-Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Germany (I.D.)
| | - Nicolle Kränkel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
| | - David M Leistner
- German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, Germany (D.M.L.)
- Department of Medicine, Cardiology and Angiology, Goethe University Hospital, Frankfurt, Germany (D.M.L.)
| | - Michael Potente
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Angiogenesis and Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.L., M.P.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.L., M.P.)
| | - Max Nieuwdorp
- Department of Internal Medicine-Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), the Netherlands (B.V., M.N.)
- Department of Vascular Medicine, Amsterdam UMC, the Netherlands (B.V., M.N.)
| | - Petra Knaus
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (V.N., P.K.)
- Berlin-Brandenburg School for Regenerative Therapies, Germany (P.K.)
- International Max-Planck Research School for Biology and Computation, Berlin, Germany (P.K.)
| | - Wolfgang M Kuebler
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (L.P., W.M.K.)
- German Center for Lung Research (DZL), Berlin, Germany (W.M.K.)
- Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, Canada (W.M.K.)
- Departments of Surgery and Physiology, University of Toronto, Canada (W.M.K.)
| | - Marc Ferrell
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Ina Nemet
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
| | - Stanley L Hazen
- Departments of Cardiovascular and Metabolic Sciences, and Cardiovascular Medicine, Cleveland Clinic, OH (M.F., I.N., S.L.H.)
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, OH (S.L.H.)
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, University of Gothenburg, Sweden (A.C., K.R.B., M.H., F.B.)
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden (F.B.)
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany (V.N., L.R., J.S., P.R.R., E.T.S., N.K., U.L., A.H.)
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany (V.N., L.R., P.R.R., L.P., E.T.S., N.K., M.P., W.M.K., U.L., A.H.)
- Friede Springe-Cardiovascular Prevention Center at Charité, Charité-Universitätsmedizin Berlin, Germany (V.N., J.S., E.S.-T., N.K., U.L., A.H.)
- University Hospital St. Josef-Hospital Bochum, Cardiology and Rhythmology, Ruhr University Bochum, Germany (V.N., L.R., A.H.)
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany (J.S., U.L., A.H.)
| |
Collapse
|
10
|
Ruan Z, Liu J, Zhao J. Causal associations between gut microbiota and type 2 diabetes mellitus subtypes: a mendelian randomization analysis. BMC Endocr Disord 2025; 25:79. [PMID: 40122799 PMCID: PMC11931760 DOI: 10.1186/s12902-025-01863-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE To investigate the causal relationships between gut microbiota and novel adult-onset type 2 diabetes mellitus(T2DM) subtypes. METHODS We conducted Mendelian randomization (MR) analyses using genome-wide association data from European populations. Initial MR analyses examined associations between gut microbiota and four T2DM subtypes, followed by validation analyses using type 1 diabetes mellitus(T1DM) and T2DM GWAS data. We also performed bidirectional MR analyses and tested for heterogeneity and pleiotropy across all analyses. RESULTS Our MR analyses revealed distinctive associations between gut microbiota and T2DM subtypes: six bacterial taxa with severe insulin-deficient diabetes (SIDD), four with severe insulin-resistant diabetes (SIRD), eight with mild obesity-related diabetes (MOD), and eight with mild age-related diabetes (MARD). These associations were distinct from T1DM findings. Six bacterial taxa were validated in T2DM analyses, with four showing directionally consistent effects: Class Clostridia (OR = 0.57, P = 0.045) and Order Clostridiales (OR = 0.57, P = 0.045) were associated with reduced MOD risk, while species Catus (OR = 1.80, P = 0.007) was associated with increased MOD risk, and genus Holdemania (OR = 2.51, P = 0.004) was associated with increased SIRD risk. No significant heterogeneity or pleiotropy was observed across analyses. CONCLUSIONS Our MR analyses reveal novel causal relationships between gut microbiota and adult-onset T2DM subtypes, though further validation studies are warranted.
Collapse
Affiliation(s)
- Zhichao Ruan
- Department of Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiangteng Liu
- Department of Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinxi Zhao
- Department of Endocrinology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
11
|
Zhang Q, Su T, Pan Y, Wang X, Zhang C, Qin H, Li M, Li Q, Li X, Guo J, Wu L, Qin L, Liu T. Malus hupehensis leaves: a functional beverage for alleviating hepatic inflammation and modulating gut microbiota in diabetic mice. Food Funct 2025. [PMID: 40126388 DOI: 10.1039/d4fo05325g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Malus hupehensis leaves (MHL), consumed as a daily beverage in Chinese folk tradition and recently recognized as a new food material, are abundant in polyphenols and bioactive compounds that demonstrate hypoglycemic, lipid-lowering, and anti-inflammatory effects. However, the antidiabetic mechanisms have not been fully elucidated. This study aimed to investigate the protective mechanisms of Malus hupehensis leaves' extract (MHLE) against type 2 diabetes mellitus (T2DM). The results showed that MHLE effectively ameliorated glucose and lipid metabolic abnormalities in db/db mice, and attenuated hepatic macrophage activation. Transcriptomic analysis of the liver revealed that MHLE primarily affects genes involved in inflammatory responses and inhibited the TLR4/MAPK pathway to reduce hepatic inflammation. Metagenomic sequencing identified changes in gut microbiota composition and showed that MHLE restored the abundance of Lachnospiraceae bacterium, Oscillospiraceae bacterium, and Clostridia bacterium while reducing the abundance of Escherichia coli, thereby ameliorating gut dysbiosis. The integrated regulation of metabolism, immune response, and the microbial environment by MHLE significantly alleviated symptoms of T2DM. This study offers strong scientific evidence for the potential use of MHL as a functional food.
Collapse
Affiliation(s)
- Qiue Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Tong Su
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Yajing Pan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaomeng Wang
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chengfei Zhang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Huizhao Qin
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Mingxiu Li
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Qingsong Li
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xiaochen Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Jiangfan Guo
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingling Qin
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Tonghua Liu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
12
|
Xu M, Li W, Xu Y, Zhang J, Xue H, Du J, Hu X. Arecoline Alleviates T2DM via Gut Microbiota Modulation and Liver Gene Regulation in Mice. Mol Nutr Food Res 2025:e70015. [PMID: 40123201 DOI: 10.1002/mnfr.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
SCOPE Arecoline, the main alkaloid in areca nut, has shown potential in modulating metabolism and gut microbiota. This study aimed to evaluate its therapeutic effects on glucose and lipid metabolism, inflammation, liver function, and potential mechanisms in a Type 2 diabetes mellitus (T2DM) mouse model. METHODS AND RESULTS T2DM was established in mice with a high-fat, high-sugar diet, and streptozotocin injections. Arecoline significantly reduced fasting blood glucose, enhanced glucose tolerance, and increased insulin sensitivity. Serum lipid profiles showed marked decreases in total cholesterol, triglycerides, and LDL-C levels. Systemic inflammation, as measured by serum levels of IL-1β, IL-6, and MCP-1, decreased significantly. Improvements in liver function were observed, as indicated by reductions in ALT and AST levels. Liver transcriptomic analysis revealed modulation of pathways related to glutathione metabolism, MAPK signaling, and cAMP signaling, which were involved in insulin signaling and oxidative stress response. Additionally, arecoline mitigated gut dysbiosis by restoring microbial diversity, altering gut microbiota composition, and regulating key pathways involved in NAD biosynthesis and fatty acid β-oxidation, which were critical for maintaining energy homeostasis. CONCLUSION Arecoline improves glucose metabolism, lipid profiles, and liver function, while modulating gut microbiota and liver metabolic pathways, showing potential as a therapeutic agent for T2DM.
Collapse
Affiliation(s)
- Meng Xu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Wanggao Li
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Yuan Xu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Hui Xue
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Juan Du
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore, Singapore
| | - Xiaosong Hu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Ding X, Chen Y, Du L, Li J, Meng X, Lv H, Tong B, Niu G, Jian T, Chen J. Benefits of inulin and fructo-oligosaccharides on high fat diet-induced type 2 diabetes mellitus by regulating the gut microbiota in mice. J Nutr Biochem 2025:109908. [PMID: 40122150 DOI: 10.1016/j.jnutbio.2025.109908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is pathologically associated with gut microbiota imbalance, which is implicated in disease progression through metabolic and inflammatory pathways. The therapeutic potential of inulin, a well-characterized prebiotic, has been explored to mitigate T2DM via microbiota modulation. However, the efficacy of this intervention, with its performance dependent on the degree of polymerization (DP), requires further investigation. This study assessed the therapeutic roles of inulin (DP3-60) and fructo-oligosaccharides (FOS, DP3-10) in T2DM management. Dietary administration of these prebiotic compounds demonstrated a significant capacity to alleviate multiple metabolic pathologies, including obesity, insulin resistance, systemic inflammation, oxidative stress, dyslipidemia and hepatic steatosis in high-fat diet (HFD)-fed induced T2DM mice. Significant superior efficacy was observed in FOS for ameliorating glucose metabolic dysregulation, adipocyte hypertrophy, liver weight, and histopathological alterations in colonic tissue, while inulin exhibited greater potency in alleviating oxidative stress. Both inulin and FOS enhanced gut microbiota diversity and richness in T2DM mice, accompanied by a significant reduction in Firmicutes/Bacteroidetes ratio. Notably, the S24-7 family emerged as a crucial microbial taxon modulated by both inulin and FOS. Furthermore, FOS demonstrated superior capacity to restore HFD-induced gut microbiota. Taxonomically significant amplicon sequence variants (ASVs), which were altered by HFD and modulated by inulin and FOS, exhibited distinct taxonomic profiles between the two compounds. This study provides preliminary evidence that the biological effects and beneficial properties of inulin-type fructans exhibit DP-dependent variations, which may enhance their efficient utilization in metabolic disorders.
Collapse
Affiliation(s)
- Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yan Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Lanlan Du
- Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Jing Li
- Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xiuhua Meng
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Han Lv
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Bei Tong
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Guanting Niu
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Tunyu Jian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China; Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
14
|
Liang T, Jiang T, Liang Z, Li L, Chen Y, Chen T, Yang L, Zhang N, Dong B, Xie X, Gu B, Wu Q. Gut microbiota-driven BCAA biosynthesis via Staphylococcus aureus -expressed acetolactate synthase impairs glycemic control in type 2 diabetes in South China. Microbiol Res 2025; 296:128145. [PMID: 40138872 DOI: 10.1016/j.micres.2025.128145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
An increase in branched-chain amino acid (BCAA) levels can result in insulin resistance at different stages of type 2 diabetes (T2D), however, the causes of this increase are unclear. We performed metagenomics and metabolomics profiling in patients with prediabetes (PDM), newly diagnosed diabetes (NDDM), and post-medication type 2 diabetes (P2DM) to investigate whether altered gut microbes and metabolites could explain the specific clinical characteristics of different disease stages of T2D. Here we identify acetolactate synthase (ALS) a BCAA biosynthesis enzyme in Staphylococcus aureus as a cause of T2D insulin resistance. Compared with healthy peoples, patients with PDM, NDDM, and P2DM groups, especially in P2DM group, have increased faecal numbers of S. aureus. We also demonstrated that insulin administration may be a risk factor for S. aureus infection in T2D. The presence of ALS-positive S. aureus correlated with the levels of BCAAs and was associated with an increased fasting blood glucose (FBG) and insulin resistance. Humanized microbiota transplantation experiment indicated that ALS contributes to disordered insulin resistance mediated by S. aureus. We also found that S. aureus phage can reduced the FBG levels and insulin resistance in db/db mice. The ALS-positive S. aureus are associated with insulin resistance in T2D, opening a new therapeutic avenue for the prevention or treatment of diabetes.
