1
|
Taba N, Fischer K, Estonian Biobank Research Team, Org E, Aasmets O. A novel framework for assessing causal effect of microbiome on health: long-term antibiotic usage as an instrument. Gut Microbes 2025; 17:2453616. [PMID: 39849320 PMCID: PMC11776458 DOI: 10.1080/19490976.2025.2453616] [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: 02/28/2024] [Revised: 07/25/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Assessing causality is undoubtedly one of the key questions in microbiome studies for the upcoming years. Since randomized trials in human subjects are often unethical or difficult to pursue, analytical methods to derive causal effects from observational data deserve attention. As simple covariate adjustment is not likely to account for all potential confounders, the idea of instrumental variable (IV) analysis is worth exploiting. Here we propose a novel framework of antibiotic instrumental variable regression (AB-IVR) for estimating the causal relationships between microbiome and various diseases. We rely on the recent studies showing that antibiotic treatment has a cumulative long-term effect on the microbiome, resulting in individuals with higher antibiotic usage to have a more perturbed microbiome. We apply the AB-IVR method on the Estonian Biobank data and show that the microbiome has a causal role in numerous diseases including migraine, depression and irritable bowel syndrome. We show with a plethora of sensitivity analyses that the identified causal effects are robust and propose ways for further methodological developments.
Collapse
Affiliation(s)
- Nele Taba
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | | | - Elin Org
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Oliver Aasmets
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
2
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
3
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [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: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
4
|
Nie P, Hu L, Feng X, Xu H. Gut Microbiota Disorders and Metabolic Syndrome: Tales of a Crosstalk Process. Nutr Rev 2025; 83:908-924. [PMID: 39504479 DOI: 10.1093/nutrit/nuae157] [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] [Indexed: 11/08/2024] Open
Abstract
The microbiota in humans consists of trillions of microorganisms that are involved in the regulation of the gastrointestinal tract and immune and metabolic homeostasis. The gut microbiota (GM) has a prominent impact on the pathogenesis of metabolic syndrome (MetS). This process is reciprocal, constituting a crosstalk process between the GM and MetS. In this review, GM directly or indirectly inducing MetS via the host-microbial metabolic axis has been systematically reviewed. Additionally, the specifically altered GM in MetS are detailed in this review. Moreover, short-chain fatty acids (SCFAs), as unique gut microbial metabolites, have a remarkable effect on MetS, and the role of SCFAs in MetS-related diseases is highlighted to supplement the gaps in this area. Finally, the existing therapeutics are outlined, and the superiority and shortcomings of different therapeutic approaches are discussed, in hopes that this review can contribute to the development of potential treatment strategies.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation Co., Ltd, Nanchang University, Nanchang 330200, China
| |
Collapse
|
5
|
Petitclerc I, Perron J, Dugas C, Mayer T, Raymond F, Di Marzo V, Veilleux A, Robitaille J. Association between gestational diabetes mellitus, maternal health and diet, and gut microbiota in mother-infant dyads. BMC Pregnancy Childbirth 2025; 25:486. [PMID: 40275186 DOI: 10.1186/s12884-025-07584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) increasingly affects women and predisposes both mothers and their infants to short- and long-term health consequences. Emerging research links GDM to maternal gut microbiota dysbiosis. However, the impact of GDM on the infant gut microbiota remains unclear. This cross-sectional study aims to explore potential associations between GDM and the gut microbiota in mothers and their infants, as well as correlations between maternal diet, cardiometabolic profile, and gut microbiota composition. METHODS Gut microbiota taxonomic composition was characterized by 16S rRNA gene sequencing on fecal samples collected at 2 months postpartum from 28 mothers, including 17 with (GDM+) and 11 without (GDM-) GDM, as well as 30 infants, 17 GDM + and 13 GDM-. Variations in overall composition and specific taxa between GDM + and GDM- were assessed. Correlations between maternal cardiometabolic profile, dietary intakes, and taxa were performed. RESULTS GDM was associated with the overall composition of gut microbiota between GDM + and GDM- in the maternal group, but not in infants. No statistically significant difference in alpha diversity between groups was found in either mothers or infants. However, 14 taxa showed significantly different abundance between GDM + and GDM- mothers, and 4 taxa differed in infants. Specific taxa at the family rank were correlated with maternal dietary and cardiometabolic variables in both mothers and infants. CONCLUSIONS GDM exposition was associated with gut microbiota composition in both mothers and infants at two months postpartum. This study enhances our understanding of how maternal health could be linked with the gut microbiota of mothers and their infants. TRIAL REGISTRATION NCT02872402 (2016-08-04, https://clinicaltrials.gov/study/NCT02872402?term=NCT02872402&rank=1 ) and NCT04263675 (2020-02-07, https://clinicaltrials.gov/study/NCT04263675?term=NCT04263675&rank=1 ).
Collapse
Affiliation(s)
- Isabelle Petitclerc
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Perron
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Camille Dugas
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Thomas Mayer
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Frédéric Raymond
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Vincenzo Di Marzo
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Quebec City, QC, G1V 4G5, Canada
| | - Alain Veilleux
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Robitaille
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada.
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada.
| |
Collapse
|
6
|
Liu X, Sun H, Han Q, Wang Z, Zeng J, Liu J, Ou S, Jin K, Shao Y, Li D, Gao Z, Wang F. Gut microbiota-derived UDCA enhanced by metformin inhibits FXR to activate autophagy against MCD diet-induced NAFLD in mice. Int Immunopharmacol 2025; 153:114471. [PMID: 40121741 DOI: 10.1016/j.intimp.2025.114471] [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: 08/19/2024] [Revised: 02/19/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a prevalent chronic liver disease, poses a substantial global health burden. Metformin is known for its protective effects in NAFLD, but the role of gut microbiota in the underlying mechanisms remains unclear. In this study, metformin was found to mitigate methionine-choline deficient (MCD) -diet induced NAFLD through reshaping the gut microbiota to increase ursodeoxycholic acid (UDCA) level, thereby inhibiting farnesoid X receptor (FXR) accompanied with activated autophagy. Specifically, using dirty cage experiments and 16S rRNA sequencing, it identified that metformin could reshape microbiota to release liver injury as confirmed by the results of histopathology and biochemical index detection. Furthermore, the bile acids were found to be altered by metformin, in which, the UDCA, a FXR natural inhibitor, was observed a significantly increase. Meanwhile, the inhibited FXR and activated autophagy in metformin-treated mice were captured using western blot, qRT-PCR and immunofluorescence analysis. In addition, the benefit of UDCA against NAFLD was demonstrated in UDCA treated mice. Further investigation with FXR siRNA introduced to HepG2 cells revealed that inhibiting FXR can reduce oleic acids induced cell injury with the autophagy activation. In conclusion, this study highlights metformin's potential to ameliorate NAFLD by reshaping gut microbiota, thereby upregulating UDCA in the liver and restoring cholesterol synthesis capacity, possibly via inhibiting FXR to activate autophagy.
Collapse
Affiliation(s)
- Xiujie Liu
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China; Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, Zhejiang, China
| | - Hongxia Sun
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Qiannian Han
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Zekai Wang
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Jingjing Zeng
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianwei Liu
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Shining Ou
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Keke Jin
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yuanyuan Shao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, Zhejiang, China
| | - Dongbing Li
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, Zhejiang, China
| | - Zhuowei Gao
- Oncology Department, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangdong 528300, China; Research Center of Translational Medicine, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangdong 528300, China.
| | - Fangyan Wang
- Institute of microbiota and host inflammation-related diseases; School of Basic Medical Science, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
7
|
Kadibalban AS, Künstner A, Schröder T, Zauleck J, Witt O, Marinos G, Kaleta C. Metabolic modelling reveals increased autonomy and antagonism in type 2 diabetic gut microbiota. Mol Syst Biol 2025:10.1038/s44320-025-00100-w. [PMID: 40263590 DOI: 10.1038/s44320-025-00100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
Type 2 diabetes (T2D) presents a global health concern, with evidence highlighting the role of the human gut microbiome in metabolic diseases. This study employs metabolic modelling to elucidate changes in host-microbiome interactions in T2D. Glucose levels, diet, 16S sequences and metadata were collected for 1866 individuals. In addition, microbial community models, and ecological interactions were simulated for the gut microbiomes. Our findings revealed a significant decrease in metabolic fluxes provided by the host's diet to the microbiome in T2D patients, accompanied by increased within-community exchanges. Moreover, the diabetic microbiomes shift towards increased exploitative ecological interactions at the expense of collaborative interactions. The reduced microbiome-to-host butyrate flux, along with decreased fluxes of amino acids (including tryptophan), nucleotides, and B vitamins from the host's diet, further highlight the dysregulation in microbial-host interactions in diabetes. In addition, microbiomes of T2D patients exhibit enrichment in energy metabolism, indicative of increased metabolic activity and antagonism. This study sheds light on the increased microbiome autonomy and antagonism accompanying diabetes, and provides candidate metabolic targets for intervention studies and experimental validation.
Collapse
Affiliation(s)
- A Samer Kadibalban
- Medical Systems Biology Group, University Hospital Schleswig-Holstein Campus Kiel & Kiel University, Kiel, Germany
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute for Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Torsten Schröder
- Perfood GmbH, Lübeck, Schleswig-Holstein, Germany
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Lübeck Campus & University of Lübeck, Lübeck, Germany
| | | | - Oliver Witt
- Perfood GmbH, Lübeck, Schleswig-Holstein, Germany
| | - Georgios Marinos
- Medical Systems Biology Group, University Hospital Schleswig-Holstein Campus Kiel & Kiel University, Kiel, Germany
- CAU Innovation GmbH, Kiel University, Kiel, Germany
| | - Christoph Kaleta
- Medical Systems Biology Group, University Hospital Schleswig-Holstein Campus Kiel & Kiel University, Kiel, Germany.
| |
Collapse
|
8
|
Su X, Tian Z, Fang Y, Zhou S, Ma S. Effects of high-dose glucocorticoids on gut microbiota in the treatment of Graves' ophthalmopathy. Microbiol Spectr 2025:e0246724. [PMID: 40261021 DOI: 10.1128/spectrum.02467-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
Many studies indicate the gut microbiome is associated with diseases caused by administering high-dose glucocorticoids (GCs), such as hypertension, hyperglycemia, and osteoporosis. However, the association between intestinal flora and the use of high-dose GCs remains elusive. We aimed to characterize gut microbiome in Graves' ophthalmopathy (GO) patients after administering high-dose GCs. In this study, 20 primary GO patients were recruited. The differences in gut microbiota of GO patients before and after administering high-dose GCs were analyzed by 16S rDNA sequencing technology. Untargeted metabolomic analysis was used to examine the differences in gut metabolites between two groups. There were significant differences in α and β diversities of gut microbiota in GO patients before and after administering high-dose GCs. The random forest analysis indicated that three intestinal bacteria (Faecalibacterium, Streptococcus, and Prevotella) could distinguish the two groups with the highest accuracy, which was proven by receiver operator characteristic curve and linear discriminant analysis effect size analysis. The short-chain fatty acid-producing ability in GO patients' gut after high-dose GC administration was significantly decreased. The 5-hydroxytryptamine levels significantly increased in the gut of GO patients after administering high-dose GCs. Our study suggests that high-dose GC administration causes the changes in gut microbiome and metabolites. Moreover, the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.IMPORTANCEFor the first time, we revealed that gut microbiome and metabolome in Graves' ophthalmopathy patients after high-dose glucocorticoid (GC) administration significantly changed, and the altered flora and metabolites are related to hypertension, hyperglycemia, and osteoporosis. These findings can help understand the development of side effects caused by high-dose GCs and can be further used to develop potential probiotics to facilitate the prevention for those side effects.
Collapse
Affiliation(s)
- Xinhuan Su
- Department of Endocrinology, Department of Geriatrics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenyu Tian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yalun Fang
- Department of Clinical Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Shengnan Zhou
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shizhan Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
Yang H, Lei C, Li D, Ma L, Zhang N, Lang Y, Wu L, Wang M, Tian H, Li C. An integrated fecal microbiome and metabolomics in type 2 diabetes mellitus rats reveal mechanism of action of Moringa oleifera Lamarck seeds polysaccharides to alleviate diabetes. Int J Biol Macromol 2025:143437. [PMID: 40274155 DOI: 10.1016/j.ijbiomac.2025.143437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Moringa oleifera Lamarck seeds (MOS) have been traditionally used in folk medicine and documented for their potential to alleviate type 2 diabetes symptoms, but the potential mechanisms are still unknown. The purpose of this article is to investigate the effects of MSAP (alkali-extracted polysaccharide from MOS) on diabetic rats by assessing its impact on the gut microbiome, diabetes-related biochemical markers, and fecal metabolomics. The results demonstrated that the fasting blood glucose, glucose tolerance, insulin tolerance, insulin level and lipopolysaccharides (LPS) level in the rats treated with MSAP were all improved. Specifically, MSAP was found to modulate the composition and diversity of the gut microbiota, increasing the ratio of Firmicutes/Bacteroidetes, which enhanced the quantity of probiotic Lactobacillus and butyrate-producing bacteria, such as Roseburia, thereby reinforcing the intestinal epithelial barrier. Furthermore, fecal metabolomics indicates that MSAP actively regulates pathways closely associated with diabetes, including sphingolipid metabolism, amino acid synthesis and catabolism, retrograde endogenous cannabinoid signaling, and the modulation of TRP channels by inflammatory mediators. By integrating microbiome and metabolomics data, this study elucidated the mechanisms through which MSAP alleviates diabetes. In conclusion, the findings suggest that polysaccharides from MOS hold potential as a medicinal and edible homologous food for diabetes management.
