1
|
Palm CL, de Wit S, Gorter TM, Rienstra M, Vos MJ, Kema IP, van der Ley CP, Bakker SJL, Bakker BM, de Boer RA, van Veldhuisen DJ, Meijers WC, Westenbrink BD. Beyond the gut: Systemic levels of short-chain fatty acids are altered in patients with heart failure. Int J Cardiol 2025; 428:133124. [PMID: 40068788 DOI: 10.1016/j.ijcard.2025.133124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND & AIM The gut microbiome produces short-chain fatty acids (SCFAs), which serve as a substantial energy source and provide a link between the microbiome and (cardiac) metabolism. It has been demonstrated that the composition of the microbiome is altered in patients with heart failure (HF), but whether circulating levels of SCFAs are altered in HF is unknown. METHODS & RESULTS Serum concentrations of the SCFAs acetate, propionate, and butyrate were measured in 205 patients with HF and in 54 healthy controls, using isotope dilution liquid chromatography-tandem mass spectrometry. Of the patients with HF, 99 had HF with a reduced ejection fraction (HFrEF) and 106 had HF with mildly-reduced or preserved ejection fraction (HFmrEF/HFpEF). Healthy controls were age and sex matched to the HFrEF patients. Serum concentrations of acetate and propionate were significantly lower in patients with HF than in healthy controls, whereas butyrate levels were higher in patients with HF. Analyses by HF type revealed that acetate and propionate levels were lower in both HFrEF and HFpEF/HFmrEF patients in comparison to healthy controls. However, butyrate levels were observed to be lower in patients with HFmrEF/HFpEF in comparison to healthy controls, while they were higher in patients with HFrEF. CONCLUSIONS In patients with HF, serum levels of acetate and propionate are lower across the HF spectrum, whereas serum butyrate levels are elevated in HFrEF, but lower in HFmrEF/HFpEF. These alterations in SCFA profiles suggest a microbiome-driven metabolic dysregulation, which appears to differ between HF subtypes.
Collapse
Affiliation(s)
- C L Palm
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Institute of Clinical Chemistry and Laboratory Medicine, Oldenburg Clinic, University of Oldenburg, Germany
| | - S de Wit
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - T M Gorter
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rienstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M J Vos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - I P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - C P van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - S J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - R A de Boer
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - D J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - W C Meijers
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - B D Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
2
|
Xia T, Nie Y, Chen Y, Zhang N, Wang Y, Liu S, Bai X, Cao H, Xu Y, Wang M. Structural and physicochemical properties and changes in vitro digestion and fermentation of soluble dietary fiber from tea residues modified by fermentation. Food Chem 2025; 473:142926. [PMID: 39884229 DOI: 10.1016/j.foodchem.2025.142926] [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: 10/23/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025]
Abstract
The aim of this study was to investigate the structure, physicochemical properties, and changes in vitro digestion and fermentation between unfermented tea residue dietary fiber (UDF) and fermented tea residue soluble dietary fiber (FSDF). The results showed that soluble dietary fiber in FSDF was increased from 2.54 % to 15.65 % after fermentation modification. Monosaccharide composition analysis showed that xylose and glucose accounted for a higher proportion in FSDF. FSDF had smaller particle size, lower crystallinity and higher thermal stability. The water holding capacity, oil holding capacity and water swelling capacity of FSDF were significantly increased. Rheological properties showed that FSDF exhibited higher viscosity and better elastic than UDF. Furthermore, FSDF generated more short-chain fatty acids, and the structure was looser than UDF, which was easier to be utilized by intestinal flora. These findings provided higher value of dietary fiber in tea residue by fermentation modification as functional products.
Collapse
Affiliation(s)
- Ting Xia
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yaning Nie
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yang Chen
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nannan Zhang
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yongqi Wang
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shunhang Liu
- Yunnan Tasly Deepure Bio-Tea Group Co, Ltd, Yunnan 665099, China
| | - Xiaoli Bai
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd, Tianjin 300410, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, 300052, China
| | - Yongquan Xu
- Yunnan Tasly Deepure Bio-Tea Group Co, Ltd, Yunnan 665099, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd, Tianjin 300410, China.
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety/Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
3
|
Qu Y, An K, Wang D, Yu H, Li J, Min Z, Xiong Y, Xue Z, Mao Z. Short-Chain Fatty Acid Aggregates Alpha-Synuclein Accumulation and Neuroinflammation via GPR43-NLRP3 Signaling Pathway in a Model Parkinson's Disease. Mol Neurobiol 2025; 62:6612-6625. [PMID: 39904963 DOI: 10.1007/s12035-025-04726-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Parkinson's disease (PD) is characterized by the aggregation of α-synuclein (α-syn) and the loss of dopaminergic (DA) neurons, with growing evidence suggesting a significant role of gut microbiota and their metabolites in the disease's pathogenesis. This study explores the effects of short-chain fatty acids (SCFAs) on PD progression, focusing on the G protein-coupled receptor 43 (GPR43) and the NLRP3 signaling pathway in both in vitro and in vivo models. Employing the1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model and SH-SY5Y cells with SCFAs-treated, this study investigated the impact of SCFAs on α-syn deposition, DA loss, and neuroinflammation. In vitro, supernatant from STC-1 cells was administered to SH-SY5Y cells, and the effects were assessed following the knockdown of NLRP3 or GPR43. In vivo, mice were treated with NLRP3 or GPR43 inhibitors after feeding with SCFAs, and the motor deficits, α-syn pathology, DA neuronal loss, and inflammatory responses were observed. SCFAs were found to exacerbate motor and gastrointestinal dysfunctions in PD models, intensifying α-syn pathology and neuroinflammation. The activation of the NLRP3 inflammasome through GPR43 emerged as a key pathological mechanism, with inhibition of these molecules mitigating the observed effects. Such interventions reduced α-syn accumulation, DA loss, and inflammatory responses, highlighting the pivotal role of the SCFA/GPR43-NLRP3 pathway in PD. The findings from this study elucidate a critical link between gut-derived metabolic changes and neuroinflammatory processes in PD via the SCFA/GPR43-NLRP3 pathway. Targeting this pathway offers a promising therapeutic strategy and enriches our understanding of the gut-brain axis' role in PD progression.
Collapse
Affiliation(s)
- Yi Qu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke An
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Danlei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haoheng Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingyi Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhe Min
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongjie Xiong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zheng Xue
- Department of General Practice, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijuan Mao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Kaulpiboon J, Rudeekulthamrong P. Maltotriosyl-erythritol, a transglycosylation product of erythritol by Thermus sp. amylomaltase and its application to prebiotic. Food Chem 2025; 472:142937. [PMID: 39827568 DOI: 10.1016/j.foodchem.2025.142937] [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: 10/30/2024] [Revised: 12/27/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
In this study, maltotriosyl-erythritol (EG3) was synthesized as a novel prebiotic candidate via transglycosylation using recombinant amylomaltase (AMase) from Thermus sp. Tapioca starch served as the glucosyl donor, and erythritol as the acceptor. High-performance liquid chromatography (HPLC) revealed an EG3 yield of 14.0 % with a concentration of 2.8 mg/mL. Mass spectrometry confirmed the molecular weight of EG3 as 608 Da, and its strucopture was verified by 1H and 13C NMR analysis. EG3 exhibited greater resistance to acid, heat, and digestive enzymes compared to erythritol glucosides (EG1-2) and significantly promoted the growth of Lactobacillus casei BCC36987. Fermentation of EG3 resulted in the highest levels of lactic acid and total short-chain fatty acids, which may contribute to reduced pH levels. These findings suggest that erythritol-receptor products formed via AMase-catalyzed reactions, particularly EG3, are promising prebiotic ingredients, with the prebiotic activity of erythritol derivatives being influenced by the length of the carbohydrate chain.
Collapse
Affiliation(s)
- Jarunee Kaulpiboon
- Division of Biochemistry, Department of Pre-Clinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Prakarn Rudeekulthamrong
- Department of Biochemistry, Phramongkutklao College of Medicine, Phramongkutklao Hospital, Bangkok 10400, Thailand.
| |
Collapse
|
5
|
Wu B, Tang Y, Zhao L, Gao Y, Shen X, Xiao S, Yao S, Qi H, Shen F. Integrated network pharmacological analysis and multi-omics techniques to reveal the mechanism of polydatin in the treatment of silicosis via gut-lung axis. Eur J Pharm Sci 2025; 207:107030. [PMID: 39929376 DOI: 10.1016/j.ejps.2025.107030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/05/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025]
Abstract
Silicosis is a pulmonary disease characterized by inflammation and progressive fibrosis. Previous studies have shown that polydatin (PD) has potential biological activity in key signaling pathways regulating inflammation and apoptosis. To investigate the effect of PD on rats with silicosis, this study used network pharmacology and molecular docking methods to determine the target of PD treatment for silicosis. The therapeutic effect of PD on silicosis was confirmed by measuring the lung injury score, hydroxyproline content, and mRNA expression levels of key targets. In addition, metagenomic sequencing and gas chromatography-mass spectrometry were used to determine the gut microbiota composition and targeted metabolomics analysis, respectively. The results showed that PD could inhibit the expression of inflammation-related indexes and apoptosis-related indexes at protein and mRNA levels. PD also regulates the diversity of the intestinal flora and the content of short-chain fatty acids. In conclusion, the current data suggest that PD has a protective effect against silica-induced lung injury and plays a protective role in regulating intestinal flora diversity and short-chain fatty acid levels through the gut-lung axis.
Collapse
Affiliation(s)
- Bingbing Wu
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China
| | - Yiwen Tang
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China
| | - Liyuan Zhao
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China
| | - Yan Gao
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China
| | - Xi Shen
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China
| | - Shuyu Xiao
- Tangshan Center of Disease Control and Prevention, Tangshan, Hebei, 063000, PR China
| | - Sanqiao Yao
- Xinxiang Medical University, Xinxiang, Henan, 453003, PR China
| | - Huisheng Qi
- Tangshan City workers' Hospital, Tangshan, Hebei, 063000, PR China.
| | - Fuhai Shen
- Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, PR China.
| |
Collapse
|
6
|
Tobias J, Heinl S, Dendinovic K, Ramić A, Schmid A, Daniel C, Wiedermann U. The benefits of Lactiplantibacillus plantarum: From immunomodulator to vaccine vector. Immunol Lett 2025; 272:106971. [PMID: 39765312 DOI: 10.1016/j.imlet.2025.106971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Probiotics have been increasingly recognized for positively influencing many aspects of human health. Lactiplantibacillus plantarum (L. plantarum), a non-pathogenic bacterium, previously known as Lactobacillus plantarum, is one of the lactic acid bacteria commonly used in fermentation. The probiotic properties of L. plantarum have highlighted its health benefits to humans when consumed in adequate amounts. L. plantarum strains primarily enter the body orally and alter intestinal microflora and modulate the immune responses in their host; thereby benefiting human health. Furthermore, the use of L. plantarum as vaccine vectors delivering mucosal antigens has been shown to be a promising strategy. These aspects, from Immunomodulation to vaccine delivery by L. plantarum in preclinical settings, are highlighted in this review. Along these lines, construction of a recombinant L. plantarum strain expressing a B cell multi-peptide, as a future vaccine to modulate immunity and confer anti-tumor effect by targeting Her-2/neu-overexpressing cancers in local and distal sites, is also presented and discussed.
