1
|
Alaguthevar R, Pawale AV, Murugesan B, Khan A, Rhim JW, Chelladurai D. Comprehensive review of food Archaeome: Exploring the understudied microbiome and health benefits of fermented foods. Microb Pathog 2025; 205:107718. [PMID: 40398637 DOI: 10.1016/j.micpath.2025.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/15/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
Extremophiles are species that thrive optimally in extreme environmental conditions, and advances in DNA-based identification methods have sparked increased interest in studying the dynamic ecosystem of archaea outside their harsh environmental niches. Most studies on the food microbiome have primarily focused on bacterial and fungal communities. In contrast, food archaea play an essential role in human-related foods, despite being largely ignored and understudied. Archaea can survive, colonize, and thrive under harsh processing conditions in food manufacturing, and they can enter the human body and modulate the human microbiome. Additionally, gut microbiomes can be influenced through vertical transmission facilitated by breast milk, organic animal products, and salty fermented foods. Early studies on foods concentrated mostly on salty and fermented items because these foods represent extreme environments. However, gut-associated archaea may be linked to disease as they produce toxic compounds and detoxify pathogen growth inhibitors. The precise mechanisms by which this pathogenicity occurs are still unknown, and further studies are necessary to illuminate potential human health implications. Despite the growing understanding of the prevalence of archaeal pathogens in extreme environments, current research on food-related archaeal communities remains limited. This review emphasizes the role of food archaeal pathogens in human health and describes new technologies for producing novel nutritious foods utilizing archaeal-derived materials in the food industry.
Collapse
Affiliation(s)
- Ramalakshmi Alaguthevar
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641 003, Tamil Nadu, India; Department of Food Process Engineering, Tamil Nadu Agricultural University, Coimbatore, 641 003, Tamil Nadu, India
| | - Amol Vishwas Pawale
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641 003, Tamil Nadu, India
| | - Balakrishnan Murugesan
- Department of Food Process Engineering, Tamil Nadu Agricultural University, Coimbatore, 641 003, Tamil Nadu, India.
| | - Ajahar Khan
- Department of Food and Nutrition, BioNanocomposite Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jong-Whan Rhim
- Department of Food and Nutrition, BioNanocomposite Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Devadharshini Chelladurai
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641 003, Tamil Nadu, India
| |
Collapse
|
2
|
Huang H, Cheng S, Long T, Fu B, Yang J, Cai C, Gu M, Niu H, Chen X, Hua W, Yang S. Plasma trimethylamine levels predict adverse cardiovascular events in sudden cardiac arrest Survivors: A prospective cohort study. Clin Biochem 2025; 137:110928. [PMID: 40228618 DOI: 10.1016/j.clinbiochem.2025.110928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The trimethylamine N-oxide (TMAO) pathway has been associated with multiple cardiovascular diseases, yet its prognostic value for sudden cardiac arrest (SCA) survivors remains unknown. METHODS Patients who survived SCA and received implantable cardioverter defibrillators (ICDs) were prospectively enrolled. We evaluated the associations between plasma concentrations of TMAO-related metabolites and long-term adverse clinical events, including recurrent lethal ventricular arrhythmia (VA). RESULTS A total of 75 SCA survivors were included in the study. During a median follow-up of 1099 days, 34 (45.3 %) patients experienced adverse clinical events, including 24 (32.2 %) with life-threatening VA, 12 (16.0 %) with heart failure rehospitalization, and 5 (6.7 %) with cardiovascular death. Trimethylamine (TMA), carnitine, choline, and creatinine showed strong correlations with clinically significant parameters such as left ventricular ejection fraction (LVEF), New York Heart Association functional class, and N-terminal pro-brain natriuretic peptide (NT-proBNP). These four metabolites demonstrated positive associations with adverse clinical events, with higher median level of TMA associated with more than a threefold increased risk after adjusting for age, sex, LVEF, kidney function, and NT-proBNP levels (hazard ratio = 3.36, 95 % confidence interval [CI]: 1.18-9.59; P = 0.024). A scoring system, VT-C3, incorporating LVEF, TMA, and the weighted sum of TMA-related metabolites (Choline, Carnitine, Creatinine), showed significant predictive capacity for both adverse events (area under the curve [AUC]: 0.75, 95 % CI: 0.64-0.85) and recurrent lethal VA (AUC: 0.73, 95 % CI: 0.62-0.84). No significant prognostic values were observed for TMAO and betaine. CONCLUSIONS Our findings suggest that plasma concentrations of TMA, choline, carnitine, and creatinine are associated with an increased risk of subsequent adverse clinical events among SCA survivors. A simple scoring system comprising LVEF and these biomarkers could enhance current risk stratification and improve secondary prevention strategies based on ICD implantation.