Collapse
Affiliation(s)
- Tingting Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Tong Jiang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Zhuang Liang
- Department of Rehabilitation Hospital Pain Ward, Xi'an Jiaotong University Affiliated Honghui Hospital, Xi'an, Shaanxi 710054, China
| | - Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ya Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Tong Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Lingshuang Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ni Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bo Dong
- Department of Rehabilitation Hospital Pain Ward, Xi'an Jiaotong University Affiliated Honghui Hospital, Xi'an, Shaanxi 710054, China.
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China.
| | - Bing Gu
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China.
| |
Collapse
|
15
|
Mamun MAA, Rakib A, Mandal M, Singh UP. Impact of a High-Fat Diet on the Gut Microbiome: A Comprehensive Study of Microbial and Metabolite Shifts During Obesity. Cells 2025; 14:463. [PMID: 40136712 PMCID: PMC11940932 DOI: 10.3390/cells14060463] [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: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Over the last few decades, the prevalence of metabolic diseases such as obesity, diabetes, non-alcoholic fatty liver disease, hypertension, and hyperuricemia has surged, primarily due to high-fat diet (HFD). The pathologies of these metabolic diseases show disease-specific alterations in the composition and function of their gut microbiome. How HFD alters the microbiome and its metabolite to mediate adipose tissue (AT) inflammation and obesity is not well known. Thus, this study aimed to identify the changes in the gut microbiome and metabolomic signatures induced by an HFD to alter obesity. To explore the changes in the gut microbiota and metabolites, 16S rRNA gene amplicon sequencing and metabolomic analyses were performed after HFD and normal diet (ND) feeding. We noticed that, at taxonomic levels, the number of operational taxonomic units (OTUs), along with the Chao and Shannon indexes, significantly shifted in HFD-fed mice compared to those fed a ND. Similarly, at the phylum level, an increase in Firmicutes and a decrease in Bacteroidetes were noticed in HFD-fed mice. At the genus level, an increase in Lactobacillus and Ruminococcus was observed, while Allobaculum, Clostridium, and Akkermansia were markedly reduced in the HFD group. Many bacteria from the Ruminococcus genus impair bile acid metabolism and restrict weight loss. Firmicutes are efficient in breaking down complex carbohydrates into short-chain fatty acids (SCFAs) and other metabolites, whereas Bacteroidetes are involved in a more balanced or efficient energy extraction. Thus, an increase in Firmicutes over Bacteroidetes enhances the absorption of more calories from food, which may contribute to obesity. Taken together, the altered gut microbiota and metabolites trigger AT inflammation, which contributes to metabolic dysregulation and disease progression. Thus, this study highlights the potential of the gut microbiome in the development of therapeutic strategies for obesity and related metabolic disorders.
Collapse
Affiliation(s)
| | | | | | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; (M.A.A.M.); (A.R.); (M.M.)
| |
Collapse
|
16
|
Yoo JY, Sarkar A, Song HS, Bang S, Shim G, Springer C, O’Brien ME, Shin Y, Ju S, Han S, Kim SS, Menon U, Choi TG, Groer ME. Gut Microbiome Alterations, Mental Health, and Alcohol Consumption: Investigating the Gut-Brain Axis in Firefighters. Microorganisms 2025; 13:680. [PMID: 40142574 PMCID: PMC11945892 DOI: 10.3390/microorganisms13030680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Firefighters across the world face higher risks of occupational hazards, such as exposure to chemicals, extreme heat, traumatic stressors, and intense physical demands, which can increase their vulnerability to a range of psychological and physiological difficulties. These challenges include the risk of developing chronic stress, depression, and post-traumatic stress disorder (PTSD), potentially leading to detrimental negative coping patterns such as alcohol abuse. The consequent health implications impact both short-term and long-term health and well-being. This study aimed to explore the relationship between mental health status, alcohol consumption patterns, and gut microbiome alterations in firefighters from two different regions-America and Korea. By investigating these relationships, we hope to gain insights into how repeated exposure to severe stressors impacts gut health. Healthy male firefighters (ages 21-50) and controls (matched sex, geography, and age) were recruited via flyers and snowball sampling in the United States and South Korea, resulting in 203 participants (102 firefighters and 101 controls). Firefighters reported significantly higher PTSD symptoms and depression and drank 2.3 times more alcohol than the control group. American firefighters reported more drinking than Koreans. There was a significant correlation between higher alcohol consumption and the likelihood of witnessing deaths by suicide. However, there were no correlations between alcohol consumption and PTSD symptom severity. There were associations between alcohol consumption patterns and aspects of the gut microbiome. This study highlights the mental health challenges faced by firefighters, including elevated rates of PTSD, depression, and alcohol consumption, with specific microbial imbalances linked to PTSD and alcohol use, emphasizing the role of the gut-brain axis.
Collapse
Affiliation(s)
- Ji Youn Yoo
- College of Nursing, University of Tennessee, Knoxville, TN 37996, USA; (A.S.); (M.E.G.)
| | - Anujit Sarkar
- College of Nursing, University of Tennessee, Knoxville, TN 37996, USA; (A.S.); (M.E.G.)
| | - Hyo-Sook Song
- Department of Paramedicine, Bucheon University, Bucheon 14632, Republic of Korea; (H.-S.S.); (S.B.)
| | - Sunghwan Bang
- Department of Paramedicine, Bucheon University, Bucheon 14632, Republic of Korea; (H.-S.S.); (S.B.)
| | - Gyusik Shim
- Department of Paramedicine, Korea Nazarene University, Cheonan 31172, Republic of Korea;
| | - Cary Springer
- Research Computing Support, Office of Innovative Technologies, University of Tennessee, Knoxville, TN 37996, USA;
| | - Morgan E. O’Brien
- Department of Public Health, University of Tennessee, Knoxville, TN 37996, USA;
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.J.); (S.H.); (S.S.K.)
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.J.); (S.H.); (S.S.K.)
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.J.); (S.H.); (S.S.K.)
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.J.); (S.H.); (S.S.K.)
| | - Usha Menon
- College of Nursing, University of South Florida, Tampa, FL 33612, USA;
- Tampa General Hospital Cancer Institute, Tampa, FL 33606, USA
| | - Tae Gyu Choi
- Department of Pathogenic Laboratory Research, Institute of Occupation and Environment, Korea Workers’ Compensation & Welfare Service, Incheon 21417, Republic of Korea;
| | - Maureen E. Groer
- College of Nursing, University of Tennessee, Knoxville, TN 37996, USA; (A.S.); (M.E.G.)
| |
Collapse
|
17
|
Ramos C, Magistro D, Walton GE, Whitham A, Camp N, Poveda C, Gibson GR, Hough J, Kinnear W, Hunter K. Assessing the gut microbiota composition in older adults: connections to physical activity and healthy ageing. GeroScience 2025:10.1007/s11357-025-01605-w. [PMID: 40095191 DOI: 10.1007/s11357-025-01605-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
The composition and functionality of the gut microbiota (GM) changes throughout the life course. As we move into older age, it starts to shift towards a less healthy one, which may lead to an imbalance in the GM community. Strategies that can reverse age-related dysbiosis are an important part of healthy aging. Little is known about the GM composition of older adults with different physical activity (PA) levels and whether it might contribute to healthy ageing. The aim of this study was to compare the GM composition of older adults with different PA levels and assess if it is associated with healthy ageing. 101 participants aged between 65-85 years undertook anthropometric measures, a 6-min walking test, wore an accelerometer for 7 days and provided a faecal sample. Faecal GM composition was analysed using 16S rRNA sequencing. We found that those who fulfilled the WHO/UK PA recommendations had higher relative abundance of several health-related bacteria such as Lactobacillus, F. prausnitzii and Roseburia intestinalis and lower abundance of disease-associated bacteria such as D.piger or Enterobacterales when compared to those who did not reach PA recommendations. These findings suggest that PA might improve the GM composition and has the potential to, at least partially, revert age-associated dysbiosis and promote healthy ageing.
Collapse
Affiliation(s)
- Catarina Ramos
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK.
| | - Daniele Magistro
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, The University of Reading, Whiteknights, Reading, UK
| | - Anya Whitham
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
| | - Nicola Camp
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, The University of Reading, Whiteknights, Reading, UK
| | - Glenn R Gibson
- Department of Food and Nutritional Sciences, The University of Reading, Whiteknights, Reading, UK
| | - John Hough
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
| | - Will Kinnear
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
| | - Kirsty Hunter
- Department of Sport Science, Sport, Health and Performance Enhancement (SHAPE) Research Centre, Nottingham Trent University, Nottingham, UK
- Reynolds Contamination Control, Lincoln, UK
| |
Collapse
|
18
|
Huang HJ, Liu Y, Li DW, Wang X, Feng NX, Li HY, Mo CH, Yang WD. Polystyrene Microplastics Can Aggravate the Damage of the Intestinal Microenvironment Caused by Okadaic Acid: A Prevalent Algal Toxin. Mar Drugs 2025; 23:129. [PMID: 40137315 PMCID: PMC11943709 DOI: 10.3390/md23030129] [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: 02/05/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
As emerging contaminants, microplastics (MPs) may pose a threat to human health. Their co-exposure with the widespread phycotoxin okadaic acid (OA), a marine toxin known to cause gastrointestinal toxicity, may exacerbate health risk and raise public safety concern. In this study, the toxicity mechanisms of MPs and OA on intestinal microenvironment was explored using human Caco-2 cells as the model, which was combined with an in vitro fecal fermentation experiment. Our results showed that co-exposure to MPs (80 μg/mL) and OA (20 ng/mL) significantly decreased cell viability, increased intracellular reactive oxygen species (ROS) production, elevated lactate dehydrogenase release, impaired ABC transporter activity, promoted OA accumulation, and triggered inflammatory response compared to the control, MPs, and OA groups, indicating that co-exposure directly compromises intestinal epithelial integrity. In vitro fermentation experiments revealed that co-exposure disrupted gut microbial composition, decreasing the relative abundance of some bacteria, such as Parasutterella and Adlercreutzia, while increasing opportunistic pathogens, such as Escherichia-Shigella, increased. These findings provide new insights into the impact and underlying mechanisms of MPs and OA co-exposure on intestinal homeostasis, highlighting the potential health risks associated with MPs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ce-Hui Mo
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (H.-J.H.)
| | - Wei-Dong Yang
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (H.-J.H.)