Collapse
Affiliation(s)
- Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Stefan Heinl
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Kristina Dendinovic
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ajša Ramić
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Anna Schmid
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Catherine Daniel
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Sangfuang N, McCoubrey LE, Awad A, Marzorati M, Ghyselinck J, Verstrepen L, Munck JD, Medts JD, Gaisford S, Basit AW. Effects of senotherapeutics on gut microbiome dysbiosis and intestinal inflammation in Crohn's disease: A pilot study. Transl Res 2025; 278:36-47. [PMID: 39986536 DOI: 10.1016/j.trsl.2025.02.004] [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: 05/30/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation in the gastrointestinal tract, and is usually accompanied by dysbiosis in the gut microbiome, a factor that contributes to disease progression. Excessive production of reactive oxygen species (ROS) because of gut microbiome dysbiosis-one of the hallmark features of IBD-promotes chronic inflammation and facilitates the transformation of normal cells into senescent cells. Cellular senescence is associated with the development of various chronic and age-related diseases. We hypothesise that senolytic agents, specifically dasatinib (D) and quercetin (Q), could have a beneficial effect on both the gut microbiome and intestinal cells in IBD. The modulatory effects of a combination of D + Q was assessed in the M-SHIME model with faecal microbiota sourced from Crohn's disease patients. D + Q significantly modulated butyrate and lactate levels in the samples from specific patients. In addition, metabolomic analysis showed that D + Q positively impacted the abundance of anti-inflammatory bacteria while also significantly reducing the several species of pathogenic bacteria. Findings from a Caco-2 cell/THP1 co-culture model of IBD demonstrated that D + Q exerted strong immunomodulatory effects on the gut epithelium, evidenced by reduced NF-kB activity, and lower levels of the pro-inflammatory markers TNF-α, CXCL-10, and MCP-1. Furthermore, D + Q induced the secretion of anti-inflammatory cytokines, including IL-6 and IL-10. However, it should be noted that D + Q also led to the secretion of the pro-inflammatory cytokines IL-8. These findings suggest that D + Q could offer a novel therapeutic approach for advanced IBD management by modulating both the gut microbiome and inflammatory pathways. The results support the potential repurposing of senotherapeutic agents as a strategy for addressing the chronic inflammation central to IBD pathogenesis.
Collapse
Affiliation(s)
| | - Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Now at Drug Product Development, GSK R&D, Ware SG12 0GX, UK
| | - Atheer Awad
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, UK
| | | | | | | | | | | | - Simon Gaisford
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
8
|
He Y, Jia D, Chen W, Liu J, Liu C, Shi X. Discussion on the treatment of diabetic kidney disease based on the "gut-fat-kidney" axis. Int Urol Nephrol 2025; 57:1233-1243. [PMID: 39549180 DOI: 10.1007/s11255-024-04283-3] [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: 05/26/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024]
Abstract
Diabetic kidney disease is the main cause of end-stage renal disease, and its prevention and treatment are still a major clinical problem. The human intestine has a complex flora of hundreds of millions of microorganisms, and intestinal microorganisms, and their derivatives are closely related to renal inflammatory response, immune response, and material metabolism. Brown adipose tissue is the main part of adaptive thermogenesis. Recent studies have shown that activating brown fat by regulating intestinal flora has good curative effects in diabetic kidney disease-related diseases. As an emerging medical concept, the "gut-fat-kidney" axis has received increasing attention in diabetic kidney disease and related diseases. However, the specific mechanism involved needs further study. A new theoretical basis for the prevention and treatment of diabetic kidney disease is presented in this article, based on the "gut-fat-kidney" axis.
Collapse
Affiliation(s)
- Yaping He
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Dengke Jia
- Lanzhou University Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wenying Chen
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Juan Liu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Congrong Liu
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaowei Shi
- Department of Endocrinology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
9
|
Yakut A. Gut microbiota in the development and progression of chronic liver diseases: Gut microbiota-liver axis. World J Hepatol 2025; 17:104167. [DOI: 10.4254/wjh.v17.i3.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
The gut microbiota (GM) is a highly dynamic ecology whose density and composition can be influenced by a wide range of internal and external factors. Thus, “How do GM, which can have commensal, pathological, and mutualistic relationships with us, affect human health?” has become the most popular research issue in recent years. Numerous studies have demonstrated that the trillions of microorganisms that inhabit the human body can alter host physiology in a variety of systems, such as metabolism, immunology, cardiovascular health, and neurons. The GM may have a role in the development of a number of clinical disorders by producing bioactive peptides, including neurotransmitters, short-chain fatty acids, branched-chain amino acids, intestinal hormones, and secondary bile acid conversion. These bioactive peptides enter the portal circulatory system through the gut-liver axis and play a role in the development of chronic liver diseases, cirrhosis, and hepatic encephalopathy. This procedure is still unclear and quite complex. In this study, we aim to discuss the contribution of GM to the development of liver diseases, its effects on the progression of existing chronic liver disease, and to address the basic mechanisms of the intestinal microbiota-liver axis in the light of recent publications that may inspire the future.
Collapse
Affiliation(s)
- Aysun Yakut
- Department of Gastroenterology, İstanbul Medipol University Sefakoy Health Practice Research Center, İstanbul 38000, Türkiye
| |
Collapse
|
10
|
Chen R, Fu Z, Feng Z, Xiao F, Wang G. Association between atherogenic index of plasma and chronic diarrhea: a cross-sectional study of the NHANES 2005-2010. BMC Gastroenterol 2025; 25:201. [PMID: 40140782 DOI: 10.1186/s12876-025-03784-4] [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: 12/24/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Chronic diarrhea (CD), a common chronic condition resulting from various mechanisms, has chronic inflammation as a primary determinant. Despite recent research exploring the potential mechanisms linking lipids and diarrhea, clinical studies on the relationship between lipids and the onset of CD are limited. This study aimed to investigate the association between the atherogenic index of plasma (AIP) and CD risk. METHODS This cross-sectional study used data from the 2005-2010 NHANES. The association between AIP and CD was examined through multiple linear regression analyses. A smooth curve-fitting algorithm was applied to assess the potential non-linear dose-response relationship between AIP and CD, and subgroup analyses were conducted. RESULTS Among 5,948 participants, 440 (7.4%) had CD. After adjusting for potential confounders, AIP was significantly associated with CD (OR, 1.57; 95% CI, 1.08-2.30; P = 0.018). The highest AIP quartile (Q4; 0.18 to 0.92) showed an adjusted OR for CD of 1.51 (95% CI, 1.10-2.07; P = 0.011) versus the lowest quartile (Q1; -1.15 to -0.25). Subgroup analyses indicated that diabetic individuals with higher AIP had a higher CD risk (OR, 3.84; 95% CI, 1.45-10.15), with an observed additive interaction (P for interaction = 0.045). CONCLUSIONS This study demonstrates a significant association between AIP and CD risk. AIP may serve as a promising indicator for assessing CD risk, offering valuable insights for prevention and treatment strategies.
Collapse
Affiliation(s)
- Rongpeng Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Haizhu District, Guangzhou, 510260, China
| | - Zexin Fu
- The Second Clinical College, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou, 511436, China
| | - Zhicheng Feng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Haizhu District, Guangzhou, 510260, China
| | - Feng Xiao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Haizhu District, Guangzhou, 510260, China
| | - Guoqiang Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Haizhu District, Guangzhou, 510260, China.
| |
Collapse
|
11
|
Zhang W, Zong Y, Huang X, Liu K, Luo Z, Shan J, Di L. Cordyceps militaris alleviates COPD by regulating amino acid metabolism, gut microbiota and short chain fatty acids. JOURNAL OF ETHNOPHARMACOLOGY 2025:119701. [PMID: 40147677 DOI: 10.1016/j.jep.2025.119701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic obstructive pulmonary disease (COPD) is a global health challenge with the high morbidity and mortality. Cordyceps militaris (CM) is a medicinal fungus that has been widely used in Asia for centuries. It has the effects of tonifying the lung and kidney, replenishing essence, resolving phlegm, and stopping bleeding. CM has been used clinically for alleviating COPD in China. However, the potential mechanism of CM in treating COPD remains indistinct. PURPOSE This article aimed to evaluate the efficacy and investigate the underlying mechanism of CM in treatment of COPD. METHODS The ingredients in CM were identified by LC Q/TOF-MS. The effect of CM in COPD was evaluated. Untargeted metabolomics assay and 16S rDNA sequencing were employed to examine the changes in metabolites and gut microbiota in COPD mice. Gut microbiota ablation experiment and quantification of short chain fatty acids (SCFAs) were integrated to elucidate the systematic mechanism of CM in treatment of COPD. RESULTS A total of 22 ingredients were identified in CM. CM alleviated COPD significantly by improving lung function and inhibiting pulmonary inflammation. Subsequently, 11 differential metabolites regulated by CM were mainly associated with amino acid metabolism. CM ameliorated the dysbiosis of intestinal microbiota in COPD mice, which contributed to the treatment of COPD. Moreover, CM increased the contents of SCFAs, including acetate, propionate, butyrate and isobutyrate. Spearman correlation indicated a close relationship among pulmonary function, differential metabolites, and gut microbiota. CONCLUSIONS This study revealed that CM alleviated COPD by regulating amino acid metabolism, ameliorating the imbalance of gut microbiota and increasing the SCFAs. These findings not only establish a foundation for the research of CM but also provide a basis for new treatment strategies of COPD.
Collapse
Affiliation(s)
- Wen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| | - Yuqi Zong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Xiao Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Kai Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Zichen Luo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| |
Collapse
|
12
|
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
|
13
|
Yang S, Liu X, Chen Y, Wang X, Zhang Z, Wang L. NNSFMDA: A new microbe-drug association prediction model based on the bounded nuclear norm minimization and the simplified transformer. J Mol Biol 2025:169086. [PMID: 40139309 DOI: 10.1016/j.jmb.2025.169086] [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: 11/15/2024] [Revised: 02/21/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025]
Abstract
Identifying potential connections between microbe-drug pairs play an important role in drug discovery and clinical treatment.Techniques like graph neural networks effectively derive accurate node representations from sparse topologies,however, they struggle with over-smoothing and over-compression, and their interpretability is relatively poor.Conversely, mathematical methods with low-rank approximations are interpretable but often get trapped in local optima.To address these issues, we propose a new prediction model named NNSFMDA,in which, the bounded nuclear norm minimization and the simplified transformer were combined to infer possible drug-microbe associations. In NNSFMDA, we first constructed a heterogeneous microbe-drug network by integrating multiple microbe and drug similarity metrics,according to which, we subsequently transformed the prediction problem to a matrix filling problem, and then, iteratively approximated the matrix by minimizing the number of bounded nuclear norm. Finally, based on the newly-filled matrix, we introduced a simplified transformer to estimate possible scores of microbe-drug pairs. Results showed that NNSFMDA could achieve reliable AUC value of 0.98, Which outperformed existing state-of-the-art competitive methods. In the experimental section, ablation experiments and modular analyses further demonstrate the superiority of the model, and case studies of microbe-drug associations confirm the validity of the model. These tests have all highlighted the potential of the NNSFMDA to predict latent microbe-drug associations in the future.
Collapse
Affiliation(s)
- Shuyuan Yang
- Technology Innovation Center of Changsha, Changsha University, Changsha 410022, China; Big Data Innovation and Entrepreneurship Education Center of Hunan Province, Changsha University, Changsha 410022,China
| | - Xin Liu
- Technology Innovation Center of Changsha, Changsha University, Changsha 410022, China; Big Data Innovation and Entrepreneurship Education Center of Hunan Province, Changsha University, Changsha 410022,China
| | - Yiming Chen
- Technology Innovation Center of Changsha, Changsha University, Changsha 410022, China; Big Data Innovation and Entrepreneurship Education Center of Hunan Province, Changsha University, Changsha 410022,China
| | - Xiangyi Wang
- School of Artificial Intelligence, The University of New South Wales, Sydney, Australia
| | - Zhen Zhang
- Technology Innovation Center of Changsha, Changsha University, Changsha 410022, China; Big Data Innovation and Entrepreneurship Education Center of Hunan Province, Changsha University, Changsha 410022,China
| | - Lei Wang
- Technology Innovation Center of Changsha, Changsha University, Changsha 410022, China; Big Data Innovation and Entrepreneurship Education Center of Hunan Province, Changsha University, Changsha 410022,China
| |
Collapse
|
14
|
Fernández-Veledo S, Grau-Bové C, Notararigo S, Huber-Ruano I. The role of microbial succinate in the pathophysiology of inflammatory bowel disease: mechanisms and therapeutic potential. Curr Opin Microbiol 2025; 85:102599. [PMID: 40132355 DOI: 10.1016/j.mib.2025.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition linked to gut microbiota dysbiosis and altered production of bacterial metabolites, including succinate, which is also a key intermediate in human mitochondrial energy metabolism in human cells. Succinate levels in the gut are influenced by microbial community dynamics and cross-feeding interactions, highlighting its dual metabolic and ecological importance. Extracellular succinate acts as a key signaling metabolite linking microbial metabolism to host physiology, with transient rises supporting metabolic regulation but chronic elevations contributing to metabolic disorders and disease progression. Succinate signals through its cognate receptor SUCNR1, which mediates adaptive metabolic responses under normal conditions but drives inflammation and fibrosis when dysregulated. IBD patients display a dysbiotic gut microbiota characterized by an increased prevalence of succinate-producing bacteria, contributing to elevated succinate levels in the gut and circulation. This imbalance drives inflammation, worsens IBD severity, and contributes to complications like Clostridioides difficile infection and fibrosis. Emerging evidence highlights the potential of intestinal and systemic succinate levels as indicators of microbial dysbiosis, with a bidirectional relationship between microbial composition and succinate metabolism. Understanding the factors influencing succinate levels and their interaction with dysbiosis shows promise in the development of therapeutic strategies to restore microbial balance. Approaches such as dietary fiber enrichment, prebiotics, and probiotics to enhance succinate-consuming bacteria, combined with targeted modulation of succinate pathways (e.g. SDH inhibitors, SUCNR1 antagonists), hold promise for mitigating inflammation and improving gut health in IBD.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department de Ciències Mèdiques Bàsiques, University Rovira i Virgili, Tarragona, Spain.