Collapse
Affiliation(s)
- Hao Huang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Sijing Cheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Tianxin Long
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Bingqi Fu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Juwei Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Chi Cai
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Min Gu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hongxia Niu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xuhua Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Wei Hua
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| | - Shengwen Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China; Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Heart Center and Beijing Key Laboratory of Hypertension, Beijing, PR China.
| |
Collapse
|
3
|
Jiang J, Zhu P, Ding X, Zhou L, Li X, Lei Y, Wang H, Chen L, Li X, Fei Y, Ouyang D, Li X, Zhang W. The microbiome-derived metabolite trimethylamine N-oxide is associated with chronic kidney disease risk. Appl Microbiol Biotechnol 2025; 109:97. [PMID: 40261397 PMCID: PMC12014799 DOI: 10.1007/s00253-025-13481-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
Previous studies have established a correlation between the microbiome-derived metabolite trimethylamine N-oxide (TMAO) and decreased renal function, but with great heterogeneity. Moreover, population-based evidence remains scarce, particularly in Chinese populations. We designed a meta-analysis and a population-based cross-sectional study in China to examine the associations between TMAO and chronic kidney disease (CKD). In meta-analysis, among 2125 pooled subjects with 1240 controls and 885 CKD patients, a significant association was observed between TMAO and CKD, with a standardized mean difference of - 0.93 (95% confidence interval: - 1.11, - 0.75). Meta-regression analysis identified gender, age, and body mass index (BMI) as significant heterogeneity factors. In our population-based study of 5584 subjects with an estimated glomerular filtration rate (eGFR) ≥ 60 mL/min/1.73 m2 from Sijing community, 100 developed CKD in 2 years. We matched 195 controls by age and gender from the 5484 non-CKD subjects. Male subjects and alcohol consumers exhibited a lower risk of CKD with adjusted odds ratio (OR) of 0.471 (P < 0.05) and 0.320 (P < 0.05), respectively. When comparing subjects in the lowest tertile of TMAO, adjusted OR reached to 1.243 (P > 0.05) for those in the middle and 2.123 (P < 0.05) in the highest tertile (P for trend < 0.05). TMAO demonstrated a moderate capacity to distinguish CKD from non-CKD subjects (AUC = 0.614, P < 0.01). Our findings indicate TMAO is significantly associated with the risk of CKD, and suggest age, gender, and BMI may confound the relationship between TMAO and CKD. KEY POINTS: • Subjects with elevated TMAO levels have an increased risk of CKD. • TMAO demonstrates a moderate capacity to distinguish CKD from non-CKD cases. • Age, gender and BMI may confound the relationship between TMAO and CKD.
Collapse
Affiliation(s)
- Junyi Jiang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China
| | - Peng Zhu
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Xiaoying Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Li Zhou
- Department of Nursing, the Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China
| | - Xiaoqiang Li
- Department of Food and Environmental Disease, Changzhou Center for Disease Control and Prevention, Changzhou, 213002, China
| | - Yuyan Lei
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University, Changsha, 410078, People's Republic of China
- Phase-Clinical Trial Laboratory, the Second Nanning Peoples Hospital, Nanning, 530000, People's Republic of China
| | - Hao Wang
- Department of Ophthalmology, Hebei Eye Hospital, Xingtai, 054001, People's Republic of China
| | - LuLu Chen
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China
| | - Xiang Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China
| | - Yunzhou Fei
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China
| | - Dongsheng Ouyang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China
| | - Xiaohui Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, 410221, People's Republic of China.
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University, Changsha, 410078, People's Republic of China.
| | - Wei Zhang
- Engineering Research Center of Applied Technology of Pharmacogenomics (Ministry of Education, China), Hunan Key Laboratory of Pharmacomicrobiomics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
- The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China.
- Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, 410013, People's Republic of China.
| |
Collapse
|
4
|
Caradonna E, Abate F, Schiano E, Paparella F, Ferrara F, Vanoli E, Difruscolo R, Goffredo VM, Amato B, Setacci C, Setacci F, Novellino E. Trimethylamine-N-Oxide (TMAO) as a Rising-Star Metabolite: Implications for Human Health. Metabolites 2025; 15:220. [PMID: 40278349 PMCID: PMC12029716 DOI: 10.3390/metabo15040220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
The intestinal microbiota, hosting trillions of microorganisms that inhabit the gastrointestinal tract, functions as a symbiotic organism that plays a crucial role in regulating health by producing biologically active molecules that can enter systemic circulation. Among them, trimethylamine-N-oxide (TMAO), an organic compound derived from dietary sources and microbial metabolism, has emerged as a critical biomarker linking diet, the gut microbiota, and the host metabolism to various pathological conditions. This comprehensive review highlights TMAO's biosynthesis, physiological functions, and clinical significance, focusing on its mechanistic contributions to cardiovascular and neurodegenerative diseases. Notably, TMAO-mediated pathways include endothelial dysfunction, inflammation via NLRP3 inflammasome activation, and cholesterol metabolism disruption, which collectively accelerate atherosclerosis and disease progression. Nonetheless, this work underscores the innovative potential of targeting TMAO through dietary, nutraceutical, and microbiota-modulating strategies to mitigate its pathological effects, marking a transformative approach in the prevention and management of TMAO-related disorders.