| |
Collapse
|
19
|
Diesse JM, Jadhav S, Tamekou SL, Simo G, Dzoyem JP, Souopgui J, Kuiate JR, Nema V. Disturbances in the gut microbiota potentially associated with metabolic syndrome among patients living with HIV-1 and on antiretroviral therapy at Bafoussam Regional Hospital, Cameroon. Diabetol Metab Syndr 2025; 17:86. [PMID: 40089790 PMCID: PMC11909933 DOI: 10.1186/s13098-025-01653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND This study investigates the gut microbiota components associated with metabolic syndrome in patients living with HIV-1 at Bafoussam Regional Hospital, West Cameroon, it focuses on gastrointestinal mucosal barrier disruption and dysbiosis, and their effects on persistent inflammation and metabolic disorders. METHODS A pilot study was conducted involving fourteen patients living with HIV-1. The patients were divided into two groups of seven in each group. One group consisted of patients with metabolic syndrome, and the other group included patients without metabolic syndrome. Gut microbiota was characterized using 16 S rRNA gene-targeted sequencing to analyze microbial diversity and composition. Beta diversity and the relative abundance of bacterial taxa were compared between patients with and without metabolic syndrome. RESULTS Patients living with HIV-1 and metabolic syndrome showed significantly altered beta diversity compared to those without metabolic syndrome. A higher relative abundance of Firmicutes and increased proliferation of Proteobacteria were observed in patients with metabolic syndrome. Additionally, a decrease in metabolically beneficial bacteria, such as Bifidobacterium sp., Lactobacillus sp., Akkermansia sp., and Faecalibacterium sp., was noted. Several beneficial bacterial species were associated with participants' metadata, suggesting potential links between gut microbiota and metabolic syndrome. CONCLUSION This preliminary study highlights that gut microbial balance, rather than the presence of specific bacteria, plays a crucial role in managing metabolic health in patients living with HIV-1. The altered gut microbiota in participants with metabolic syndrome emphasizes the need for further research into the optimal gut microbial structure. Understanding the interaction between gut microbiota changes and the chemical environment in these patients could guide targeted interventions to improve metabolic outcomes.
Collapse
Affiliation(s)
- Joël Martial Diesse
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Sushama Jadhav
- Division of Molecular Biology, Indian Council of Medical Research - National Institute of Translational Virology and AIDS Research , 73 G MIDC Bhosari, Pune, 411026, India
| | | | - Gustave Simo
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Jean Paul Dzoyem
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Jacob Souopgui
- Laboratory of Embryology & Biotechnology DBM-IBMM, "Université Libre de Bruxelles", Brussels, Belgium
| | - Jules-Roger Kuiate
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Vijay Nema
- Division of Molecular Biology, Indian Council of Medical Research - National Institute of Translational Virology and AIDS Research , 73 G MIDC Bhosari, Pune, 411026, India.
| |
Collapse
|
20
|
Cheng H, Guo H, Wen C, Sun G, Tang F, Li Y. The dual role of gut microbiota in pancreatic cancer: new insights into onset and treatment. Ther Adv Med Oncol 2025; 17:17588359251324882. [PMID: 40093983 PMCID: PMC11909682 DOI: 10.1177/17588359251324882] [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: 08/25/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Pancreatic cancer ranks among the most lethal digestive malignancies, exhibiting a steadily increasing incidence and mortality worldwide. Despite significant advances in cancer research, the 5-year survival rate remains below 10%, predominantly due to delayed diagnosis and limited therapeutic options. Concurrently, the gut microbiota-an integral component of host physiology-has emerged as a crucial player in the pathogenesis of pancreatic cancer. Mounting evidence indicates that alterations in gut microbial composition and function may influence tumor initiation, progression, and response to therapy. This review provides an in-depth examination of the intricate interplay between the gut microbiome and pancreatic cancer, highlighting potential diagnostic biomarkers and exploring microbiome-targeted therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Huijuan Cheng
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Hongkai Guo
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Chengming Wen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Guodong Sun
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Affairs, Lanzhou University First Hospital, Lanzhou, Gansu, P.R. China
| | - Futian Tang
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Yumin Li
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
21
|
Inan S, Wilson RP, Tükel Ç. IUPHAR review: From gut to brain: The role of gut dysbiosis, bacterial amyloids, and metabolic disease in Alzheimer's disease. Pharmacol Res 2025; 215:107693. [PMID: 40086611 DOI: 10.1016/j.phrs.2025.107693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Gut microbial dysbiosis, or altered gut microbial communities, in Alzheimer's Disease suggests a pathogenic role for gut inflammation and microbial products in shaping a neuroinflammatory environment. Similarly, metabolic diseases, such as obesity and diabetes, are also associated with an increased risk of Alzheimer's Disease. As the metabolic landscape shifts during gut inflammation, and gut inflammation in turn impacts metabolic processes, we explore how these interconnected pathways may contribute to the progression of Alzheimer's Disease. Additionally, we discuss the role of bacterial amyloids produced by gut microbes, which may exacerbate amyloid aggregation in the brain and contribute to neurodegenerative processes. Furthermore, we highlight potential therapeutic strategies aimed at reducing gut inflammation, improving metabolic health, and decreasing amyloid content as a means to mitigate Alzheimer's Disease progression. These approaches, targeting the gut-brain-metabolic axis, could offer promising avenues for delaying or preventing cognitive decline in affected individuals.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| | - R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Yang SJ, Nguyen TTM, Jin X, Zheng Q, Park SJ, Yi GS, Yi TH. A PRISMA Systematic Review of Sexual Dysfunction and Probiotics with Pathophysiological Mechanisms. BIOLOGY 2025; 14:286. [PMID: 40136541 PMCID: PMC11940792 DOI: 10.3390/biology14030286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Sexual dysfunction, influenced by hormonal imbalances, psychological factors, and chronic diseases, affects a significant portion of the population. Probiotics, known for their beneficial effects on gut microbiota, have emerged as potential therapeutic agents for improving sexual health. This systematic review evaluates the impact of probiotics on sexual function, hormonal regulation, and reproductive outcomes. A comprehensive search identified 3308 studies, with 12 meeting the inclusion criteria-comprising 10 randomized controlled trials (RCTs) and 2 in vivo and in vitro studies. Probiotic interventions were shown to significantly improve sexual function, particularly in women undergoing antidepressant therapy (p < 0.05). Significant improvements in Female Sexual Function Index (FSFI) scores were observed, with combined treatments such as Lactofem with Letrozole and Lactofem with selective serotonin reuptake inhibitors (SSRIs) demonstrating a 10% biochemical and clinical pregnancy rate compared to 0% in the control group (p = 0.05). Probiotic use was also associated with a 66% reduction in menopausal symptoms, increased sperm motility (36.08%), viability (46.79%), and morphology (36.47%). Probiotics also contributed to favorable hormonal changes, including a reduced luteinizing hormone (LH) to follicle-stimulating hormone (FSH) ratio (from 3.0 to 2.5, p < 0.05) and increased testosterone levels. Regarding reproductive outcomes, probiotic use was associated with higher pregnancy rates in women undergoing fertility treatments and improvements in sperm motility, viability, and morphology in men. This review highlights the promising role of probiotics in addressing sexual dysfunction and reproductive health, suggesting their potential as adjunctive treatments for conditions such as depression and infertility. Further research is needed to better understand the underlying mechanisms of these beneficial effects.
Collapse
Affiliation(s)
- Su-Jin Yang
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (S.-J.Y.); (T.T.M.N.); (Q.Z.); (S.-J.P.)
| | - Trang Thi Minh Nguyen
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (S.-J.Y.); (T.T.M.N.); (Q.Z.); (S.-J.P.)
| | - Xiangji Jin
- Department of Dermatology, School of Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dong-daemun, Seoul 02447, Republic of Korea;
| | - Qiwen Zheng
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (S.-J.Y.); (T.T.M.N.); (Q.Z.); (S.-J.P.)
| | - Se-Jig Park
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (S.-J.Y.); (T.T.M.N.); (Q.Z.); (S.-J.P.)
| | - Gyeong-Seon Yi
- Department of Biopharmaceutical Biotechnology, Graduate School, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea;
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (S.-J.Y.); (T.T.M.N.); (Q.Z.); (S.-J.P.)
| |
Collapse
|
23
|
Dai R, Zhang J, Liu F, Xu H, Qian JM, Cheskis S, Liu W, Wang B, Zhu H, Pronk LJU, Medema MH, de Jonge R, Pieterse CMJ, Levy A, Schlaeppi K, Bai Y. Crop root bacterial and viral genomes reveal unexplored species and microbiome patterns. Cell 2025:S0092-8674(25)00198-9. [PMID: 40081368 DOI: 10.1016/j.cell.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/14/2024] [Accepted: 02/16/2025] [Indexed: 03/16/2025]
Abstract
Reference genomes of root microbes are essential for metagenomic analyses and mechanistic studies of crop root microbiomes. By combining high-throughput bacterial cultivation with metagenomic sequencing, we constructed comprehensive bacterial and viral genome collections from the roots of wheat, rice, maize, and Medicago. The crop root bacterial genome collection (CRBC) significantly expands the quantity and phylogenetic diversity of publicly available crop root bacterial genomes, with 6,699 bacterial genomes (68.9% from isolates) and 1,817 undefined species, expanding crop root bacterial diversity by 290.6%. The crop root viral genome collection (CRVC) contains 9,736 non-redundant viral genomes, with 1,572 previously unreported genus-level clusters in crop root microbiomes. From these, we identified conserved bacterial functions enriched in root microbiomes across soils and host species and uncovered previously unexplored bacteria-virus connections in crop root ecosystems. Together, the CRBC and CRVC serve as valuable resources for investigating microbial mechanisms and applications, supporting sustainable agriculture.
