1
|
Yang H, Yang Y, Cui G, Xu Y, Zhao R, Le G, Xie Y, Li P. Dietary methionine restriction ameliorates atherosclerosis by remodeling the gut microbiota in apolipoprotein E-knockout mice. Food Funct 2025. [PMID: 40421996 DOI: 10.1039/d5fo00841g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Dietary methionine restriction (MR) has been shown to reduce the risk of atherosclerosis, but the specific regulatory effects and mechanisms remain unclear. This research intends to investigate the effects of MR on atherosclerosis in apolipoprotein E-knockout (ApoE-/-) mice fed a high-fat, high-cholesterol, high-choline diet and their mechanisms. ApoE-/- mice were fed a normal diet (0.86% methionine + 4.5% fat + 0% cholesterol + 0.2% choline), a high-fat, high-cholesterol, high-choline diet (0.86% methionine + 20% fat + 1% cholesterol + 1% choline), or a high-fat, high-cholesterol, high-choline + MR diet (0.17% methionine + 20% fat + 1% cholesterol + 1% choline) for 8 consecutive weeks. The results show that MR reduced body weight, fat mass, fat deposition in the liver and adipocytes, and plasma lipid levels; improved the morphological structure of the aorta; and reduced the aortic lesion area and lipid levels. In addition, MR downregulated aortic pro-inflammatory cytokine levels, upregulated aortic anti-inflammatory cytokine levels, and improved aortic oxidative stress. Moreover, metagenomic sequencing results suggested that MR improved the gut microbiota composition, particularly through increased relative abundance of short-chain fatty acid (SCFA)-producing bacteria, and changed the relative abundance of inflammation-, lipid metabolism-, and bile acid metabolism-related bacteria at the species level. Furthermore, MR promoted SCFA production and bile acid metabolism, and reduced cell adhesion molecules and foam cell formation in the aorta. Thus, our findings indicated that MR improved the gut microbiota composition, especially increased SCFA production, and ameliorated oxidative stress and inflammation in the aorta, thereby preventing atherosclerosis.
Collapse
Affiliation(s)
- Hao Yang
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Yuhui Yang
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Guifang Cui
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Yuncong Xu
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Renyong Zhao
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Guowei Le
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yanli Xie
- Henan Key Laboratory of Cereal and Oil Food Safety and Nutrition, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China.
| | - Peng Li
- Institute for Complexity Science, Henan University of Technology, Zhengzhou 450001, Henan, China
| |
Collapse
|
2
|
Eslami M, Naderian R, Bahar A, Babaeizad A, Rezanavaz Gheshlagh S, Oksenych V, Tahmasebi H. Microbiota as diagnostic biomarkers: advancing early cancer detection and personalized therapeutic approaches through microbiome profiling. Front Immunol 2025; 16:1559480. [PMID: 40406094 PMCID: PMC12095362 DOI: 10.3389/fimmu.2025.1559480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
The important function of microbiota as therapeutic modulators and diagnostic biomarkers in cancer has been shown by recent developments in microbiome research. The intricate interplay between the gut microbiota and the development of cancer, especially in colorectal and breast cancers, emphasizes how microbial profiling may be used for precision treatment and early diagnosis. Important microbial signatures, including Bacteroides fragilis and Fusobacterium nucleatum, have been linked to the development and progression of cancer, providing important information on the processes behind carcinogenesis. Additionally, the influence of microbiota on the effectiveness of treatments such as immunotherapy and chemotherapy highlights its dual function in improving treatment outcomes and reducing side effects. To optimize treatment results, strategies including dietary changes and fecal microbiota transplantation (FMT) are being investigated. Despite these developments, there are still issues, such as individual variations in microbial composition, a lack of standardized procedures, and the requirement for reliable biomarkers. Integrating microbiome-based diagnostics with conventional approaches, such as liquid biopsies and machine learning algorithms, could revolutionize cancer detection and management. This review provides an overview of the current understanding of the host-microbe immunological axis and discusses emerging therapeutic strategies centered on microbiota modulation to support human health. Further research is essential to overcome existing challenges and fully realize the promise of microbiota-driven innovations in oncology.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Aisa Bahar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Babaeizad
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | | | | | - Hamed Tahmasebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
3
|
Jin J, Sun X, Wang L. Association of dietary index of gut microbiota with cardiovascular disease risk: new evidence from NHANES 2007-2018. BMC Cardiovasc Disord 2025; 25:332. [PMID: 40295908 PMCID: PMC12039086 DOI: 10.1186/s12872-025-04776-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The dietary index of gut microbiota (DI-GM) is a newly proposed index for assessing dietary quality, and studies on its association with cardiovascular disease (CVD) are limited. This study aimed to investigate the association between DI-GM and the prevalence of CVD. METHODS We utilized data from the National Health and Nutrition Examination Survey (NHANES). Logistic regression analyses were performed to examine the association between DI-GM and CVD. Smoothed curve fitting was employed to explore potential nonlinear relationships. Additionally, subgroup analyses were conducted to assess the stability of the results. RESULTS The study included 22,590 participants, of whom 20,216 had no CVD and 2,374 had CVD. After adjusting for all covariates, the DI-GM score was significantly negatively associated with CVD risk, with a 4% reduction in CVD risk for each unit increase in DI-GM score (OR = 0.96, 95% CI: 0.94-0.99, P = 0.015). Notably, the highest DI-GM score group (6-12) had a 13% lower risk of CVD compared to the lowest DI-GM score group (0-3) (OR = 0.87, 95% CI: 0.76-1.00, P = 0.048). CONCLUSION The research results indicate that a higher DI-GM score protects against CVD, providing crucial empirical support for dietary intervention strategies based on gut microbiota modulation. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jiameng Jin
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xingang Sun
- Department of Cardiology Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang Province, China.
| | - Lihong Wang
- Department of Cardiology Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
4
|
Ye F, Li L, Wang J, Yang H. Advances in gut-lung axis research: clinical perspectives on pneumonia prevention and treatment. Front Immunol 2025; 16:1576141. [PMID: 40330490 PMCID: PMC12052896 DOI: 10.3389/fimmu.2025.1576141] [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: 02/13/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
In recent years, the study of the interaction between gut microbiota and distant organs such as the heart, lungs, brain, and liver has become a hot topic in the field of gut microbiology. With a deeper understanding of its immune regulation and mechanisms of action, these findings have increasingly highlighted their guiding value in clinical practice. The gut is not only the largest digestive organ in the human body but also the habitat for most microorganisms. Imbalances in gut microbial communities have been associated with various lung diseases, such as allergic asthma and cystic fibrosis. Furthermore, gut microbial communities have significant impacts on metabolic function and immune responses. Their metabolites not only regulate gastrointestinal immune systems but may also affect distant organs such as the lungs and brain. As one of the most common types of respiratory system diseases worldwide, pulmonary infections have high morbidity and mortality rates. Pulmonary infections caused by immune dysfunction can lead to gastrointestinal problems like diarrhea, further resulting in imbalances within complex interactions that are associated with abnormal manifestations under disequilibrium conditions. Meanwhile, clinical interventions can significantly modulate the composition of gut microbiota, and alteration in gut microbiota may subsequently indicate susceptibility to pulmonary infections and even contribute to the prevention or regulation of their progression. This review delves into the interaction between gut microbiota and pulmonary infections, elucidating the latest advancements in gut-lung axis research and providing a fresh perspective for the treatment and prevention of pneumonia.
Collapse
Affiliation(s)
| | | | | | - Hongfeng Yang
- Department of Critical Care Medicine, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
5
|
Mo X, Cheng R, Shen L, Sun Y, Wang P, Jiang G, Wen L, Li X, Peng X, Liao Y, He R, Yan H, Liu L. High-fat diet induces sarcopenic obesity in natural aging rats through the gut-trimethylamine N-oxide-muscle axis. J Adv Res 2025; 70:405-422. [PMID: 38744403 PMCID: PMC11976414 DOI: 10.1016/j.jare.2024.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION The lack of suitable animal models for sarcopenic obesity (SO) limits in-depth research into the disease. Emerging studies have demonstrated that gut dysbiosis is involved in the development of SO. As the importance of microbial metabolites is starting to unveil, it is necessary to comprehend the specific metabolites associated with gut microbiota and SO. OBJECTIVES We aimed to investigate whether high-fat diet (HFD) causes SO in natural aging animal models and specific microbial metabolites that are involved in linking HFD and SO. METHODS Young rats received HFD or control diet for 80 weeks, and obesity-related metabolic disorders and sarcopenia were measured. 16S rRNA sequencing and non-targeted and targeted metabolomics methods were used to detect fecal gut microbiota and serum metabolites. Gut barrier function was evaluated by intestinal barrier integrity and intestinal permeability. Trimethylamine N-oxide (TMAO) treatment was further conducted for verification. RESULTS HFD resulted in body weight gain, dyslipidemia, impaired glucose tolerance, insulin resistance, and systemic inflammation in natural aging rats. HFD also caused decreases in muscle mass, strength, function, and fiber cross-sectional area and increase in muscle fatty infiltration in natural aging rats. 16S rRNA sequencing and nontargeted and targeted metabolomics analysis indicated that HFD contributed to gut dysbiosis, mainly characterized by increases in deleterious bacteria and TMAO. HFD destroyed intestinal barrier integrity and increased intestinal permeability, as evaluated by reducing levels of colonic mucin-2, tight junction proteins, goblet cells and elevating serum level of fluorescein isothiocyanate-dextran 4. Correlation analysis showed a positive association between TMAO and SO. In addition, TMAO treatment aggravated the development of SO in HFD-fed aged rats through regulating the ROS-AKT/mTOR signaling pathway. CONCLUSION HFD leads to SO in natural aging rats, partially through the gut-microbiota-TMAO-muscle axis.