Collapse
Affiliation(s)
- Hongru Yang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Chongbin Lei
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Dongyao Li
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Lei Ma
- College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Na Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; College of Biochemistry and Environmental Engineering, Baoding University, Baoding, Hebei 071000, China
| | - Yumiao Lang
- College of Public Health, Hebei University, Baoding, Hebei 071000, China
| | - Liping Wu
- College of Nursing, Hebei University, Baoding, Hebei 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co. Ltd., Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China
| | - Hongtao Tian
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; National Engineering Research Center for Agriculture in Northern Mountainous Areas, Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China.
| | - Chen Li
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China; Hebei Technology Innovation Center of Probiotic Functional Dairy Product, Baoding, Hebei 071000, China.
| |
Collapse
|
10
|
Zhu Y, Tian Q, Huang Q, Wang J. Bile-processed Rhizoma Coptidis alleviates type 2 diabetes mellitus through modulating the gut microbiota and short-chain fatty acid metabolism. Int Immunopharmacol 2025; 156:114645. [PMID: 40253770 DOI: 10.1016/j.intimp.2025.114645] [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: 11/10/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Bile-Processed Rhizoma Coptidis (BPRC) is a processed products of Rhizoma Coptidis (RC) commonly used to treat type 2 diabetes mellitus (T2DM). However, the synergistic mechanism of its processing remains unknown. Current research indicates that the gut microbiota and its metabolites, such as short-chain fatty acids (SCFAs), are closely associated with the progression of T2DM. PURPOSE This study aims to investigate the effects of BPRC on the gut microbiota and its metabolite SCFAs in T2DM rats. METHODS T2DM rat model was induced by a high-fat diet (HFD) combined with streptozotocin (STZ), followed by a 4-week treatment with BPRC to observe its therapeutic effects. The impact of BPRC on the gut microbiota was studied through metagenomic sequencing. Quantitative analysis of SCFAs was conducted using GC-MS. Western blot and quantitative real-time PCR (qRT-PCR) were conducted to investigate the potential mechanisms of BPRC. RESULTS BPRC significantly improved insulin resistance in T2DM rats, downregulated levels of pancreatic cell apoptosis factors, and upregulated the abundance of Bacteroides uniformis, Bacteroides sp A1C1, Anaerostipes caccae, Alistipes finegoldii and Blautia sp.N6H1-15 in T2DM rats. Additionally, BPRC increased the levels of seven SCFAs in the intestines of T2DM rats. It activated intestinal TGR5, GPR41, GPR43, and GPR109a receptors, collectively upregulating GLP-1 protein expression, and exerted therapeutic effects on T2DM. CONCLUSION The results indicate that the synergistic mechanism of BPRC in treating T2DM is associated with modulating the gut microbiota, increasing SCFAs content in the intestines, and regulating intestinal GLP-1 production.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingqing Tian
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qinwan Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Shi L, Wang Y, Guan Y, Men L, Sun J, Yuan G. To establish a new quality assessment method based on the regulation of intestinal microbiota in type 2 diabetes by lignans of Schisandra chinensis (Turcz.) Baill. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119822. [PMID: 40250638 DOI: 10.1016/j.jep.2025.119822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The mature fruit of Schisandra chinensis, a traditional Chinese medicinal herb, is primarily utilized for the management of diabetes. Its principal bioactive constituents include lignans, polysaccharides, and organic acids. Nonetheless, a standardized quality control methodology grounded in the therapeutic efficacy of Schisandra chinensis (Turcz.) Baill. for diabetes treatment has yet to be developed. AIM OF THE STUDY Due to the diverse origins, intricate composition, and multiple therapeutic targets of Schisandra chinensis Fructus, relying on a single index component for quality control is challenging. Consequently, this study proposes a novel quality evaluation approach, integrating pharmacological activity and intestinal microbiota analysis, to assess the efficacy of Schisandra chinensis Fructus in diabetes management. MATERIALS AND METHODS Twelve batches of Schisandra chinensis Fructus from diverse origins were selected for lignan content analysis, and 3 representative batches were subsequently chosen for pharmacological experiments pertaining to diabetes. The relationship between lignan content in Schisandra chinensis Fructus and its pharmacological efficacy was assessed by evaluating the recovery rates of eleven serum biochemical markers affected by Schisandra lignans. Furthermore, 16S rRNA gene sequencing was utilized to explore the association between the gut microbiota in diabetic rats and the lignan content in Schisandra chinensis Fructus. RESULTS The results of serum biochemistry analyses demonstrate that Schisandra lignans significantly decrease bone gamma-carboxyglutamate protein (BGP), blood lipid levels, and oxidative stress in diabetic rats, thereby conferring hepatoprotective effects. Correlation analysis between the constituents and pharmacological effects revealed a positive relationship between the anti-diabetic efficacy of Schisandra chinensis Fructus and its total lignan content. Furthermore, Schisandra lignans were observed to enhance gut microbiota diversity in diabetic rats, mitigate the dysbiosis induced by Type 2 Diabetes Mellitus (T2DM), and increase the abundance of beneficial bacterial species. CONCLUSIONS The observed variations in the efficacy of Schisandra chinensis Fructus from different sources may be attributed to differences in total lignan content. The higher the total lignan content in Schisandra chinensis Fructus, the stronger its protective effect. Based on the analysis of component-efficacy-microbiota correlation this study has identified a chisandrin content of ≥3.5 mg/g and a total lignan content of≥17 mg/g as quality evaluation indicators for the improvement of T2DM by Schisandra chinensis Fructus.
Collapse
Affiliation(s)
- Liqiang Shi
- College of Pharmacy, Beihua University, Jilin, Jilin, 132013, PR China.
| | - Yutong Wang
- College of Pharmacy, Beihua University, Jilin, Jilin, 132013, PR China.
| | - Yunhui Guan
- College of Pharmacy, Beihua University, Jilin, Jilin, 132013, PR China.
| | - Lihui Men
- College of Basic Medical Sciences, Jilin University, Changchun, 130021, PR China.
| | - Jinghui Sun
- College of Pharmacy, Beihua University, Jilin, Jilin, 132013, PR China.
| | - Guangxin Yuan
- College of Pharmacy, Beihua University, Jilin, Jilin, 132013, PR China.
| |
Collapse
|
12
|
Fang X, Zhang Y, Huang X, Miao R, Zhang Y, Tian J. Gut microbiome research: Revealing the pathological mechanisms and treatment strategies of type 2 diabetes. Diabetes Obes Metab 2025. [PMID: 40230225 DOI: 10.1111/dom.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025]
Abstract
The high prevalence and disability rate of type 2 diabetes (T2D) caused a huge social burden to the world. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. With in-depth research on the pathogenesis of T2D and growing advances in microbiome sequencing technology, the association between T2D and gut microbiota has been confirmed. The gut microbiota participates in the regulation of inflammation, intestinal permeability, short-chain fatty acid metabolism, branched-chain amino acid metabolism and bile acid metabolism, thereby affecting host glucose and lipid metabolism. Interventions focusing on the gut microbiota are gaining traction as a promising approach to T2D management. For example, dietary intervention, prebiotics and probiotics, faecal microbiota transplant and phage therapy. Meticulous experimental design and choice of analytical methods are crucial for obtaining accurate and meaningful results from microbiome studies. How to design gut microbiome research in T2D and choose different machine learning methods for data analysis are extremely critical to achieve personalized precision medicine.
Collapse
Affiliation(s)
- Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyue Huang
- First Clinical Medical College, Changzhi Medical College, Shanxi, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Deng XR, Zhai YJ, Shi XY, Tang SS, Fang YY, Heng HY, Zhao LY, Yuan HJ. Characteristic dysbiosis in patients with type 2 diabetes and hyperuricemia, and the effect of empagliflozin on gut microbiota. World J Diabetes 2025; 16:102970. [PMID: 40236847 PMCID: PMC11947907 DOI: 10.4239/wjd.v16.i4.102970] [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: 11/05/2024] [Revised: 01/04/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Gut microbiota play a crucial role in metabolic diseases, including type 2 diabetes (T2DM) and hyperuricemia (HUA). One-third of uric acid is excreted into the intestinal tract and further metabolized by gut microbiota. Thus, the gut microbiota might be a new therapeutic target for HUA. Empagliflozin significantly lowers serum uric acid levels and contributes to cardiovascular benefits which are partly attributed to altered gut microbiota. We hypothesize that gut dysbiosis in patients with diabetes and HUA, and the reduction of uric acid by empagliflozin, may be mediated by gut microbiota. AIM To investigate dysbiosis in patients with T2DM and HUA, and the effect of empagliflozin on gut microbiota associated with purine metabolism. METHODS In this age and sex-matched, case-control study, we recruited 30 patients with T2DM and HUA; 30 with T2DM; and 30 healthy controls at the Henan Provincial People's Hospital between February 2019 and August 2023. Nine patients with T2DM and HUA were treated with empagliflozin for three months. Gut microbiota profiles were assessed using the 16S rRNA gene. RESULTS Patients with T2DM and HUA had the highest total triglycerides (1.09 mmol/L in heathy control vs 1.56 mmol/L in T2DM vs 2.82 mmol/L in T2DM + HUA) and uric acid levels (302.50 μmol/L in heathy control vs 288.50 μmol/L in T2DM vs 466.50 μmol/L in T2DM + HUA) among the three groups. The composition of the gut microbiota differed significantly between patients with T2DM and HUA, and those with T2DM/healthy controls (P < 0.05). Notably, patients with T2DM and HUA demonstrated a deficiency of uric acid-degrading bacteria such as Romboutsia, Blautia, Clostridium sensu stricto 1 (P < 0.05). Empagliflozin treatment was associated with significantly reduced serum uric acid levels and purine metabolism-related pathways and genes in patients with T2DM and HUA (P < 0.05). CONCLUSION Gut dysbiosis may contribute to the pathogenesis of HUA in T2DM, and empagliflozin may partly restore the gut microbiota related to uric acid metabolism.