Collapse
Affiliation(s)
- Rui Dai
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Gene Function and Modulation Research, Peking-Tsinghua-NIBS Graduate Program, School of Life Sciences, Peking University, Beijing 100871, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jingying Zhang
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Gene Function and Modulation Research, Peking-Tsinghua-NIBS Graduate Program, School of Life Sciences, Peking University, Beijing 100871, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fang Liu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Haoran Xu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jing-Mei Qian
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Shani Cheskis
- Department of Plant Pathology and Microbiology, Institute of Environmental Science, The Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Weidong Liu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Binglei Wang
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Gene Function and Modulation Research, Peking-Tsinghua-NIBS Graduate Program, School of Life Sciences, Peking University, Beijing 100871, China
| | - Honghui Zhu
- State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Lotte J U Pronk
- Bioinformatics Group, Wageningen University & Research, 6708 PB Wageningen, the Netherlands
| | - Marnix H Medema
- Bioinformatics Group, Wageningen University & Research, 6708 PB Wageningen, the Netherlands
| | - Ronnie de Jonge
- Plant-Microbe Interactions, Department of Biology, Science for Life, Utrecht University, 3584 CH Utrecht, the Netherlands; AI Technology for Life, Department of Information and Computing Sciences, Science for Life, Utrecht University, 3584 CC Utrecht, the Netherlands
| | - Corné M J Pieterse
- Plant-Microbe Interactions, Department of Biology, Science for Life, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Asaf Levy
- Department of Plant Pathology and Microbiology, Institute of Environmental Science, The Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Klaus Schlaeppi
- Department of Environmental Sciences, University of Basel, Basel 4056, Switzerland
| | - Yang Bai
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Gene Function and Modulation Research, Peking-Tsinghua-NIBS Graduate Program, School of Life Sciences, Peking University, Beijing 100871, China; CAS-JIC Centre of Excellence for Plant and Microbial Science, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
24
|
Bariod L, Fuentes E, Millet M, Jacquiod S, White J, Moreau J, Monceau K. Direct and indirect effects of pesticide exposure on the gut microbiota of a farmland raptor. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136857. [PMID: 39708601 DOI: 10.1016/j.jhazmat.2024.136857] [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: 09/19/2024] [Revised: 11/20/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Recent studies in humans have shown that certain pesticides could affect the composition and functions of the gut microbiota, an essential modulator of vertebrate physiology, leading to potential dysbiosis. However, this relationship remains largely unknown in wild birds despite the implications of pesticides in the current decline of farmland species. The present study sought to fill this gap by providing data on the association between pesticide concentrations in blood and gut microbiota characteristics in relation to individual traits in a farmland raptor, the Montagu's harrier (Circus pygargus). Results showed that females with higher body condition and higher pesticide load exhibited greater gut bacterial richness and diversity, while the relationship was opposite in males with higher body condition. In terms of taxonomic composition, Proteobacteria emerged as the dominant phylum across all nestlings. Differences in the abundance of specific phyla and genera were observed according to pesticide load, with higher levels of Bacteroidota and Leifsonia, but lower levels of Bulkholderia, in nestlings with higher pesticide concentrations in their blood. This study highlights differences in microbiota and contamination by several pesticides according to the phenotypic characteristics of a wild raptor, and shows that farmland birds can represent relevant biosentinels for assessing the health/proper functioning of ecosystems (One Health approach).
Collapse
Affiliation(s)
- Léa Bariod
- UMR 7372, Centre d'Études Biologiques de Chizé, La Rochelle Université & CNRS, Villiers en Bois 79360, France.
| | - Elva Fuentes
- UMR 7372, Centre d'Études Biologiques de Chizé, La Rochelle Université & CNRS, Villiers en Bois 79360, France.
| | - Maurice Millet
- Université de Strasbourg, CNRS-UMR 7515, ICPEES, 67087 Strasbourg cedex 2, France.
| | - Samuel Jacquiod
- Agroécologie, Institut Agro Dijon, INRAE, Université Bourgogne, Université Bourgogne Franche-Comté, Dijon, France.
| | - Joël White
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR 5300, CNRS-IRD-UT3-INPT 5174, 118 Route de Narbonne, Toulouse F-31062, France; Ecole Nationale Supérieure de Formation de l'Enseignement Agricole, Castanet-Tolosan, 31320, France.
| | - Jérôme Moreau
- UMR 7372, Centre d'Études Biologiques de Chizé, La Rochelle Université & CNRS, Villiers en Bois 79360, France; Biogéosciences, UMR 6282 CNRS, Université de Bourgogne, 6 Boulevard Gabriel, Dijon 21000, France.
| | - Karine Monceau
- UMR 7372, Centre d'Études Biologiques de Chizé, La Rochelle Université & CNRS, Villiers en Bois 79360, France; LTSER "Zone Atelier Plaine & Val de Sèvre", CNRS, Villiers-en-Bois 79360, France.
| |
Collapse
|
25
|
Yoshimura Y, Wakabayashi H, Nagano F, Matsumoto A, Shimazu S, Shiraishi A, Kido Y, Bise T, Hamada T, Yoneda K, Maeda K. Gut microbiome diversity is associated with muscle mass, strength and quality in post-stroke patients. Clin Nutr ESPEN 2025; 67:25-33. [PMID: 40049396 DOI: 10.1016/j.clnesp.2025.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The gut microbiome has emerged as a potential influencer of muscle health; however, its role in hospitalized patients remains unclear. This study aimed to investigate the association between gut microbiome diversity and skeletal muscle mass, strength, and quality in hospitalized post-stroke patients. METHODS We conducted a cross-sectional study of post-stroke patients admitted to a rehabilitation facility. Gut microbiome diversity was assessed using 16S ribosomal ribonucleic acid (rRNA) gene sequencing, calculating Operational Taxonomic Unit (OTU) Richness, Faith's Phylogenetic Diversity (PD), and Shannon index. Muscle health was evaluated using skeletal muscle index (SMI) for muscle mass, handgrip strength (HGS) for muscle strength, and bioimpedance analysis-derived phase angle (PhA) for muscle quality. Multiple linear regression analyses were performed, adjusting for potential confounders. RESULTS A total of 156 patients (mean age 78.4 years; 55.7 % male) were analyzed. OTU Richness showed significant positive associations with SMI (β = 0.197, p = 0.025), HGS (β = 0.180, p = 0.005), and PhA (β = 0.178, p = 0.022). The Shannon index was also positively associated with SMI (β = 0.120, p = 0.041), HGS (β = 0.140, p = 0.028), and PhA (β = 0.164, p = 0.032). Faith's PD did not demonstrate significant associations with muscle health parameters. CONCLUSIONS Higher gut microbiome diversity, assessed by OTU Richness and Shannon index, is associated with better muscle mass, strength, and quality in post-stroke patients. These findings suggest a potential role for gut microbiota in muscle health during stroke rehabilitation.
Collapse
Affiliation(s)
- Yoshihiro Yoshimura
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Hidetaka Wakabayashi
- Department of Rehabilitation Medicine, Tokyo Women's Medical University Hospital, Japan.
| | - Fumihiko Nagano
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Ayaka Matsumoto
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Sayuri Shimazu
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Ai Shiraishi
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Yoshifumi Kido
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Takahiro Bise
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Takenori Hamada
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Kouki Yoneda
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan.
| | - Keisuke Maeda
- Nutrition Therapy Support Center, Aichi Medical University Hospital, Japan; Department of Geriatric Medicine, Hospital, National Center for Geriatrics and Gerontology, Japan.
| |
Collapse
|
26
|
Zhang Y, Liu R, Chen Y, Cao Z, Liu C, Bao R, Wang Y, Huang S, Pan S, Qin L, Wang J, Ning G, Wang W. Akkermansia muciniphila supplementation in patients with overweight/obese type 2 diabetes: Efficacy depends on its baseline levels in the gut. Cell Metab 2025; 37:592-605.e6. [PMID: 39879980 DOI: 10.1016/j.cmet.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Akkermansia muciniphila is a promising target for managing obesity and type 2 diabetes (T2D), but human studies are limited. We conducted a 12-week randomized, double-blind, placebo-controlled trial involving 58 participants with overweight or obese T2D, who received A. muciniphila (AKK-WST01) or placebo, along with routine lifestyle guidance. Both groups showed decreases in body weight and glycated hemoglobin (HbA1c), without significant between-group differences. In participants with low baseline A. muciniphila, AKK-WST01 supplementation showed high colonization efficiency and significant reductions in body weight, fat mass, and HbA1c, which were not found in the placebo group. However, AKK-WST01 supplementation showed poor colonization and no significant clinical improvements in participants with high baseline A. muciniphila. These findings were verified in germ-free mice receiving feces with low or high A. muciniphila. Our study indicates that metabolic benefits of A. muciniphila supplementation could depend on its baseline intestinal levels, supporting the potential for gut microbiota-guided probiotic supplementation. (ClinicalTrials.gov number, NCT04797442).
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiwen Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Cong Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Riqiang Bao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shan Huang
- Department of Endocrinology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Shijia Pan
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Li Qin
- Department of Endocrinology, Chongming hospital affiliated to Shanghai University of Health & Medicine Sciences, Shanghai 202150, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Center for Metabolic Diseases (Shanghai), Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
27
|
Ponsuksili S, Hadlich F, Li S, Trakooljul N, Reyer H, Oster M, Abitew YA, Sommerfeld V, Rodehutscord M, Wimmers K. DNA methylation dynamics in the small intestine of egg-selected laying hens along egg production stages. Physiol Genomics 2025; 57:125-139. [PMID: 39869094 DOI: 10.1152/physiolgenomics.00063.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
Decades of artificial selection have markedly enhanced egg production efficiency, yet the epigenetic underpinnings, notably DNA methylation dynamics in the gut, remain largely unexplored. Here, we investigate how breeds and developmental stages influence DNA methylation profiles in laying hens, and their potential relationship to laying performance and gut health. We compared two highly selected laying hen strains, Lohmann Brown-Classic (LB) and Lohmann Selected Leghorn-Classic (LSL), which exhibited similar egg production but divergent physiological, metabolic, and immunological characteristics. Our sampling encompassed key developmental stages: the pullet stage (10 and 16 wk old), peak production (24 and 30 wk old), and later stage (60 wk old) (n = 99; 10 per group), allowing us to elucidate the temporal dynamics of epigenetic regulation. Our findings highlight a crucial window of epigenetic modulation during the prelaying period, characterized by stage-specific methylation alterations and the involvement of predicted transcription factor motifs within methylated regions. This observation was consistent with the expression patterns of DNA methyltransferases (DNMTs), including DNMT1, DNMT3A, and DNMT3B. In addition, a higher methylation level was observed in specific loci or regions in the LSL compared with the LB strain. Notably, we uncover strain-specific differences in methylation levels, particularly pronounced in genomic regions associated with intestinal integrity, inflammation, and energy homeostasis. Our research contributes to the multidisciplinary framework of epigenetics and egg-laying performance, offering valuable implications for poultry production and welfare.NEW & NOTEWORTHY Our study reveals key methylation changes in the jejunum mucosa of laying hens across developmental stages and between strains, with implications for gut health, immune function, and egg production. These findings highlight a crucial role of epigenetic regulation in optimizing performance.
Collapse
Affiliation(s)
| | - Frieder Hadlich
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Shuaichen Li
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Nares Trakooljul
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Henry Reyer
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Michael Oster
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
| | | | - Vera Sommerfeld
- Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | | | - Klaus Wimmers
- Research Institute for Farm Animal Biology, Dummerstorf, Germany
- Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| |
Collapse
|
28
|
Futakuchi T, Furuhashi H, Isshi K, Hara Y, Ono S, Kurokawa R, Takayasu L, Suda W, Sumiyama K. Ex Vivo Analysis of the Effect of Endoscopic Premedications on the Microbiota Profile in Gastric Juice. JGH Open 2025; 9:e70141. [PMID: 40114860 PMCID: PMC11924131 DOI: 10.1002/jgh3.70141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
Background and Aim Dimethicone (GAS), lidocaine (XYL), and protease (PRO) are commonly used as premedications during esophagogastroduodenoscopy (EGD). However, the effects of these drugs on the gastric microbiota remain unexplored. Therefore, we aimed to investigate the effects of these premedications on gastric juice collected from patients undergoing EGD. Methods Gastric juice was endoscopically aspirated from six patients and divided into six aliquots for in vitro analysis. The samples were mixed with premedications in corresponding treatment sets: GAS, XYL, PRO, MIX (a mixture of GAS, XYL, and PRO), and control (CTL1 and 2; no medication treatment). After extraction of microbial DNA from the treated samples, the 16S rRNA amplicon sequence was analyzed to determine the microbiota profile in terms of (1) the amount of genomic DNA (gDNA), (2) α-diversity indices, Shannon index, number of observed operational taxonomic units (OTUs), and Chao1 index, (3) weighted and unweighted UniFrac distances, and (4) the relative abundance of phyla and genera. Results The total amount of extracted gDNA did not significantly differ between the six groups. The α-diversity indices did not significantly differ between treatment groups. Although GAS, PRO, and MIX differed significantly from the technical replicates in the weighted UniFrac distance (p = 0.03 all), no significant difference was observed in the unweighted UniFrac distance. However, significant differences were observed in the relative abundance of several bacterial microbiota at the phylum and genus levels. Conclusions Premedications affect the microbiota profile of specific phylum- and genus-level bacterial groups. Trial Registration: University Hospital Medical Information Network Clinical Trials Registry: UMIN-CTR 000040192 and UMIN-CTR 000051289.