Collapse
Affiliation(s)
- Xiaoxing Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ruijie Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Lihui Shen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Yunhong Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Pei Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Guanhua Jiang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Lin Wen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Xiaobo Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ruikun He
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
6
|
Ramos IM, Navajas-Porras B, Delgado-Osorio A, Rufián-Henares JÁ, Poveda JM. Bioactive Compounds and Antioxidant Properties of Sheep's Milk Yogurt Enriched with a Postbiotic Extract from Lactiplantibacillus plantarum UCLM56: Effects of In Vitro Digestion and Fermentation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7325-7334. [PMID: 40085732 DOI: 10.1021/acs.jafc.4c12744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Lactic acid bacteria present in yogurts produce bioactive compounds, such as gamma-aminobutyric acid (GABA) and short-chain fatty acids (SCFAs). Sheep's milk yogurt is an excellent medium to enhance their bioactive properties. In this study, the impact of adding an extract derived from the GABA-producing strain Lactiplantibacillus plantarum UCLM56 to sheep's milk yogurt was evaluated in terms of its bioactive and antioxidant properties. Samples were subjected to in vitro digestion and fermentation to simulate the effects of the gastrointestinal tract. GABA, SCFAs, amino acids, biogenic amines, antioxidant capacity, and organic acid levels were analyzed. Yogurt enriched with UCLM56 postbiotic extract showed higher levels of GABA and propionic acid, with increases of more than 360 and 260%, respectively, along with an improved antioxidant capacity (FRAP and DPPH methods) compared to conventional yogurt. After in vitro fermentation, the levels of lactic and propionic acids were significantly higher in the enriched yogurt (50 and 41% increases, respectively), as well as the antioxidant capacity (more than 200% improvement). In conclusion, the use of Lactiplantibacillus plantarum UCLM56 extract enhances the bioactive properties of sheep's milk yogurt, making it a promising option for developing functional dairy products with added value.
Collapse
Affiliation(s)
- Inés María Ramos
- Departamento de Química Analítica y Tecnología de los Alimentos. Instituto Regional de Investigación Científica Aplicada (IRICA)/Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. Camilo José Cela, s/n, 13071 Ciudad Real, Spain
| | - Beatriz Navajas-Porras
- Servicio de Endocrinología y Nutrición, Hospital Universitario Doctor Peset, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), 46017 Valencia, Spain
| | - Adriana Delgado-Osorio
- Centro de Investigación Biomédica, Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Universidad de Granada, 18071 Granada, Spain
| | - José Ángel Rufián-Henares
- Centro de Investigación Biomédica, Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de Alimentos, Universidad de Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Universidad de Granada, 18014 Granada, Spain
| | - Justa María Poveda
- Departamento de Química Analítica y Tecnología de los Alimentos. Instituto Regional de Investigación Científica Aplicada (IRICA)/Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avda. Camilo José Cela, s/n, 13071 Ciudad Real, Spain
| |
Collapse
|
7
|
Zhang L, Yin Y, Jin S. Gut microbial metabolites: The bridge connecting diet and atherosclerosis, and next-generation targets for dietary interventions. Microbiol Res 2025; 292:128037. [PMID: 39752807 DOI: 10.1016/j.micres.2024.128037] [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: 10/08/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Mounting evidence indicates that gut microbial metabolites are central hubs linking the gut microbiota to atherosclerosis (AS). Gut microbiota enriched with pathobiont bacteria responsible for producing metabolites like trimethylamine N-oxide and phenylacetylglutamine are related to an increased risk of cardiovascular events. Furthermore, gut microbiota enriched with bacteria responsible for producing short-chain fatty acids, indole, and its derivatives, such as indole-3-propionic acid, have demonstrated AS-protective effects. This study described AS-related gut microbial composition and how microbial metabolites affect AS. Summary findings revealed gut microbiota and their metabolites-targeted diets could benefit AS treatment. In conclusion, dietary interventions centered on the gut microbiota represent a promising strategy for AS treatment, and understanding diet-microbiota interactions could potentially be devoted to developing novel anti-AS therapies.
Collapse
Affiliation(s)
- Liyin Zhang
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Yao Yin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China.
| |
Collapse
|
8
|
Sudi S, Suresh SD, Kolli T, Porras AM. Trymethylamine-N-oxide, a gut-derived metabolite, induces myofibroblastic activation of valvular interstitial cells through endoplasmic reticulum stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636980. [PMID: 39975143 PMCID: PMC11839121 DOI: 10.1101/2025.02.06.636980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Calcific aortic valve disease currently lacks effective treatments beyond surgical valve replacement, due to an incomplete understanding of its pathogenesis. Emerging evidence suggests that the gut microbiome influences cardiovascular health through the production of metabolites derived from dietary components. Among them, trimethylamine-N-oxide (TMAO) has been identified as a potential causal factor for several cardiovascular conditions. However, its role in the development of aortic valve disease remains poorly understood. This study sought to investigate the impact of TMAO on valvular interstitial cells (VICs), the most abundant cell type in the aortic valve. Here, we demonstrate that TMAO activates VICs towards a myofibroblastic profibrotic phenotype. Using an in vitro protocol to generate quiescent VICs, we found that TMAO induces the upregulation of myofibroblastic markers in a sex-independent manner. These quiescent VICs were more sensitive to TMAO than conventionally cultured VICs. Treatment with TMAO also elevated extracellular matrix production and oxidative stress, phenotypic hallmarks of an activated profibrotic state. Finally, inhibition of the endoplasmic reticulum stress kinase prior to TMAO treatment blocked all effects of this metabolite. These findings suggest that TMAO contributes to the early stages of valve disease by promoting VIC activation through endoplasmic reticulum stress mechanisms. Understanding the role of TMAO and other gut-derived metabolites in the pathogenesis of valve disease could inform the development of novel preventive or therapeutic strategies to modify or delay disease progression. Furthermore, these insights underscore the importance of host-microbiome interactions and highlight the potential for targeted dietary interventions to mitigate cardiovascular disease risk.
Collapse
Affiliation(s)
- Samanvitha Sudi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Sai Drishya Suresh
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Tanmayee Kolli
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Ana Maria Porras
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
9
|
de Cuevillas B, Riezu-Boj JI, Milagro FI, Galera Alquegui S, Babio N, Pastor-Villaescusa B, Gil-Campos M, Leis R, De Miguel-Etayo P, Moreno LA, Salas-Salvadó J, Martínez JA, Navas-Carretero S. Parent-child microbiota relationships involved in childhood obesity: A CORALS ancillary study. Nutrition 2025; 130:112603. [PMID: 39550838 DOI: 10.1016/j.nut.2024.112603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVES Childhood obesity continues to rise worldwide. Family gut microorganisms may be associated with childhood obesity. The aim of the study was to analyze bacterial similarities in fecal microbiota composition between parent-offspring pairs as linked to body weight. METHODS A total of 146 father/mother and offspring pairs were categorized into four groups according to the weight status of the parent-child pair as follows: group 1, parent and child with normal weight; group 2, parent and child with overweight/obesity; group 3, parent with normal weight and child with overweight/obesity; group 4, parent with overweight/obesity and child with normal weight. Anthropometric measurements and lifestyle assessments were performed in all participants. Microbiota characteristics were determined by 16S ribosomal RNA gene sequencing. Logistic regression models were performed to determine whether the abundance of any bacteria was able to predict childhood obesity. Moreover, receiver operating characteristic curves were fitted to define the relative diagnostic strength of bacterial taxa for the correct identification of childhood obesity. RESULTS The absence/abundance of Catenibacterium mitsuokai, Prevotella stercorea, Desulfovibrio piger, Massiliprevotella massiliensis, and Phascolarctobacterium succinatutens was involved in body weight family associations. A positive relationship between P. succinatutens richness from parents and M. massiliensis from children was observed with regard to body weight status (odds ratio, 1.14, P = 0.013). CONCLUSIONS This study describes five potential gut bacteria that may be putatively involved in family weight status relationships and appear to be useful for predicting obesity.