| | - Carme Grau-Bové
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; SucciPro, S.L, Barcelona, Spain
| | - Sara Notararigo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; SucciPro, S.L, Barcelona, Spain
| | - Isabel Huber-Ruano
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; SucciPro, S.L, Barcelona, Spain.
| |
Collapse
|
15
|
Bagheri R, Daneshi SS, Bina S, Haghshenas M, Khoshnoud MJ, Asadi-Yousefabad SL, Khodaei F, Rashedinia M. Metformin Mitigates the Impact of Arsenic Exposure on the Maternal and Offspring Reproductive System of Female Mice. Biol Trace Elem Res 2025:10.1007/s12011-025-04577-2. [PMID: 40119994 DOI: 10.1007/s12011-025-04577-2] [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: 01/09/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Exposure to arsenic causes health problems and is associated with adverse effects on fertility and development. Humans are facing increasing exposure to arsenic from multiple sources, such as drinking water, food products, and industrial processes. The mechanisms behind arsenic-induced reproductive toxicity and its impact on fertility and the development of future generations are investigated by the protective role of metformin (200 mg/kg) against arsenic-induced (20 ppm As2O3) ovarian damage in both maternal and offspring generations. Results showed arsenic exposure caused significant weight loss, increased mortality, reduced serum anti-Mullerian hormone (AMH) levels, and heightened oxidative stress, indicated by increased reactive oxygen species (ROS), malondialdehyde (MDA), and reduced ovarian antioxidant activity. Gene expression changes related to apoptosis and inflammation, such as BAX, Bcl-2, Bcl-2, caspase-3, tumor necrosis factor-alpha (TNF-α), and interleukin-1 (IL-1), were also noted, along with a decrease in HO-1 expression. Arsenic exposure led to a reduction in ovarian follicles and an increase in atretic follicles and uterine thickness. However, metformin significantly reduced ROS and MDA levels, enhanced antioxidant capacity, and protected ovarian tissue by upregulating heme oxygenase-1 (HO-1) and Bcl-2, modulating apoptotic and inflammatory genes, and preserving AMH levels. The possible protective role of metformin against arsenic-induced toxicity and its detrimental effects aims to improve therapeutic approaches to alleviate the harmful consequences of environmental pollutants, especially arsenic.
Collapse
Affiliation(s)
- Razieh Bagheri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Sajad Daneshi
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samaneh Bina
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Marziyeh Haghshenas
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Khoshnoud
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Forouzan Khodaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Rashedinia
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
16
|
Rodríguez-Arellano SN, González-Gómez JP, Gomez-Gil B, González-Ávila M, Palomera-Hernández JR, Barrón-Cabrera E, Vergara-Jiménez MDJ, Chaidez C. A Two-Phage Cocktail Modulates Gut Microbiota Composition and Metabolic Profiles in an Ex Vivo Colon Model. Int J Mol Sci 2025; 26:2805. [PMID: 40141446 DOI: 10.3390/ijms26062805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Bacteriophage therapy is a promising approach for targeting antibiotic-resistant bacteria and modulating gut microbiota in metabolic diseases such as obesity. This study evaluated the impact of a two-phage cocktail on an ex vivo colonic simulation model of gut microbiota derived from obese individuals, both in its normalized state and after enrichment with Enterobacter cloacae, an obesity-related bacteria. Microbiological analyses confirmed that the phage cocktail remained active throughout the colonic regions over three digestion cycles and effectively reduced enterobacterial populations in the enriched microbiota. Metabarcoding of the 16S rRNA gene revealed that phage therapy did not significantly alter the abundance of dominant genera, but selectively reduced E. cloacae across all colonic regions. Alpha diversity was significantly affected only in the enriched microbiota, while beta diversity analysis indicated significant compositional shifts during therapy, with reduced dispersion in the final treatment stage. Short-chain fatty acid profiling demonstrated region- and group-specific metabolic responses, with increased lactic and butyric acid concentrations in the ascending colon of the enriched microbiota following phage treatment. This study provides the first ex vivo evidence that a two-phage cocktail can selectively eliminate E. cloacae while preserving overall microbiota structure and functionality. These findings establish a foundation for future in vivo studies exploring the role of phage therapy in reshaping gut microbial communities and metabolic profiles, highlighting its potential as a precision tool for managing gut dysbiosis in metabolic disorders.
Collapse
Affiliation(s)
| | - Jean Pierre González-Gómez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Culiacan 80110, Sinaloa, Mexico
| | - Bruno Gomez-Gil
- Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlan 82112, Sinaloa, Mexico
| | - Marisela González-Ávila
- Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Juan Ramón Palomera-Hernández
- Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Guadalajara 44270, Jalisco, Mexico
| | - Elisa Barrón-Cabrera
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacan 80019, Sinaloa, Mexico
| | | | - Cristobal Chaidez
- Laboratorio Nacional para la Investigación en Inocuidad Alimentaria (LANIIA), Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Culiacan 80110, Sinaloa, Mexico
| |
Collapse
|
17
|
Zhu B, Gu Z, Hu H, Huang J, Zeng Z, Liang H, Yuan Z, Huang S, Qiu Y, Sun XD, Liu Y. Altered Gut Microbiota Contributes to Acute-Respiratory-Distress-Syndrome-Related Depression through Microglial Neuroinflammation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0636. [PMID: 40110391 PMCID: PMC11919824 DOI: 10.34133/research.0636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Acute respiratory distress syndrome (ARDS) survivors often suffer from long-term psychiatric disorders such as depression, but the underlying mechanisms remain unclear. Here, we found marked alterations in the composition of gut microbiota in both ARDS patients and mouse models. We investigated the role of one of the dramatically changed bacteria-Akkermansia muciniphila (AKK), whose abundance was negatively correlated with depression phenotypes in both ARDS patients and ARDS mouse models. Specifically, while fecal transplantation from ARDS patients into naive mice led to depressive-like behaviors, microglial activation, and intestinal barrier destruction, colonization of AKK or oral administration of its metabolite-propionic acid-alleviated these deficits in ARDS mice. Mechanistically, AKK and propionic acid decreased microglial activation and neuronal inflammation through inhibiting the Toll-like receptor 4/nuclear factor κB signaling pathway. Together, these results reveal a microbiota-dependent mechanism for ARDS-related depression and provide insight for developing a novel preventative strategy for ARDS-related psychiatric symptoms.
Collapse
Affiliation(s)
- Bowen Zhu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zheng Gu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongbin Hu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Huang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoxuan Liang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Ziyi Yuan
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Shiwei Huang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yuetan Qiu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiang-Dong Sun
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
18
|
Bañares C, Paterson S, Gómez-Garre D, Ortega-Hernández A, Sánchez-González S, Cueva C, de la Fuente MÁ, Hernández-Ledesma B, Gómez-Cortés P. Modulation of Gut Microbiota and Short-Chain Fatty Acid Production by Simulated Gastrointestinal Digests from Microalga Chlorella vulgaris. Int J Mol Sci 2025; 26:2754. [PMID: 40141395 DOI: 10.3390/ijms26062754] [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: 02/18/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Chlorella vulgaris is a source of potential bioactive compounds that can reach the large intestine and interact with colonic microbiota. However, the effects of consumption of this microalga on gastrointestinal function have scarcely been studied. This paper simulates, for the first time, the passage of C. vulgaris through the gastrointestinal tract, combining the INFOGEST method and in vitro colonic fermentation to evaluate potential effects on the human colonic microbiota composition by 16S rRNA gene sequencing and its metabolic functionality. The results show that the presence of this microalga increased the release of short-chain fatty acids (SCFAs), such as acetic, propionic, butyric, and isobutyric fatty acids, after 48 h colonic fermentation, being indicators of gut health. In correlation with the release of SCFAs, a significant reduction in bacterial groups causing intestinal imbalance, such as Enterobacteriaceae, Enterococcus spp., and Staphylococcus spp., was observed. In addition, digests from C. vulgaris favored intestinal health-related taxa, such as Akkermansia and Lactobacillus. C. vulgaris is, therefore, a promising food ingredient for good intestinal health and the maintenance of a balanced colonic microbiota.
Collapse
Affiliation(s)
- Celia Bañares
- Institute of Food Science Research (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Samuel Paterson
- Institute of Food Science Research (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Dulcenombre Gómez-Garre
- Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), C/ Prof. Martín Lagos, 28040 Madrid, Spain
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Adv. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal, 28040 Madrid, Spain
| | - Adriana Ortega-Hernández
- Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), C/ Prof. Martín Lagos, 28040 Madrid, Spain
| | - Silvia Sánchez-González
- Microbiota and Vascular Biology Laboratory, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), C/ Prof. Martín Lagos, 28040 Madrid, Spain
| | - Carolina Cueva
- Institute of Food Science Research (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Miguel Á de la Fuente
- Institute of Food Science Research (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain
| | | | - Pilar Gómez-Cortés
- Institute of Food Science Research (CIAL, CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain
| |
Collapse
|
19
|
dos Santos PP, Fujimori ASS, Polegato BF, Okoshi MP. The Therapeutic Potential of Orange Juice in Cardiac Remodeling: A Metabolomics Approach. Metabolites 2025; 15:198. [PMID: 40137162 PMCID: PMC11944373 DOI: 10.3390/metabo15030198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Cardiovascular diseases are a leading cause of death worldwide, and the process of cardiac remodeling lies at the core of most of these diseases. Sustained cardiac remodeling almost unavoidably ends in progressive muscle dysfunction, heart failure, and ultimately death. Therefore, in order to attenuate cardiac remodeling and reduce mortality, different therapies have been used, but it is important to identify adjuvant factors that can help to modulate this process. One of these factors is the inclusion of affordable foods in the diet with potential cardioprotective properties. Orange juice intake has been associated with several beneficial metabolic changes, which may influence cardiac remodeling induced by cardiovascular diseases. Current opinion highlights how the metabolites and metabolic pathways modulated by orange juice consumption could potentially attenuate cardiac remodeling. It was observed that orange juice intake significantly modulates phospholipids, energy metabolism, endocannabinoid signaling, amino acids, and gut microbiota diversity, improving insulin resistance, dyslipidemia, and metabolic syndrome. Specifically, modulation of phosphatidylethanolamine (PE) metabolism and activation of PPARα and PPARγ receptors, associated with improved energy metabolism, mitochondrial function, and oxidative stress, showed protective effects on the heart. Furthermore, orange juice intake positively impacted gut microbiota diversity and led to an increase in beneficial bacterial populations, correlated with improved metabolic syndrome. These findings suggest that orange juice may act as a metabolic modulator, with potential therapeutic implications for cardiac remodeling associated with cardiovascular diseases.