Collapse
Affiliation(s)
- Toshiki Futakuchi
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
| | - Hiroto Furuhashi
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
| | - Kimio Isshi
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
- Isshi Gastro-Intestinal Clinic Tokyo Japan
| | - Yuko Hara
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
| | - Shingo Ono
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
| | - Rina Kurokawa
- Laboratory for Symbiotic Microbiome Sciences RIKEN Center for Integrative Medical Sciences Kanagawa Japan
| | - Lena Takayasu
- Laboratory for Symbiotic Microbiome Sciences RIKEN Center for Integrative Medical Sciences Kanagawa Japan
| | - Wataru Suda
- Laboratory for Symbiotic Microbiome Sciences RIKEN Center for Integrative Medical Sciences Kanagawa Japan
| | - Kazuki Sumiyama
- Department of Endoscopy The Jikei University School of Medicine Tokyo Japan
| |
Collapse
|
29
|
Lei P, Yu H, Jiang T, Ma J, Du J, Fang Y, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D. Development of a sodium hyaluronate-enriched therapeutic formulation with stevia glycoside and mogroside V for the comprehensive management of diabetes and its complications. Int J Biol Macromol 2025; 293:139487. [PMID: 39756763 DOI: 10.1016/j.ijbiomac.2025.139487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Diabetes prevalence continues to increase as a result of people's increasing sugar intake. Diabetes mellitus and its complications (dry skin, constipation, depression, and dental caries), as well as the prohibition of sweets ingestion, seriously affect patients' physical and mental health. Therefore, it is crucial to develop a long-term food for special medical purposes (FSMP) that aids in managing diabetes and its complications. To ensure effective biomedical function and taste, we developed a FSMP beverage formulation containing stevia glycoside, mogroside V, and sodium hyaluronate (SMH-B), each at a concentration of 0.1 mg/mL. Meanwhile, this study verified that SMH-B is an environmentally friendly and biocompatible formulation. Furthermore, both in vivo and in vitro studies have demonstrated that SMH-B significantly lowers blood glucose and lipid levels, enhances skin moisture and elasticity, prevents dental caries, alleviates constipation, reduces oxidative stress, and mitigates depressive symptoms. Notably, the SMH-B compound formula exhibits a more effective adjuvant therapeutic effect compared to single-ingredient formulation composed of stevia glycosides, mogroside V, and sodium hyaluronate. Moreover, SMH-B provides the sweetness desired by diabetic patients without affecting blood glucose levels, while also offering an auxiliary therapeutic role, making it a potential FSMP for diabetes management.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China; MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
30
|
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] [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
|
31
|
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
|
32
|
Nohesara S, Mostafavi Abdolmaleky H, Pettinato G, Pirani A, Thiagalingam S, Zhou JR. IUPHAR review: Eating disorders, gut microbiota dysbiosis and epigenetic aberrations. Pharmacol Res 2025; 213:107653. [PMID: 39970995 DOI: 10.1016/j.phrs.2025.107653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Eating disorders (EDs) are a heterogeneous class of increasing mental disorders that are characterized by disturbances in eating behaviors, body weight regulation, and associated psychological dysfunctions. These disorders create physiological imbalances that alter the diversity and composition of the gut microbiota. While evidence suggests that EDs can arise from epigenetic aberrations, alterations in gut microbial communities may also contribute to the development and/or persistence of EDs through epigenetic mechanisms. Understanding the interplay among gut microbial communities, epigenetic processes, and the risk of EDs provides opportunities for designing preventive and/or therapeutic interventions through gut microbiome modulation. This review highlights how microbiome-based therapeutics and specific dietary interventions can contribute to improving various subtypes of EDs by modulating gut microbial communities and mitigating epigenetic aberrations. First, we briefly review the literature on links between epigenetic aberrations and the pathophysiology of EDs. Second, we examine the potential role of the gut microbiome in the pathogenesis of EDs through epigenetic mechanisms. Next, we explore the associations between EDs and other psychiatric disorders, and examine the potential roles of the microbiome in their pathogenesis. Finally, we present evidence supporting the potential of microbiome-based therapeutics and specific dietary interventions to improve EDs through epigenetic modifications.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Nutrition/Metabolism laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Giuseppe Pettinato
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ahmad Pirani
- Mental Health Research Center, Psychosocial Health Research Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Zhang L, Luo Y, Lv S, Liu Y, Wang R, Wang Y, Lin K, Liu L. Performance of electro-assisted ecological floating bed in antibiotics and conventional pollutants degradation: Mechanisms and microbial response. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 376:124393. [PMID: 39919574 DOI: 10.1016/j.jenvman.2025.124393] [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: 10/14/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
Electro-assisted technology is promising for enhancing plant activity, optimizing functional microbial communities, and significantly strengthening pollutant removal efficiency. In this study, four reactors were designed as control group (CG), Hydrocotyle vulgaris L. ecological floating bed (PEFB), microbial fuel cell (MFC), and Hydrocotyle vulgaris L. ecological floating bed-microbial fuel cell (PEFB-MFC) to investigate the efficiency and mechanisms for the synchronous removal of conventional and antibiotic contaminants. Results showed that PEFB-MFC hold superior removal performance for sulfamethoxazole (61%), tetracycline (61%), CODCr (65%), NH4+-N (86%), TN (41%), and TP (51%). High-throughput sequencing indicated that Pseudomonadota and Actinomycetota were the predominant phyla in the different reactors. Metagenomic sequencing results showed that pollutant degradation-related metabolic functions, such as those involved in carbohydrate and amino acid metabolism in PEFB-MFC exhibited superior abundance compared to the other reactors. LC-MS analysis revealed sulfamethoxazole degradation occurred through active-site cleavage, and tetracycline underwent demethylation, aldehyde formation, dehydroxylation. This study offers a deeper insight into electro-enhanced PEFB on decontamination performance and functional microbial communities.
Collapse
Affiliation(s)
- Liangjing Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China; School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yan Luo
- Ningbo Research Institute of Ecological and Environmental Sciences, Ningbo, 315012, China
| | - Shucong Lv
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China
| | - Yunlong Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China
| | - Rui Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China
| | - Yu Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China
| | - Kuixuan Lin
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China
| | - Lusan Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Science, Beijing, 10012, China; School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
34
|
Amiri P, Hosseini SA, Saghafi-Asl M, Roshanravan N, Tootoonchian M. Expression of PGC-1α, PPAR-α and UCP1 genes, metabolic and anthropometric factors in response to sodium butyrate supplementation in patients with obesity: a triple-blind, randomized placebo-controlled clinical trial. Eur J Clin Nutr 2025; 79:249-257. [PMID: 39448815 DOI: 10.1038/s41430-024-01512-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVES There is increasing evidence that gut metabolites have a role in the etiology of obesity. This study aimed to investigate the effects of sodium butyrate (NaB) supplementation on the expression of peroxisome proliferator-activated receptor (PPAR) gamma coactivator-1α (PGC-1α), PPAR-α, and uncoupling protein-1 (UCP-1) genes, as well as on the metabolic parameters and anthropometric indices in persons with obesity. METHODS In this triple-blind placebo-controlled randomized clinical trial, 50 individuals with obesity were randomly assigned to NaB (600 mg/day) + hypo-caloric diet or placebo group + hypo-caloric diet for 8 weeks. The study measured the participants' anthropometric characteristics, food consumption, and feelings of hunger in addition to the serum levels of metabolic indices and the mRNA expression of the PGC-1α, PPAR-α, and UCP-1 genes in peripheral blood mononuclear cells (PBMCs). RESULTS PGC-1α and UCP-1 genes expression significantly increased in NaB group compared to the placebo at the endpoint. A significant decrease in weight, BMI, and waist circumference (WC) was observed in NaB group. Among the metabolic factors, NaB significantly decreased fasting blood sugar (FBS) (P = 0.04), low-density lipoprotein cholesterol (LDL-C) (P = 0.038) and increased high-density lipoprotein cholesterol (HDL-C) (P = 0.016). NaB could not significantly change serum GLP-1 level. CONCLUSIONS This study unveiled NaB supplementation alone cannot have significant beneficial effects on anthropometric, and biochemical factors. NaB could affect anthropometric and metabolic risk variables associated with obesity only when prescribed, along with calorie restriction. CLINICAL TRIAL REGISTRATION This study was registered in the Iranian Registry of Clinical Trials ( https://en.irct.ir/trial/53968 ) on 31 January 2021 (registry number IRCT20190303042905N2).
Collapse
Affiliation(s)
- Parichehr Amiri
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Nutrition and Metabolic Diseases Research Center, Clinical Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Ahmad Hosseini
- Nutrition and Metabolic Diseases Research Center, Clinical Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Maryam Saghafi-Asl
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mitra Tootoonchian
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Xie C, Qi C, Zhang J, Wang W, Meng X, Aikepaer A, Lin Y, Su C, Liu Y, Feng X, Gao H. When short-chain fatty acids meet type 2 diabetes mellitus: Revealing mechanisms, envisioning therapies. Biochem Pharmacol 2025; 233:116791. [PMID: 39894305 DOI: 10.1016/j.bcp.2025.116791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Evidence is accumulating that short-chain fatty acids (SCFAs) produced by the gut microbiota play pivotal roles in host metabolism. They contribute to the metabolic regulation and energy homeostasis of the host not only by preserving intestinal health and serving as energy substrates but also by entering the systemic circulation as signaling molecules, affecting the gut-brain axis and neuroendocrine-immune network. This review critically summarizes the current knowledge regarding the effects of SCFAs in the fine-tuning of the pathogenesis of type 2 diabetes mellitus (T2DM) and insulin resistance, with an emphasis on the complex relationships among diet, microbiota-derived metabolites, T2DM inflammation, glucose metabolism, and the underlying mechanisms involved. We hold an optimistic view that elucidating how diet can influence gut bacterial composition and activity, SCFA production, and metabolic functions in the host will advance our understanding of the mutual interactions of the intestinal microbiota with other metabolically active organs, and may pave the way for harnessing these pathways to develop novel personalized therapeutics for glucometabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Cong Qi
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Jianwen Zhang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Wei Wang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Xing Meng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Aifeila Aikepaer
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Yuhan Lin
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Chang Su
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730124 China
| | - Yunlu Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700 China
| | - Xingzhong Feng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| | - Huijuan Gao
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| |
Collapse
|
36
|
Wu J, Ji K, Kang G, Zhang M, Wang J, Wang L, Gao M, Jia X, Lu X, Wang Y, Gao X, Guo Y, Zhu Z, Wang Q, Zhao Z, Liu Q, Huang H. Butyrate-engineered yeast activates Nppa and Sgcg genes and reduces radiation-induced heart damage via the gut-heart axis. Pharmacol Res 2025; 213:107642. [PMID: 39909125 DOI: 10.1016/j.phrs.2025.107642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Radiotherapy is a method of treating cancer through radiation aimed at killing cancer cells or inhibiting their growth. However, radiotherapy has numerous side effects because it kills tumors while causing damage to normal cells or tissues. The literature shows that radiation can cause damage to heart tissue. This study found that engineered yeast that produced butyrate can maintain small intestinal barrier function by recovering GPR109A to reduce intestinal damage caused by abdominal irradiation in mice. We unexpectedly found that engineered yeast could mitigate irradiation-induced heart damage via the gut-heart axis. Mechanistically, engineered yeast enhanced taurine and nicotinamide metabolism by increasing the relative abundance of Akkermansia and Lachnospiraceae_NK4A136; then, yeast modulated cardiac function by activating the Sgcg and Nppa genes to attenuate cardiac damage induced by abdominal irradiation. Finally, we confirmed that engineered yeast mitigated cardiac damage caused by total body irradiation, which protected other vital organs through the intestinal tract. This study has a profound impact on cancer treatment, the emergence of engineered yeast will alleviate radiotherapy side effects and benefit patients.