Collapse
Affiliation(s)
- Begoña de Cuevillas
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Jose I Riezu-Boj
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain
| | - Fermín I Milagro
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Galera Alquegui
- Department of Personalized Medicine, Navarra Services and Technologies, Government of Navarra, Pamplona, Spain
| | - Nancy Babio
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Food, Nutrition, Development and Mental Health Research Group, Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Belén Pastor-Villaescusa
- Metabolism and Investigation Unit, Maimónides Institute of Biomedicine Research of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Mercedes Gil-Campos
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Metabolism and Investigation Unit, Maimónides Institute of Biomedicine Research of Córdoba, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Rosaura Leis
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Pediatric Service, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain; Pediatric Nutrition Research Group, Unit of Investigation in Nutrition, Growth and Human Development of Galicia-USC, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Pilar De Miguel-Etayo
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, Instituto de Investigación Sanitaria Aragón, University of Zaragoza, Zaragoza, Spain
| | - Luis A Moreno
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Growth, Exercise, Nutrition and Development Research Group, Instituto Agroalimentario de Aragón, Instituto de Investigación Sanitaria Aragón, University of Zaragoza, Zaragoza, Spain
| | - Jordi Salas-Salvadó
- Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Food, Nutrition, Development and Mental Health Research Group, Unitat de Nutrició Humana, Departament de Bioquímica i Biotecnologia, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain; Institut d'Investigació Sanitària Pere Virgili, Reus, Spain
| | - J Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Navas-Carretero
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research, Pamplona, Spain; Consorcio Centro de Investigación Biomédica en Red, M. P. Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
10
|
Yao L, Zhou X, Jiang X, Chen H, Li Y, Xiong X, Tang Y, Zhang H, Qiao P. High-fat diet promotes gestational diabetes mellitus through modulating gut microbiota and bile acid metabolism. Front Microbiol 2025; 15:1480446. [PMID: 39935515 PMCID: PMC11810896 DOI: 10.3389/fmicb.2024.1480446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/27/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Gestational diabetes mellitus (GDM) is a condition characterized by glucose intolerance during pregnancy, estimated to affect approximately 20% of the whole pregnancies and is increasing in prevalence globally. However, there is still a big gap in knowledge about the association between gut microbiota associated metabolism alterations and GDM development. Methods All the participants accomplished the validated internet-based dietary questionnaire for Chinese and serum, fecal samples were collected. HFD, control diet or colesevelam intervention was fed to GDM mice models or Fxr-/- mice models, with or without antibiotics cocktail treatment. Fecal microbiota transplantation were used for further validation. Gut microbiota and metabolites were detected by metagenomic sequencing and high-performance liquid chromatography-mass spectrometry, respectively. Bile acids of serum, fecal samples from human and mice were analysised. Body weight, average feed intake, blood glucose, insulin levels and oral glucose tolerance test was performed among each groups. Expression levels of Fxr, Shp and Fgf15 mRNA and protein were detected by quantitative reverse transcription polymerase chain reaction and western blot, respectively. Results Our data indicated that high fat diet (HFD) was linked with higher prevalence of GDM, and HFD was positively associated with poor prognosis in GDM patients. Moreover, compared with normal diet (ND) group, GDM patients from HFD group performed a loss of gut microbiota diversity and enrichment of Alistipes onderdonkii, Lachnospiraceae bacterium 1_7_58FAA, and Clostridium aspaaragiforme while ruduction of Akkermansiaceae, Paraprevotell xylaniphila, and Prevotella copri. Additionally, HFD aggravated GDM in mice and gut microbiota depletion by antibiotics crippled the effect of excess fat intake. BAs profile altered in HFD GDM patients and mice models. Fecal microbiota transplantation (FMT) further confirmed that gut microbiota contributed to bile acids (BAs) metabolic dysfunction during HFD-associated GDM development. Mechanically, HFD-FMT administration activated Fxr, Shp, and Fgf15 activity, disturbed the glucose metabolism and aggravated insulin resistance but not in HFD-FMT Fxr-/- mice and ND-FMT Fxr-/- mice. Furthermore, colesevelam intervention alleviated HFD-associated GDM development, improved BAs metabolism, suppressed Fxr, Shp, and Fgf15 activity only in WT mice but not in the Fxr-/- HFD + Colesevelam group and Fxr-/- HFD group. HFD induced GDM and contributed to poor prognosis in GDM parturients through inducing gut microbial dysbiosis and metabolic alteration, especially appeared in BAs profile. Moreover, Fxr pathway participated in regulating HFD-associated gut microbiota disordered BAs metabolites and aggravating GDM in mice. Discussion Modulating gut microbiota and BAs metabolites could be a potential therapeutic strategy in the prevention and treatment of HFD-associated GDM.
Collapse
Affiliation(s)
- Lei Yao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuefei Zhou
- Department of Gynaecology and Obstetrics, The Center of Red Cross Hospital of Harbin, Harbin, China
| | - Xianqi Jiang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chen
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanliang Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Xiong
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Tang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haogang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengfei Qiao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Leng X, Wei X, Wang J, Yao X, Zhang M, Sun D, Liang J, Chi L, Cheng Y. Impacts of intestinal microbiota metabolite trimethylamine N-oxide on cardiovascular disease: a bibliometric analysis. Front Microbiol 2025; 15:1491731. [PMID: 39834376 PMCID: PMC11743947 DOI: 10.3389/fmicb.2024.1491731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Background Trimethylamine N-oxide (TMAO), a metabolite dependent on intestinal microbiota, is closely related to the emergence, progression, and prognosis of cardiovascular disease (CVD), and has received increasing attention in recent years. Objective The current research hotspots and future development trends in TMAO and CVD field are found through bibliometrics analysis, which provides reference for further study. Methods The bibliometrics tools VOSviewer and CiteSpace were used to analyze the publications from the Web of Science Core Collection (WOSCC) database. The articles published from 2004 to 2024 about the relationship between TMAO and CVD were retrieved. Bibliometric analysis includes annual publications, countries/regions, institutions, authors and co-cited authors, journals and cited-journals, references and keywords. Results After searching and screening, 1,466 publications were included for subsequent bibliometric analysis. Since 2014, the number of publications exposing the relationship between TMAO and CVD has increased rapidly, as has the frequency of citations. China, USA and Italy are the countries that publish the most relevant research. Cleveland Clinic is the leading institution in this field. Stanley L Hazen, Zeneng Wang and W H Wilson Tang are the most prolific authors in this field, and the latter two have the closest academic cooperation. American Journal of Clinical Nutrition and Journal of the American Heart Association are influential journals that publish research in this field. "Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk" is the most frequently cited article. Keyword analysis shows that gut microbiota, metabolism, phosphatidylcholine and atherosclerosis (AS) are the hotspots in this field. Conclusion This study summarizes the research situation of TMAO and CVD in the past 20 years, focusing on the effect of TMAO on pathogenesis of AS, predictive value of TMAO on CVD risk, and dietary and drug intervention for TMAO. Probiotics and natural products may be the research focus of preventing and treating CVD by intervening TMAO in the future.
Collapse
Affiliation(s)
- Xiaohui Leng
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Yantai Yuhuangding Hospital, Yantai, China
| | - Xiunan Wei
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jun Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaodong Yao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Miaomiao Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dajuan Sun
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Junwei Liang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Chi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Cheng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
Yang Q, Wang Z, Liu M, Gan L. Causal Relationship Between Gut Microbiota and Leukemia: Future Perspectives. Oncol Ther 2024; 12:663-683. [PMID: 39217582 PMCID: PMC11573970 DOI: 10.1007/s40487-024-00300-8] [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: 05/25/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota plays a crucial role in maintaining homeostasis in the human gastrointestinal tract. Numerous studies have shown a strong association between the gut microbiota and the emergence and progression of various diseases. Leukemia is one of the most common hematologic malignancies. Although standardized protocols and expert consensus have been developed for routine diagnosis and treatment, limitations remain due to individual differences. Nevertheless, a large number of studies have established a link between the gut microbiota and leukemia, with disturbances in the gut microbiota directly or indirectly affecting the development of leukemia. However, the causal relationship between the two remains unclear, and studying and exploring the causal relationship may open up entirely new avenues and protocols for use in the prevention and/or treatment of leukemia, offering new insights into diagnosis and treatment. In this review, the intricate relationship between the gut microbiota and leukemia is explored in depth, including causal associations, metabolite effects, therapeutic applications, and complications. Based on the characteristics of the gut microbiota, the future applications and prospects of gut microbiota are discussed to provide useful information for clinical treatment of leukemia.
Collapse
Affiliation(s)
- Qiang Yang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Zexin Wang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China.
| | - Miao Liu
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Lingling Gan
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| |
Collapse
|
13
|
Guo Y, Cai W, Xie W, Xu Y, Li X, Yu C, Wu Q. Sodium houttuyfonate modulates the lung Th1/Th2 balance and gut microbiota to protect against pathological changes in lung of ovalbumin-induced asthmatic mice. J Asthma 2024; 61:1759-1771. [PMID: 39021077 DOI: 10.1080/02770903.2024.2380525] [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: 04/25/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVE The gut-lung axis involves microbial and product interactions between the lung and intestine. Antibiotics for chronic asthma can cause intestinal dysbiosis, disrupting this axis. Sodium houttuyfonate (SH) has diverse biological activities, including modifying gut microbiota, antibacterial, and anti-inflammatory. This study aims to explore the relationship between SH, CD4+ T cells, and gut microbiota. METHODS Allergic asthma was experimentally induced in mice through injection and inhalation of ovalbumin. After the administration of different amounts of SH, ELISA was utilized to ascertain the levels of inflammatory cytokines in the serum, flow cytometry was used to examine the levels of Th1/Th2 cytokines in CD4+ cells from lung tissues. The expression of T-bet and GATA3 in lung tissue was determined by Western blotting and quantitative real-time PCR assay. Gut microbiota was determined by 16S rRNA gene sequencing. RESULTS The results showed that SH can alleviate pulmonary injury in asthmatic mice, reducing serum levels of IL-4, IL-5, and IL-13 while simultaneously increasing IFN-γ. Furthermore, SH has been observed to modulate the balance of Th1/Th2 cells by up-regulating the mRNA and protein expression of T-bet but down-regulating GATA3 in the lung tissues of asthmatic mice, thereby promoting the differentiation of Th1 cells. Additionally, SH can regulate the variety and composition of gut microbiota especially genus Akkermansia in asthmatic mice. CONCLUSION SH can alleviate asthma through the regulation of Th1/Th2 cells and gut microbiota.