Collapse
Affiliation(s)
- Xin-Ru Deng
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Yu-Jia Zhai
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Xiao-Yang Shi
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Sha-Sha Tang
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Yuan-Yuan Fang
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Hong-Yan Heng
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Ling-Yun Zhao
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| | - Hui-Juan Yuan
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
14
|
Cao X, Yao F, Liu W, Wang Y, Zhang Z, Zhang C, Dong Z, Zhang B, He R, Sun X. Vaccinium myrtillus L. ameliorates diabetic nephropathy via modulating metabolites and gut microbiota in rats. Front Pharmacol 2025; 16:1541947. [PMID: 40264677 PMCID: PMC12011793 DOI: 10.3389/fphar.2025.1541947] [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: 12/09/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Diabetic nephropathy (DN), one of the serious complications in the diabetes, has a high mortality in the diabetic patients. Bilberry (Vaccinium myrtillus L.) have received much attention for their health benefits in alleviating metabolic diseases, which are rich in anthocyanins. However, the anti-DN ability of bilberry has not been fully studied. The aim of this study was to investigate the effect and mechanism of Vaccinium myrtillus L. extract (VCE) on diabetic nephropathy in vivo and in vitro. Methods Streptozocin (STZ) combined with high fat induced DN model was established in rats. Biochemical indicators, histopathology, 16s third generation sequencing and serum metabolomics were used to evaluate the effects of VCE on DN. Subsequently, a cell model of advanced glycation end products (AGEs) induced podocyte injury was established to verify which compounds in VCE played the main anti-diabetic nephropathy function and the mechanism of action. Finally, in vitro experiments were conducted to verify the effect of characteristic metabolites screened by serum metabolomics on improving diabetic nephropathy. Results Insulin resistance index, lipid metabolism, oxidative stress and inflammatory response indexes of DN rats were significantly improved after 8 weeks of VCE treatment. In addition, intake of VCE modulates gut microbiota composition and reverses the abundance of Lactobacillus, Bifidobacterium and Ruminococcus. Supplementation with VCE altered serum metabolite levels, including uridine and phenylacetylglycine. Pretreatment with VCE and its anthocyanins inhibited the expression of LDH, IL-6 and TNF-α, reduced the levels of p38-MAPK, IĸBα, IKKβ, and NF-κB in podocyte cells. In addition, pretreatment with serum metabolite uridine also reduced the expression of LDH and mitochondrial ROS, and inhibited cell apoptosis. Conclusion Our findings suggest that the improvement of gut microbiota and metabolic function were related to the anti-DN potential of VCE, and the underlying mechanism may be related to the inhibition of MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xinxin Cao
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Fan Yao
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Wenxiu Liu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yufang Wang
- Innovation Research and Development Center, BY HEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Zhen Zhang
- Innovation Research and Development Center, BY HEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Chongyang Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Zhengqi Dong
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Ruikun He
- Innovation Research and Development Center, BY HEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Diabetes Research Center, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Zhang G, Lu Y, Wang Z, Ma R, Jin H, Zhang J, Liu F, Ding Y. Causal relationship between gut microbiota and ageing: A multi-omics Mendelian randomization study. Arch Gerontol Geriatr 2025; 131:105765. [PMID: 39988416 DOI: 10.1016/j.archger.2025.105765] [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: 11/08/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Observational studies and clinical trials have suggested a connection between gut microbiota and aging. However, the causal relationship between them remains undetermined. OBJECTIVES This study aimed to use bidirectional two-sample Mendelian randomization (TSMR) analysis to explore the causal relationship between gut microbiota and aging. METHODS Summary statistics from genome-wide association studies (GWAS) on gut microbiota and seven aging-related phenotypes were employed for TSMR analysis. Reverse Mendelian randomization (MR) analysis was performed to assess the potential for reverse causality. Additionally, the relationship between Akkermansia muciniphila and inflammation-related proteins and metabolites was further investigated. The effects of Akkermansia muciniphila on aging were also examined in Caenorhabditis elegans by measuring both lifespan and healthspan. RESULTS MR analysis of 207 microbial taxa and seven aging phenotypes revealed 44 causal relationships between the gut microbiota and aging. Akkermansia muciniphila was found to be causally linked to several aging-related traits, including mvAge, appendicular lean mass, and grip strength (P < 0.05). Reverse MR analysis identified 23 causal relationships, but no bidirectional causality was observed. Moreover, Akkermansia muciniphila is causally related to ST1A1, taurine bile acid, and mannose (P < 0.05). In Caenorhabditis elegans, treatment with Akkermansia muciniphila significantly extended lifespan (P < 0.05) and improved mobility in aging nematodes. CONCLUSION TSMR analysis uncovers multiple potential causal links between gut microbiota and aging, particularly Akkermansia muciniphila. Experimental results support its role in alleviating aging. This study provides a strong foundation for future research on gut microbiota's role in aging.
Collapse
Affiliation(s)
- Guolin Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuqing Lu
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhen Wang
- Department of General Surgery, The First Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ruicong Ma
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hongjin Jin
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingsi Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Fengyi Liu
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanchun Ding
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
16
|
Jiao X, Li Z. Impact of streptozotocin-induced type 1 and type 2 diabetes on ocular surface microbial signatures in C57BL/6J mice. Exp Eye Res 2025; 253:110282. [PMID: 39955022 DOI: 10.1016/j.exer.2025.110282] [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: 11/09/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
The ocular surface (OS), like other mucosal sites, hosts a diverse microbiome. However, the impact of hyperglycemia associated with diabetes on OS microbial composition remains poorly understood. In this study, we established type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) models in C57BL/6J mice by administering high-dose streptozotocin (STZ) for T1DM and low-dose STZ combined with a high-fat diet for T2DM. The OS microbiome was characterized and analyzed using 16S rRNA sequencing. The results showed that neither T1DM nor T2DM significantly affected microbial richness compared to normal mice; however, T2DM led to a significant reduction in microbial diversity. This reduction in microbial diversity in T2DM is consistent with known microbial dysbiosis in diabetes, which may contribute to the pathogenesis of ocular complications such as dry eye disease and diabetic retinopathy. Community composition analysis identified Proteobacteria, Pelagibacterium, and Aliihoeflea as the core OS bacteria in normal mice. Diabetes significantly altered the OS microbial composition at various taxonomic levels. Specifically, T1DM was associated with 9 signature bacterial species, including Oceanospirillales, Bacillales, Halomonas, unclassified_f_Lachnospiraceae, and Anoxybacillus. T2DM exhibited 17 bacterial markers, including Firmicutes, Staphylococcus, Corynebacterium, and Parasutterella. Functional prediction of the microbiota using PICRUSt2 indicated potential impairments in carbohydrate metabolism due to diabetes. In conclusion, diabetic mice exhibit severe dysregulation of their OS microbiota, and restoring microbial balance in diabetic patients may represent a promising strategy for preventing and treating diabetic OS pathologies.
Collapse
Affiliation(s)
- Xinwei Jiao
- Department of Ophthalmology, The Affiliated Hospital of Shandong Second Medical University, Weifang, China; Department of Pathology, Jinan University Medical School, Guangzhou, China
| | - Zhijie Li
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China.
| |
Collapse
|
17
|
Zhang Y, Chen S, Chen G, Zhou L, Zhou G, Yu X, Yuan L, Deng W, Wang Z, Li J, Tu Y, Zhang D, li Y, Sammad A, Zhu X, Yin K. The Type III Secretion System (T3SS) of Escherichia Coli Promotes Atherosclerosis in Type 2 Diabetes Mellitus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413296. [PMID: 39807021 PMCID: PMC12005784 DOI: 10.1002/advs.202413296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Large-scale studies indicate a strong relationship between the gut microbiome, type 2 diabetes mellitus (T2DM), and atherosclerotic cardiovascular disease (ASCVD). Here, a higher abundance of the type III secretion system (T3SS) virulence factors of Enterobacteriaceae/Escherichia-Shigella in patients with T2DM-related-ASCVD, which correlates with their atherosclerotic stenosis is reported. Overexpression of T3SS via Citrobacter rodentium (CR) infection in Apoe-/- T2DM mice exacerbated atherosclerotic lesion formation and increased gut permeability. Non-targeted metabolomic and proteomic analysis of mouse serum showed that T3SS caused abnormal glycerophospholipid metabolism in mice. Proteomics, RNA sequencing, and functional analyses showed that T3SS induced ferroptosis in intestinal epithelial cells, partly due to increased expression of ferritin heavy chains (FTH1). This findings first demonstrated that T3SS increases ferroptosis in intestinal epithelial cells, via disrupting the intestinal barrier and upregulation of phosphatidylcholine, thereby exacerbating T2DM-related ASCVD.
Collapse
Affiliation(s)
- Yao‐Yuan Zhang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
| | - Song‐Tao Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Gang Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Le Zhou
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Guo‐Liang Zhou
- Department of CardiologyThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| | - Xin‐Yuan Yu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Long Yuan
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Wei‐Qian Deng
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Zhen‐Bo Wang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Jing Li
- Department of Imaging DiagnosisZhujiang Hospital of Southern Medical UniversityGuangzhou510515China
| | - Yi‐Fu Tu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Da‐Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of PediatricsFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaT6G 2R3Canada
| | - Yuan li
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Abdul Sammad
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Xiao Zhu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
| | - Kai Yin
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
- Guangxi Clinical Research Center for Diabetes and Metabolic DiseasesThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| |
Collapse
|
18
|
Dai S, Long J, Han W, Zhang L, Chen B. Alleviative effect of probiotics and prebiotics on dry eye in type 2 diabetic mice through the gut-eye axis. Ocul Surf 2025; 36:244-260. [PMID: 39922458 DOI: 10.1016/j.jtos.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] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Diabetes Mellitus (DM) is a metabolic disease that manifests as a state of "chronic low-grade inflammation". Patients with DM have a disorder of intestinal flora. There is a discernible correlation between this disorder of intestinal flora and the onset and progression of eye diseases, which offers novel insights into treating eye diseases through the modulation of intestinal flora. Here, we demonstrated that a high-fat diet and streptozotocin injection-induced intestinal microbiota dysbiosis can lead to dry eye-like manifestations in T2DM mice. Probiotic and prebiotic treatments not only alleviated intestinal inflammation and barrier disruption, but also mitigated damage to the lacrimal barrier and suppressed immune cell infiltration and inflammatory responses. Additional mechanism investigation found that probiotics and prebiotics inhibited the TLR4/NF-κB signaling pathway and its downstream pro-inflammatory products both in the lacrimal gland and colon. 16S RNA sequencing identified a reduction in the bacterial genera Akkermansia and Lactobacillus in the fecal samples of DM mice. By contrast, treatment with probiotics and prebiotics led to a reshaping of the intestinal microbial community and a reduction in bile acid metabolites, such as taurocholic acid and deoxycholic acid. Our current study demonstrates that probiotic and prebiotic treatments can ameliorate dry eye-like symptoms and associated pathological changes in T2DM mice. Moreover, we proved that a high-fat diet and STZ-induced microbiota dysbiosis were involved in diabetic dry eye through the gut-eye axis.
Collapse
Affiliation(s)
- Shirui Dai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Jianfeng Long
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Wentao Han
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Liwei Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
19
|
Chen Q, Gao Y, Li F, Yuan L. The role of gut-islet axis in pancreatic islet function and glucose homeostasis. Diabetes Obes Metab 2025; 27:1676-1692. [PMID: 39916498 PMCID: PMC11885102 DOI: 10.1111/dom.16225] [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: 09/03/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 03/08/2025]
Abstract
The gastrointestinal tract plays a vital role in the occurrence and treatment of metabolic diseases. Recent studies have convincingly demonstrated a bidirectional axis of communication between the gut and islets, enabling the gut to influence glucose metabolism and energy homeostasis in animals strongly. The 'gut-islet axis' is an essential endocrine signal axis that regulates islet function through the dialogue between intestinal microecology and endocrine metabolism. The discovery of glucagon-like peptide-1 (GLP-1), gastric inhibitory peptide (GIP) and other gut hormones has initially set up a bridge between gut and islet cells. However, the influence of other factors remains largely unknown, such as the homeostasis of the gut microbiota and the integrity of the gut barrier. Although gut microbiota primarily resides and affect intestinal function, they also affect extra-intestinal organs by absorbing and transferring metabolites derived from microorganisms. As a result of this transfer, islets may be continuously exposed to gut-derived metabolites and components. Changes in the composition of gut microbiota can damage the intestinal barrier function to varying degrees, resulting in increased intestinal permeability to bacteria and their derivatives. All these changes contribute to the severe disturbance of critical metabolic pathways in peripheral tissues and organs. In this review, we have outlined the different gut-islet axis signalling mechanisms associated with metabolism and summarized the latest progress in the complex signalling molecules of the gut and gut microbiota. In addition, we will discuss the impact of the gut renin-angiotensin system (RAS) on the various components of the gut-islet axis that regulate energy and glucose homeostasis. This work also indicates that therapeutic approaches aiming to restore gut microbial homeostasis, such as probiotics and faecal microbiota transplantation (FMT), have shown great potential in improving treatment outcomes, enhancing patient prognosis and slowing down disease progression. Future research should further uncover the molecular links between the gut-islet axis and the gut microbiota and explore individualized microbial treatment strategies, which will provide an innovative perspective and approach for the diagnosis and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
20
|
Liu W, Jin R, Ma F, Zhao P, Su Y, Wang J, Zhang Y, Wang R, Zhu J, Liu X. Effects of Dioscorea opposita polysaccharides on insulin resistance and gut microbiota in high-fat-diet induced type 2 diabetic rats. Int J Biol Macromol 2025; 304:141004. [PMID: 39952518 DOI: 10.1016/j.ijbiomac.2025.141004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
This study investigates the effects Dioscorea opposita polysaccharides (DOP) on insulin resistance, lipid metabolism, oxidative stress, and intestine microbiota in high-fat-diet and streptozotocin induced type 2 diabetes (T2DM) rats. Low dose (DOP-L, 200 mg/kg BW), high dose (DOP-H, 400 mg/kg BW) and D. opposita powder (DO, 400 mg/kg BW) were oral-administrated to T2DM rats. After 6 weeks of treatment, supplementation of DOP-H and DO improved body weight and glucose/lipid metabolism-related indicators, including glucagon-like peptide 1, total cholesterol and high-density lipoprotein cholesterol. DOP-H and DO suppressed liver oxidative stress through increasing the level of superoxide dismutase, catalase, glutathione and reducing malondialdehyde. DOP attenuated the pathological change in liver, such as hepatic steatosis, and thus improved the liver function. Furthermore, the anti-diabetic effects of DOP was correlated with alterations of the gut microbiota, including an increase in Firmicutes and Bacteroidetes and a decrease in Actinobacteria and Proteobacteria, which promoted a healthier gut environment. Further analysis of short-chain fatty acids and metabolites provided evidences of DOP's regulatory effects on cecal contents in T2DM rats. Therefore, DOP-H present decent effects on T2DM, suggesting that DOP can ameliorate the insulin resistance and restore blood lipid level of T2DM rats with high-fat-diet by regulating intestinal microbiota.