Collapse
Affiliation(s)
- Priscila Portugal dos Santos
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, Brazil; (A.S.S.F.); (B.F.P.); (M.P.O.)
| | | | | | | |
Collapse
|
20
|
Liu C, Li J, Liu R, Zhao G. The role of intestinal homeostasis in sevoflurane-induced myelin development and cognitive impairment in neonatal mice. Front Cell Infect Microbiol 2025; 15:1541757. [PMID: 40144591 PMCID: PMC11936920 DOI: 10.3389/fcimb.2025.1541757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background Inhalational anesthetic sevoflurane is commonly used in pediatric anesthesia. Multiple exposures to sevoflurane in early postnatal life have been associated with long-term abnormalities in myelin development and cognitive and memory impairments, although the underlying mechanisms remain incompletely elucidated. Disruption of gut microbiota is recognized as an important contributor to neurological diseases. Here, we explore the potential mechanisms underlying the abnormal myelin development induced by multiple sevoflurane exposures in neonatal rats by analyzing gut homeostasis. Methods Six-day-old (P6) C57BL/6 mice were exposed to 3% sevoflurane for 2 hours per day for three consecutive days. Mice exposed to a mixture of 60% nitrogen and oxygen under the same conditions and duration served as controls. Behavioral tests were conducted between P32 and P42. At P9 (24 hours after the last sevoflurane exposure) and P42 (after the completion of behavioral tests), intestinal and brain examinations were performed to investigate the effects of sevoflurane exposure during the lactation and adolescent periods on gut homeostasis and myelin development in mice. Subsequently, the ameliorative effects of butyrate supplementation on sevoflurane-induced abnormalities in myelin development and cognitive and memory impairments were observed. Results After repeated exposure to sevoflurane, neonatal mice developed persistent gut microbiota imbalance accompanied by a decrease in short-chain fatty acids. Short-term intestinal inflammation emerged, with damage to the mucus layer and barrier function. In the hippocampus and prefrontal cortex, the expression of genes and transcription factors related to oligodendrocyte differentiation and myelin development was significantly affected, and these changes persisted even after the exposure ended. There was a reduction in proteins associated with oligodendrocytes and myelin formation, which had a certain impact on memory and cognitive behavior. This study also explored the potential connections between microbiota, metabolism, the gut, the brain, and behavior. Timely supplementation with butyrate could effectively reverse these changes, indicating that gut homeostasis is crucial for brain neurodevelopment. Conclusion Multiple exposures to sevoflurane in neonatal mice disrupt gut homeostasis and affect oligodendrocyte differentiation and myelin development in the hippocampus and prefrontal cortex, inducing cognitive and memory impairments. Supplementation with butyrate can alleviate these changes.
Collapse
Affiliation(s)
- Chang Liu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinjie Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ruizhu Liu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Guoqing Zhao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin University, Changchun, China
| |
Collapse
|
21
|
Xie Y, Chen Q, Shan D, Pan X, Hu Y. Unraveling the role of the gut microbiome in pregnancy disorders: insights and implications. Front Cell Infect Microbiol 2025; 15:1521754. [PMID: 40125520 PMCID: PMC11925892 DOI: 10.3389/fcimb.2025.1521754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
The gut microbiota is the collective term for the microorganisms that reside in the human gut. In recent years, advances in sequencing technology and bioinformatics gradually revealed the role of gut microbiota in human health. Dramatic changes in the gut microbiota occur during pregnancy due to hormonal and dietary changes, and these changes have been associated with certain gestational diseases such as preeclampsia (PE) and gestational diabetes mellitus (GDM). Modulation of gut microbiota has also been proposed as a potential treatment for these gestational diseases. The present article aims to review current reports on the association between gut microbiota and gestational diseases, explore possible mechanisms, and discuss the potential of probiotics in gestational diseases. Uncovering the link between gut microbiota and gestational diseases could lead to a new therapeutic approach.
Collapse
Affiliation(s)
- Yupei Xie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Dan Shan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| | - Xiongfei Pan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- West China Second University Hospital, Sichuan University, Shuangliu Institute of Women’s and Children’s Health, Shuangliu Maternal and Child Health Hospital, Chengdu, China
| | - Yayi Hu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology, Qingbaijiang Maternal and Child Health Hospital, Chengdu, China
| |
Collapse
|
22
|
Sun M, Huang H, Tang H, Chen J, Chen W, Yang D. Effects of simulated digestion and prebiotics properties of polysaccharides extracted from Imperatae Rhizoma based on different pilot processes. Front Microbiol 2025; 16:1544261. [PMID: 40124890 PMCID: PMC11925942 DOI: 10.3389/fmicb.2025.1544261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Recent studies have highlighted the prebiotic potential of natural plant polysaccharides, demonstrating their role in promoting beneficial gut microbiota and improving health. However, research on the digestive properties and prebiotic activities of Imperatae Rhizoma Polysaccharides (IRPs) remains limited. This study investigated fresh Imperatae Rhizoma as the research object. After processing, dry Imperatae Rhizoma and carbonized Imperatae Rhizoma were prepared. Three polysaccharides from the fresh, dry, and carbonized Imperatae Rhizoma were extracted with traditional hot water. And another polysaccharide was obtained by cold water extraction from fresh Imperatae Rhizoma. Total four IRPs were extracted and named: IRPs-F, IRPs-D, IRPs-C, and IRPs-J. This study evaluated the prebiotic activity of four polysaccharides derived from the roots of thatch, demonstrating their resistance to digestion, their ability to promote probiotic growth, and their enhancement of short-chain fatty acid (SCFA) production. The final results show that four IRPs exhibit strong resistance to digestion and IRPs-F ability to promote the growth of beneficial probiotics, making it a promising candidate for functional foods aimed at improving intestinal health, immune regulation, and metabolic benefits. This research is highly relevant to food microbiology and holds significant potential for application in the functional food and gut health sectors.
Collapse
Affiliation(s)
- Mengge Sun
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
- College of Life Science, Jilin University, Changchun, China
| | - Haotian Huang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
- College of Life Science, Jilin University, Changchun, China
| | - Haibao Tang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
- College of Life Science, Jilin University, Changchun, China
| | - Jiajie Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
- College of Life Science, Jilin University, Changchun, China
| | - Wei Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| | - Dongsheng Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| |
Collapse
|
23
|
Li J, Hu X, Tao X, Li Y, Jiang W, Zhao M, Ma Z, Chen B, Sheng S, Tong J, Zhang H, Shen B, Gao X. Deconstruct the link between gut microbiota and neurological diseases: application of Mendelian randomization analysis. Front Cell Infect Microbiol 2025; 15:1433131. [PMID: 40115072 PMCID: PMC11922733 DOI: 10.3389/fcimb.2025.1433131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/21/2025] [Indexed: 03/23/2025] Open
Abstract
Background Recent research on the gut-brain axis has deepened our understanding of the correlation between gut bacteria and the neurological system. The inflammatory response triggered by gut microbiota may be associated with neurodegenerative diseases. Additionally, the impact of gut microbiota on emotional state, known as the "Gut-mood" relationship, could play a role in depression and anxiety disorders. Results This review summarizes recent data on the role of gut-brain axis in the pathophysiology of neuropsychiatric and neurological disorders including epilepsy, schizophrenia, Alzheimer's disease, brain cancer, Parkinson's disease, bipolar disorder and stroke. Also, we conducted a Mendelian randomization study on seven neurological disorders (Epilepsy, schizophrenia, Alzheimer's disease, brain cancer, Parkinson's disease, bipolar disorder and stroke). MR-Egger and MR-PRESSO tests confirmed the robustness of analysis against horizontal pleiotropy. Conclusions By comparing the protective and risk factors for neurological disorders found in our research and other researches, we can furtherly determine valuable indicators for disease evolution tracking and potential treatment targets. Future research should explore extensive microbiome genome-wide association study datasets using metagenomics sequencing techniques to deepen our understanding of connections and causality between neurological disorders.
Collapse
Affiliation(s)
- Jingqiu Li
- Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Xinyang Hu
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Xinyu Tao
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yuming Li
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Wan Jiang
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mingtao Zhao
- Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Zhehui Ma
- Second Clinical Medical College, Anhui Medical University, Hefei, China
| | - Bangjie Chen
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Shuyan Sheng
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Jiaye Tong
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Haibo Zhang
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| | - Bing Shen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiaomei Gao
- Frist Clinical Medical College, Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Fu J, Liang Z, Chen Z, Zhou Y, Xiong F, Liang Q, Gao H. Deciphering the Therapeutic Efficacy and Underlying Mechanisms of Dendrobium officinale Polysaccharides in the Intervention of Alzheimer's Disease Mice: Insights from Metabolomics and Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5635-5648. [PMID: 39536176 DOI: 10.1021/acs.jafc.4c07913] [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: 11/16/2024]
Abstract
As a traditional drug-food homologous plant, Dendrobium officinale is widely recognized for its nutritional and medicinal value. Specifically, D. officinale polysaccharide (DOP) has garnered attention as a potential prebiotic for its protective effects on gut microbiota and the nervous system. However, the underlying mechanism by which DOP improves cognitive dysfunction in Alzheimer's disease (AD) remains unclear. This study intends to elucidate the beneficial effects of DOP on AD mice from the perspectives of metabolomics and the intestinal microbiome. The results showed that DOP significantly ameliorated cognitive dysfunction, attenuated hippocampal neuronal damage and Aβ plaque deposition, and restored intestinal barrier integrity in AD mice. The antibiotic-cocktail-induced germ-free mouse model confirmed that the neuroprotective effect of DOP was dependent on gut microbiota. Further investigations demonstrated that DOP influenced the composition of gut microbiota and restored its diversity. Additionally, DOP reshaped metabolic profile disorders in AD mice and increased the short-chain fatty acids (SCFAs) content. Correlation analysis further highlighted that specific gut microbiota was associated with the metabolism of AD mice. In conclusion, this study sheds light on the positive impact of DOP in reshaping the gut microbiota and enhancing cognitive function, offering important perspectives for the possible advancement and utilization of DOP.
Collapse
Affiliation(s)
- Jun Fu
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhaohan Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Zihao Chen
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Yiyang Zhou
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Fen Xiong
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongchang Gao
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
25
|
Ku JY, Lee MJ, Jung Y, Choi HJ, Park J. Changes in the gut microbiome due to diarrhea in neonatal Korean indigenous calves. Front Microbiol 2025; 16:1511430. [PMID: 40109976 PMCID: PMC11921620 DOI: 10.3389/fmicb.2025.1511430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Studies on gut microbiome changes in neonatal Korean indigenous calves with diarrhea are rare. In this study, 14 normal calves and 11 calves with diarrhea were selected from Korean indigenous calves up to 30 days of age and classified into three groups at 10-day intervals (1-10, 11-20, and 21-30 days). Feces from 25 calves were collected, and the diversity, similarity, structure, and correlation of the gut microbiome were analyzed. Firmicutes, Bacteroidetes, and Proteobacteria were predominant in the taxonomic composition of the gut microbiome of the calves regardless of the presence of diarrhea. However, Proteobacteria increased and Bacteroidetes and Actinobacteria decreased in calves with diarrhea. In addition, calves with diarrhea showed a significant decrease in the diversity of the gut microbiome, especially for anaerobic microorganisms Faecalibacterium prausnitzii, Gemmiger formicilis, and Collinsella aerofaciens. The microbial communities in calves with diarrhea and normal calves were distinct. By analyzing the microorganisms that showed correlation with diarrhea and age using linear discriminant analysis effect size, at the genus level, Prevotella and Lachnospiraceae_uc were significantly related in the normal (11-20 days) group whereas Enterobacterales, Gammaproteobacteria, Enterobacteriaceae, Escherichia, and Proteobacteria were significantly associated with diarrhea in the 11-20 days group. Futhermore, the normal (21-30 days) group showed significant correlation with Blautia, Provotellaceae, Muribaculaceae, Christensenellaceae, and Catenella, whereas the diarrhea (21-30 days) group showed significant correlation with Dorea. The microorganisms associated with diarrhea in calves were mainly known as harmful microorganisms, we confirmed that there is a relationship between the increase in harmful bacteria and diarrhea. These results show that diarrhea significantly affects the gut microbiome of Korean indigenous calves. The changes in the gut microbiome of Korean indigenous calves observed in this study could be helpful in predicting and managing diarrhea calves, and furthermore, in establishing preventive measures for calf diarrhea through management of gut microbiome.