Collapse
Affiliation(s)
- Yanping Guo
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Cai
- School of Pharmacy, Zhejiang Pharmaceutical University, Ningbo, China
| | - Wei Xie
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Yunlu Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuejian Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Chenhuan Yu
- Experimental animal platform, Hangzhou Institute of Medicine Chinese Academy of Sciences, Hangzhou, China
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Qiaofeng Wu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Hemmati MA, Monemi M, Asli S, Mohammadi S, Foroozanmehr B, Haghmorad D, Oksenych V, Eslami M. Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk. Cells 2024; 13:1987. [PMID: 39682735 PMCID: PMC11640725 DOI: 10.3390/cells13231987] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
The gut microbiota significantly impacts human health, influencing metabolism, immunological responses, and disease prevention. Dysbiosis, or microbial imbalance, is linked to various diseases, including cancer. It is crucial to preserve a healthy microbiome since pathogenic bacteria, such as Escherichia coli and Fusobacterium nucleatum, can cause inflammation and cancer. These pathways can lead to the formation of tumors. Recent advancements in high-throughput sequencing, metagenomics, and machine learning have revolutionized our understanding of the role of gut microbiota in cancer risk prediction. Early detection is made easier by machine learning algorithms that improve the categorization of cancer kinds based on microbiological data. Additionally, the investigation of the microbiome has been transformed by next-generation sequencing (NGS), which has made it possible to fully profile both cultivable and non-cultivable bacteria and to understand their roles in connection with cancer. Among the uses of NGS are the detection of microbial fingerprints connected to treatment results and the investigation of metabolic pathways implicated in the development of cancer. The combination of NGS with machine learning opens up new possibilities for creating customized medicine by enabling the development of diagnostic tools and treatments that are specific to each patient's microbiome profile, even in the face of obstacles like data complexity. Multi-omics studies reveal microbial interactions, biomarkers for cancer detection, and gut microbiota's impact on cancer progression, underscoring the need for further research on microbiome-based cancer prevention and therapy.
Collapse
Affiliation(s)
- Mohammad Amin Hemmati
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran; (M.A.H.); (B.F.)
| | - Marzieh Monemi
- Department of Basic Science, Faculty of Pharmacy and Pharmaceutical Science, Tehran Medical Science, Islamic Azad University, Tehran 19395-1495, Iran;
| | - Shima Asli
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran; (S.A.); (S.M.)
| | - Sina Mohammadi
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran; (S.A.); (S.M.)
| | - Behina Foroozanmehr
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran; (M.A.H.); (B.F.)
| | - Dariush Haghmorad
- Department of Immunology, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7028 Trondheim, Norway
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Majid Eslami
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
15
|
Mokoena H, Mabhida SE, Choshi J, Sekgala MD, Nkambule BB, Ndwandwe D, Mchiza ZJ, Kengne AP, Dludla PV, Hanser S. Soluble P-selectin as an inflammatory mediator potentially influencing endothelial activation in people living with HIV in sub-rural areas of Limpopo, South Africa. PLoS One 2024; 19:e0310056. [PMID: 39602436 PMCID: PMC11602056 DOI: 10.1371/journal.pone.0310056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/19/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVES There is a growing need to understand the potential role of soluble platelet selectin (sP-selectin) in sustained endothelial activation through increased levels of soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular adhesion-1 (sVCAM-1) in people living with HIV (PLWH) on highly active antiretroviral therapy (HAART). METHODOLOGY This was a cross-sectional study involving PLWH on HAART (n = 55), in comparison to PLWH not on treatment (HAART-naïve) (n = 29), and (iii) HIV negative controls (n = 48) from the Mankweng area in the Limpopo province, South Africa. We quantified serum levels of sP-selectin, together with sICAM-1 and sVCAM-1. Most of the HAART-exposed group were on treatment for <5 years. We further performed frequency distribution and descriptive statistics for categorical variables. RESULTS Soluble P-selectin was positively correlated with sVCAM-1 (r = 0.469; p<0.001) in PLWH on HAART, even after adjusting for confounding factor such as age, BMI, and total cholesterol (r = 0.467; p<0.001). Moreover, in PLWH on HAART sP-selecting was independently associated with the release of sVCAM-1 (β = 0.445; p<0.001), even after adjusting for confounders (β = 0.475; p = 0.001). Serum levels of low-density lipoprotein cholesterol (LDL-C) (p = 0.004) and total cholesterol (p<0.001) were significantly higher in PLWH on HAART as compared to the HAART-naïve group. CONCLUSION There is a need for more studies to investigate the role of sP-selectin in promoting endothelial activation and CVD-risk in PLWH on HAART, especially within the sub-Saharan Africa region.
Collapse
Affiliation(s)
- Haskly Mokoena
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, South Africa
| | - Sihle E. Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, South Africa
| | - Joel Choshi
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, South Africa
| | - Machoene D. Sekgala
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, South Africa
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Tygerberg, South Africa
| | - Zandile J. Mchiza
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, South Africa
- School of Public Health, University of the Western Cape, Bellville, South Africa
| | - André P. Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Phiwayinkosi V. Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Sidney Hanser
- Department of Physiology and Environmental Health, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
16
|
Tao E, Lang D. Unraveling the gut: the pivotal role of intestinal mechanisms in Kawasaki disease pathogenesis. Front Immunol 2024; 15:1496293. [PMID: 39664384 PMCID: PMC11633670 DOI: 10.3389/fimmu.2024.1496293] [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: 09/14/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Kawasaki disease (KD), an acute systemic vasculitis that primarily affects children under 5 years of age, is the leading cause of acquired heart disease in this age group. Recent studies propose a novel perspective on KD's etiology, emphasizing the gastrointestinal (GI) tract, particularly the role of gut permeability. This review delves into how disruptions in gut barrier function trigger systemic inflammatory responses, exacerbate vascular inflammation, and contribute to coronary artery aneurysms. Evidence suggests that children with KD often exhibit increased gut permeability, leading to an imbalance in gut immunity and subsequent gut barrier damage. These changes impact vascular endothelial cells, promoting platelet aggregation and activation, thereby advancing severe vascular complications, including aneurysms. Additionally, this review highlights the correlation between GI symptoms and increased resistance to standard treatments like intravenous immunoglobulin (IVIG), indicating that GI involvement may predict therapeutic outcomes. Advocating for a new paradigm, this review calls for integrated research across gastroenterology, immunology, and cardiology to examine KD through the lens of GI health. The goal is to develop innovative therapeutic interventions targeting the intestinal barrier, potentially revolutionizing KD management and significantly improving patient outcomes.
Collapse
Affiliation(s)
- Enfu Tao
- Department of Neonatology and Neonatal Intensive Care Unit (NICU), Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang, China
| | - Dandan Lang
- Department of Pediatrics, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong, China
| |
Collapse
|
17
|
Wang Y, Wang Z. Association between ideal cardiovascular health and bowel conditions among US adults. Front Nutr 2024; 11:1473531. [PMID: 39574525 PMCID: PMC11580258 DOI: 10.3389/fnut.2024.1473531] [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: 07/31/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Objective The aim of this study is to explore the relationship between ideal cardiovascular health (CVH), as assessed using the Life's Essential 8 (LE8), and bowel conditions. Methods This cross-sectional study selected 11,108 participants aged ≥20 years from 2005 to 2010 National Health and Nutrition Examination Survey. LE8 scores (range 0-100) were measured according to American Heart Association definitions and were divided into health behavior and health factor scores. Bowel conditions including chronic diarrhea, constipation, and fecal incontinence were diagnosed by the Bowel Health Questionnaire. Weighted logistic regression and restricted cubic spline models were used for correlation analysis. Results Logistic regression results showed that LE8 scores were negatively associated with chronic diarrhea and fecal incontinence, but the difference with chronic constipation was not statistically significant. The health behaviors subscale was also negatively correlated with chronic diarrhea, chronic constipation, and fecal incontinence, but health factors were negatively related to chronic diarrhea and fecal incontinence and positively related to chronic constipation. The RCS was consistent with the trend of the logistic regression findings. Sensitivity analyses reconfirmed these outcomes. Conclusion LE8 is highly associated with chronic diarrhea and fecal incontinence, not with chronic constipation. Encouraging optimization of CVH levels may be beneficial for bowel disorders, and prevention of bowel disorders may enhance CVH.