Collapse
Affiliation(s)
- Wei Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China; Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rumeng Jin
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Fanyi Ma
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| | - Peng Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yuting Su
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jiani Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Yun Zhang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Ruijiao Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Jinhua Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China
| | - Xiuhua Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, School of Pharmacy, State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou 450046, China.
| |
Collapse
|
21
|
Zhang B, Xu K, Deng W, Liu C, Xu Q, Sheng H, Feng J, Yuan Q. Protective effects of Sulforaphene on kidney damage and gut dysbiosis in high-fat diet plus streptozotocin-induced diabetic mice. Food Chem 2025; 469:142558. [PMID: 39709924 DOI: 10.1016/j.foodchem.2024.142558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Diabetic nephropathy (DN) is one of the most serious and prevalent complications associated with diabetes. Consequently, antidiabetic drugs or foods potentially protecting the kidneys are of significant therapeutic value. Sulforaphene (SFE) is a natural isothiocyanate derived from radish seeds, known for its anti-inflammatory and antioxidant properties. However, no studies have investigated on the ability of SFE to prevent or treat DN. This study established a high-fat diet combined with a streptozotocin-induced type II diabetes mellitus mouse model. We administered SFE treatment to examine its protective effects on renal and intestinal homeostasis in DN mice. After 4 weeks of treatment, SFE (50 mg/kg b.w.) not only reduced blood glucose concentration (20.3 %, P < 0.001), kidney to body weight ratio (26.2 %, P < 0.01), and levels of serum total cholesterol (40.6 %, P < 0.001), triglycerides (38.2 %, P < 0.01), creatinine (36.7 %, P < 0.01), and urea nitrogen (45.0 %, P < 0.001) in DN mice compared to control mice but also increased the kidney superoxide dismutase (72.7 %, P < 0.001), catalase (51.1 %, P < 0.001), and glutathione peroxidase activities (31.6 %, P < 0.01), as well as glutathione levels (39.2 %, P < 0.01) in comparison to DN mice. Furthermore, SFE decreased levels of reactive oxygen species (55.4 %, P < 0.01), 4-hydroxyalkenals (36.9 %, P < 0.001), malondialdehyde (42.6 %, P < 0.001), and 8-hydroxy-deoxyguanosine (26.3 %, P < 0.001), accompanied by a meliorating kidney morphological abnormalities. Notably, a reduction in renal inflammatory factors was also observed in SFE-treated DN mice compared to untreated DN mice, particularly in the C-X-C motif chemokine ligand 8 factors (54.8 %, P < 0.001). Western blotting results indicated that SFE significantly down-regulated the protein expression of TLR4 and MyD88 (1.9, 1.7-fold, P < 0.001). Additionally, SFE improved gut microbiota (GM) dysbiosis and intestinal homeostasis, as evidenced by increased expression of antimicrobial peptides and tight junction proteins in colon tissue. SFE appeared to enhance the proliferation of probiotics, such as Bacteroidota, Lachnospiraceae_NK4A136_group and norank_f__Muribaculaceae, while also decreasing harmful bacteria to a greater extent compared to STZ treatment. These findings suggest that SFE modulates GM and improves intestinal homeostasis, providing a theoretical basis for its use in the treatment of DN.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Kang Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wenlei Deng
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ce Liu
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qianmin Xu
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huakang Sheng
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jialu Feng
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
22
|
Tahmasebi A, Beheshti R, Mahmoudi M, Jalilzadeh M, Salehi-Pourmehr H. Alterations in gut microbial community structure in obstructive sleep apnea /hypopnea syndrome (OSAHS): A systematic review and meta-analysis. Respir Med 2025; 241:108077. [PMID: 40158663 DOI: 10.1016/j.rmed.2025.108077] [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: 11/23/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVES This systematic review investigates gut bacterial diversity and composition in patients with Obstructive Sleep Apnea-Hypopnea Syndrome (OSAHS) and examines how these changes may contribute to cardiovascular complications. METHODS A comprehensive search was conducted in PubMed, Web of Science, and Scopus up to March 2025. After removing duplicates, titles and abstracts were screened by two reviewers, and full texts were assessed for inclusion. Data extraction on study characteristics and outcomes was performed. Methodological quality was evaluated using the Joanna Briggs Institute checklist. α-diversity was assessed using richness and diversity indices, while β-diversity examined community structure differences. Meta-analysis was conducted using standardized mean differences (SMD) and confidence intervals (CIs), and heterogeneity was assessed with the Cochrane I2 test. RESULTS The review included 18 studies (16 adults, 2 pediatrics) examining the gut microbiome in OSAHS. Meta-analysis revealed significant reductions in α-diversity indices (Shannon, Chao1, observed species, ACE) in OSAHS patients, while Simpson's index showed no difference. β-diversity analyses showed distinct gut microbiome differences in OSA. Key differential bacteria included Bacteroides, Proteobacteria, Faecalibacterium, Ruminococcaceae, Megamonas, Oscillibacter, Dialister, Roseburia, and Lachnospira. Study quality was medium to high. CONCLUSION OSAHS is associated with significant gut microbiome alterations, including a reduction in beneficial bacteria and an increase in LPS-producing bacteria, leading to intestinal barrier dysfunction. These changes may contribute to systemic inflammation and elevate the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Ali Tahmasebi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasa Beheshti
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadsina Mahmoudi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahan Jalilzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Iranian EBM Center: A Joanna Briggs Institute Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
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
|
24
|
Xu M, Li W, Xu Y, Zhang J, Xue H, Du J, Hu X. Arecoline Alleviates T2DM via Gut Microbiota Modulation and Liver Gene Regulation in Mice. Mol Nutr Food Res 2025:e70015. [PMID: 40123201 DOI: 10.1002/mnfr.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
SCOPE Arecoline, the main alkaloid in areca nut, has shown potential in modulating metabolism and gut microbiota. This study aimed to evaluate its therapeutic effects on glucose and lipid metabolism, inflammation, liver function, and potential mechanisms in a Type 2 diabetes mellitus (T2DM) mouse model. METHODS AND RESULTS T2DM was established in mice with a high-fat, high-sugar diet, and streptozotocin injections. Arecoline significantly reduced fasting blood glucose, enhanced glucose tolerance, and increased insulin sensitivity. Serum lipid profiles showed marked decreases in total cholesterol, triglycerides, and LDL-C levels. Systemic inflammation, as measured by serum levels of IL-1β, IL-6, and MCP-1, decreased significantly. Improvements in liver function were observed, as indicated by reductions in ALT and AST levels. Liver transcriptomic analysis revealed modulation of pathways related to glutathione metabolism, MAPK signaling, and cAMP signaling, which were involved in insulin signaling and oxidative stress response. Additionally, arecoline mitigated gut dysbiosis by restoring microbial diversity, altering gut microbiota composition, and regulating key pathways involved in NAD biosynthesis and fatty acid β-oxidation, which were critical for maintaining energy homeostasis. CONCLUSION Arecoline improves glucose metabolism, lipid profiles, and liver function, while modulating gut microbiota and liver metabolic pathways, showing potential as a therapeutic agent for T2DM.
Collapse
Affiliation(s)
- Meng Xu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Wanggao Li
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Yuan Xu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Hui Xue
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Juan Du
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology, Singapore, Singapore
| | - Xiaosong Hu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Ding X, Chen Y, Du L, Li J, Meng X, Lv H, Tong B, Niu G, Jian T, Chen J. Benefits of inulin and fructo-oligosaccharides on high fat diet-induced type 2 diabetes mellitus by regulating the gut microbiota in mice. J Nutr Biochem 2025; 141:109908. [PMID: 40122150 DOI: 10.1016/j.jnutbio.2025.109908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is pathologically associated with gut microbiota imbalance, which is implicated in disease progression through metabolic and inflammatory pathways. The therapeutic potential of inulin, a well-characterized prebiotic, has been explored to mitigate T2DM via microbiota modulation. However, the efficacy of this intervention, with its performance dependent on the degree of polymerization (DP), requires further investigation. This study assessed the therapeutic roles of inulin (DP3-60) and fructo-oligosaccharides (FOS, DP3-10) in T2DM management. Dietary administration of these prebiotic compounds demonstrated a significant capacity to alleviate multiple metabolic pathologies, including obesity, insulin resistance, systemic inflammation, oxidative stress, dyslipidemia and hepatic steatosis in high-fat diet (HFD)-fed induced T2DM mice. Significant superior efficacy was observed in FOS for ameliorating glucose metabolic dysregulation, adipocyte hypertrophy, liver weight, and histopathological alterations in colonic tissue, while inulin exhibited greater potency in alleviating oxidative stress. Both inulin and FOS enhanced gut microbiota diversity and richness in T2DM mice, accompanied by a significant reduction in Firmicutes/Bacteroidetes ratio. Notably, the S24-7 family emerged as a crucial microbial taxon modulated by both inulin and FOS. Furthermore, FOS demonstrated superior capacity to restore HFD-induced gut microbiota. Taxonomically significant amplicon sequence variants (ASVs), which were altered by HFD and modulated by inulin and FOS, exhibited distinct taxonomic profiles between the two compounds. This study provides preliminary evidence that the biological effects and beneficial properties of inulin-type fructans exhibit DP-dependent variations, which may enhance their efficient utilization in metabolic disorders.
Collapse
Affiliation(s)
- Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yan Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Lanlan Du
- Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Jing Li
- Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xiuhua Meng
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Han Lv
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Bei Tong
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Guanting Niu
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Tunyu Jian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China; Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
26
|
Wang Y, Qian X, Chen L, Yong Y, Wu M, Li Y, Ni Z, Li L, Shao Y, Chen A. Structural characteristics of a polysaccharide isolated from Lactaruis volemus Fr. and its anti-diabetic effects regulated by the modulation of gut microbiota and metabolites. Int J Biol Macromol 2025; 307:142294. [PMID: 40118396 DOI: 10.1016/j.ijbiomac.2025.142294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
As natural bioactive compounds, polysaccharides have promising effects in the treatment of type 2 diabetes mellitus (T2DM) owing to their changes to the intestinal microenvironment; however, the mechanisms underlying their effects have not been elucidated. In the present study, a polysaccharide LV-P4-1 isolated from Lactaruis volemus Fr was purified and characterized. The hyperglycemic function and the regulatory effect of LV-P4-1 on the gut microbiota and its metabolites were investigated in a T2DM mouse model. LV-P4-1 was mainly consisted of Fuc, Gal, Glc, Man, and GlcA, with a molecular weight of 5.89 kDa. The functional groups and glycosyl linkage types of LV-P4-1 were investigated using Fourier transform infrared spectroscopy, methylation and nuclear magnetic resonance analyses. Oral administration of 400 mg/kg LV-P4-1 increased glucose metabolism and alleviated tissue damage in mice with T2DM. Moreover, LV-P4-1 significantly regulated the abundances of gut microbiota, changed metabolite levels, and altered some metabolic pathways involved in T2DM development. Spearman analysis showed that the alterations in the gut microbiota were closely related to the differential metabolites. These results suggest that LV-P4-1 may alleviate hyperglycemia by influencing the structure of the intestinal microbiota and regulating the metabolic profile by altering the activity of certain metabolic pathways.
Collapse
Affiliation(s)
- Yanan Wang
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China; Jiangsu Key Construction Laboratory of Food Resource Development and Quality Safe, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Xinyi Qian
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Lingzhi Chen
- School of Biotechnology, Jiangnan University, 214122 Wuxi, Jiangsu, China
| | - Yidan Yong
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Mengmeng Wu
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Yihao Li
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Zaizhong Ni
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China; Jiangsu Key Construction Laboratory of Food Resource Development and Quality Safe, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Lulu Li
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China; Jiangsu Key Construction Laboratory of Food Resource Development and Quality Safe, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Ying Shao
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China; Jiangsu Key Construction Laboratory of Food Resource Development and Quality Safe, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China
| | - Anhui Chen
- College of Food and Bioengineering, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China; Jiangsu Key Construction Laboratory of Food Resource Development and Quality Safe, Xuzhou University of Technology, 221018 Xuzhou, Jiangsu, China.
| |
Collapse
|
27
|
Liu J, Li F, Yang L, Luo S, Deng Y. Gut microbiota and its metabolites regulate insulin resistance: traditional Chinese medicine insights for T2DM. Front Microbiol 2025; 16:1554189. [PMID: 40177494 PMCID: PMC11963813 DOI: 10.3389/fmicb.2025.1554189] [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: 01/06/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
The gut microbiota is closely associated with the onset and development of type 2 diabetes mellitus (T2DM), characterized by insulin resistance (IR) and chronic low-grade inflammation. However, despite the widespread use of first-line antidiabetic drugs, IR in diabetes and its complications continue to rise. The gut microbiota and its metabolic products may promote the development of T2DM by exacerbating IR. Therefore, regulating the gut microbiota has become a promising therapeutic strategy, with particular attention given to probiotics, prebiotics, synbiotics, and fecal microbiota transplantation. This review first examines the relationship between gut microbiota and IR in T2DM, summarizing the research progress of microbiota-based therapies in modulating IR. We then delve into how gut microbiota-related metabolic products contribute to IR. Finally, we summarize the research findings on the role of traditional Chinese medicine in regulating the gut microbiota and its metabolic products to improve IR. In conclusion, the gut microbiota and its metabolic products play a crucial role in the pathophysiological process of T2DM by modulating IR, offering new insights into potential therapeutic strategies for T2DM.