Collapse
Affiliation(s)
- Ji-Yeong Ku
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| | - Mi-Jin Lee
- Department of Veterinary Nursing, College of Health Science, Wonkwang University, Iksan, Republic of Korea
| | - Youngwoo Jung
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| | - Hak-Jong Choi
- Technology Innovation Research Division, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Jinho Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| |
Collapse
|
26
|
Xu P, Fan C, Yan M, Liu J, Zhang X. Remnant cholesterol and suicide attempts in untreated first-episode major depressive disorder. Front Psychiatry 2025; 16:1493509. [PMID: 40104336 PMCID: PMC11913861 DOI: 10.3389/fpsyt.2025.1493509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025] Open
Abstract
Objective The objective of this research was to investigate the relationship between remnant cholesterol (RC) levels and suicide attempts (SA) made by Chinese patients with untreated first-episode major depressive disorder (UFE MDD). Methods This study included 1718 patients with UFE MDD. Demographic, clinical characteristics, and blood lipid parameters were collected. The 17-item Hamilton Depression Rating Scale (HAMD), the 14-item Hamilton Anxiety Rating Scale (HAMA), and the positive subscale of the Positive and Negative Syndrome Scale (PANSS) were used to assess their depression, anxiety, and psychotic symptoms, respectively. Multivariable binary logistic regression analysis was used to estimate the association between RC and the risk of SA. A two-piecewise linear regression model was used to investigate the threshold effects if non-linear associations existed. Results Univariate logistic regression analysis showed a significant positive correlation between RC and SA, but after controlling for confounding factors, the association between them was not statistically significant. After dividing the RC into quartiles, only the RC in the Q4 group was significantly positively correlated with suicide attempts (OR = 1.73, 95% CI: 1.13-2.65, P = 0.012, vs. Q1) in a fully adjusted model. Curve fitting analysis also showed a nonlinear relationship between RC and suicide attempts with an inflection point at 1.99 mmol/L in RC. On the left of the inflection point, a significant positive correlation was observed between RC and SA (OR: 1.36, 95% CI: 1.09-1.69, p=0.006). However, on the right of the inflection point, no significant correlation was found (OR: 0.79, 95% CI: 0.55-1.14, p=0.214). Conclusion This study demonstrates a non-linear association between RC levels and SA in patients with untreated first-episode major depressive disorder. When RC was less than 1.99 mmol/L, they showed a significant positive correlation.
Collapse
Affiliation(s)
- Ping Xu
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Cheng Fan
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Mingxing Yan
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Junjun Liu
- Department of Psychiatry, Nanjing Meishan Hospital, Nanjing, China
- Medical College of Soochow University, Suzhou, China
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xiangyang Zhang
- Chinese Academy of Sciences (CAS) Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
- Min Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yan Liu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Li Zhong
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Fang Wu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Jinjin Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
29
|
Chen Y, Zheng K, Leng Y, Zhang Z, Li X, Li X, Ou H, Wen M, Qiu F, Yu H. Alleviating effect of Lactobacillus fermentum E15 on hyperlipidemia and hepatic lipid metabolism in zebrafish fed by a high-fat diet through the production of short-chain fatty acids. Front Nutr 2025; 12:1522982. [PMID: 40098735 PMCID: PMC11911183 DOI: 10.3389/fnut.2025.1522982] [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: 11/05/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Hyperlipidemia is regarded as one of the crucial factors leading to atherosclerosis and other cardiovascular diseases. Gut microbiota plays an important role in regulating host lipid metabolism. Nevertheless, the exact mechanisms behind this remain unclear. Methods In the present study, a hyperlipidemic zebrafish model was established using a high-cholesterol diet (HCD) to evaluate the anti-hyperlipidemic effects of Lactobacillus fermentum E15 (L. fermentum E15). Results Results showed that L. fermentum E15 effectively reduced lipid accumulation in the blood vessels and liver of HCD-fed zebrafish larvae. Meanwhile, L. fermentum E15 improved abnormal lipid levels, and normalized liver enzyme activity. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis revealed that L. fermentum E15 downregulated the expression of sterol regulatory element-binding factor (SREBP-1), peroxisome proliferator-activated receptor-gamma (PPAR-γ), and fatty acid synthase (Fasn), while upregulated peroxisome proliferator-activated receptor-alpha (PPAR-α). Additionally, metabolomic analysis revealed that L. fermentum E15 produced a series of short-chain fatty acids (SCFAs), including acetic acid, propionic acid, butyric acid, and isovaleric acid. Notably, isovaleric acid contributed to the reduction of lipid droplet accumulation in the liver and blood vessels of HCD-fed zebrafish larvae. In contrast, blocking G-protein coupled receptor 43 (GPR43) with pertussis toxin (PTX) abolished the effects of L. fermentum E15 and isovaleric acid on reducing lipid accumulation in HCD-fed zebrafish larvae. RT-qPCR results further suggested that both L. fermentum E15 and isovaleric acid promoted the expression of GPR43 and leptin A, which was inhibited by PTX. Conclusion These findings suggested that L. fermentum E15 alleviates HCD-induced hyperlipidemia by activating GPR43 through SCFAs.
Collapse
Affiliation(s)
- Yishu Chen
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Kangdi Zheng
- Guangdong Longseek Testing Co., Ltd., Guangzhou, China
| | - Yang Leng
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Zhao Zhang
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
- Guangdong Longseek Testing Co., Ltd., Guangzhou, China
| | - Xiaoling Li
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Xiaoyan Li
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Huajun Ou
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Muhao Wen
- Department of Laboratory Medicine, the Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Feng Qiu
- Department of Laboratory Medicine, the Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Huajun Yu
- Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
30
|
Li Y, Lyu L, Ding H. The potential roles of gut microbiome in porto-sinusoidal vascular disease: an under-researched crossroad. Front Microbiol 2025; 16:1556667. [PMID: 40099185 PMCID: PMC11911366 DOI: 10.3389/fmicb.2025.1556667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Accumulating evidence indicates that patients with liver diseases exhibit distinct microbiological profiles, which can be attributed to the bidirectional relationship of the gut-liver axis. Porto-sinusoidal vascular disease (PSVD) has recently been introduced to describe a group of vascular diseases of the liver, involving the portal venules and sinusoids. Although the pathophysiology of PSVD is not yet fully understood, several predisposing conditions, including immunodeficiency, inflammatory bowel disease, abdominal bacterial infections are associated with the increasing in intestinal permeability and microbial translocation, supporting the role of altered gut microbiota and gut-derived endotoxins in PSVD etiopathogenesis. Recent studies have proposed that the gut microbiome may play a crucial role in the pathophysiology of intrahepatic vascular lesions, potentially influencing the onset and progression of PSVD in this context. This review aims to summarize the current understanding of the gut microbiome's potential role in the pathogenesis of hepatic microvascular abnormalities and thrombosis, and to briefly describe their interactions with PSVD. The insights into gut microbiota and their potential influence on the onset and progression of PSVD may pave the way for new diagnostic, prognostic, and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing Youan Hospital Affiliated with Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Chalabi S, Loonen L, Boekhorst J, Li H, Fang L, Harrison PW, Lakhal W, Lluch J, Sokolov A, Djebali S, Rau A, Giuffra E, Wells J. Differences in maternal diet fiber content influence patterns of gene expression and chromatin accessibility in fetuses and piglets. Genomics 2025; 117:110995. [PMID: 39814241 DOI: 10.1016/j.ygeno.2025.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
This study investigates the impact of maternal gestation diets with varying fiber contents on gene expression and chromatin accessibility in fetuses and piglets fed a low fiber diet post weaning. High-fiber maternal diets, enriched with sugar beet pulp or pea internal fiber, were compared to a low-fiber maternal diet to evaluate their effects on liver and muscle tissues. The findings demonstrate that maternal high-fiber diets significantly alter chromatin accessibility, predicted transcription factor activity and transcriptional landscape in both fetuses and piglets. A gene set enrichment analysis revealed over-expression of gene ontology terms related to metabolic processes and under-expression of those linked to immune responses in piglets from sows given the high-fiber diets during gestation. This suggests better metabolic health and immune tolerance of the fetus and offspring, in line with the documented epigenetic effects of short chain fatty acids on immune and metabolic pathways. A deconvolution analysis of the bulk RNA-seq data was performed using cell-type specific markers from a single cell transcriptome atlas of adult pigs. These results confirmed that the transcriptomic and chromatin accessibility data do not reflect different cell type compositions between maternal diet groups but rather phenotypic changes triggered by maternal nutrition in shaping the epigenetic and transcriptional environment of fetus and offspring. Our findings have implications for improving animal health and productivity as well as broader implications for human health, suggesting that optimizing maternal diet with high-fiber content could enhance metabolic health and immune function in the formative years after birth and potentially to adulthood.
Collapse
Affiliation(s)
- Smahane Chalabi
- Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Linda Loonen
- Microbe Interactomics Group, Dept. Animal Sciences, Wageningen University & Research (WUR), Wageningen, the Netherlands
| | - Jos Boekhorst
- Microbe Interactomics Group, Dept. Animal Sciences, Wageningen University & Research (WUR), Wageningen, the Netherlands
| | - Houcheng Li
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus 8000, Denmark
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus 8000, Denmark
| | - Peter W Harrison
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, United Kingdom
| | - Wassim Lakhal
- Diagenode, Liège Science Park, Rue du Bois Saint-Jean 3, 4102 Liège, Belgium
| | - Jerome Lluch
- INRAE, US 1426, GeT-PlaGe, Genotoul, Castanet-Tolosan, France
| | - Alexey Sokolov
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, United Kingdom
| | - Sarah Djebali
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Andrea Rau
- Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France
| | - Elisabetta Giuffra
- Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France.
| | - Jerry Wells
- Microbe Interactomics Group, Dept. Animal Sciences, Wageningen University & Research (WUR), Wageningen, the Netherlands.
| |
Collapse
|
32
|
Berzack S, Galor A. Microbiome-based therapeutics for ocular diseases. Clin Exp Optom 2025; 108:115-122. [PMID: 39617011 PMCID: PMC11875938 DOI: 10.1080/08164622.2024.2422479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 12/08/2024] Open
Abstract
The relationship between the gut microbiome and ocular health has garnered increasing attention within the scientific community. Recent research has focused on the gut-eye axis, examining whether imbalances within the gut microbiome can influence the development, progression and severity of ocular diseases, including dry eye disease, uveitis, and glaucoma. Dysbiosis within the gut microbiome is linked to immune dysregulation, chronic inflammation, and epithelial barrier dysfunction, all of which contribute to ocular pathology. This review synthesises current evidence on these associations, exploring how gut microbiome alterations drive disease mechanisms. Furthermore, it examines the therapeutic potential of microbiome-targeted interventions, including antibiotics, prebiotics, probiotics, and faecal microbiota transplantation, all of which aim to restore microbial balance and modulate immune responses. As the prevalence of these conditions continues to rise, a deeper understanding of the gut-eye axis may facilitate the development of novel, targeted therapies to address unmet needs in the management of ocular diseases.
Collapse
Affiliation(s)
- Shannan Berzack
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
33
|
Costa de Almeida T, Sabino YNV, Brasiel PGDA, Rocha BMDO, de Cássia Ávila Alpino G, Rocha VN, Dias VC, Diniz CG, Paiva AD, Silva VLD, Dutra Medeiros J, Potente Dutra Luquetti SC, Barbosa Ferreira Machado A. Maternal kefir intake during lactation impacts the breast milk and gut microbiota of the Wistar rat's offspring. Int J Food Sci Nutr 2025; 76:179-193. [PMID: 39895284 DOI: 10.1080/09637486.2025.2461142] [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: 05/14/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Environmental factors can play fundamental role in health in childhood and adulthood during critical developmental periods like lactation. The maternal intake of probiotics like kefir during lactation could benefit newborns' intestinal health. This study aimed to evaluate the effects of maternal kefir intake during lactation on bacterial breast milk composition and the gut microbiota of offspring Wistar male rats at weaning. Lactating Wistar rats and their pups were divided into four groups based on litter size and maternal kefir intake. Sequencing of the 16S rRNA gene in breast milk revealed the predominance of the Proteobacteria, Firmicutes, and Actinobacteriota phyla. Offspring gut microbiota exhibited clustering tendencies in kefir groups with varying genus abundance. Additionally, maternal kefir intake led to increased levels of butyrate acid in offspring faeces (> +30%, p > 0.05). These findings show that the lactation period could be a window of opportunity to program intestinal health through microbiota modulation.