Collapse
Affiliation(s)
- Jiahao Wu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Guangbo Kang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Manman Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lina Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Mengxue Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Xiaoxiao Jia
- Department of Anatomy, Shandong Second Medical University, Weifang 261053, China
| | - Xinran Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Xinran Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Yufei Guo
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhixin Zhu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Qinghua Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhenyu Zhao
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - He Huang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
37
|
Bibi A, Zhang F, Shen J, Din AU, Xu Y. Behavioral alterations in antibiotic-treated mice associated with gut microbiota dysbiosis: insights from 16S rRNA and metabolomics. Front Neurosci 2025; 19:1478304. [PMID: 40092066 PMCID: PMC11906700 DOI: 10.3389/fnins.2025.1478304] [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: 08/09/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The gut and brain interact through various metabolic and signaling pathways, each of which influences mental health. Gut dysbiosis caused by antibiotics is a well-known phenomenon that has serious implications for gut microbiota-brain interactions. Although antibiotics disrupt the gut microbiota's fundamental structure, the mechanisms that modulate the response and their impact on brain function are still unclear. It is imperative to comprehend and investigate crucial regulators and factors that play important roles. We aimed to study the effect of long-term antibiotic-induced disruption of gut microbiota, host metabolomes, and brain function and, particularly, to determine the basic interactions between them by treating the C57BL/6 mice with two different, most commonly used antibiotics, ciprofloxacin and amoxicillin. Anxiety-like behavior was confirmed by the elevated plus-maze test and open field test. Gut microbes and their metabolite profiles in fecal, serum, and brain samples were determined by 16S rRNA sequencing and untargeted metabolomics. In our study, long-term antibiotic treatment exerted anxiety-like effects. The fecal microbiota and metabolite status revealed that the top five genera found were Lactobacillus, Bacteroides, Akkermansia, Ruminococcus_gnavus_group, and unclassified norank_f_Muribaculaceae. The concentration of serotonin, L-Tyrosine, 5-Hydroxy-L-tryptophan, L-Glutamic acid, L-Glutamate, 5-Hydroxyindole acetic acid, and dopaminergic synapsis was comparatively low, while adenosine was high in antibiotic-treated mice. The KEGG enrichment analysis of serum and brain samples showed that amino acid metabolism pathways, such as tryptophan metabolism, threonine metabolism, serotonergic synapsis, methionine metabolism, and neuroactive ligand-receptor interaction, were significantly decreased in antibiotic-treated mice. Our study demonstrates that long-term antibiotic use induces gut dysbiosis and alters metabolic responses, leading to the dysregulation of brain signaling molecules and anxiety-like behavior. These findings highlight the complex interactions between gut microbiota and metabolic functions, providing new insights into the influence of microbial communities on gut-brain communication.
Collapse
Affiliation(s)
- Asma Bibi
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Famin Zhang
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jilong Shen
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ahmad Ud Din
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Yuanhong Xu
- The Key Laboratory of Microbiology and Parasitology Anhui, School of Basic Medical Sciences, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Clinical Laboratory Diagnostics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
38
|
Chen B, Li Y, Li Z, Hu X, Zhen H, Chen H, Nie C, Hou Y, Zhu S, Xiao L, Li T. Vitamin E ameliorates blood cholesterol level and alters gut microbiota composition: A randomized controlled trial. Nutr Metab Cardiovasc Dis 2025:103964. [PMID: 40087044 DOI: 10.1016/j.numecd.2025.103964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND AND AIMS Antioxidants, including vitamin E (VE) and grape seed extract (GSE), as anti-aging supplementation have been widely used to improve human health. The gut microbiota plays a crucial role in health and affects the treatment effect of various interventions. However, the role of gut microbiota in VE remains unclear. This study aimed to assess the longitudinal impact of VE treatment on body health and the gut microbiota. METHODS AND RESULTS A randomized controlled trial was conducted with 90 healthy individuals. The participants were randomly assigned to three groups: a treatment group receiving VE, another antioxidant treatment group receiving GSE, and a control group receiving a placebo. We found that VE ameliorated blood cholesterol levels by reducing the levels of low-density lipoprotein cholesterol (LDL-C) in healthy volunteers. After the intervention, there was an increase in the relative abundance of short-chain fatty acid (SCFA)-producing bacteria and bile acid metabolizers. Specifically, the abundances of Lachnospira sp. and Faecalibacterium spp. increased in the VE. Interestingly, the gut microbiota of poor responders harbored a greater proportion of disease-associated bacterial species. CONCLUSIONS VE could promote health by lowering LDL-C, partly and indirectly by affecting gut bacteria with the ability to produce SCFAs or metabolize bile acids. REGISTRATION NUMBER FOR CLINICAL TRIALS The clinical trial was registered on August 28, 2021. Registration number was ChiCTR2100050567 (https://www.chictr.org.cn).
Collapse
Affiliation(s)
- Bangwei Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China; BGI Genomics, Shenzhen, China
| | - Yaxin Li
- BGI Genomics, Shenzhen, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Xiaojie Hu
- Department of Gastrointestinal Surgery, Hebei General Hospital, Shijiazhuang, China
| | | | | | | | | | | | | | - Tao Li
- BGI Genomics, Shenzhen, China; BGI Research, Shenzhen, China.
| |
Collapse
|
39
|
Li Z, Chen S, Yin B, Wei J, Wang D, Zhou H, Sun Z. Intermittent fasting regulates gut microbiota and serum metabolome profiles in middle-aged mice fed high-fat diet. Nutr Metab (Lond) 2025; 22:16. [PMID: 40001132 PMCID: PMC11863773 DOI: 10.1186/s12986-025-00904-5] [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: 08/22/2024] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Intermittent fasting (IF) has received wide attention as an effective diet strategy. Existing studies showed that IF is a promising approach for weight control, improving insulin sensitivity and reducing type 2 diabetes mellitus (T2DM) prevalence. METHODS Twenty-eight 8-month-old male C57BL/6J mice were randomly divided into a normal control group (NC), a high-fat diet group (HFD) and an HFD + IF group. Body weight (BW) and food intake were monitored weekly. After 20 weeks, the intraperitoneal glucose tolerance test (IPGTT), oral glucose tolerance test (OGTT), and intraperitoneal insulin tolerance test (IPITT) were performed weekly in sequence. Fresh faeces were collected to examine changes in gut microbiota, and serum untargeted metabolite profiling was conducted on serum samples. RESULTS IF significantly reduced weight gain, fat mass and liver weight, improved glucose tolerance and insulin sensitivity in middle-aged mice fed with high-fat diet. 16 S rRNA gene sequencing revealed that IF significantly reduced the Firmicutes/Bacteroidetes (F/B) ratio by increased Muribaculaceae, Bacteroides, Parabacteroides, and decreased Bilophila, Colidextribacter, Oscillibacter. The serum untargeted metabolomics revealed that IF could modulate differential metabolites and metabolic pathways associated with glycolipid metabolism. Spearman's correlation analysis indicated that key differential microbiota were strongly correlated with glucose metabolism-related indicators and serum metabolites such as stearic acid, obeticholic acid, and N-acetylglycine. CONCLUSIONS IF improves glucose metabolism, regulates gut microbiota, and alters serum metabolites in middle-aged mice fed a high-fat diet. This provides a new pathway for trials testing diabetes prevention in middle-aged and elderly patients.
Collapse
Affiliation(s)
- Ziru Li
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Sufang Chen
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Bingbing Yin
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiacun Wei
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Duofei Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Huoxiang Zhou
- Laboratory of Microbiology and Immunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhi Sun
- Department of Pharmacy, Henan Engineering Research Center of Clinical Mass Spectrometry for Precision Medicine, Zhengzhou Key Laboratory of Clinical Mass Spectrometry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
40
|
Chen M, Li Y, Zhai Z, Wang H, Lin Y, Chang F, Ge S, Sun X, Wei W, Wang D, Zhang M, Chen R, Yu H, Feng T, Huang X, Cheng D, Liu J, Di W, Hao Y, Yin P, Tang P. Bifidobacterium animalis subsp. lactis A6 ameliorates bone and muscle loss via modulating gut microbiota composition and enhancing butyrate production. Bone Res 2025; 13:28. [PMID: 40000617 PMCID: PMC11862215 DOI: 10.1038/s41413-024-00381-1] [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: 11/17/2023] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 02/27/2025] Open
Abstract
Systematic bone and muscle loss is a complex metabolic disease, which is frequently linked to gut dysfunction, yet its etiology and treatment remain elusive. While probiotics show promise in managing diseases through microbiome modulation, their therapeutic impact on gut dysfunction-induced bone and muscle loss remains to be elucidated. Employing dextran sulfate sodium (DSS)-induced gut dysfunction model and wide-spectrum antibiotics (ABX)-treated mice model, our study revealed that gut dysfunction instigates muscle and bone loss, accompanied by microbial imbalances. Importantly, Bifidobacterium animalis subsp. lactis A6 (B. lactis A6) administration significantly ameliorated muscle and bone loss by modulating gut microbiota composition and enhancing butyrate-producing bacteria. This intervention effectively restored depleted butyrate levels in serum, muscle, and bone tissues caused by gut dysfunction. Furthermore, butyrate supplementation mitigated musculoskeletal loss by repairing the damaged intestinal barrier and enriching beneficial butyrate-producing bacteria. Importantly, butyrate inhibited the NF-κB pathway activation, and reduced the secretion of corresponding inflammatory factors in T cells. Our study highlights the critical role of dysbiosis in gut dysfunction-induced musculoskeletal loss and underscores the therapeutic potential of B. lactis A6. These discoveries offer new microbiome directions for translational and clinical research, providing promising strategies for preventing and managing musculoskeletal diseases.