Collapse
Affiliation(s)
- Jing Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fuxing Li
- Ningxiang Traditional Chinese Medicine Hospital, Changsha, China
| | - Le Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Shengping Luo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihui Deng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
28
|
Liu S, Li F, Cai Y, Sun L, Ren L, Yin M, Cui H, Pan Y, Gang X, Wang G. Gout drives metabolic dysfunction-associated steatotic liver disease through gut microbiota and inflammatory mediators. Sci Rep 2025; 15:9395. [PMID: 40102566 PMCID: PMC11920238 DOI: 10.1038/s41598-025-94118-7] [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: 11/25/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
This study explores the relationship between gout and metabolic dysfunction-associated steatotic liver disease (MASLD), two metabolic conditions linked to worsening health outcomes. While hyperuricemia's association with MASLD is established, the specific connection between gout and MASLD remains less explored. Using data from the UK Biobank, the study employs COX proportional hazard models, multi-state survival analysis, and Mendelian randomization to assess the independent and mutual risks of gout and MASLD. Findings indicate a mutual risk increase: male gout patients, those younger than 60, and those with high BMI are particularly susceptible to MASLD, while female MASLD patients are at heightened risk for gout. Shared risk factors for both conditions include high BMI, hypertension, diabetes, and hyperuricemia. The study further identifies a bidirectional causal link, with gout leading to MASLD, mediated by gut microbiota Ruminococcaceae and proteins like IL-2 and GDF11, implicating specific metabolic pathways. The findings highlight a clinical and mechanistic correlation, emphasizing the need for targeted interventions to address these overlapping metabolic pathways in future treatments.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Fan Li
- Department of Hepatobiliary and Pancreatic Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yunjia Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Mengsha Yin
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Huijuan Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yujie Pan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
29
|
Zhu D, Li S, Xu Z, Kulyar MF, Bai X, Wang Y, Wang B, Khateeb E, Deng D, Wang L, Chen Y, Guo A, Shen Y. Comparative analysis of gut microbiota in healthy and diarrheic foals. Microbiol Spectr 2025:e0087124. [PMID: 40105330 DOI: 10.1128/spectrum.00871-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/01/2025] [Indexed: 03/20/2025] Open
Abstract
Diarrhea presents a substantial risk of high morbidity and mortality among foals. Although studies have shown connections between gut microbiota and several gastrointestinal diseases, there is still inadequate information on gut microbial alterations in foals during diarrhea. In this study, we conducted 16S rRNA and ITS gene amplicon sequencing to investigate gut bacterial and fungal differences between healthy and diarrheic foals. The results unveiled significant reductions in gut bacterial and fungal diversities among foals experiencing diarrhea, accompanied by notable shifts in the composition of gut microbial communities. A considerable decrease was observed in the relative abundance of 30 bacterial and 34 fungal genera. Moreover, two bacterial and eight fungal genera were utterly undetectable in the gut microbiota of diarrheic foals. Some decreased genera, such as Bifidobacterium and Saccharomyces, were deemed beneficial and recognized as probiotics. The study revealed significant alterations in foals' gut bacterial and fungal communities during diarrhea, which enriched our comprehension of gut microbial dynamics in foals across varying health statuses. These findings offer valuable insights for managing diarrhea through gut microbiota modulation, suggesting that probiotics may be superior to antibiotics in preventing and controlling foal diarrhea.IMPORTANCEThis research advances the understanding of gut bacterial and fungal dynamics in foals, highlighting gut microbiota dysbiosis as a potential contributor to foal diarrhea. Additionally, we observed that many altered bacteria and fungi were downregulated during diarrhea, including some probiotic strains. Consequently, our findings provide evidence that probiotics may offer superior efficacy compared with antibiotics as potential candidates for preventing and treating foal diarrhea.
Collapse
Affiliation(s)
- Di Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siyu Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Md F Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xu Bai
- China Horse Industry Association, Beijing, China
| | - Yu Wang
- China Horse Industry Association, Beijing, China
| | - Boya Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Emaan Khateeb
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dandan Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lidan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuji Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
30
|
Peng C, Lei P, Qi H, Zhu Q, Huang C, Fu J, Zhao C. Effect of fecal microbiota transplantation on diabetic wound healing through the IL-17A-mTOR-HIF1α signaling axis. Appl Environ Microbiol 2025; 91:e0201924. [PMID: 40019272 PMCID: PMC11921319 DOI: 10.1128/aem.02019-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Diabetes is the third most common chronic disorder worldwide. Diabetic wounds are a severe complication that is costly and often results in non-traumatic lower limb amputation. Recent investigations have demonstrated that the gut microbiota as a "virtual organ" can regulate metabolic diseases like diabetes. Fecal microbiota transplantation (FMT) is an innovative therapeutic approach for promoting wound healing, but its function remains incompletely defined. A diabetes model was established by supplying mice with a high-fat diet and performing an intraperitoneal injection of streptozotocin. Diabetic wounds were then created, followed by bacterial transplantation. The relevant indexes of wound healing were evaluated to verify the promoting effect of FMT on the diabetic wounds. Human skin keratinocytes were also cultured, and cell scratch experiments were conducted to further investigate the underlying mechanism. The FMT regulated the levels of specific bacteria in the diabetic mice and helped restore the balance of intestinal microbes. This transplantation also enhanced wound healing in the diabetic mice by augmenting the closure rate, accelerating re-epithelialization, and boosting collagen deposition in skin wounds. Furthermore, FMT promoted the production of IL-17A, which significantly enhanced the growth and movement of human keratinocytes. Inhibiting molecules related to the IL-17A-mTOR-HIF1α signaling axis were shown to hinder wound re-epithelialization.This study clarifies the function of the IL-17A-mTOR-HIF1α signaling axis in the utilization of FMT in diabetic wound healing, providing a new therapeutic method and target for promoting the healing of diabetic wounds. IMPORTANCE The Intestinal microbiota, as the organ with the largest number of microorganisms in the body, plays a crucial role in the physiological functions of the human body. Normal microbiota can be involved in various functions such as energy absorption, metabolism, and immunity of the body, and microbiota imbalance is related to many diseases such as obesity and diabetes. Diabetes, as one of the world's three major chronic diseases, is a significant health issue that troubles more than a billion people globally. Diabetic wounds are a problem that all diabetic patients must confront when undergoing surgery, and it is an important cause of non-traumatic amputations. Exploring the role of intestinal microorganisms in the wound-healing process of diabetic mice can offer the possibility of using microorganisms as a therapeutic means to intervene in clinically related diseases.
Collapse
Affiliation(s)
- Chenmei Peng
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Pan Lei
- Department of General Practice Medicine, Qinghai University Affiliated Hospital, Xining, China
| | - Hongying Qi
- Department of Endocrinology, Qinghai University Affiliated Hospital, Xining, China
| | - Qianjun Zhu
- Department of Endocrinology, Qinghai Province People’s Hospital, Xining, China
| | - Chushun Huang
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Ju Fu
- Qinghai University Affiliated Hospital, Qinghai University, Xining, China
| | - Chengyu Zhao
- Department of Geriatrics, Qinghai University Affiliated Hospital, Xining, China
| |
Collapse
|
31
|
Shi H, Li J. MAGs-based genomic comparison of gut significantly enriched microbes in obese individuals pre- and post-bariatric surgery across diverse locations. Front Cell Infect Microbiol 2025; 15:1485048. [PMID: 40171165 PMCID: PMC11958714 DOI: 10.3389/fcimb.2025.1485048] [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: 08/23/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Obesity, a pressing global health issue, is intricately associated with distinct gut microbiota profiles. Bariatric surgeries, such as Laparoscopic Sleeve Gastrectomy (LSG), Sleeve Gastrectomy (SG), and Roux-en-Y Gastric Bypass (RYGB), induce substantial weight loss and reshape gut microbiota composition and functionality, yet their comparative impacts remain underexplored. Methods This study integrated four published metagenomic datasets, encompassing 500 samples, and employed a unified bioinformatics workflow for analysis. We assessed gut microbiota α-diversity, identified species biomarkers using three differential analysis approaches, and constructed high-quality Metagenome-Assembled Genomes (MAGs). Comparative genomic, functional profiling and KEGG pathway analyses were performed, alongside estimation of microbial growth rates via Peak-to-Trough Ratios (PTRs). Results RYGB exhibited the most pronounced enhancement of gut microbiota α-diversity compared to LSG and SG. Cross-cohort analysis identified 39 species biomarkers: 27 enriched in the non-obesity group (NonOB_Enrich) and 12 in the obesity group (OB_Enrich). Among the MAGs, 177 were NonOB_Enrich and 14 were OB_Enrich. NonOB_Enrich MAGs displayed enriched carbohydrate degradation profiles (e.g., GH105, GH2, GH23, GH43, and GT0 families) and higher gene diversity in fatty acid biosynthesis and secondary metabolite pathways, alongside significant enrichment in amino acid metabolism (KEGG analysis). Post-surgery, Akkermansia muciniphila and Bacteroides uniformis showed elevated growth rates based on PTRs. Discussion These findings underscore RYGB's superior impact on gut microbiota diversity and highlight distinct microbial functional adaptations linked to weight loss, offering insights for targeted therapeutic strategies.
Collapse
Affiliation(s)
| | - Jia Li
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
32
|
Nie LJ, Cheng Z, He YX, Yan QH, Sun YH, Yang XY, Tian J, Zhu PF, Yu JY, Zhou HP, Zhou XQ. Role of duodenal mucosal resurfacing in controlling diabetes in rats. World J Diabetes 2025; 16:102277. [PMID: 40093272 PMCID: PMC11885968 DOI: 10.4239/wjd.v16.i3.102277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 01/03/2025] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND The duodenum plays a significant role in metabolic regulation, and thickened mucous membranes are associated with insulin resistance. Duodenal mucosal resurfacing (DMR), a new-style endoscopic procedure using hydrothermal energy to ablate this thickened layer, shows promise for enhancing glucose and lipid metabolism in type 2 diabetes (T2D) patients. However, the mechanisms driving these improvements remain largely unexplored. AIM To investigate the mechanisms by which DMR improves metabolic disorders using a rat model. METHODS Rats with T2D underwent a revised DMR procedure via a gastric incision using a specialized catheter to abrade the duodenal mucosa. The duodenum was evaluated using histology, immunofluorescence, and western blotting. Serum assays measured glucose, lipid profiles, lipopolysaccharide, and intestinal hormones, while the gut microbiota and metabolomics profiles were analyzed through 16S rRNA gene sequencing and ultra performance liquid chromatography-mass spectrum/mass spectrum, severally. RESULTS DMR significantly improved glucose and lipid metabolic disorders in T2D rats. It increased the serum levels of cholecystokinin, gastric inhibitory peptide, and glucagon-like peptide 1, and reduced the length and depth of duodenal villi and crypts. DMR also enhanced the intestinal barrier integrity and reduced lipopolysaccharide translocation. Additionally, DMR modified the gut microbiome and metabolome, particularly affecting the Blautia genus. Correlation analysis revealed significant links between the gut microbiota, metabolites, and T2D phenotypes. CONCLUSION This study illustrates that DMR addresses metabolic dysfunctions in T2D through multifaceted mechanisms, highlighting the potential role of the Blautia genus on T2D pathogenesis and DMR's therapeutic impact.