Collapse
Affiliation(s)
| | - Zhigang Wang
- Xi’an International Medical Center Hospital Affiliated to Northwest University, Xi’an, China
| |
Collapse
|
18
|
Chen X, Wu J, Zhou B, Zhu M, Zhang J, Zhou N, Zhu YZ, Zhang X, Duan X, Men K. Bacterial Lysate-Based Bifunctional mRNA Nanoformulation for Efficient Colon Cancer Immunogene Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56580-56598. [PMID: 39397736 DOI: 10.1021/acsami.4c07684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
mRNA-based nonviral gene therapy has played an important role in cancer therapy, however, the limited delivery efficiency and therapeutic capacity still require further exploration and enhancement. Immunogene therapy provides a strategy for cancer treatment. Bacteria are tiny single-celled living organisms, many of which can be found in and on the human body and are beneficial to humans. Lactobacillus reuteri is a bacterial member of the gut flora, and recent research has shown that it can reduce intestinal inflammation by stimulating an immunomodulatory response. L. reuteri lysate represents an ideal resource for constructing advanced mRNA delivery systems with immune stimulation potential. Here, we prepared a bifunctional mRNA delivery system DMP-Lac (DOTAP-mPEG-PCL-L. reuteri lysate), which successfully codelivered L. reuteri lysate and IL-23A mRNA, exhibited a high mRNA delivery efficiency of 75.56% ± 0.85%, and strongly promoted the maturation and activation of the immune system in vivo. Both the CT26 abdominal metastasis model and the lung metastasis model also exhibited a good therapeutic effect, and the tumor inhibition rate of DMP-Lac/IL-23A group reached 97.92%. Protein chip technology verified that DMP acted as an immune adjuvant, demonstrating that the L. reuteri lysate could regulate the related immune cells, while IL-23 mRNA caused changes in downstream factors, thus producing the corresponding tumor treatment effect. The DMP-Lac/IL-23A complex exhibited strong anticancer immunotherapeutic effects. Our results demonstrated that this bifunctional mRNA formulation served as a tumor-specific nanomedicine, providing an advanced strategy for colon cancer immunogene therapy.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Na Zhou
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Yi Zhun Zhu
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xin Zhang
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
19
|
Shi M, Guo K, Liu Y, Cao F, Fan T, Deng Z, Meng Y, Bu M, Ma Z. Role of macrophage polarization in periodontitis promoting atherosclerosis. Odontology 2024; 112:1209-1220. [PMID: 38573421 DOI: 10.1007/s10266-024-00935-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Periodontitis is a chronic inflammatory destructive disease occurring in periodontal supporting tissues. Atherosclerosis(AS) is one of the most common cardiovascular diseases. Periodontitis can promote the development and progression of AS. Macrophage polarization is closely related to the development and progression of the above two diseases, respectively. The purpose of this animal study was to evaluate the effect of periodontitis on aortic lesions in atherosclerotic mice and the role of macrophage polarization in this process. 45 ApoE-/-male mice were randomly divided into three groups: control (NC), atherosclerosis (AS), and atherosclerosis with periodontitis (AS + PD). Micro CT, serological testing and pathological testing(hematoxylin-eosin staining, oil red O staining and Masson staining) were used for Evaluate the modeling situation. Immunohistochemistry(IHC) and immunofluorescence(IF) were performed to evaluate macrophage content and macrophage polarization in plaques. Cytokines associated with macrophage polarization were analyzed using quantitative real-time polymerase chain reaction(qRT-PCR) and enzyme-linked immunosorbent assay(Elisa). The expression of macrophages in plaques was sequentially elevated in the NC, AS, and AS + PD groups(P < 0.001). The expression of M1 and M1-related cytokines showed the same trend(P < 0.05). The expression of M2 and M2-related cytokines showed the opposite trend(P < 0.05). The rate of M1/M2 showed that AS + PD > AS > NC. Our preliminary data support that experimental periodontitis can increase the content of macrophage in aortic plaques to exacerbate AS. Meanwhile, experimental periodontitis can increase M1 macrophages, and decrease M2 macrophages, increasing M1/M2 in the plaque.
Collapse
Affiliation(s)
- Mingyue Shi
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Kaili Guo
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Yue Liu
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Fengdi Cao
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Tiantian Fan
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Zhuohang Deng
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Yuhan Meng
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Mingyang Bu
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China
| | - Zhe Ma
- Department of Preventive Dentistry, Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, No.383, Zhongshan East Road, Changan District, Shijiazhuang, Hebei, China.
| |
Collapse
|
20
|
Asar TO, Al-Abbasi FA, Sheikh RA, Zeyadi MAM, Nadeem MS, Naqvi S, Kumar V, Anwar F. Metformin's dual impact on Gut microbiota and cardiovascular health: A comprehensive analysis. Biomed Pharmacother 2024; 178:117128. [PMID: 39079259 DOI: 10.1016/j.biopha.2024.117128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/25/2024] Open
Abstract
Cardiovascular diseases (CVD) cause significant global morbidity, mortality and public health burden annually. CVD alters richness, diversity, and composition of Gut microbiota along with RAS and histopathological differences. Present study explores Metformin role in mitigating doxorubicin induced cardiovascular toxicity/remodeling. Animals were divided into 4 groups with n=6: Group I (N. Control) free access to diet and water; Group II (MET. Control) on oral Metformin (250 mg/kg) daily; Group III (DOX. Control) alternate day intraperitoneal Doxorubicin (3 mg/kg) totaling 18 mg/kg; Group IV (DOX. MET. Control) received both daily oral Metformin (250 mg/kg) and alternate day Doxorubicin (3 mg/kg). Gut microbial analysis was made from stool before animals were sacrificed for biochemical and histopathological analysis. Significant alterations were observed in ɑ and β-diversity with new genus from Firmicutes, specifically Clostridia_UCG-014, Eubacterium ruminantium, and Tunicibacter, were prevalent in both the DOX. Control and DOX.MET groups. Proteobacteria, represented by Succinivibrio, were absent in all groups. Additionally, Parabacteroides from the Bacteroidia phylum was absent in all groups except the N. control. In the DOX.MET Control group, levels of Angiotensin II ( 7.75± 0.49 nmol/min, p<0.01) and Renin (2.60±0.26 ng/ml/hr) were significantly reduced. Conversely, levels of CK-MB, Fibrinogen, Troponin, CRP ( p < 0.0001), and TNFɑ (p < 0.05) were elevated. Histopathological examination revealed substantial cardiac changes, including Fibrinogen and fat deposition and eosinophilic infiltration, as well as liver damage characterized by binucleated cells and damaged hepatocytes, along with altered renal tissues in the DOX.MET.Control group. The findings suggest that MET. significantly modifies gut microbiota, particularly impacting the Firmicutes and Proteobacteria phyla. The reduction in Angiotensin II levels, alongside increased inflammatory markers and myocardial damage, highlights the complex interactions and potential adverse effects associated with MET therapy on cardiovascular health.
Collapse
Affiliation(s)
- Turky Omar Asar
- Department of Biology, College of Science and Arts at Alkamil, University of Jeddah, Jeddah, Saudi Arabia.
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ryan Adnan Sheikh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | | | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Salma Naqvi
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| | - Vikas Kumar
- Natural Product Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, SHUATS, Prayagraj, India.
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
21
|
Flori L, Benedetti G, Martelli A, Calderone V. Microbiota alterations associated with vascular diseases: postbiotics as a next-generation magic bullet for gut-vascular axis. Pharmacol Res 2024; 207:107334. [PMID: 39103131 DOI: 10.1016/j.phrs.2024.107334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The intestinal microbiota represents a key element in maintaining the homeostasis and health conditions of the host. Vascular pathologies and other risk factors such as aging have been recently associated with dysbiosis. The qualitative and quantitative alteration of the intestinal microbiota hinders correct metabolic homeostasis, causing structural and functional changes of the intestinal wall itself. Impairment of the intestinal microbiota, combined with the reduction of the barrier function, worsen the pathological scenarios of peripheral tissues over time, including the vascular one. Several experimental evidence, collected in this review, describes in detail the changes of the intestinal microbiota in dysbiosis associated with vascular alterations, such as atherosclerosis, hypertension, and endothelial dysfunction, the resulting metabolic disorders and how these can impact on vascular health. In this context, the gut-vascular axis is considered, for the first time, as a merged unit involved in the development and progression of vascular pathologies and as a promising target. Current approaches for the management of dysbiosis such as probiotics, prebiotics and dietary modifications act mainly on the intestinal district. Postbiotics, described as preparation of inanimate microorganisms and/or their components that confers health benefits on the host, represent an innovative strategy for a dual management of intestinal dysbiosis and vascular pathologies. In this context, this review has the further purpose of defining the positive effects of the supplementation of bacterial strains metabolites (short‑chain fatty acids, exopolysaccharides, lipoteichoic acids, gallic acid, and protocatechuic acid) restoring intestinal homeostasis and acting directly on the vascular district through the gut-vascular axis.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| |
Collapse
|
22
|
Yu J, Song H, Zhou L, Wang S, Liu X, Liu L, Ma Y, Li L, Wen S, Luo Y, Zhang X, Li W, Niu X. (-)-Epicatechin gallate prevented atherosclerosis by reducing abnormal proliferation of VSMCs and oxidative stress of AML 12 cells. Cell Signal 2024; 121:111276. [PMID: 38936786 DOI: 10.1016/j.cellsig.2024.111276] [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: 04/08/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
(-)-Epicatechin gallate (ECG) is beneficial to the treatment of cardiovascular diseases (CVDs), especially atherosclerosis (AS) through antioxidant stress, but there is a lack of detailed mechanism research. In this study, the therapeutic target of ECG was determined by crossing the drug target and disease target of CVDs and AS. The combination ability of ECG with important targets was verified by Discovery Studio software. The abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by Ang-II and the oxidative damage of AML 12 induced by H2O2 were established to verify the reliability of ECG intervention on the target protein. A total of 120 ECG targets for the treatment of CVDs-AS were predicted by network pharmacology. The results of molecular docking showed that ECG has strong binding force with VEGFA, MMP-9, CASP3 and MMP-2 domains. In vitro experiments confirmed that ECG significantly reduced the expression of VEGFA, MMP-9, CASP3 and MMP-2 in Ang-II-induced VSMCs, and also blocked the abnormal proliferation, oxidative stress and inflammatory reaction of VSMCs by inhibiting the phosphorylation of PI3K signaling pathway. At the same time, ECG also interfered with H2O2-induced oxidative damage of AML 12 cells, decreased the expression of ROS and MDA and cell foaming, and increased the activities of antioxidant enzymes such as SOD, thus playing a protective role.