Collapse
Affiliation(s)
- Thaís Costa de Almeida
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Yasmin Neves Vieira Sabino
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | - Beatriz Macedo de Oliveira Rocha
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | - Vinícius Novaes Rocha
- Department of Veterinary Medicine, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Vanessa Cordeiro Dias
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Cláudio Galuppo Diniz
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Vânia Lúcia da Silva
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | | | | |
Collapse
|
34
|
Dominguez-Mozo MI, López-Mecández D, Villar LM, Costa-Frossard L, Villarrubia N, Aladro Y, Pilo B, Montalbán X, Comabella M, Casanova-Peño I, González-Suárez I, Martínez-Ginés ML, García-Domínguez JM, García-Calvo E, Machuca-Marcos A, Luque-Garcia JL, Garcia-Martinez MA, Arroyo R, Alvarez-Lafuente R. Short-chain fatty acids in multiple sclerosis: Associated with disability, number of T2 lesions, and inflammatory profile. Ann Clin Transl Neurol 2025; 12:478-490. [PMID: 40033709 PMCID: PMC11920722 DOI: 10.1002/acn3.52259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 03/05/2025] Open
Abstract
OBJECTIVE An alteration in the composition of the intestinal microbiota has been observed in patients with multiple sclerosis (pwMS) with respect to healthy controls (HC). Microorganism-derived metabolites such as short-chain fatty acids (SCFA) have been suggested to play a role in the disease. Thus, to analyze the association of SCFA with clinical and radiological parameters of the disease and with those related to the inflammatory response of the immune system. METHODS Multicentric observational retrospective cross-sectional study. In addition 161 pwMS and 130 HC were included. The following plasma SCFA were analyzed using liquid chromatography coupled to mass spectrometry: acetate (AA), propionate (PA) and butyrate (BA). Blood cell subpopulations and cytokine expression were analyzed by flow cytometry. RESULTS Plasma PA and PA/AA ratio was lower in pwMS than in HC (P = 0.0001, and P = 0.00005, respectively). PA/AA and BA/AA ratios were lower in pwMS with higher disability (P = 0.001, and P = 0.001, respectively). T2 lesion load inversely correlated with PA/AA (r = -0.353; P = 0.002) and BA/AA (r = -0.322; P = 0.005) ratios. Plasma PA/AA and/or BA/AA ratios negatively correlated with the following pro-inflammatory cytokines producing cells: GM-CSF+CD4+T, GM-CSF+CD8+T, TNF-alpha+CD4+T, TNF-alpha+CD8+T, IFN-gamma+CD4+T, IFN-gamma+CD8+T, and TNF-alpha+B cells. INTERPRETATION In MS, plasma PA/AA and BA/AA ratios are unbalanced, promoting an environment that could be boosting the mechanisms underlying the pathogenesis of the disease. Since we have found statistical significant associations with the EDSS and the number of T2 lesions, but not with the number of relapses or gadolinium enhancing lesions, PA/AA and BA/AA ratios could be more associated with those mechanisms of the disease related to the neurodegenerative processes than those related with the activity of the disease.
Collapse
Affiliation(s)
- Maria Inmaculada Dominguez-Mozo
- Grupo de Investigación de Factores ambientales en enfermedades degenerativas, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| | - Daniel López-Mecández
- Department of Clinical Analysis, Hospital Clínico San Carlos, Instituto de Medicina del Laboratorio (IML), Madrid, Spain
| | - Luisa María Villar
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| | - Lucienne Costa-Frossard
- Servicio de Neurología, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| | - Noelia Villarrubia
- Servicio de Inmunología, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| | - Yolanda Aladro
- Servicio de Neurología, Hospital Universitario de Getafe, Spain
| | - Belén Pilo
- Servicio de Neurología, Hospital Universitario de Getafe, Spain
| | - Xavier Montalbán
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ignacio Casanova-Peño
- Department of Neurology, Torrejón de Ardoz, España, School of Medicine, University Hospital Torrejón, Francisco de Vitoria University, Madrid, Spain
| | - Inés González-Suárez
- Unidad de enfermedades desmielinizantes, Hospital Álvaro Cunqueiro, Red de Enfermedades Inflamatorias (REI), Vigo, Spain
| | - María Luisa Martínez-Ginés
- Servicio de Neurología, Hospital General Universitario Gregorio Marañón/Red de Enfermedades Inflamatorias (REI), Madrid, Spain
| | - Jose Manuel García-Domínguez
- Servicio de Neurología, Hospital General Universitario Gregorio Marañón/Red de Enfermedades Inflamatorias (REI), Madrid, Spain
| | - Estefanía García-Calvo
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - Andrés Machuca-Marcos
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose Luis Luque-Garcia
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - María Angel Garcia-Martinez
- Grupo de Investigación de Factores ambientales en enfermedades degenerativas, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| | - Rafael Arroyo
- Departamento de Neurología, Hospital Universitario Quironsalud Madrid, Madrid, Spain
| | - Roberto Alvarez-Lafuente
- Grupo de Investigación de Factores ambientales en enfermedades degenerativas, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), Red Española de Esclerosis Múltiple, Madrid, Spain
| |
Collapse
|
35
|
Xu S, Xiong J, Qin X, Ma M, Peng Y, Cheng J, Nie X, Fan X, Deng Y, Ju Y, Liu J, Zhang L, Liu B, Zhang Y, Li L. Association between gut microbiota and perinatal depression and anxiety among a pregnancy cohort in Hunan, China. Brain Behav Immun 2025; 125:168-177. [PMID: 39736365 DOI: 10.1016/j.bbi.2024.12.150] [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: 06/22/2024] [Revised: 12/11/2024] [Accepted: 12/21/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Perinatal depression and anxiety pose significant risks to maternal health and may lead to suicide. The gut microbiota may play a crucial role in perinatal depression and anxiety. However, the relationship between the alterations in gut microbiota and perinatal depression and anxiety remains unclear. This study aimed to investigate the dynamic changes of gut microbiota over various perinatal stages and their associations with perinatal depression and anxiety symptoms, especially suicide ideation. METHODS A total of 177 pregnant and 19 postpartum women were recruited in this study, with 48 of them participating longitudinally. Maternal depression and anxiety symptoms were assessed using the Edinburgh Postnatal Depression Scale (EPDS), 9-item Patient Health Questionnaire (PHQ-9), and 7-item Generalized Anxiety Disorder Scale (GAD-7). Fecal samples collected during the perinatal period were analyzed using 16S rRNA gene sequencing. RESULTS Significant changes in microbial diversity and multi-taxonomic levels were observed during pregnancy. The random forest regression model showed significant associations of some gut microbial features with depression and anxiety symptoms. Several genera were significantly associated with gestation age and perinatal depression and anxiety, such as Akkermansia, Bifidobacterium and Streptococcus. In addition, Erysipelotrichaceae_UCG-003 and Eubacterium_hallii_group were positively associated with suicidal ideation. The glycine biosynthesis pathway might act as a mediator between Eubacterium_hallii_group and suicidal ideation (ab = 3.27, p < 0.05). CONCLUSION The gut microbiota undergoes a programmed shift during pregnancy, which may play a critical role in perinatal depression and anxiety. Our findings underscore the impact of certain bacterial genera and metabolic pathways on perinatal mental health, which may help to develop new diagnostic tools and targeted interventions to reduce perinatal mental disorders and improve the outcomes for both mothers and infants.
Collapse
Affiliation(s)
- Shuyin Xu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Jintao Xiong
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Xuemei Qin
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Mohan Ma
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Yilin Peng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Junzhe Cheng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Xueqing Nie
- Changsha Hospital for Maternal and Child Health Care, Changsha 410007, Hunan, China
| | - Xing Fan
- Changsha Hospital for Maternal and Child Health Care, Changsha 410007, Hunan, China
| | - Yali Deng
- Department of Obstetrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yumeng Ju
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Jin Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Li Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China
| | - Bangshan Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China.
| | - Yan Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China.
| | - Lingjiang Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Technology Institute of Psychiatry, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha 410011, Hunan, China.
| |
Collapse
|
36
|
Zhang J, Zhang X, Li G, Ge J, Feng X. Loureirin B Ameliorates Glycolipid Metabolism Disorders in Ob/ob Mice by Regulating Bile Acid Levels and Modulating Gut Microbiota Composition. Chem Biodivers 2025; 22:e202401793. [PMID: 39431713 DOI: 10.1002/cbdv.202401793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/22/2024]
Abstract
Loureirin B (LB), an active component of Resina Draconis, exhibits hypoglycemic and hypolipidemic effects; however, its mode of action remains unclear. Here, ob/ob mice were utilized to investigate the effects of LB on the regulation of glucolipid metabolism disorders. Non-targeted metabolomics and 16S rDNA sequencing were performed to elucidate the potential mechanisms involved. Results indicated that LB treatment (45 mg/kg) significantly improved glucose intolerance and insulin resistance, reduced lipid levels, and alleviated hepatic steatosis. Non-targeted metabolomics analysis revealed that LB treatment regulated bile acid levels. Quantification of liver bile acids demonstrated that LB treatment significantly decreased the ratio of 12α-OH to non-12α-OH bile acids in the liver. 16S rDNA sequencing results showed that LB treatment increased the abundance of short-chain fatty acid-producing microbiota while decreasing the abundance of bile salt hydrolase (BSH) enzyme-producing microbiota. In conclusion, LB ameliorates glucolipid metabolism disorders by regulating liver bile acid levels and modulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Junyang Zhang
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West District, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Xiaoyan Zhang
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West District, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Gen Li
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West District, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Jun Ge
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West District, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Xinchi Feng
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West District, Tuanbo New Town, Jinghai District, Tianjin, China
| |
Collapse
|
37
|
Sun X, Wang C, Li S, Liu X, Li Y, Wang Y, Niu Y, Ren Z, Yang X, Yang X, Liu Y. Folic acid alleviates the negative effects of dexamethasone induced stress on production performance in Hyline Brown laying hens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:54-65. [PMID: 39949729 PMCID: PMC11821403 DOI: 10.1016/j.aninu.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/24/2024] [Accepted: 11/28/2024] [Indexed: 02/16/2025]
Abstract
Multiple stressors are believed to deteriorate production performance and cause substantial economic losses in commercial poultry farming. Folic acid (FA) is an antioxidant compound that can improve oocyte function and regulate gut microbiota composition. The current study was conducted to investigate the role of FA in alleviating stress and improving production performance. Sixty Hyline Brown laying hens at 21 weeks of age were randomly divided into three groups, with 10 replicates in each group and each replicate containing two chickens. Each group received basic diet and saline injection (Con group), basic diet with dexamethasone (DXM) injection (DXM group), or basic diet supplemented with FA (13 mg/kg in the premix) with DXM injection (FA group). The feeding trial lasted five weeks. Birds in the DXM and FA groups receiving subcutaneous DXM injections at a dosage of 4.50 mg/kg per day during the first seven days of the trial. Results showed that the levels of corticosterone, triglyceride, total cholesterol, and malondialdehyde in serum were significantly increased in the DXM group (P < 0.05), while the concentrations of FA and 5-methyltetrahydrofolate were decreased in the DXM group (P < 0.05). Laying hens in the DXM group had lower laying rates and egg quality, including egg weight, eggshell thickness, eggshell strength, albumen height, and Haugh units (P < 0.05). Conversely, FA alleviated these negative impacts. Through transcriptome analysis, a total of 247 and 151 differentially expressed genes were identified among the three groups, and 32 overlapped genes were further identified. Moreover, 44 and 59 differential metabolites were influenced by DXM and FA, respectively. Kyoto Encyclopedia of Genes and Genomes enrichment from the transcriptome and metabolomics showed that the reduced production performance may be due to the disturbance of oocyte production, calcium metabolism, and oxidative stress. Analysis of 16S rRNA gene amplicon sequences revealed the differential microbial composition and potential functional changes among the different groups. LEfSe analysis showed that Mucispirillum and Nautella were the predominant bacteria in the DXM group, while Clostridium was the predominant bacteria in the FA group. Functional prediction demonstrated that stressors enhanced fatty acid biosynthesis, while betaine biosynthesis and retinol metabolism were elevated in the FA group. Dietary FA reversed the elevated levels of bile acids (BA), including cholic acid, taurodeoxycholic acid, and taurochenodeoxycholic acid (P < 0.05). The DXM group showed an overall decrease in short-chain fatty acids (SCFA), but FA restored the concentrations of acetic acid, propionic acid, and isobutyric acid (P < 0.05). In conclusion, this study reveals that dietary FA can alleviate the degradation of production performance caused by stress through improving circulating antioxidant capacity, maintaining intestinal microbiota homeostasis, and regulating SCFA and BA biosynthesis. Thus, highlighting the prominent role of gut microbe-host interactions in alleviating multi-stresses.