Collapse
Affiliation(s)
- Li-Juan Nie
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhe Cheng
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yi-Xian He
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Qian-Hua Yan
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Yao-Huan Sun
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Xin-Yi Yang
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Jie Tian
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Peng-Fei Zhu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Jiang-Yi Yu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| | - Hui-Ping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Xi-Qiao Zhou
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
33
|
Wang Y, He X, Gao Y, Xue M, Zhang H, Sun L, He Q, Jin J. Disorders of gut microbiota and fecal-serum metabolic patterns are associated with pulmonary tuberculosis and pulmonary tuberculosis comorbid type 2 diabetes mellitus. Microbiol Spectr 2025:e0177224. [PMID: 40084872 DOI: 10.1128/spectrum.01772-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/31/2025] [Indexed: 03/16/2025] Open
Abstract
Pulmonary tuberculosis (PTB) and diabetes mellitus (DM) are prevalent chronic diseases with substantial implications for human health. DM patients are more susceptible to PTB, which exacerbates diabetes-related complications. However, the complex molecular mechanisms underlying the enhanced susceptibility of DM patients to PTB infection remain poorly understood. In this study, α- and β-diversity of gut microbiota was significantly reduced in PTB patients and PTB-DM patients. The abundances of families Lachnospiraceae and Ruminococcaceae in the the Firmicutes phylum were reduced in PTB patients and further diminished in PTB-DM patients. On the other hand, untargeted metabolomics in frozen serum and stool samples indicated that phenylalanine, tyrosine, and tryptophan biosynthesis, metabolites of arginine, proline, tryptophan, and histidine were consistently altered in PTB patients and PTB-DM patients, with significant upregulation of most metabolites. Amino acids like serine, proline, and histidine were both remarkably elevated in PTB and PTB-DM patients. The correlation network analysis reveals the relationships between the shared microbial biomarkers and the shared metabolic pathways. This research contributes to the exploration of pivotal diagnostic biomarkers for both patients with PTB and PTB accompanied by diabetes. Specifically, shared reductions were identified in the genera g-Roseburia, g-Ruminococcaceae_UCG.013, g-Ruminococcaceae_NK4A214, g-Lachnospiraceae_unclassified, and g-Firmicutes_unclassified in addition to notable regulation of amino acids, like glycine, serine, and histidine in patients with PTB and PTB-DM. Our study expands the comprehension of the intricate connections linking gut microbiota, fecal metabolites, and serum metabolites in PTB and PTB-DM patients. IMPORTANCE This study expands the understanding of the complex links between gut microbiota, fecal metabolites, and serum metabolites in patients with PTB and PTB-DM through multi-omics techniques. It is helpful for us to understand the complex molecular mechanism of increased susceptibility to PTB infection in diabetic patients.
Collapse
Affiliation(s)
- Yunguang Wang
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Xinxin He
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Yixuan Gao
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Mengjiao Xue
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Hua Zhang
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Lifang Sun
- Department of Tuberculosis, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Qiang He
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| | - Juan Jin
- Department of Nephrology, the First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Research and Translation for Kidney Deficiency-Stasis-Turbidity Disease, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Barakat H, Aljutaily T. Role of γ-Aminobutyric Acid (GABA) as an Inhibitory Neurotransmitter in Diabetes Management: Mechanisms and Therapeutic Implications. Biomolecules 2025; 15:399. [PMID: 40149935 PMCID: PMC11940341 DOI: 10.3390/biom15030399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
GABA (γ-Aminobutyric Acid), a well-established inhibitory neurotransmitter in the central nervous system, has garnered considerable interest for its potential role in diabetes management, particularly due to its presence in pancreatic islets. This review aims to explore the therapeutic role of GABA in diabetes management and its potential mechanisms for antidiabetic effects. Relevant studies were searched across databases such as PubMed and ScienceDirect, applying strict eligibility criteria focused on GABA administration methods and diabetic models. The collective results showed that the administration of GABA in diabetic models resulted in remarkable enhancements in glucose and insulin homeostasis, favorable modifications in lipid profiles, and amelioration of dysfunctions across neural, hepatic, renal, and cardiac systems. The findings from the literature demonstrated that GABAergic signaling within pancreatic tissues can significantly contribute to the stimulation of β cell proliferation through the facilitation of a sustained trans-differentiation process, wherein glucagon-secreting α cells are converted into insulin-secreting β-like cells. In addition, activated GABAergic signaling can trigger the initiation of the PI3K/AKT signaling pathway within pancreatic tissues, leading to improved insulin signaling and maintained glucose homeostasis. GABAergic signaling can further function within hepatic tissues, promoting inhibitory effects on the expression of genes related to gluconeogenesis and lipogenesis. Moreover, GABA may enhance gut microbiota diversity by attenuating gut inflammation, attributable to its anti-inflammatory and immunomodulatory properties. Furthermore, the neuroprotective effects of GABA play a significant role in ameliorating neural disorders associated with diabetes by facilitating a substantial reduction in neuronal apoptosis. In conclusion, GABA emerges as a promising candidate for an antidiabetic agent; however, further research is highly encouraged to develop a rigorously designed framework that comprehensively identifies and optimizes the appropriate dosages and intervention methods for effectively managing and combating diabetes.
Collapse
Affiliation(s)
- Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | | |
Collapse
|
35
|
Yi HM, Won S, Pak J, Park SE, Kim MR, Kim JH, Park EY, Hwang SY, Lee MH, Son HS, Kwak S. Fecal Microbiome and Urine Metabolome Profiling of Type 2 Diabetes. J Microbiol Biotechnol 2025; 35:e2411071. [PMID: 40147938 PMCID: PMC11985407 DOI: 10.4014/jmb.2411.11071] [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: 11/28/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Type 2 diabetes is a prevalent metabolic disorder with serious health consequences, necessitating both enhanced diagnostic methodologies and comprehensive elucidation of its pathophysiological mechanisms. We compared fecal microbiome and urine metabolome profiles in type 2 diabetes patients versus healthy controls to evaluate their respective diagnostic potential. Using a cohort of 94 subjects (48 diabetics, 46 controls), this study employed 16S rRNA amplicon sequencing for fecal microbiome analysis and GC-MS for urinary metabolomics. While fecal microbiome alpha diversity showed no significant differences between groups, urinary metabolomics demonstrated distinct structural patterns and higher evenness in type 2 diabetes patients. The study identified several diabetes-associated urinary metabolites, including elevated levels of glucose and inositol, along with decreased levels of 6 urine metabolites including glycolic acid, hippurate, and 2-aminoethanol. In the fecal microbiome, genera such as Escherichia-Shigella showed positive correlation with type 2 diabetes, while Lacticaseibacillus demonstrated negative correlation. Receiver operating characteristic curve analyses revealed that urinary metabolites exhibited superior diagnostic potential compared to fecal microbiome features, with an area under the curve of 0.90 for the combined metabolite model versus 0.82 for the integrated bacterial taxa model. These findings suggest that urinary metabolomics may offer a more reliable approach for type 2 diabetes diagnosis compared to fecal 16S metabarcoding, while highlighting the potential of multi-marker panels for enhanced diagnostic accuracy.
Collapse
Affiliation(s)
- Hye-Min Yi
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
- Dangbom Korean Medicine Clinic, Seoul 03192, Republic of Korea
| | - Seok Won
- Department of Bio and Fermentation Convergence Technology, College of Science and Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Juhan Pak
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Seong-Eun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Mi-Ri Kim
- Dangbom Korean Medicine Clinic, Seoul 03192, Republic of Korea
| | - Ji-Hyun Kim
- Dangbom Korean Medicine Clinic, Seoul 03192, Republic of Korea
| | - Eun-Young Park
- Dangbom Korean Medicine Clinic, Seoul 03192, Republic of Korea
| | - Sun-Young Hwang
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
| | - Hong-Seok Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Suryang Kwak
- Department of Bio and Fermentation Convergence Technology, College of Science and Technology, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
36
|
Huang H, Zhao T, Ma W. Omega-3 polyunsaturated fatty acids attenuates cognitive impairment via the gut-brain axis in diabetes-associated cognitive dysfunction rats. Brain Behav Immun 2025; 127:147-169. [PMID: 40068791 DOI: 10.1016/j.bbi.2025.03.015] [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: 07/09/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Diabetes-related cognitive dysfunction (DACD) is a comorbidity of type 2 diabetes that has a negative effect on patients' quality of life. Research has indicated that disruption of the gut microbiota (GM) may be linked to dementia with altered cognitive performance. Conversely, omega-3 polyunsaturated fatty acids (n-3 PUFAs) may reverse DACD. The present study aimed to assess the effects of an n-3 PUFA intervention and fecal microbiota transplantation (FMT) on high-fat and streptozotocin-induced DACD model rats. In DACD rats, n-3 PUFA treatment restored fasting blood glucose (FBG) levels and cognitive function, increased the expression of anti-inflammatory cytokines and downregulated the expression of proinflammatory cytokines in the cortex and colon. Additionally, the expression of the postsynaptic density protein-95 mRNA and protein varied with n-3 PUFA treatment. Treatment with n-3 PUFAs also increased the expression of tight junction proteins. Beneficial and short-chain fatty acid-producing bacteria were more abundant when rats were exposed to n-3 PUFAs. After FMT from the rats with DACD symptoms that were improved by the n-3 PUFA dietary intervention into another batch of DACD rats, we observed recovery in recipient DACD rats. These results indicated that the alleviation of DACD symptoms by n-3 PUFAs was attributed to gut microbiota remodeling.
Collapse
Affiliation(s)
- Hongying Huang
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Nanchang Institute of Disease Control and Prevention, China Railway Nanchang Bureau Group Co., Ltd., Nanchang, 330003, People's Republic of China
| | - Tong Zhao
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China.
| |
Collapse
|
37
|
Misnikova I, Kovaleva Y, Shokur S, LeBaron TW, Povarova O, Medvedev O. Hydrogen and Methane Detection in Breath in Response to Two Different Types of Dietary Fiber and Its Relationship to Postprandial Glucose Concentration in Obese Patients with Type 2 Diabetes and Normoglycemic Subjects. Nutrients 2025; 17:917. [PMID: 40077785 PMCID: PMC11902166 DOI: 10.3390/nu17050917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Background: The aim of this study was to investigate the relationship between postprandial glycemic levels based on flashmonitoring and the production of intestinal hydrogen (H2) and methane (CH4) gases based on the measurement of the amount of these gases in exhaled air. Materials and Methods: We studied 14 subjects with type 2 diabetes mellitus (T2DM) and 14 individuals without diabetes (control) with two food load tests, including two types of dietary fiber (inulin and guar gum), with the simultaneous determination of gases in exhaled air and the assessment of glucose levels. Results: All subjects in the control group had a significant increase in exhaled H2. OR for increased hydrogen production in patients with T2DM was 0.17 (95% CI 0.031-0.93, p = 0.043). The level of H2 in exhaled breath after food load in patients with T2DM was lower than in normoglycemic subjects. There was an inverse correlation between maximum glucose rise and maximum H2 in exhaled air after food load in normoglycemic subjects (r = -0.569, p = 0.034). Patients with T2DM had direct correlations between the level of CH4 in exhaled air and the parameters of postprandial glycemia in the lactulose test (p < 0.05). Conclusions: The confirmation of a causal relationship between decreased H2 production, increased intestinal CH4 production, and more severe postprandial glycemia may identify new therapeutic targets in the correction of postprandial glycemia in patients with T2DM.
Collapse
Affiliation(s)
- Inna Misnikova
- M.F. Vladimirski Moscow Regional Research and Clinical Institute, Schepkina 61/2, 129110 Moscow, Russia; (I.M.); (Y.K.)
| | - Yulia Kovaleva
- M.F. Vladimirski Moscow Regional Research and Clinical Institute, Schepkina 61/2, 129110 Moscow, Russia; (I.M.); (Y.K.)
| | - Svetlana Shokur
- M.F. Vladimirski Moscow Regional Research and Clinical Institute, Schepkina 61/2, 129110 Moscow, Russia; (I.M.); (Y.K.)
| | - Tyler W. LeBaron
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA;
- Molecular Hydrogen Institute, Enoch, UT 84721, USA
| | - Oxana Povarova
- Department of Pharmacology, M. V Lomonosov Moscow State University, Lomonosovsky Prospect 27-1, 119991 Moscow, Russia;
| | - Oleg Medvedev
- Department of Pharmacology, M. V Lomonosov Moscow State University, Lomonosovsky Prospect 27-1, 119991 Moscow, Russia;
- National Medical Research Center of Cardiology, Laboratory of Experimental Pharmacology, Academician Chazov Str., 15a, 121552 Moscow, Russia
| |
Collapse
|
38
|
Li S, Liu J, Zhang X, Gu Q, Wu Y, Tao X, Tian T, Pan G, Chu M. The Potential Impact of Antibiotic Exposure on the Microbiome and Human Health. Microorganisms 2025; 13:602. [PMID: 40142495 PMCID: PMC11944296 DOI: 10.3390/microorganisms13030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Antibiotics are a cornerstone of modern medicine, saving countless lives. However, their widespread use presents two major challenges. First, antibiotic-induced changes in the microbiome can disrupt immune function, increasing the susceptibility to diseases associated with these alterations. Second, prolonged antibiotic use fosters the proliferation of antibiotic resistance genes, leading to the emergence of resistant strains and threatening our ability to control infections. These challenges highlight an urgent global health crisis, necessitating in-depth investigation into the multifaceted effects of antibiotic exposure on microbiome dynamics and human health. In this review, we explore the potential effects of antibiotic exposure on the microbiome and its implications for overall health. Additionally, we examine the role of emerging technologies in addressing these challenges and in shaping future antibiotic development. Our goal is to provide insights that will inform more effective public health strategies and interventions aimed at mitigating the adverse consequences of antibiotic use, restoring microbial balance, and improving overall health outcomes.