Collapse
Affiliation(s)
- Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingyi Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingli Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Sha Wen
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yuzhi Luo
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinya Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
23
|
Mabrok HB, Ramadan AA, Hamed IM, Mohamed DA. Obesity as Inducer of Cognitive Function Decline via Dysbiosis of Gut Microbiota in Rats. Brain Sci 2024; 14:807. [PMID: 39199499 PMCID: PMC11353248 DOI: 10.3390/brainsci14080807] [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: 07/10/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
Diet-induced obesity is a global phenomenon that affects the population worldwide with manifestations at both the phenotypic and genotypic levels. Cognitive function decline is a major global health challenge. The relation between obesity and cognitive function is a debatable issue. The main goal of the current research was to study the implications of obesity on cognitive function and gut microbiota diversity and its impact on plasma and brain metabolic parameters in rats. Obesity was induced in rats by feeding on a high-fat (HF) or a high-fat/high-sucrose (HFHS) diet. The results reveal that both the HF (0.683) and HFHS (0.688) diets were effective as obesity inducers, which was confirmed by a significant increase in the body mass index (BMI). Both diet groups showed dyslipidemia and elevation of oxidative stress, insulin resistance (IR), and inflammatory markers with alterations in liver and kidney functions. Obesity led to a reduction in cognitive function through a reduction in short-term memory by 23.8% and 30.7% in the rats fed HF and HFHS diets, respectively, and learning capacity and visuo-spatial memory reduced by 8.9 and 9.7 s in the rats fed an HF or HFHS diet, respectively. Bacteroidetes, Firmicutes, Proteobacteria, Fusobacteria, and Spirochaetes phyla were detected. The Firmicutes/Bacteroidetes ratio (F/B) significantly decreased in the HF group, while it increased in the HFHS group compared to the normal control. The two species, Bacteroides acidifaciens and Bacteroides ovatus, which are associated with IR, were drastically compromised by the high-fat/high-sucrose diet. Some species that have been linked to reduced inflammation showed a sharp decrease in the HFHS group, while Prevotella copri, which is linked to carbohydrate metabolism, was highly enriched. In conclusion: Obesity led to cognitive impairment through changes in short-term and visuo-spatial memory. A metagenomic analysis revealed alterations in the abundance of some microbial taxa associated with obesity, inflammation, and insulin resistance in the HF and HFHS groups.
Collapse
Grants
- a626035bfd925943, 4c6c6a0dc9645904, 175e6bf937114ef5, 18dca4e8f29e587c, aaf09103eb8bd6ee, 3740a1d4a23d772f, 1b07773fd3c8c954, 4f8fa1a570a3a4b7, 490e7e4e51713e71, 1e87a07edec11a96, 7642f29d62c1068b, c06bc3bf279a8491, c78b30a55528e880, e160d996ffb69ed4, 133 Discount Vouchers
Collapse
Affiliation(s)
| | | | | | - Doha A. Mohamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo 12622, Egypt; (H.B.M.); (A.A.R.); (I.M.H.)
| |
Collapse
|
24
|
Caradonna E, Nemni R, Bifone A, Gandolfo P, Costantino L, Giordano L, Mormone E, Macula A, Cuomo M, Difruscolo R, Vanoli C, Vanoli E, Ferrara F. The Brain-Gut Axis, an Important Player in Alzheimer and Parkinson Disease: A Narrative Review. J Clin Med 2024; 13:4130. [PMID: 39064171 PMCID: PMC11278248 DOI: 10.3390/jcm13144130] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are severe age-related disorders with complex and multifactorial causes. Recent research suggests a critical link between neurodegeneration and the gut microbiome, via the gut-brain communication pathway. This review examines the role of trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite, in the development of AD and PD, and investigates its interaction with microRNAs (miRNAs) along this bidirectional pathway. TMAO, which is produced from dietary metabolites like choline and carnitine, has been linked to increased neuroinflammation, protein misfolding, and cognitive decline. In AD, elevated TMAO levels are associated with amyloid-beta and tau pathologies, blood-brain barrier disruption, and neuronal death. TMAO can cross the blood-brain barrier and promote the aggregation of amyloid and tau proteins. Similarly, TMAO affects alpha-synuclein conformation and aggregation, a hallmark of PD. TMAO also activates pro-inflammatory pathways such as NF-kB signaling, exacerbating neuroinflammation further. Moreover, TMAO modulates the expression of various miRNAs that are involved in neurodegenerative processes. Thus, the gut microbiome-miRNA-brain axis represents a newly discovered mechanistic link between gut dysbiosis and neurodegeneration. MiRNAs regulate the key pathways involved in neuroinflammation, oxidative stress, and neuronal death, contributing to disease progression. As a direct consequence, specific miRNA signatures may serve as potential biomarkers for the early detection and monitoring of AD and PD progression. This review aims to elucidate the complex interrelationships between the gut microbiota, trimethylamine-N-oxide (TMAO), microRNAs (miRNAs), and the central nervous system, and the implications of these connections in neurodegenerative diseases. In this context, an overview of the current neuroradiology techniques available for studying neuroinflammation and of the animal models used to investigate these intricate pathologies will also be provided. In summary, a bulk of evidence supports the concept that modulating the gut-brain communication pathway through dietary changes, the manipulation of the microbiome, and/or miRNA-based therapies may offer novel approaches for implementing the treatment of debilitating neurological disorders.
Collapse
Affiliation(s)
- Eugenio Caradonna
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| | - Raffaello Nemni
- Unit of Neurology, Centro Diagnostico Italiano S.p.A., Milan Fondazione Crespi Spano, 20011 Milan, Italy;
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Angelo Bifone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Patrizia Gandolfo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Lucy Costantino
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Luca Giordano
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Elisabetta Mormone
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Anna Macula
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Colleretto Giacosa, 10010 Turin, Italy;
- Department of Physics, University of Torino, 10124 Torino, Italy
| | - Mariarosa Cuomo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | | | - Camilla Vanoli
- Department of Clinical Psychology, Antioch University Los Angeles, Culver City, CA 90230, USA
| | - Emilio Vanoli
- School of Nursing, Cardiovascular Diseases, University of Pavia, 27100 Pavia, Italy;
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| |
Collapse
|
25
|
Ondondo B. Editorial: Overcoming challenges in microbial immunology: 2022. Front Immunol 2024; 15:1436631. [PMID: 38953029 PMCID: PMC11215133 DOI: 10.3389/fimmu.2024.1436631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- Beatrice Ondondo
- Immunology Department, University Hospitals of Leicester National Health Service (NHS) Trust, Leicester, United Kingdom
| |
Collapse
|
26
|
Abrignani V, Salvo A, Pacinella G, Tuttolomondo A. The Mediterranean Diet, Its Microbiome Connections, and Cardiovascular Health: A Narrative Review. Int J Mol Sci 2024; 25:4942. [PMID: 38732161 PMCID: PMC11084172 DOI: 10.3390/ijms25094942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
The Mediterranean diet (MD), rich in minimally processed plant foods and in monounsaturated fats but low in saturated fats, meat, and dairy products, represents one of the most studied diets for cardiovascular health. It has been shown, from both observational and randomized controlled trials, that MD reduces body weight, improves cardiovascular disease surrogates such as waist-to-hip ratios, lipids, and inflammation markers, and even prevents the development of fatal and nonfatal cardiovascular disease, diabetes, obesity, and other diseases. However, it is unclear whether it offers cardiovascular benefits from its individual components or as a whole. Furthermore, limitations in the methodology of studies and meta-analyses have raised some concerns over its potential cardiovascular benefits. MD is also associated with characteristic changes in the intestinal microbiota, mediated through its constituents. These include increased growth of species producing short-chain fatty acids, such as Clostridium leptum and Eubacterium rectale, increased growth of Bifidobacteria, Bacteroides, and Faecalibacterium prausnitzii species, and reduced growth of Firmicutes and Blautia species. Such changes are known to be favorably associated with inflammation, oxidative status, and overall metabolic health. This review will focus on the effects of MD on cardiovascular health through its action on gut microbiota.