Collapse
Affiliation(s)
- Xi Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Chaohui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Sijing Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaoying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yun Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yumeng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yuxin Niu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhouzheng Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
38
|
Vayena E, Fuchs L, Peyhani HM, Lagoda K, Nguyen B, Hardt WD, Hatzimanikatis V. Metabolic network reconstruction as a resource for analyzing Salmonella Typhimurium SL1344 growth in the mouse intestine. PLoS Comput Biol 2025; 21:e1012869. [PMID: 40067815 PMCID: PMC11925469 DOI: 10.1371/journal.pcbi.1012869] [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: 06/11/2024] [Revised: 03/20/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Nontyphoidal Salmonella strains (NTS) are among the most common foodborne enteropathogens and constitute a major cause of global morbidity and mortality, imposing a substantial burden on global health. The increasing antibiotic resistance of NTS bacteria has attracted a lot of research on understanding their modus operandi during infection. Growth in the gut lumen is a critical phase of the NTS infection. This might offer opportunities for intervention. However, the metabolic richness of the gut lumen environment and the inherent complexity and robustness of the metabolism of NTS bacteria call for modeling approaches to guide research efforts. In this study, we reconstructed a thermodynamically constrained and context-specific genome-scale metabolic model (GEM) for S. Typhimurium SL1344, a model strain well-studied in infection research. We combined sequence annotation, optimization methods and in vitro and in vivo experimental data. We used GEM to explore the nutritional requirements, the growth limiting metabolic genes, and the metabolic pathway usage of NTS bacteria in a rich environment simulating the murine gut. This work provides insight and hypotheses on the biochemical capabilities and requirements of SL1344 beyond the knowledge acquired through conventional sequence annotation and can inform future research aimed at better understanding NTS metabolism and identifying potential targets for infection prevention.
Collapse
Affiliation(s)
- Evangelia Vayena
- Laboratory of Computational Systems Biotechnology, EPFL, Lausanne, Switzerland
| | - Lea Fuchs
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | | - Konrad Lagoda
- Laboratory of Computational Systems Biotechnology, EPFL, Lausanne, Switzerland
| | - Bidong Nguyen
- Institute of Microbiology, D-BIOL, ETH Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
39
|
Yang Q, Kang Y, Tang W, Li M, Zhao C. Interplay of gut microbiota in Kawasaki disease: role of gut microbiota and potential treatment strategies. Future Microbiol 2025; 20:357-369. [PMID: 40013895 PMCID: PMC11938985 DOI: 10.1080/17460913.2025.2469432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Kawasaki disease (KD) is an acute systemic immune vasculitis with predominant involvement of the medium and small arteries. It mostly affects pediatric patients, representing the most common form of pediatric vasculitis in children less than 5 years old. Numerous diseases, especially those related to the immune system, have established links with the intestinal flora. Recent studies have investigated the intestinal flora changes throughout the management of KD. There was gut microbiota dysbiosis in pediatric KD at the acute phase, particularly the downregulation of short-chain fat acids-producing microbiota and the over-proliferation of opportunistic pathogens. The relationship between the response to therapies in individuals with KD and specific microbiota remains uncertain. Targeted microbial supplements and dietary regulation may serve as potential measures to alleviate KD complications and thus improve prognosis. This review provides an overview of the current understanding of the interplay of the gut microbiota and KD. Furthermore, it discusses the possibility of altering the gut microbiota to reinstate a healthy condition.
Collapse
Affiliation(s)
- Qing Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Yaqing Kang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Tang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Cuifen Zhao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
40
|
Tadese DA, Mwangi J, Luo L, Zhang H, Huang X, Michira BB, Zhou S, Kamau PM, Lu Q, Lai R. The microbiome's influence on obesity: mechanisms and therapeutic potential. SCIENCE CHINA. LIFE SCIENCES 2025; 68:657-672. [PMID: 39617855 DOI: 10.1007/s11427-024-2759-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
In 2023, the World Obesity Atlas Federation concluded that more than 50% of the world's population would be overweight or obese within the next 12 years. At the heart of this epidemic lies the gut microbiota, a complex ecosystem that profoundly influences obesity-related metabolic health. Its multifaced role encompasses energy harvesting, inflammation, satiety signaling, gut barrier function, gut-brain communication, and adipose tissue homeostasis. Recognizing the complexities of the cross-talk between host physiology and gut microbiota is crucial for developing cutting-edge, microbiome-targeted therapies to address the global obesity crisis and its alarming health and economic repercussions. This narrative review analyzed the current state of knowledge, illuminating emerging research areas and their implications for leveraging gut microbial manipulations as therapeutic strategies to prevent and treat obesity and related disorders in humans. By elucidating the complex relationship between gut microflora and obesity, we aim to contribute to the growing body of knowledge underpinning this critical field, potentially paving the way for novel interventions to combat the worldwide obesity epidemic.
Collapse
Affiliation(s)
- Dawit Adisu Tadese
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James Mwangi
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Luo
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Zhang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xiaoshan Huang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Brenda B Michira
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengwen Zhou
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peter Muiruri Kamau
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiumin Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
41
|
Tashkent Y, Choo JM, Richard A, Wang Z, Calzadilla‐Bertot L, Vasil E, Miller S, Taylor SL, Ivey KL, Woodman R, Adler B, Ayonrinde OT, Olynyk JK, Beilin LJ, Mori TA, Wigg AJ, Muller KR, Adams LA, Rogers GB. Steatotic Liver Disease in Younger Adults is Associated With Altered Gut Microbiology. Liver Int 2025; 45:e70032. [PMID: 39999013 PMCID: PMC11855901 DOI: 10.1111/liv.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD) is a leading cause of chronic liver disease worldwide. As SLD pathogenesis has been linked to gut microbiome alterations, we aimed to identify SLD-associated gut microbiome features early in SLD development by utilising a highly characterised cohort of community-dwelling younger adults. METHODS AND RESULTS At age 27 years, 588 participants of the Raine Study Generation 2 underwent cross-sectional assessment. Hepatic steatosis was quantified using a validated magnetic resonance imaging (MRI) volumetric liver fat fraction (VLFF) equation (HepaFat). Of the 588 participants, 488 (83%) were classified as having 'no SLD' (VLFF ≤ 3.55%), 76 (12.9%) with 'mild-moderate' SLD (VLFF: 3.56%-13.4%) and 24 (4.10%) with 'severe' SLD (VLFF > 13.4%). Stool microbiome profiling identified an association between severe SLD and lower microbiota alpha diversity (observed features [p = 0.015], Pielou evenness [p = 0.001] and Shannon diversity [p = 0.002]) compared to no SLD. Faecal microbiota composition differed significantly between no SLD and both mild-moderate (p = 0.004) and severe SLD groups (p = 0.001). There was no significant difference in microbiota dispersion between SLD groups. Reduced relative abundance of short-chain fatty acid producing bacteria, and higher levels of proinflammatory bacterial taxa, were both significantly associated with severe SLD (q < 0.05). CONCLUSIONS SLD in younger adults is associated with reduced intestinal microbial diversity and a pattern of bacterial taxa depletion that is consistent with other chronic inflammatory conditions. Our characterisation of gut microbiome characteristics in early SLD development provides a potential basis for risk identification and reduction. TRIAL REGISTRATION The Raine Study is registered in the Australian New Zealand Clinical Trials Registry (ACTRN12617001599369).
Collapse
Affiliation(s)
- Yasmina Tashkent
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Jocelyn M. Choo
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Alyson Richard
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Zhengyi Wang
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Luis Calzadilla‐Bertot
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of HepatologySir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Egi Vasil
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Sophie Miller
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Steven L. Taylor
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Kerry L. Ivey
- Division of Aging, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Richard Woodman
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Brendan Adler
- Envision Medical ImagingWembleyWestern AustraliaAustralia
| | - Oyekoya T. Ayonrinde
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of Gastroenterology and HepatologyFiona Stanley HospitalMurdochWestern AustraliaAustralia
- Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - John K. Olynyk
- Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - Lawrence J. Beilin
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Trevor A. Mori
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Alan J. Wigg
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Kate R. Muller
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
- South Australian Hepatology and Transplant Medicine UnitSouthern Adelaide Local Health NetworkBedford ParkSouth AustraliaAustralia
| | - Leon A. Adams
- Medical SchoolThe University of Western AustraliaPerthWestern AustraliaAustralia
- Department of HepatologySir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Geraint B. Rogers
- Microbiome and Host Health ProgramSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| |
Collapse
|
42
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
43
|
Lin Q, Ouyang X, Pan Q, Huang J, Zhang Z, Yang Y, Wang H, Yang L, Zhu X, Li X, Zhang R. Extracts of Drynariae Rhizoma Promote Bone Formation in OVX Rats through Modulating the Gut Microbiota. PLANTA MEDICA 2025; 91:127-141. [PMID: 39500341 PMCID: PMC11928296 DOI: 10.1055/a-2462-4844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Drynariae Rhizoma has been commonly used as a preventive and therapeutic agent for bone diseases. However, its pharmacological mechanisms have not been fully elucidated. Here, we aimed to investigate the effects of Drynariae Rhizoma in a bilateral ovariectomized rat model and explore the correlation with gut microbiome. We established an ovariectomized rat model, which we treated with different doses of Drynariae Rhizoma (Drynariae Rhizoma-Low, 0.27 g/kg/day; Drynariae Rhizoma-Middle, 0.81 g/kg/day; Drynariae Rhizoma-High, 2.43 g/kg/day) through intragastric administration for 12 weeks. Results showed that Drynariae Rhizoma alleviated body weight, moderated bone microstructure, and promoted the expression of bone formation-related factors in ovariectomized rats, in which Drynariae Rhizoma-High showed the most significant effects among the three doses. Furthermore, the effects of Drynariae Rhizoma on promoting bone formation were correlated to the changes in microbial richness and the restorations of several genera, among which Ruminiclostridium and Ruminococcaceae_UCG_007 were positively correlated with the bone formation-related factors, and both were enriched in the Drynariae Rhizoma-High group as biomarkers. Moreover, CMP-legionaminate biosynthesis I might be a crucial pathway of Drynariae Rhizoma to regulate gut microbiota. The content of serum short-chain fatty acids in the ovariectomized rats were regulated by Drynariae Rhizoma. Our results demonstrate that Drynariae Rhizoma promotes bone formation in ovariectomized rats, and is related to the regulation of the gut microbiota structure.