Collapse
Affiliation(s)
- Ming Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Zhengyuan Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Hui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yuan Lin
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feifan Chang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Siliang Ge
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xinyu Sun
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Wei Wei
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Duanyang Wang
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingming Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ruijing Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Haikuan Yu
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Taojin Feng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Dongliang Cheng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Jiang Liu
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenxuan Di
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Pengbin Yin
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| |
Collapse
|
41
|
Namted S, Chailaor P, Bunchasak C. Effects of drinking water fructo-oligosaccharide supplementation on broiler chicken growth performance, blood glucose level, white blood cell count, carcass yield, meat quality, and cecal microbiota. Poult Sci 2025; 104:104901. [PMID: 40024010 PMCID: PMC11919399 DOI: 10.1016/j.psj.2025.104901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025] Open
Abstract
This study investigated the effects of fructo-oligosaccharides (FOS) supplementation on the growth performance, blood glucose level, white blood cell count, carcass yield, meat quality, and cecal microbiota of Ross 308 broiler chickens. A completely randomized design was employed; FOS was supplemented in the drinking water at concentrations of 0 %, 0.25 %, and 0.50 %. From 11 to 24 d of age, 0.25 % FOS supplementation significantly increased feed intake (FI), while feed cost per gain (FCG) was significantly reduced at 0.50 % FOS (P < 0.05). During the overall period (1-36 d of age), FOS supplementation significantly improved the European Production Efficiency Factor (EPEF) (P < 0.01), and slowed down the reduction in blood glucose levels after the re-feeding period (2, 3, 4, and 5 h) (P < 0.01). Furthermore, FOS supplementation decreased the heterophil/lymphocyte (H:L) ratio (P < 0.05). However, it had no significant effect on breast meat yield or abdominal fat, but 0.50 % FOS supplementation tended to increase the percentage of cecal weight (P = 0.08). Supplementation with FOS (0.25 % and 0.50 %) significantly reduced breast meat cooking loss (P < 0.05). Regarding cecal microbiota, the FOS-supplemented groups showed increased abundances of Lactobacillaceae and Acidaminococcaceae, whereas the abundances of Lachnospiraceae and Barnesiellaceae were reduced (P < 0.05). In conclusion, drinking water FOS supplementation had a beneficial effect on the overall productive performance and cooking loss of broiler chickens via stress reduction, which may involve an improvement in the gut microbiota.
Collapse
Affiliation(s)
- Siriporn Namted
- Department of Agriculture, Faculty of Agriculture Technology, Valaya Alongkorn Rajabhat University Under the Royal Patronage, Pathum Thani 13180, Thailand
| | | | - Chaiyapoom Bunchasak
- Department of Animal Science, Faculty of Agriculture, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
42
|
Jiang Y, Jin H, Liang Q, Zhu X. Causal relationship between gut microbiota and dental caries: A Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e41555. [PMID: 39993094 PMCID: PMC11856918 DOI: 10.1097/md.0000000000041555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/02/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
The onset of dental caries is associated with multiple factors, including oral microbiota, dietary sugars, the defensive mechanisms of saliva and teeth, oral hygiene practices, and socioeconomic factors. However, its relationship with the gut microbiota remains to be further explored. It remains crucial to establish a definitive causal link between the gut microbiota and the development of dental caries. This study aimed to investigate the causal relationship between gut microbiota and the risk of dental caries, focusing on identifying specific microbial communities potentially implicated in its pathogenesis. Gut microbiota data from genome-wide association studies (GWAS) conducted by the MiBioGen consortium were utilized as the exposure variable, with dental caries as the outcome variable. A Mendelian randomization (MR) approach was employed, leveraging comprehensive, publicly available GWAS summary data from European populations. The primary analytical method was the inverse variance weighted method, supplemented by additional techniques such as the weighted median model, MR-Egger, simple mode, and weighted mode, to ensure the robustness of the results. Heterogeneity was evaluated using Cochran Q test, and potential pleiotropy was assessed through MR-Egger regression. Sensitivity analyses were performed using the leave-one-out method to further validate the findings. The results revealed that a higher relative abundance of Christensenellaceae, FamilyXIII, Ruminococcaceae, and Senegalimassilia was associated with a reduced risk of dental caries. In contrast, a higher relative abundance of Erysipelotrichia, Erysipelotrichales, Pasteurellales, Erysipelotrichaceae, Pasteurellaceae, Methanobrevibacter, Roseburia, and Terrisporobacter was linked to an elevated risk of dental caries. This study provides compelling evidence for a causal relationship between gut microbiota and the development of dental caries, offering novel insights into the potential role of specific gut microbial communities in the pathogenesis of dental caries.
Collapse
Affiliation(s)
- Yongyuan Jiang
- Department of Stomatology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, P.R. China
| | - Huan Jin
- Department of Emergency, Qingdao Municipal Hospital, Qingdao, Shandong, P.R. China
| | - Qian Liang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Zhu
- Department of Stomatology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, P.R. China
| |
Collapse
|
43
|
Wu Z, Gong C, Wang B. The relationship between dietary index for gut microbiota and diabetes. Sci Rep 2025; 15:6234. [PMID: 39979448 PMCID: PMC11842723 DOI: 10.1038/s41598-025-90854-y] [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: 12/11/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
This study aims to explore the relationship between the Dietary Index for Gut Microbiota (DI-GM) and diabetes. In recent years, there has been increasing attention to the role of the gut microbiome in regulating host metabolism. However, the relationship between DI-GM and the risk of diabetes has not been sufficiently studied. This study utilized relevant data from the National Health and Nutrition Examination Survey (NHANES) 2007-2018. Multiple logistic regression analysis was conducted to explore the relationship between DI-GM and the risk of diabetes. The dose-response relationship between DI-GM and the risk of diabetes was observed using restricted cubic splines (RCS). Threshold effect analysis was performed based on RCS results. Subgroup analyses were used to conduct a sensitivity analysis of the relationship between DI-GM and the risk of diabetes. The results from multiple logistic regression analysis indicated a significant negative correlation between DI-GM and the risk of diabetes (OR, 0.954, 95%CI, 0.918-0.991). RCS results also showed a significant nonlinear negative relationship between DI-GM and the risk of diabetes (P < 0.001, P for nonlinear = 0.010). The threshold effect analysis revealed that when DI-GM was below 6.191, there was a significant negative correlation between DI-GM and the risk of diabetes (OR, 0.921, 95% CI, 0.876-0.969). However, when DI-GM exceeded 6.191, the relationship between DI-GM and the risk of diabetes was no longer significant. Subgroup analysis revealed that the negative correlation between DI-GM and the risk of diabetes remained significant in Whites, participants with a poverty-income ratio > 3.5, body mass index > 24, current drinkers, never or current smokers, and those without chronic kidney disease (P < 0.05). This study demonstrates a nonlinear negative correlation between DI-GM and the risk of diabetes. Maintaining DI-GM above 6.191 may help prevent diabetes.
Collapse
Affiliation(s)
- Zhe Wu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changle Gong
- Department of Dermatology, Jinan Hospital of Traditional Chinese Medicine, Jinan, China
| | - Bin Wang
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
44
|
Ohlsson C, Lawenius L, Jiang Y, Horkeby K, Wu J, Nilsson KH, Koskela A, Tuukkanen J, Movérare-Skrtic S, Henning P, Sjögren K. The beneficial effects of a probiotic mix on bone and lean mass are dependent on the diet in female mice. Sci Rep 2025; 15:6182. [PMID: 39979617 PMCID: PMC11842756 DOI: 10.1038/s41598-025-91056-2] [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: 11/19/2024] [Accepted: 02/18/2025] [Indexed: 02/22/2025] Open
Abstract
Bone mass and lean mass decrease with age and these changes are associated with increased fracture risk and sarcopenia. Previous studies demonstrated that a probiotic mixture of Lacticaseibacillus paracasei DSM13434, Lactiplantibacillus plantarum DSM 15312 and DSM 15313 (L. Mix) prevents bone loss in ovariectomized (ovx) female mice. The purpose of the present study is to test if the beneficial effect of L. Mix is modified by the diet. Female mice were fed either a high-fat (HFD, 60% kcal from fat) or a low-fat (LFD, 10% kcal from fat) diet and subjected to either sham or ovx surgery and treated with L. Mix for 12 weeks. L. Mix treatment increased total body bone mineral density (p ≤ 0.01), by increasing cortical bone area, and total body lean mass (p = 0.035) in mice on LFD but not in mice on HFD. Metagenome sequencing of cecal content showed that L. Mix treatment increased the relative abundance of Lacticaseibacillus paracasei and, Lactiplantibacillus plantarum, demonstrating successful treatment. In addition, the probiotic treatment affected the overall gut microbiota composition and functionality. These findings demonstrate that the L. Mix in combination with a healthy diet is beneficial for musculoskeletal health in female mice.
Collapse
Affiliation(s)
- Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lina Lawenius
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yiwen Jiang
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Horkeby
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jianyao Wu
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin H Nilsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| | - Sofia Movérare-Skrtic
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Klara Sjögren
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
45
|
Li Y, Zhang Z(J, Saarela O, Sharma D, Xu W. Mediation CNN (Med-CNN) Model for High-Dimensional Mediation Data. Int J Mol Sci 2025; 26:1819. [PMID: 40076446 PMCID: PMC11899585 DOI: 10.3390/ijms26051819] [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: 11/28/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 03/14/2025] Open
Abstract
Complex biological features such as the human microbiome and gene expressions play a crucial role in human health by mediating various biomedical processes that influence disease progression, such as immune responses and metabolic processes. Understanding these mediation roles is essential for gaining insights into disease pathogenesis and improving treatment outcomes. However, analyzing such high-dimensional mediation features presents challenges due to their inherent structural and correlations, such as the hierarchical taxonomic structures in microbial operational taxonomic units (OTUs), gene-pathway relationships, and the high dimensionality of the datasets, which complicates mediation analysis. We propose the Med-CNN model, an iterative approach using Convolutional Neural Networks (CNNs) to incorporate the complex biological network of the mediation features. The output values from network-specific CNN models are condensed into an integrative mediation metric (IMM), which captures essential biological information for estimating mediation effects. Our approach is designed to handle high-dimensional data and accommodate their unique structures and non-linear interactive mediation effects. Through comprehensive simulation studies, we evaluated the performance of our algorithm across different scenarios, including various mediation effects, effect sizes, and sample sizes, and we compared it to conventional methods. Our simulations demonstrated consistently lower biases in mediation effect estimates, with values ranging from 0.17 to 0.56, which were lower than other established methods ranging from 0.24 to 13.27. In a real data application, our method identified a mediation effect of 0.06 between ethnicity and vaginal pH levels.