Collapse
Affiliation(s)
- Vincenzo Abrignani
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Andrea Salvo
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Gaetano Pacinella
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, University of Palermo, 90127 Palermo, Italy; (V.A.); (A.S.); (G.P.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
27
|
Zhang J, Ling L, Xiang L, Li W, Bao P, Yue W. Role of the gut microbiota in complications after ischemic stroke. Front Cell Infect Microbiol 2024; 14:1334581. [PMID: 38644963 PMCID: PMC11026644 DOI: 10.3389/fcimb.2024.1334581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Ischemic stroke (IS) is a serious central nervous system disease. Post-IS complications, such as post-stroke cognitive impairment (PSCI), post-stroke depression (PSD), hemorrhagic transformation (HT), gastrointestinal dysfunction, cardiovascular events, and post-stroke infection (PSI), result in neurological deficits. The microbiota-gut-brain axis (MGBA) facilitates bidirectional signal transduction and communication between the intestines and the brain. Recent studies have reported alterations in gut microbiota diversity post-IS, suggesting the involvement of gut microbiota in post-IS complications through various mechanisms such as bacterial translocation, immune regulation, and production of gut bacterial metabolites, thereby affecting disease prognosis. In this review, to provide insights into the prevention and treatment of post-IS complications and improvement of the long-term prognosis of IS, we summarize the interaction between the gut microbiota and IS, along with the effects of the gut microbiota on post-IS complications.
Collapse
Affiliation(s)
- Jinwei Zhang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Ling Ling
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Lei Xiang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Wenxia Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Pengnan Bao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Wei Yue
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
28
|
Wang T, Zhuang Y, Yu C, Wang Z, Liu Y, Xu Q, Liu K, Li Y. D-beta-hydroxybutyrate up-regulates Claudin-1 and alleviates the intestinal hyperpermeability in lipopolysaccharide-treated mice. Tissue Cell 2024; 87:102343. [PMID: 38442546 DOI: 10.1016/j.tice.2024.102343] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
The hyperpermeability of intestinal epithelium is a key contributor to the occurrence and development of systemic inflammation. Although D-beta-hydroxybutyrate (BHB) exhibits various protective effects, whether it affects the permeability of intestinal epithelium in systemic inflammation has not been clarified. In this study, we investigated the effects of BHB on the intestinal epithelial permeability, the epithelial marker E-cadherin and the tight junction protein Claudin-1 in colon in the lipopolysaccharide (LPS)-induced systemic inflammation mouse model. Intraperitoneal injection of LPS was used to induce systemic inflammation and BHB was given by oral administration. The permeability of intestinal epithelium, the morphological changes of colonic epithelium, the distribution and generation of colon E-cadherin, and the Claudin-1 generation and its epithelial distribution in colon were detected. The results confirmed the intestinal epithelial hyperpermeability and inflammatory changes in colonic epithelium, with disturbed E-cadherin distribution in LPS-treated mice. Besides, colon Claudin-1 generation was decreased and its epithelial distribution in colon was weakened in LPS-treated mice. However, BHB treatments alleviated the LPS-induced hyperpermeability of intestinal epithelium, attenuated the colonic epithelial morphological changes and promoted orderly distribution of E-cadherin in colon. Furthermore, BHB up-regulated colon Claudin-1 generation and promoted its colonic epithelial distribution and content in LPS-treated mice. In conclusion, BHB may alleviate the hyperpermeability of intestinal epithelium via up-regulation of Claudin-1 in colon in LPS-treated mice.
Collapse
Affiliation(s)
- Ting Wang
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China
| | - Yuchen Zhuang
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China
| | - Chenglong Yu
- Teaching laboratory center, Hebei Medical University, Hebei, People's Republic of China
| | - Zhaobo Wang
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China
| | - Yuan Liu
- Department of Ophthalmology, First Central Hospital of Baoding, Hebei, People's Republic of China
| | - Qian Xu
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China
| | - Kun Liu
- Teaching laboratory center, Hebei Medical University, Hebei, People's Republic of China.
| | - Yanning Li
- Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Hebei, People's Republic of China.
| |
Collapse
|
29
|
Tian Y, Yao G, Skudder-Hill L, Xu G, Qian Y, Tang F, Wang Q, Bao Q, Li L. Gut microbiota's causative relationship with peripheral artery disease: a Mendelian randomization study. Front Microbiol 2024; 15:1340262. [PMID: 38505559 PMCID: PMC10948605 DOI: 10.3389/fmicb.2024.1340262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction The relationship between gut microbiota and peripheral artery disease (PAD) remains understudied. While traditional risk factors like smoking and hyperlipidemia are well-understood, our study aims to determine the potential causative association of gut microbiota with PAD using Mendelian Randomization. Methods Data from the International MiBioGen Consortium and the FinnGen research project were used to study 211 bacterial taxa. Instrumental variables, comprising 2079 SNPs, were selected based on significance levels and linkage disequilibrium. Analyses were conducted utilizing the inverse-variance weighted (IVW) method and other statistical MR techniques to mitigate biases, processed in R (v4.3.1) with the TwosampleMR package. Results Three bacterial taxa, namely genus Coprococcus2, RuminococcaceaeUCG004, and RuminococcaceaeUCG010, emerged as protective factors against PAD. In contrast, family. FamilyXI and the genus Lachnoclostridium and LachnospiraceaeUCG001 were identified as risk factors. Conclusion Our findings hint at a causative association between certain gut microbiota and PAD, introducing new avenues for understanding PAD's etiology and developing effective treatments. The observed associations now warrant further validation in varied populations and detailed exploration at finer taxonomic levels.
Collapse
Affiliation(s)
- Yu Tian
- Vascular Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guanqun Yao
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | | | - Guangyang Xu
- Vascular Department, Beijing Hua Xin Hospital (1st Hospital of Tsinghua University), Beijing, China
| | - Yuxuan Qian
- Vascular Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Feng Tang
- Vascular Department, Beijing Hua Xin Hospital (1st Hospital of Tsinghua University), Beijing, China
| | - Qian Wang
- Vascular Department, Beijing Hua Xin Hospital (1st Hospital of Tsinghua University), Beijing, China
| | - Qianhui Bao
- School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Lei Li
- School of Clinical Medicine, Tsinghua University, Beijing, China
- Vascular Department, Beijing Hua Xin Hospital (1st Hospital of Tsinghua University), Beijing, China
| |
Collapse
|
30
|
Luqman A, Hassan A, Ullah M, Naseem S, Ullah M, Zhang L, Din AU, Ullah K, Ahmad W, Wang G. Role of the intestinal microbiome and its therapeutic intervention in cardiovascular disorder. Front Immunol 2024; 15:1321395. [PMID: 38343539 PMCID: PMC10853344 DOI: 10.3389/fimmu.2024.1321395] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
The gut microbiome is a heterogeneous population of microbes comprising viruses, bacteria, fungi, and protozoa. Such a microbiome is essential for sustaining host equilibrium, and its impact on human health can be altered by a variety of factors such as external variables, social behavior, age, nutrition, and genetics. Gut microbes' imbalances are related to a variety of chronic diseases including cancer, obesity, and digestive disorders. Globally, recent findings show that intestinal microbes have a significant role in the formation of cardiovascular disease (CVD), which is still the primary cause of fatalities. Atherosclerosis, hypertension, diabetes, inflammation, and some inherited variables are all cardiovascular risk variables. However, studies found correlations between metabolism, intestinal flora, and dietary intake. Variations in the diversity of gut microbes and changes in their activity are thought to influence CVD etiology. Furthermore, the gut microbiota acts as an endocrine organ, producing bioactive metabolites such as TMA (trimethylamine)/TMAO (trimethylamine N-oxide), SCFA (short-chain fatty acids), and bile acids, which have a substantial impact on host wellness and disease by multiple mechanisms. The purpose of this overview is to compile current evidence highlighting the intricate links between gut microbiota, metabolites, and the development of CVD. It focuses on how intestinal dysbiosis promotes CVD risk factors such as heart failure, hypertension, and atherosclerosis. This review explores the normal physiology of intestinal microbes and potential techniques for targeting gut bacteria for CVD treatment using various microbial metabolites. It also examines the significance of gut bacteria in disease treatment, including supplements, prebiotics, probiotics, antibiotic therapies, and fecal transplantation, which is an innovative approach to the management of CVD. As a result, gut bacteria and metabolic pathways become increasingly attractive as potential targets for CVD intervention.