Collapse
Affiliation(s)
- Siqi Li
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Jiahao Liu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xinyang Zhang
- School of Medical, Nantong University, Nantong 226019, China;
| | - Qihong Gu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Yutong Wu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xiaobo Tao
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Tian Tian
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Gongbu Pan
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7005, Australia
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| |
Collapse
|
39
|
Huang H, Liu S, Peng Z, Wang B, Zhan S, Huang S, Li W, Liu D, Yang X, Zhu Y, Xiao W. Comparative effects of different sugar substitutes: Mogroside V, stevioside, sucralose, and erythritol on intestinal health in a type 2 diabetes mellitus mouse. Food Funct 2025; 16:2108-2123. [PMID: 39969196 DOI: 10.1039/d4fo04446k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Intestinal health disorders significantly contribute to the development of type 2 diabetes mellitus (T2DM). Sugar substitutes such as mogroside V (MOG), stevioside (ST), sucralose (TGS), and erythritol (ERT), are increasingly used in T2DM management as alternatives to sucrose (SUC). However, their effects on intestinal health in T2DM have not been fully compared. In the present study, we established a T2DM mouse model using a high-fat diet and streptozotocin injection. These mice were treated with equal doses of SUC, MOG, ST, TGS, or ERT for 4 weeks to evaluate the effects of these sugar substitutes on intestinal health in T2DM. T2DM mice exhibited increased intestinal permeability, reduced goblet cell numbers, elevated pro-inflammatory cytokine levels, and alterations in both gut microbiota and metabolite composition. After 4 weeks of treatment, MOG showed the most significant benefits. MOG activates the PI3K/AKT pathway, enhancing the expression of tight junction proteins, which improves intestinal barrier function and reduces permeability. This is accompanied by NF-κB inhibition, leading to reduced pro-inflammatory cytokine production and increased mucus secretion. These changes help maintain healthy gut microbiota and metabolites, preventing pathogenic bacteria from entering the bloodstream. ST downregulates NF-κB to alleviate intestinal inflammation and improves gut microbiota and metabolic homeostasis in T2DM. ERT has less beneficial effects. TGS and SUC reduce intestinal inflammation and have a better effect on the duodenum. However, TGS has a negative effect on the colon microbiota and metabolites, whereas SUC has a negative effect on the colon microbiota alone. MOG improved intestinal health in T2DM by modulating the PI3K/AKT and NF-κB pathways, whereas ST primarily modulated NF-κB to alleviate intestinal inflammation. Both treatments were effective, with MOG showing the best performance. Therefore, MOG can be considered a viable alternative to SUC for T2DM management.
Collapse
Affiliation(s)
- Huaxue Huang
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sha Liu
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Zhi Peng
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Bin Wang
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Shuang Zhan
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sirui Huang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Wei Li
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Dai Liu
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Xiulian Yang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Yizhun Zhu
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
| | - Wenjun Xiao
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
40
|
Zhang S, Niu H, Zhu J. Personalized nutrition studies of human gut microbiome-polyphenol interactions utilizing continuous multistaged in vitro fermentation models-a narrative review. Nutr Res 2025; 135:101-127. [PMID: 39999639 DOI: 10.1016/j.nutres.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
The gut microbiota, a complex community of microorganisms primarily inhabiting the human large intestine, plays a crucial role in human health. Gut dysbiosis, characterized by an imbalance in gut bacterial populations, has been increasingly recognized as a significant factor in the pathogenesis of metabolic diseases such as type 2 diabetes, inflammatory bowel disease, and colorectal cancer. Polyphenols are critical modulators of gut microbial composition and metabolism. However, the extent of polyphenol-induced modulation of the gut microbiome remains largely unexplored. In vitro models offer a convenient and ethical alternative to in vivo studies for investigating nutrient-gut microbiome interactions, facilitating easy sampling and controlled experimental conditions. Among these, continuous multistaged in vitro fermentation models, which simulate different sections of the human gastrointestinal tract (e.g., proximal colon, transverse colon, and distal colon), provide a more accurate representation of the human gut environment compared to single-batch fermentation. Various configurations of these multistaged models have been developed and widely employed in studies examining the effects of polyphenols on the gut microbiome. This review aims to summarize the different configurations of multistaged in vitro fermentation models and recent advancements in their development, highlight key aspects of experimental design, outline commonly used analytical workflows with complementary analyses, and review the restorative effects of polyphenol interventions on dysregulated gut microbiota.
Collapse
Affiliation(s)
- Shiqi Zhang
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Hanmeng Niu
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
41
|
Xiao J, Chen B, Chen L, Wang Q, Tan S, Yuan H, Xiang D, Zhang B, Li X, Huang S, Tan Y, Cheng Y, Xie W, Xu P. Interpretable time-series neural turing machine for prognostic prediction of patients with type 2 diabetes in physician-pharmacist collaborative clinics. Int J Med Inform 2025; 195:105737. [PMID: 39626597 DOI: 10.1016/j.ijmedinf.2024.105737] [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: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 02/12/2025]
Abstract
BACKGROUND Type 2 diabetes (T2D) has become a serious health threat globally. However, the existing approaches for diabetes prediction mainly had difficulty in addressing multiple time-series features. This study aims to provide an adjunctive tool for the clinical identification of patients in physician-pharmacist collaborative clinics at high risk of poor prognosis. METHODS This study proposes a novel interpretable time-series Neural Turing Machine (ITS-NTM) to form patient characteristics into feature matrixes to simulate one's disease and treatment process, predicting the prognosis of patients with T2D and alerting early interventions. Model robustness was verified by 10-fold cross-validation, external validation and multi-model comparisons. We also conducted dynamic prediction and feature importance analysis to explore its interpretability. RESULTS The study population included patients with T2D attending physician-pharmacist collaborative clinics over 12 months in primary healthcare centers, while clinical features and behavioral indicators at baseline, 3rd, 6th, 9th and 12th months were used to reflect the fluctuation of disease control over time. Compared with five state-of-the-art prediction models, the ITS-NTM obtains 92.0 % in accuracy and 91.8 % F1-score, demonstrating the superiority performance. Feature importance demonstrated that the top 5 features were glycosylated hemoglobin, fasting blood glucose, medication adherence scores, 2-hour postprandial blood glucose and waist-to-hip ratio, which had the greatest impact on the performance of the predictive model. CONCLUSIONS Proposed ITS-NTM could be used to promote the implementation of physician-pharmacist collaborative clinics, and further prompt the application of artificial intelligence to optimize the allocation of medical resources and improve the quality of care in under-resourced areas.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bin Chen
- KylinSoft Corporation Ltd, Changsha, China; School of Automatic, Central South University, Changsha, China
| | - Lei Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qing Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shenglan Tan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyan Yuan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xia Li
- Department of Endocrine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuting Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuhan Tan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yining Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenzheng Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China; Institute of Clinical Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
42
|
Wang K, Liu Z, Tang R, Sha Y, Wang Z, Chen Y, Chen G. Gallstones in the Era of Metabolic Syndrome: Pathophysiology, Risk Prediction, and Management. Cureus 2025; 17:e80541. [PMID: 40225487 PMCID: PMC11993725 DOI: 10.7759/cureus.80541] [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] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Gallstone disease (GSD) and metabolic syndrome (MetS) are increasingly prevalent conditions with significant global health implications. Recent evidence highlights a strong epidemiological association between these disorders, driven by shared pathophysiological mechanisms. This review provides a comprehensive analysis of the intricate relationship between MetS and GSD, focusing on the role of insulin resistance, dyslipidemia, obesity, and gut microbiota dysbiosis in gallstone formation. An integrated pathophysiological model is proposed, linking metabolic disturbances to bile cholesterol supersaturation, gallbladder dysmotility, and chronic inflammation. The review also explores clinical implications, including risk prediction models based on metabolic parameters, early detection biomarkers, and targeted interventions such as lifestyle modifications, pharmacological therapies, and microbiome modulation. By addressing the metabolic underpinnings of GSD, this synthesis offers a foundation for developing preventive and therapeutic strategies to mitigate the burden of these interconnected conditions. Future research directions are outlined to refine mechanistic insights and improve clinical outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Surgery, Wannan Medical College, Wuhu, CHN
| | - Zhigang Liu
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| | - Rongmei Tang
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| | | | | | - Yisheng Chen
- General Surgery, Wuhu Guangji Hospital, Wuhu, CHN
| | - Guangbin Chen
- Hepatobiliary Surgery, The Second People's Hospital of Wuhu, Wuhu Hospital Affiliated to East China Normal University, Wuhu, CHN
| |
Collapse
|
43
|
Ma N, Li R, Zhang GF, Gao RH, Zhang DJ. Fermentation-enriched quinoa β-glucan ameliorates disturbed gut microbiota and metabolism in type 2 diabetes mellitus mice. Int J Biol Macromol 2025; 306:141666. [PMID: 40032090 DOI: 10.1016/j.ijbiomac.2025.141666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Quinoa β-glucan (QBG) has shown potential benefits in treating type 2 diabetes mellitus (T2DM); however, comprehensive evaluations of its effects remain limited. This study investigates the impact of QBG-derived from hot water extraction (Q-) and microbial fermentation enrichment (Q+)-on serum glucose levels, lipid profiles, appetite-regulating hormones, fecal short-chain fatty acids (SCFAs), and gut microbiota composition and function in streptozotocin/high-fat diet (STZ/HFD)-induced T2DM mice. The results indicate that QBG treatment significantly reduced fasting blood glucose, insulin levels, triglycerides (TG) and total cholesterol (TC), while concurrently increasing high-density lipoprotein cholesterol (HDLC) levels. Additionally, liver and pancreatic function improved, as evidenced by decreased levels of malondialdehyde (MDA), aspartate transaminase (AST), and alanine transaminase (ALT). SCFA levels were significantly higher in QBG-treated groups compared to MC group. QBG treatment also reduced the abundance of Firmicutes and Patescibacteria, along with the Firmicutes/Bacteroidota ratio, while increasing levels of Bacteroidota and Actinobacteria. These findings suggest that QBG can regulate the dysbiosis of SCFAs production in T2DM mice and may indirectly modulate the secretion of appetite-regulating hormones by influencing gut microbiota composition. Furthermore, PICRUSt analysis revealed that QBG treatment, particularly Q + _H, could enhance disrupted metabolism and improve gut microbiota functions, helping restore normal physiological function.
Collapse
Affiliation(s)
- Nan Ma
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Heilongjiang Bayi Agricultural University Mudanjiang Institute of Food and Biotechnology, Mudanjiang 157000, China; Daqing Center of Inspection and Testing for Rural Affairs Agricultural Products and Processed Products, Ministry of Agriculture and Rural Affairs, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Rong Li
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Gui-Fang Zhang
- National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ruo-Han Gao
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Dong-Jie Zhang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Daqing Center of Inspection and Testing for Rural Affairs Agricultural Products and Processed Products, Ministry of Agriculture and Rural Affairs, Heilongjiang Bayi Agricultural University, Daqing 163319, China; National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China; Key Laboratory of Agro-Products Processing and Quality Safety of Heilongjiang Province, Daqing 163319, China.
| |
Collapse
|
44
|
Li X, Wen H, Ke J, Zhao D. Association of constipation with all-cause mortality among individuals with type 2 diabetes: A retrospective cohort study. J Diabetes Investig 2025; 16:501-509. [PMID: 39718116 PMCID: PMC11871400 DOI: 10.1111/jdi.14375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Constipation is a common complication in type 2 diabetes mellitus (T2DM), yet its impact on mortality remains unclear. This study aimed to investigate the association between constipation and all-cause mortality in patients with T2DM. METHODS We conducted a retrospective cohort study using data from the National Health and Nutrition Examination Survey (NHANES) 2005-2010. Mortality outcomes were ascertained through linkage to National Death Index records until December 31, 2019. The association between constipation and all-cause mortality was assessed using weighted Cox proportional hazards regression models. Kaplan-Meier curves were then employed to visualize survival probabilities. Effect modification was explored through stratified analyses and interaction tests. RESULTS Of 1,339 participants with T2DM, 146 (10.90%) reported constipation. During a median follow-up of 10.75 years, 411 deaths occurred (57 in the constipation group, 354 in the non-constipation group). Fully adjusted weighted Cox regression analysis revealed that constipation was associated with increased all-cause mortality (HR 1.50, 95% CI 1.01-2.22, P = 0.04). Kaplan-Meier analysis demonstrated a significantly lower survival probability in patients with constipation (log-rank P < 0.05). Stratified analyses and interaction tests corroborated these findings across various subgroups. CONCLUSIONS Constipation is associated with elevated all-cause mortality risk in T2DM patients. These findings suggest that constipation management may be an important consideration in improving long-term outcomes for individuals with T2DM.