Collapse
Affiliation(s)
- Yao Li
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A1, Canada; (Y.L.); (Z.Z.); (O.S.); (D.S.)
| | - Zhongyuan (Jasper) Zhang
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A1, Canada; (Y.L.); (Z.Z.); (O.S.); (D.S.)
| | - Olli Saarela
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A1, Canada; (Y.L.); (Z.Z.); (O.S.); (D.S.)
| | - Divya Sharma
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A1, Canada; (Y.L.); (Z.Z.); (O.S.); (D.S.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A1, Canada; (Y.L.); (Z.Z.); (O.S.); (D.S.)
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| |
Collapse
|
46
|
Francini E, Badillo Pazmay GV, Fumarola S, Procopio AD, Olivieri F, Marchegiani F. Bi-Directional Relationship Between Bile Acids (BAs) and Gut Microbiota (GM): UDCA/TUDCA, Probiotics, and Dietary Interventions in Elderly People. Int J Mol Sci 2025; 26:1759. [PMID: 40004221 PMCID: PMC11855466 DOI: 10.3390/ijms26041759] [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: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
The gut microbiota (GM), the set of microorganisms that colonizes our intestinal tract, can undergo many changes, some of which are age related. Several studies have shown the importance of maintaining a healthy GM for a good quality of life. In the elderly, maintaining a good GM may become a real defense against infection by pathogens, such as C. difficile. In addition to the GM, bile acids (BAs) have been shown to provide an additional defense mechanism against the proliferation of pathogenic bacteria and to regulate bacterial colonization of the gut. BAs are molecules produced in the host liver and secreted with the bile into the digestive tract, and they are necessary for the digestion of dietary lipids. In the gut, host-produced BAs are metabolized by commensal bacteria to secondary BAs. In general GM and host organisms interact in many ways. This review examines the relationship between GM, BAs, aging, and possible new approaches such as dietary interventions, administration of ursodesoxycholic acid/tauroursodesoxycholic acid (UDCA/TUDCA), and probiotics to enrich the microbial consortia of the GM in the elderly and achieve a eubiotic state necessary for maintaining good health. The presence of Firmicutes and Actinobacteria together with adequate levels of secondary BAs would provide protection and improve the frailty state in the elderly. In fact, an increase in secondary BAs has been observed in centenarians who have reached old age without serious health issues, which may justify their active role in achieving longevity.
Collapse
Affiliation(s)
- Emanuele Francini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
| | - Gretta V. Badillo Pazmay
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
| | - Stefania Fumarola
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
| | - Antonio Domenico Procopio
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Fabiola Olivieri
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Francesca Marchegiani
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
| |
Collapse
|
47
|
Jia F, Zhu G, Ding M, Song J, Ruan X, Zhou G, Yuan M, Saleem S, Wang J, Gao Q, Ali A, Wu X, Chen X. Flavonoids of Pigmented Rice Lower Blood Glucose Levels by Regulating the Glucose Metabolism Pathway and Gut Microbiota Composition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4094-4104. [PMID: 39913671 DOI: 10.1021/acs.jafc.4c09675] [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: 02/20/2025]
Abstract
Black and red rice have many physiological effects, including antioxidant capacity and decreasing blood glucose, because they are flavonoid-rich. Here, our study found that Hongcaomi (a red rice variety) of five examined rice cultivars contained the highest total polyphenol content and total flavonoid content and the strongest antioxidant activity, with values of 5.83 mg GAE/g, 1.57 mg RE/g, and 72.63 μmol Trolox/g, respectively. The decreased blood glucose effect showed that the strongest effect was shown by the two red rice cultivars, followed by the black rice cultivar Ashamurasaki, the white rice cultivar Yixiang1B, and the black rice cultivar Chongxuehei which had no effect. The study further found that the strongest physiological effect in red rice was derived from catechin. In addition, our study reveals that pigmented rice could decrease blood glucose levels through many pathways, including repairing liver morphology, improving the expression level of genes involved in gluconeogenesis and glycogenesis pathways, and regulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Feng Jia
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Guoxu Zhu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Mengru Ding
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jianguo Song
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinxin Ruan
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Gege Zhou
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Meng Yuan
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Saira Saleem
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jian Wang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Qingyi Gao
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Asif Ali
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xianjun Wu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoqiong Chen
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
48
|
Zeng Y, Wu Q, Guo M, Teng F, Jiang C, Chen J, Tan X, Zeng C, Long Y, Law BYK, Xu Y. Gut microbiota-derived imidazole propionate: an emerging target for the prevention and treatment of cardiometabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1409119. [PMID: 40034229 PMCID: PMC11872695 DOI: 10.3389/fendo.2025.1409119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Despite significant advancements in prevention and treatment, cardiometabolic diseases continue to pose a high burden of incidence and mortality. The chronic progression of these diseases necessitates the identification of early and complementary therapeutic targets to elucidate and mitigate residual risks in patient care. The gut microbiota acts as a sentinel between internal and external environments, transmitting modified risks associated with these factors to the host. Imidazole propionate (ImP), a histidine metabolite originating from the gut microbiota, gained attention after being found to impair glucose tolerance and insulin signaling several years ago. Epidemiological studies over the past five years have demonstrated a robust correlation between ImP and an increased risk of onset of type 2 diabetes (T2D) and obesity, exacerbation of kidney traits in chronic kidney disease (CKD), progression of atherosclerotic plaques, and elevated mortality rates in heart failure (HF). These findings suggest that ImP may serve as a pivotal target for the prevention and treatment of cardiometabolic diseases. Mechanistic insights have uncovered associations between ImP and insulin resistance, impaired glucose metabolism, chronic inflammation, and intestinal barrier damage. This review provides a comprehensive summary of the current evidence regarding the association between ImP and cardiometabolic impairment, highlighting its potential in advancing personalized approaches to disease prevention and management, and exploring the intricate interplay of diet, gut microbiota, and ImP in cardiovascular metabolic impairment. Overall, this review offers valuable insights into the multifaceted roles of ImP in cardiometabolic diseases, identifies current knowledge gaps, and discusses future research directions.
Collapse
Affiliation(s)
- Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, The Afiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fangyuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxia Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jiao Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, China, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
49
|
Li Y, Feng T, Zhao Y, Zhang X, Chen H, Xia P, Yang D, Liang Z. Medicinal and edible homologous poly/oligo-saccharides: Structural features, effect on intestinal flora and preventing and treating type 2 diabetes, and their applications: A review. Int J Biol Macromol 2025; 305:141031. [PMID: 39965679 DOI: 10.1016/j.ijbiomac.2025.141031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is the third most common chronic metabolic disorder worldwide and seriously dangerous. Novel therapeutics are sought due to the paucity of safe and effective metabolic disorder-related diabetes medicines. Intestinal flora impacts glucose and lipid balance, making it a unique T2DM therapeutic target. Due to gut fermentation, poly/oligo-saccharides are highly beneficial prebiotic carbohydrates for intestinal health. Moreover, supplementation with naturally occurring medicinal and edible homologous traditional Chinese medicines (MEHTCM) poly/oligo-saccharides has significant antidiabetic effects with few side effects. Now, a comprehensive review of research developments of MEHTCM poly/oligo-saccharides was presented to explore their prospects. We outlined the structural characteristics, structure classification, and structure-activity relationships. Notably, structure-activity relationships illustrated that molecular weight, monosaccharide composition, and glycosidic bond type could influence the hypoglycemic activity and prebiotic effect of MEHTCM poly/oligo-saccharides. Additionally, the review systematically summarized the effect and potential mechanism of MEHTCM poly/oligo-saccharide on T2DM, focusing on gut microbiota. The potential applications in formulations for special medical purposes, common food, health care product, agriculture and other fields have also been summarized. This review emphasizes MEHTCM poly/oligo-saccharides' potential as prebiotics for T2DM treatment. This information provides new insights and a theoretical foundation for MEHTCM poly/oligo-saccharide nutritional and medicinal research.
Collapse
Affiliation(s)
- Yuan Li
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Tinghui Feng
- College of Life Sciences, Northwest A & F University, Xi'an 710000, China
| | - Yaxin Zhao
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xiaodan Zhang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Haimin Chen
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Pengguo Xia
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Dongfeng Yang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zongsuo Liang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
50
|
Park W, Lee SK, Gwack J, Lee SY, Cho YG, Kang SB, Park J. Dysbiosis of Bile Microbiota in Cholangiocarcinoma Patients: A Comparison with Benign Biliary Diseases. Int J Mol Sci 2025; 26:1577. [PMID: 40004041 PMCID: PMC11855699 DOI: 10.3390/ijms26041577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Dysbiosis in the bile microbiota of cholangiocarcinoma (CCA) patients suggests a potential role for microbial alterations in the pathogenesis of CCA. This study aimed to investigate bile microbial communities in patients with CCA and compare them to those in individuals with benign biliary diseases as a control (CTR) group. Microbial profiling was conducted using next-generation sequencing (NGS), targeting the V3-V4 regions of the 16S rRNA gene, followed by bioinformatics analysis using the VSEARCH and EzBioCloud platforms. Alpha and beta diversity analyses were performed to assess microbial richness and structural differences. The linear discriminant analysis effect size (LEfSe) was utilized to identify potential microbial biomarkers. Results: This study identified distinct microbial profiles in the two groups at both the phylum and genus levels. In the CTR group, Pseudomonadota (65%) was the dominant phyla, while Bacillota (49%) was more abundant in the CCA group. At the genus level, Escherichia (29%), Enterobacteriaceae (12%), Enterococcus (8%), Ralstonia (8%), and Clostridium (5%) were more prevalent in the CTR group, whereas Streptococcus (34%), Ralstonia (8%), and Veillonella (5%) were dominant in the CCA group. Although an alpha diversity analysis showed no statistically significant differences in species richness or diversity between groups, a beta diversity analysis revealed significant structural differences associated with disease severity. Our comparative microbiome study using LEfSe analysis suggested a statistically significant inhibition of normal intestinal bacterial flora in patients with CCA who had not received any treatment. These findings suggest that microbial dysbiosis may play a role in the pathogenesis of CCA. Specific microbial taxa were identified as potential biomarkers for distinguishing benign from malignant diseases. These results underscore the potential role of microbial dysbiosis in CCA pathogenesis and highlight the bile microbiota's utility as a diagnostic marker for biliary diseases.
Collapse
Affiliation(s)
- Wonsuk Park
- Division of Gastroenterology, Department of Internal Medicine, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Sang Kuon Lee
- Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jin Gwack
- Department of Prevention Medicine, College of Medicine, Jeonbuk National University, Jeonju 54907, Republic of Korea;
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (S.Y.L.); (Y.G.C.)
| | - Seung Yeob Lee
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (S.Y.L.); (Y.G.C.)
- Department of Laboratory Medicine, Jeonbuk National University College of Medicine and Hospital, Jeonju 54907, Republic of Korea
| | - Yong Gon Cho
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (S.Y.L.); (Y.G.C.)
- Department of Laboratory Medicine, Jeonbuk National University College of Medicine and Hospital, Jeonju 54907, Republic of Korea
| | - Sang-Bum Kang
- Division of Gastroenterology, Department of Internal Medicine, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Joonhong Park
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (S.Y.L.); (Y.G.C.)
- Department of Laboratory Medicine, Jeonbuk National University College of Medicine and Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|