Collapse
Affiliation(s)
- Qing Lin
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China
| | - Xinchen Ouyang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Qi Pan
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiajia Huang
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Zhifen Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Yumei Yang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Haoyu Wang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Li Yang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Xiaofeng Zhu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Ronghua Zhang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
44
|
Peñalver Bernabé B, Oliveira ML, Wolf PG, McLeod A, Gabel K, Cares K, Robinson N, DiPiazza B, Varady K, Tussing-Humphreys L. Intermittent Fasting: Implications for Obesity-Related Colorectal Tumorigenesis. Endocrinol Metab Clin North Am 2025; 54:61-83. [PMID: 39919878 DOI: 10.1016/j.ecl.2024.10.007] [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] [Indexed: 02/09/2025]
Abstract
Obesity is associated with metabolic and immune perturbations (ie, insulin resistance, increased inflammation, and oxidative stress), circadian rhythm dysregulation, and gut microbial changes that can promote colorectal tumorigenesis. Colorectal cancer (CRC) is the third most incident cancer in the United States. This narrative review examines the effects of intermittend fasting on factors influencing colon tumorigenesis, such as body weight, metabolic and immune markers, circadian rythm, and the gut microbiota in humans. Findings suggest that intermittent fasting regimens can lead to weight loss and shifts in metabolic markers, which could be preventive for CRC but effects on the gut microbiota composition and functions still remains elusive.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois Chicago, 851 South Morgan Street, Chicago, IL, USA; Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Manoela Lima Oliveira
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA
| | - Patricia G Wolf
- Department of Nutrition Science, Purdue University, 700 Mitch Daniels Boulevard, West Lafayette, IN, USA; Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Andrew McLeod
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA
| | - Kelsey Gabel
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; Department of Nutrition Science, Purdue University, 700 Mitch Daniels Boulevard, West Lafayette, IN, USA
| | - Kate Cares
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Nadia Robinson
- College of Nursing, University of Illinois Chicago, 845 South Damen Avenue, MC 802, Chicago, IL, USA
| | - Brittany DiPiazza
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Krista Varady
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
45
|
Petrut SM, Bragaru AM, Munteanu AE, Moldovan AD, Moldovan CA, Rusu E. Gut over Mind: Exploring the Powerful Gut-Brain Axis. Nutrients 2025; 17:842. [PMID: 40077713 PMCID: PMC11901622 DOI: 10.3390/nu17050842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Background: The human gastrointestinal tract is home to a wide variety of microorganisms. For some decades now, bacteria known as probiotics have been added to various foods because of their beneficial effects for human health. Evidence indicates that probiotics significantly regulate gut microbiota, which is vital for digestion, metabolism, immune function, and mental health. Methods: We conducted a narrative review of available original research published in PubMed for the past ten years focusing on recent advancements that provide a thorough understanding of the relationship between the gastrointestinal system and the brain. Results: Recent advances in research have focused on the importance of gut microbiota in influencing mental health. The microbiota-gut-brain axis is a complex, bidirectional communication network linking the central nervous system and the gastrointestinal tract, which highlights how the gut and brain are deeply interconnected and influence each other in ways that affect our overall health, emotions, and behavior. This powerful link is a major area of research as scientists discover more about how gut health can impact mental well-being. Conclusions: A comprehensive understanding of microbiota composition and mechanisms involved in these interactions between the gut and the brain could shape future medical and therapeutic approaches. It would balance scientific explanation with clinical relevance, offering insights into how understanding the brain-gut axis can revolutionize our approach to treating mental health and gastrointestinal disorders.
Collapse
Affiliation(s)
- Stefana-Maria Petrut
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| | - Alexandra Maria Bragaru
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
| | - Alice Elena Munteanu
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of Cardiology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Adina-Diana Moldovan
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
- MedLife SA, 010719 Bucharest, Romania
| | - Cosmin-Alec Moldovan
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of General Surgery, Witting Clinical Hospital, 010243 Bucharest, Romania
| | - Elena Rusu
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| |
Collapse
|
46
|
Zhang X, Yang Y, Wen M, Zhong F, Shu X, Xu R, Xiong P, Zhou Z, He X, Tang X, Wang B, Zhou L, Shen T. Supplementary Hesperidin Alleviated CPT-11-Induced Diarrhea by Modulating Gut Microbiota and Inhibiting the IL-17 Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40017447 DOI: 10.1021/acs.jafc.4c09602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Irinotecan (CPT-11) is a chemotherapy agent commonly used for the treatment of gastrointestinal tumors, with diarrhea being a frequent adverse effect. Hesperidin is a flavonoid abundant in citrus fruits and has shown potential in managing CPT-11-induced diarrhea (CID). However, the mechanisms underlying its effects remain unclear. This study established a mouse model of CID using CPT-11 administration to evaluate the effects of hesperidin on diarrhea severity, intestinal pathology, gut microbiota composition, and metabolite profiles by conducting biochemical analysis, histopathology, immunohistochemistry, 16S rRNA sequencing, and untargeted metabolomics. In addition, transcriptomic analysis, molecular docking, and molecular dynamics simulations were conducted to investigate potential mechanisms of action. Hesperidin supplementation was found to significantly alleviate CID in mice. Analysis of gut microbiota using 16S rRNA sequencing revealed that hesperidin improved microbial composition, with key taxa such as Alistipes, Limosilactobacillus, Rikenella, and Mucispirillum playing a central role in ameliorating CID. Furthermore, hesperidin enhanced intestinal barrier function by upregulating tight junction proteins, mitigating epithelial damage, and reducing the expression of IL-17A, TARF6, p38, phosphorylated-p38 (P-p38), and AP-1 proteins in the colon. These findings suggest that hesperidin supplementation mitigates CID by modulating gut microbiota and inhibiting the IL-17 signaling pathway, thereby improving intestinal barrier integrity.
Collapse
Affiliation(s)
- Xiaobo Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yang Yang
- Institute of Traditional Chinese Medicine of Sichuan Academy of Chinese Medicine Sciences, Chengdu 610031, China
| | - Mingchao Wen
- Wenjiang District Traditional Chinese Medicine Hospital, Wenjiang 611130, China
| | - Fanghui Zhong
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyao Shu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruitong Xu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peiyu Xiong
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zubing Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaoyan He
- College of public health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xuehua Tang
- Academic Department, Chengdu hemoyunyin medical laboratory Co., Ltd., Wenjiang 611135, China
| | - Baojia Wang
- Academic Department, Chengdu hemoyunyin medical laboratory Co., Ltd., Wenjiang 611135, China
| | - Liping Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
47
|
Li B, Chen Z, Wang G, Chen Y, Hou X, Lu B, Ning S. Biliary-intestinal anastomosis leads to alterations in intestinal flora and its flora metabolites and increases the risk of long-term postoperative complications: a case-control study. Front Microbiol 2025; 16:1531955. [PMID: 40078541 PMCID: PMC11900546 DOI: 10.3389/fmicb.2025.1531955] [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: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Pancreaticoduodenectomy (PD) is a major surgical intervention that encompasses the resection of multiple organs and the reconstruction of the digestive tract, with reconstructive procedures including pancreatico-enteric, bilioenteric, and gastroenteric anastomoses. Prior research has documented a high incidence of long-term complications following PD, which significantly impact patient prognosis and survival, however, the underlying mechanisms remain elusive. Evidence from previous studies suggests that biliary-intestinal anastomosis modifies biliary tract anatomy, altering bile flow into the gut and potentially affecting the gut microbiota and its metabolites. Given the close association between biliary tract infections and alterations in gut microbiota, we hypothesize that changes in intestinal flora and its metabolites post-PD may be a critical factor in the development of long-term complications. The objective of this study is to investigate whether biliary-intestinal anastomosis during PD induces changes in the intestinal microbiota and its metabolites, which in turn may increase the risk of long-term postoperative complications. Methods This study included 17 patients who underwent the procedure (group T) and 20 sex- and age-matched controls who did not (group N), patients in group T were stratified into those with (complication group) and without (non-complication group) long-term postoperative complications. Faecal samples were collected from all subjects and DNA was extracted from the samples using 16S rRNA gene sequencing to analyse the composition of the faecal flora and detect flora metabolites. Results 1. Alpha diversity analysis of the two sample groups indicated a trend towards lower microbial abundance in Group T relative to Group N, however, no significant differences were observed in the Shannon and Simpson diversity indices. 2. At the genus level, Group T patients exhibited markedly higher levels of Escherichia-Shigella, Veillonella, and Enterobacter, while showing significantly lower abundance of Blautia and Bifidobacterium compared to Group N subjects. Analysis of Spearman's correlation and degree of correlation between genera showed a significant negative correlation between Escherichia shigella and Blautia. Veillonella showed a significant positive correlation with both Escherichia shigella and Enterobacter. In addition, Blautia and Bifidobacterium showed a significant positive correlation with each other. 3. Subsequent comparative analysis of the bacterial flora between the complication and non-complication groups revealed a significantly elevated abundance of Escherichia-Shigella in the complication group as compared to the non-complication group. 4. Faecal metabolomic analysis revealed that L-palmitoylcarnitine, arachidic acid and PG 13:0_15:0 were significantly increased in the T group compared to the N group, whereas 3-isopropylmalic acid was significantly decreased in the T group. 5. KEGG pathway analysis identified nine crucial metabolic pathways associated with these microbial shifts: alterations in starch and sucrose metabolism, steroid hormone biosynthesis, caffeine metabolism, the citric acid cycle, riboflavin metabolism, sulfur metabolism, and the biosynthesis of valine, leucine, and isoleucine, as well as pyruvate metabolism and ABC transporter protein pathways. Conclusion 1. The biliary-intestinal anastomosis, which is performed as part of a pancreaticoduodenectomy, induces significant shifts in the intestinal flora. 2. Increased abundance of Escherichia-Shigella may promote long-term complications after biliary-intestinal anastomosis. 3. Biliary-intestinal anastomosis leads to alterations in the metabolites of the patient's intestinal flora. 4. Intestinal flora and their metabolites in patients after biliary-intestinal anastomosis may contribute to the development of long-term complications through nine metabolic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shili Ning
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
48
|
Hiremath S, Viswanathan P. Harnessing the Power of Donkey's Milk and Homemade Pickles: Unveiling Oxalate-Degrading Probiotics and Their Heat-Killed Cells as Antiadipogenic Agents in 3T3-L1 Adipocytes. Curr Microbiol 2025; 82:155. [PMID: 40009235 DOI: 10.1007/s00284-025-04146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Gut microbial dysbiosis is associated with the development of critical clinical conditions of metabolic syndrome (obesity, type II diabetes), and calcium oxalate kidney stones. The human gut microbial eubiosis with functional probiotics and their heat-killed cells of lactic acid bacteria (LAB) is considered the current therapy for metabolic syndrome (MS). In accordance with this, our study aimed to isolate oxalate-degrading, cholesterol-lowering, and anti-adipogenic bacterial strains from raw donkey's milk and homemade fermented pickles. Nine LAB strains with potential in vitro oxalate degrading, α-glucosidase inhibiting, and cholesterol-lowering activities were pre-screened from fourteen isolates. Further, the heat-killed cells of selected strains were evaluated for anti-adipogenic activity in murine 3T3-L1 adipocytes. This activity was examined by studying the lipid storage, gene, and protein expression of adipogenic and lipogenic transcription factors. Subsequently, four potential isolates demonstrated a significant reduction in lipid storage by limiting adipogenesis (reducing C/EBPα, PPARγ expression), lipid transportation (downregulating aP2 expression), and lipogenesis (reducing PLIN-1 expression). These effective isolates were characterized using 16S rRNA molecular sequencing, and were identified as closest relatives to the Enterococcus (RRLA5, RRLA1, and RRLD6) and Lactobacillus (RRLM2) genera. Further, they displayed good survivability under in vitro gastric conditions and non-haemolytic activity. Taken together, the live cells of effective isolates depicted significant in vitro oxalate degradation, and their heat-killed cells demonstrated anti-adipogenic activity through downregulating the adipogenesis and lipogenesis. Moreover, future preclinical animal model studies on the synergistic role of probiotics and their heat-killed cells in disease prevention through gut microbial modulation could provide evidence as a biotherapeutic agent.
Collapse
Affiliation(s)
- Shridhar Hiremath
- School of Bio Sciences and Technology, Vellore Institute of Technology, #412, Renal Research Laboratory, Pearl Research Park, Vellore, Tamil Nadu, 632014, India
| | - Pragasam Viswanathan
- School of Bio Sciences and Technology, Vellore Institute of Technology, #412, Renal Research Laboratory, Pearl Research Park, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
49
|
Macom RV, Brown CM. Gastrointestinal Dysfunction and Dysbiosis in Ischemic Stroke: Opportunities for Therapeutic Intervention. Pharmaceuticals (Basel) 2025; 18:320. [PMID: 40143100 DOI: 10.3390/ph18030320] [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: 11/11/2023] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 03/28/2025] Open
Abstract
Although strokes originate in the brain, it is now widely appreciated that peripheral organ systems are also impacted by stroke. The gastrointestinal system is one peripheral organ system that is impaired during ischemic stroke. This impairment results in numerous complications, which impede post-stroke recovery. Many of the gastrointestinal mechanisms that contribute to the pathophysiology of ischemic stroke remain poorly understood. This review will highlight the molecular and cellular mechanisms underlying gastrointestinal outcomes in stroke by focusing on the complex interactions that largely occur in the small intestine. The final portion of this review will focus on therapeutic interventions that target the gut as a strategy to prevent or delay functional impairment and cognitive disability in stroke patients.
Collapse
Affiliation(s)
- Rhiannon V Macom
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Candice M Brown
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| |
Collapse
|
50
|
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
|