Collapse
Affiliation(s)
- Xianhua Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Lu He HospitalCapital Medical UniversityBeijingChina
| | - Haibin Wen
- Department of NephrologyJiang Bin Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Lu He HospitalCapital Medical UniversityBeijingChina
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Lu He HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
45
|
Ma C, Ju B, Liu J, Wen L, Zhao Y, Yang J, Hu J. Phenylethanol Glycosides from Cistanche tubulosa Modulate the Gut Microbiota and Cecal Metabolites to Ameliorate Diabetic Nephropathy Induced by Streptozotocin Combined with High-Fat Diet in Rats. J Med Food 2025; 28:219-231. [PMID: 39401174 DOI: 10.1089/jmf.2024.k.0175] [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: 03/19/2025] Open
Abstract
Diabetic nephropathy (DN) is a prevalent complication and serious microvascular of diabetes mellitus. After previous studies, we found that phenylethanol glycosides (CPhGs) derived from Cistanche tubulosa (Schenk) Wight exerts antidiabetic and renoprotective effects. However, the effects of CPhGs on DN remain incompletely understood. The study aimed to examine the effects of CPhGs on DN in rats and explore the underlying mechanism involved. A DN rat model was established by streptozotocin (STZ) combined with a high-fat diet. Reagent kits were used to assess the extent to which CPhGs ameliorate hyperglycemia, insulin resistance (IR), renal dysfunction, kidney oxidative stress, and peripheral inflammation. Histology and immunohistochemical staining were used to detect the changes in renal tissue structure and the expression levels of α-smooth muscle actin (α-SMA) and collagen I. Furthermore, we analyzed the cecal contents of DN rats to investigate the effect of CPhGs on gut microbiota by using 16S rRNA sequencing and broad-spectrum metabolite profiling. The results showed that CPhGs demonstrated a range of advantageous outcomes in DN, encompassing the enhancement of kidney function and alleviation of hyperglycemia, IR, renal injury, oxidative stress, and peripheral inflammatory reactions. In addition, CPhGs regulated the abundance of the [Eubacterium]_coprostanoligenes_group, Oscillospiraceae_UCG-005, etc. to modulate the gut microbiota. CPhGs significantly upregulated the content of vitamin B6 and tyrosyl-tryptophan and downregulated histamine, L-methionine, etc. In summary, the therapeutic efficacy of CPhGs on DN rats may be achieved by modulating the gut microbiota and cecal metabolites to restore the metabolic disorders of vitamin B6, histidine, etc.
Collapse
Affiliation(s)
- Chong Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Bowei Ju
- Department of Pharmacy, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jiangyun Liu
- College of Pharmacy, Department of Pharmacy, Soochow University, Jiangsu, China
| | - Limei Wen
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yao Zhao
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Jianhua Yang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
46
|
Ciobârcă D, Cătoi AF, Gavrilaș L, Banc R, Miere D, Filip L. Natural Bioactive Compounds in the Management of Type 2 Diabetes and Metabolic (Dysfunction)-Associated Steatotic Liver Disease. Pharmaceuticals (Basel) 2025; 18:279. [PMID: 40006091 PMCID: PMC11859434 DOI: 10.3390/ph18020279] [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: 11/18/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Type 2 diabetes (T2D) and metabolic (dysfunction)-associated steatotic liver disease (MASLD) affect a growing number of individuals worldwide. T2D and MASLD often coexist and substantially elevate the risk of adverse hepatic and cardiovascular clinical outcomes. Several common pathogenetic mechanisms are responsible for T2D and MASLD onset and progression, including insulin resistance, oxidative stress, and low-grade inflammation, among others. The latter can also be induced by gut microbiota and its derived metabolites. Natural bioactive compounds (NBCs) have been reported for their therapeutic potential in both T2D and MASLD. A large amount of evidence obtained from clinical trials suggests that compounds like berberine, curcumin, soluble fibers, and omega-3 fatty acids exhibit significant hypoglycemic, hypolipidemic, and hepatoprotective activity in humans and may be employed as adjunct therapy in T2D and MASLD management. In this review, the role of the most studied NBCs in the management of T2D and MASLD is discussed, emphasizing recent clinical evidence supporting these compounds' efficacy and safety. Also, prebiotics that act against metabolic dysfunction by modulating gut microbiota are evaluated.
Collapse
Affiliation(s)
- Daniela Ciobârcă
- Department 2, Faculty of Nursing and Health Sciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania; (D.C.); (L.G.)
| | - Adriana Florinela Cătoi
- Department of Pathophysiology, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 2-4 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Laura Gavrilaș
- Department 2, Faculty of Nursing and Health Sciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania; (D.C.); (L.G.)
| | - Roxana Banc
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania; (R.B.); (D.M.); (L.F.)
| | - Doina Miere
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania; (R.B.); (D.M.); (L.F.)
| | - Lorena Filip
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania; (R.B.); (D.M.); (L.F.)
- Academy of Romanian Scientists (AOSR), 3 Ilfov Street, 050044 Bucharest, Romania
| |
Collapse
|
47
|
Zhong H, Yu Y, Abdullah, Zhang H, Du J, Sun J, Chen L, Feng F, Guan R. Lactiplantibacillus plantarum N1 derived lipoteichoic acid alleviates insulin resistance in association with modulation of the gut microbiota and amino acid metabolism. Food Funct 2025; 16:1371-1388. [PMID: 39877991 DOI: 10.1039/d4fo06100d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
This study aimed to investigate the effects of heat-killed Lactiplantibacillus plantarum N1 (HK-N1) and lipoteichoic acid (LTA) derived from it on alleviating insulin resistance by modulating the gut microbiota and amino acid metabolism. High-fat diet (HFD)-fed mice were administered live bacteria or HK-N1, and the results demonstrated that HK-N1 significantly reduced epididymal adipocyte size and serum low density lipoprotein-cholesterol, and improved insulin resistance by increasing the YY peptide and glucagon-like peptide levels. HK-N1 also modulated the gut microbiome composition, enhancing microbiota uniformity and reducing the abundance of Ruminococcus, Oscillospira and norank_f_Mogibacteriaceae. Three main active substances obtained from HK-N1 (membrane protein, peptidoglycan, and lipoteichoic acid) were also used to investigate their potential effects in hyperglycemic zebrafish. Only LTA reduced blood sugar and altered the gut microbiome, particularly reducing Aeromonas, which is positively related to hyperglycemia. Untargeted metabolomics revealed that LTA improved vitamin and amino acid metabolism, thereby alleviating metabolic disorders in zebrafish. Collectively, our findings indicate that HK-N1, primarily through LTA, modulated insulin sensitivity by regulating the gut microbiota and amino acid metabolism, offering a potential therapeutic strategy for insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Yufen Yu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Abdullah
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Haoxuan Zhang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Juan Du
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
- Hangzhou Kangyuan Food Science & Technology Co., Ltd, Hangzhou 310012, China
| | - Jiangwei Sun
- Sanya Branch of Hainan Academy of Inspection and Testing, Shanghai 201700, China
| | - Ling Chen
- Sanya Branch of Hainan Food and Drug Inspection Institute, San Ya, 572011, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
48
|
Movahednasab M, Dianat-Moghadam H, Khodadad S, Nedaeinia R, Safabakhsh S, Ferns G, Salehi R. GLP-1-based therapies for type 2 diabetes: from single, dual and triple agonists to endogenous GLP-1 production and L-cell differentiation. Diabetol Metab Syndr 2025; 17:60. [PMID: 39962520 PMCID: PMC11834518 DOI: 10.1186/s13098-025-01623-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/02/2025] [Indexed: 02/20/2025] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin peptide hormone mainly secreted by enteroendocrine intestinal L-cells. GLP-1 is also secreted by α-cells of the pancreas and the central nervous system (CNS). GLP-1 secretion is stimulated by nutrient intake and exerts its effects on glucose homeostasis by stimulating insulin secretion, gastric emptying confiding the food intake, and β-cell proliferation. The insulinotropic effects of GLP-1, and the reduction of its effects in type 2 diabetes mellitus (T2DM), have made GLP-1 an attractive option for the treatment of T2DM. Furthermore, GLP-1-based medications such as GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, have been shown to improve diabetes control in preclinical and clinical trials with human subjects. Importantly, increasing the endogenous production of GLP-1 by different mechanisms or by increasing the number of intestinal L-cells that tend to produce this hormone may be another effective therapeutic approach to managing T2DM. Herein, we briefly describe therapeutic agents/compounds that enhance GLP-1 function. Then, we will discuss the approaches that can increase the endogenous production of GLP-1 through various stimuli. Finally, we introduce the potential of L-cell differentiation as an attractive future therapeutic approach to increase GLP-1 production as an attractive therapeutic alternative for T2DM.
Collapse
Affiliation(s)
- Maedeh Movahednasab
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sana Khodadad
- Department of Genetics and Molecular Biology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeid Safabakhsh
- Micronesian Institute for Disease Prevention and Research, 736 Route 4, Suite 103, Sinajana, GU, 96910, USA
| | - Gordon Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
49
|
Zhang L, Su H, Wang S, Fu Y, Wang M. Gut Microbiota and Neurotransmitter Regulation: Functional Effects of Four Traditional Chinese Fermented Soybean (Sojae Semen Praeparatum). Foods 2025; 14:671. [PMID: 40002115 PMCID: PMC11854601 DOI: 10.3390/foods14040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
This study aims to evaluate the potential disease prevention and treatment functions of four types of traditional Chinese fermented Sojae Semen Praeparatum (SSP) by analyzing their nutritional active components and their effects on the gut microbiota. Raw soybeans and the four SSPs were administered as dietary supplements to normal SD rats for 6 weeks. Fecal samples were collected at weeks 0, 2, and 6 to assess changes in the gut microbiota. Our results revealed that different fermentation methods resulted in variations in soybean isoflavone content. Fermented soybeans promoted the growth of beneficial microorganisms associated with short-chain fatty acid production in the gut microbiota, such as Christensenellaceae_R_7_group, compared to unfermented soybeans. Supplementation with SSPs fermented with different processes increased the diversity of the rat gut microbiota, except for the fermented group of qingwenjiedu decoction (QW). The dominant gut microbiota in the fermented group of Artemisia Annuae Herba and Mori Folium (QS) exhibited anti-inflammatory effects, while the dominant gut microbiota in the fermented group of Ephedrae Herba and Perillae Folium (MZ) showed antidepressant effects. In the neurotransmitter analysis, MZ reduced gamma-aminobutyric acid (GABA) levels, the fermented group without Chinese medicine (DD) decreased dopamine levels, and both QS and QW increased norepinephrine levels. Correlation analysis highlighted connections between gut microbiota, neurotransmitters, and chemical levels. The results indicate that SSPs may contribute uniquely to health by maintaining intestinal balance and improving neurological disorders while predicting a potential association between neurotransmitters and gut microbiota by correlation analysis.
Collapse
Affiliation(s)
| | | | | | | | - Manyuan Wang
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
50
|
Wang Y, Jiang D, Pan X, Sun K, Li T, Cao X, Zhu X. Gut Microbiota in T2DM Patients with Microvascular Complications: A 16S rRNA Sequencing Study. Diabetes Metab Syndr Obes 2025; 18:373-381. [PMID: 39963193 PMCID: PMC11831918 DOI: 10.2147/dmso.s493720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/24/2024] [Indexed: 02/20/2025] Open
Abstract
Objective This study aims to investigate the characteristics of gut microbiota in patients with microvascular complications of Type 2 Diabetes Mellitus (T2DM) using 16S rRNA high-throughput sequencing technology. Methods Patients diagnosed with T2DM were enrolled as study subjects. Based on the presence of microvascular complications, subjects were divided into a study group, a control group. Clinical fecal samples from the two groups were subjected to diversity analysis using the Illumina MiSeq high-throughput sequencing technology, comparing the richness and diversity of the gut microbiota between the two groups. The Tax4Fun software was utilized for the functional prediction of differential microbiota. Results A total of 3727 operational taxonomic units (OTUs) were identified, with 1311 OTUs common to both groups, and 1363 and 1053 OTUs unique to the study group and the control group, respectively. The study group exhibited a significant increase in the relative abundance of Clostridia and Negativicutes, and a marked decrease in Gammaproteobacteria, Bacilli, and Verrucomicrobia compared to the control group. LefSe analysis revealed significant differences in the relative abundance at two phyla, two classes, two orders, three families, and two genera levels between the groups. KEGG pathway analysis of differential microbiota identified 10 pathways with statistically significant differences (P<0.05). Conclusion This study reveals significant disparities in gut microbiota abundance between T2DM patients and those with microvascular complications of T2DM, suggesting potential microbial markers for diagnosing and treating microvascular complications of T2DM.
Collapse
Affiliation(s)
- Yuying Wang
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Dongmei Jiang
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Xia Pan
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Ke Sun
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Tingting Li
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Xin Cao
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| | - Xiaohui Zhu
- Department of Endocrinology, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng City, Jiangsu Province, People’s Republic of China
| |
Collapse
|