Collapse
Affiliation(s)
- Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| | - Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Mehraj Ullah
- School of Fermentation Engineering Tianjin University of Science and Technology, Tianjin, China
| | | | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, United States
| | - Kamran Ullah
- Department of Biology, The University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Waqar Ahmad
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| |
Collapse
|
31
|
Wang L, Nan Y, Zhu W, Wang S. Effect of TMAO on the incidence and prognosis of cerebral infarction: a systematic review and meta-analysis. Front Neurol 2024; 14:1287928. [PMID: 38259655 PMCID: PMC10801906 DOI: 10.3389/fneur.2023.1287928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Objective This study aimed to evaluate the effect of trimethylamine oxide (TMAO) on the incidence and prognosis of cerebral infarction. Methods We searched PubMed, Embase, and Cochrane databases for all clinical studies on the association of TMAO with cerebral infarction incidence and prognosis from inception to April 2023. A systematic review and meta-analysis were conducted using the meta-analysis of observational studies in epidemiology (MOOSE) declaration list. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the study. This study protocol was registered on the PROSPERO database with the ID: CRD42023459661. The extracted data included the OR value of the effect of TMAO on the incidence and prognosis of cerebral infarction, the HR value between TMAO and underlying diseases, the RR value, 95% confidence intervals, and the AUC value of TMAO in the prediction model of cerebral infarction. Results Fifteen studies including 40,061 patients were included. All the patients were from China or Germany. The TMAO level was significantly correlated with the Modified Rankin Score (mRS) 3 months after the onset of cerebral infarction (OR, 1.581; 95% CI, 1.259-1.987; p < 0.01). The TMAO level was significantly correlated with the rate of first-time incidence and recurrence of cerebral infarction (OR, 1.208; 95% CI, 1.085-1.344; p < 0.01 and HR, 1.167; 95% CI, 1.076-1.265; p < 0.01, respectively). The TMAO level was also highly correlated with disease severity at onset (National Institutes of Health Stroke Scale, NIHSS >5) (OR, 5.194; 95% CI, 1.206-22.363; p < 0.05), but had no significant correlation with mortality after cerebral infarction (p > 0.05). Correlation analysis of TMAO with underlying diseases in the population indicated that TMAO had a significant correlation with histories of hypertension, diabetes mellitus, coronary artery disease, and cerebral infarction (p < 0.05), but not with hyperlipidemia (p > 0.05). Six risk prediction models of TMAO for cerebral infarction reported in four studies were systematically evaluated; five of them had good predictive value (AUC ≥ 0.7). Conclusion TMAO is an independent risk factor affecting the onset, prognosis, and severity of cerebral infarction.
Collapse
Affiliation(s)
- Lin Wang
- Traditional Chinese Medicine Department, Beijing Tiantan Hospital, Beijing, China
| | - Yinan Nan
- International Department, China-Japan Friendship Hospital, Beijing, China
| | - Wenhao Zhu
- Department of Encephalopathy, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Shaoqing Wang
- Traditional Chinese Medicine Department, Beijing Tiantan Hospital, Beijing, China
| |
Collapse
|
32
|
菲尔顿· 吐, 张 为, 伊力哈木江· 克, 张 明, 王 莽. [Correlation Between Plasma Trimethylamine N-Oxide and Lipid Levels in Hyperlipidemic Patients]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1030-1034. [PMID: 37866964 PMCID: PMC10579080 DOI: 10.12182/20230960109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Indexed: 10/24/2023]
Abstract
Objective To investigate the correlation between the plasma levels of trimethylamine N-oxide (TMAO) and lipid levels in hyperlipidemic patients. Methods A total of 130 patients who received treatment and underwent coronary angiography at the Heart Center, the First Affiliated Hospital of Xinjiang Medical University between March 2019 and March 2021 were enrolled. Patients' plasma TMAO levels were determined by stable isotope liquid chromatography/mass spectrometry. The correlation between plasma TMAO levels and the levels of lipids, including triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C), was analyzed. Results Patients with hyperlipidemia had higher plasma TMAO levels (μmol/L) than patients without hyperlipidemia did (6.73±5.40 vs. 3.82±2.65), with the difference being statistically significant ( P<0.05). After controlling for the effects of age and body mass index, partial correlation analysis revealed that plasma TMAO levels were positively correlated with plasma TG ( r=0.286, P<0.001) and negatively correlated with HDL-C ( r=-0.366, P<0.001). Conclusion There is a correlation between plasma TMAO levels and lipid levels in hyperlipidemic patients.
Collapse
Affiliation(s)
- 吐尔洪 菲尔顿·
- 新疆医科大学第一附属医院 心脏中心 心脏外科 (乌鲁木齐 830000)Department of Cardiac Surgery, Heart Center, The First Affiliated Hospital of Xinjiang Medical University, Urumuqi 830000, China
| | - 为民 张
- 新疆医科大学第一附属医院 心脏中心 心脏外科 (乌鲁木齐 830000)Department of Cardiac Surgery, Heart Center, The First Affiliated Hospital of Xinjiang Medical University, Urumuqi 830000, China
| | - 克尤木 伊力哈木江·
- 新疆医科大学第一附属医院 心脏中心 心脏外科 (乌鲁木齐 830000)Department of Cardiac Surgery, Heart Center, The First Affiliated Hospital of Xinjiang Medical University, Urumuqi 830000, China
| | - 明明 张
- 新疆医科大学第一附属医院 心脏中心 心脏外科 (乌鲁木齐 830000)Department of Cardiac Surgery, Heart Center, The First Affiliated Hospital of Xinjiang Medical University, Urumuqi 830000, China
| | - 莽原 王
- 新疆医科大学第一附属医院 心脏中心 心脏外科 (乌鲁木齐 830000)Department of Cardiac Surgery, Heart Center, The First Affiliated Hospital of Xinjiang Medical University, Urumuqi 830000, China
| |
Collapse
|
33
|
Masenga SK, Povia JP, Lwiindi PC, Kirabo A. Recent Advances in Microbiota-Associated Metabolites in Heart Failure. Biomedicines 2023; 11:2313. [PMID: 37626809 PMCID: PMC10452327 DOI: 10.3390/biomedicines11082313] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Heart failure is a risk factor for adverse events such as sudden cardiac arrest, liver and kidney failure and death. The gut microbiota and its metabolites are directly linked to the pathogenesis of heart failure. As emerging studies have increased in the literature on the role of specific gut microbiota metabolites in heart failure development, this review highlights and summarizes the current evidence and underlying mechanisms associated with the pathogenesis of heart failure. We found that gut microbiota-derived metabolites such as short chain fatty acids, bile acids, branched-chain amino acids, tryptophan and indole derivatives as well as trimethylamine-derived metabolite, trimethylamine N-oxide, play critical roles in promoting heart failure through various mechanisms. Mainly, they modulate complex signaling pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells, Bcl-2 interacting protein 3, NLR Family Pyrin Domain Containing inflammasome, and Protein kinase RNA-like endoplasmic reticulum kinase. We have also highlighted the beneficial role of other gut metabolites in heart failure and other cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| | - Joreen P. Povia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Propheria C. Lwiindi
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone 10101, Zambia; (J.P.P.); (P.C.L.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA
| |
Collapse
|
34
|
Martín Giménez VM, Modrego J, Gómez-Garre D, Manucha W, de las Heras N. Gut Microbiota Dysbiosis in COVID-19: Modulation and Approaches for Prevention and Therapy. Int J Mol Sci 2023; 24:12249. [PMID: 37569625 PMCID: PMC10419057 DOI: 10.3390/ijms241512249] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Inflammation and oxidative stress are critical underlying mechanisms associated with COVID-19 that contribute to the complications and clinical deterioration of patients. Additionally, COVID-19 has the potential to alter the composition of patients' gut microbiota, characterized by a decreased abundance of bacteria with probiotic effects. Interestingly, certain strains of these bacteria produce metabolites that can target the S protein of other coronaviruses, thereby preventing their transmission and harmful effects. At the same time, the presence of gut dysbiosis can exacerbate inflammation and oxidative stress, creating a vicious cycle that perpetuates the disease. Furthermore, it is widely recognized that the gut microbiota can metabolize various foods and drugs, producing by-products that may have either beneficial or detrimental effects. In this regard, a decrease in short-chain fatty acid (SCFA), such as acetate, propionate, and butyrate, can influence the overall inflammatory and oxidative state, affecting the prevention, treatment, or worsening of COVID-19. This review aims to explore the current evidence regarding gut dysbiosis in patients with COVID-19, its association with inflammation and oxidative stress, the molecular mechanisms involved, and the potential of gut microbiota modulation in preventing and treating SARS-CoV-2 infection. Given that gut microbiota has demonstrated high adaptability, exploring ways and strategies to maintain good intestinal health, as well as an appropriate diversity and composition of the gut microbiome, becomes crucial in the battle against COVID-19.
Collapse
Affiliation(s)
- Virna Margarita Martín Giménez
- Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, Universidad Católica de Cuyo, San Juan 5400, Argentina;
| | - Javier Modrego
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Dulcenombre Gómez-Garre
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos-Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Plaza Ramón y Cajal, s/n. Universidad Complutense, 28040 Madrid, Spain
| | - Walter Manucha
- Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina;
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Mendoza 5500, Argentina
| | - Natalia de las Heras
- Departamento de Fisiología, Facultad de Medicina, Plaza Ramón y Cajal, s/n. Universidad Complutense, 28040 Madrid, Spain
| |
Collapse
|