Collapse
Affiliation(s)
- Eugenio Caradonna
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Federico Abate
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy;
| | - Elisabetta Schiano
- Inventia Biotech-Healthcare Food Research Center S.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy;
| | - Francesca Paparella
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy (F.F.)
| | - Emilio Vanoli
- School of Nursing, Cardiovascular Diseases, University of Pavia, 27100 Pavia, Italy;
| | | | - Vito Maria Goffredo
- Department of Interdisciplinary Medicine, Università Degli Studi di Bari, 70124 Bari, Italy;
| | - Bruno Amato
- Department of Public Health, Università Degli Studi di Napoli Federico II, 80138 Naples, Italy;
| | - Carlo Setacci
- Vascular and Endovascuar Surgery Unit, “Le Scotte” Hospital of Siena, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy;
| | - Francesco Setacci
- Vascular Surgery Unit, Università degli Studi di Enna “Kore”, 94100 Enna, Italy;
| | - Ettore Novellino
- Department of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| |
Collapse
|
5
|
McCerery R, Hall L, Omanakuttan J, Chai HZ, Lawrence J, Woodward J, Pearce DA. Preparation protocol for epifluorescence microscopy when working close to the detection limit. Lett Appl Microbiol 2025; 78:ovaf026. [PMID: 39987441 DOI: 10.1093/lambio/ovaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/15/2025] [Accepted: 02/21/2025] [Indexed: 02/24/2025]
Abstract
Working with low density, low biomass material can be challenging, especially when working near the detection limit. Although background contamination is a universal consideration in microbiological research, its impact is increased when the cells under assessment approach the same concentration as the background contamination. The aim of this work was to identify and remove laboratory sources of background contamination in the cell mounting process for epifluorescence microscopy to improve the reliability of cell counting for low biomass samples. Microscope slides and coverslips were assessed before and after autoclaving, washing with detergent and rinsing with ethanol solution. The solutions used in sample mounting; 4',6-diamidino-2-phenylindole, phosphate buffered saline, and immersion oil, were tested before and after autoclaving as well as both single and triple filtering with a 0.2 µm membrane filter. Using a combination of detergent and ethanol rinses of glassware and triple filtering of all solutions, we were able to reduce the background contamination by almost two orders of magnitude, down from 1 × 104(±4.3 × 103) cells to 302(±312) cells per filter paper. This method was then validated with low biomass glacial sediment samples from Renardbreen, Svalbard, with cell concentrations of 1.8 × 105(±2.9 × 104) cells g-1, close to the reported detection limit of epifluorescence microscopy.
Collapse
Affiliation(s)
- Rebecca McCerery
- Department of Geography and Environmental Sciences, Faculty of Engineering and Environment, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - Lewis Hall
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - Jithin Omanakuttan
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - Hui Zhi Chai
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - James Lawrence
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - John Woodward
- Department of Geography and Environmental Sciences, Faculty of Engineering and Environment, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
| | - David A Pearce
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle-upon-Tyne, NE1 8ST, United Kingdom
- British Antarctic Survey, Natural Environment Research Council, Cambridge CB3 OET, United Kingdom
| |
Collapse
|
6
|
Zheng X, Fan J, Yin J, Chu Y. The role of gut microbiota and plasma metabolites in ulcerative colitis: Insights from Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e41710. [PMID: 40020117 PMCID: PMC11875619 DOI: 10.1097/md.0000000000041710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
Emerging research suggests that alterations in gut microbiota composition may play a significant role in the pathogenesis of ulcerative colitis (UC). Plasma metabolites, which are influenced by gut microbiota, have also been implicated, but their role in UC remains unclear. This study aims to determine whether specific plasma metabolites mediate the causal relationship between gut microbiota and UC using Mendelian randomization (MR) analysis. This study employed publicly available summary-level data from genome-wide association studies and metagenomic datasets. Gut microbiota data were derived from the FINRISK cohort (5959 participants), plasma metabolite data from the Canadian Longitudinal Study on Aging (8299 individuals), and UC data from multiple consortia (17,030 cases and 883,787 controls). Forward and reverse MR analyses, supplemented by linkage disequilibrium score regression (LDSC), were conducted to assess causal relationships. Mediation effects of plasma metabolites between gut microbiota and UC were analyzed using the product of coefficients method. Various sensitivity analyses, including MR-Egger and MR-PRESSO, were applied to detect pleiotropy and ensure robust results. The study identified 20 bacterial taxa and 93 plasma metabolites linked to UC. Forward MR analysis showed that Clostridium S felsineum increased UC risk via reduced carnitine levels, with a mediation proportion of 39.77%. Eubacterium callanderi was associated with decreased UC risk through the tryptophan to pyruvate ratio (16.02% mediation). Additionally, species CAG-590 sp000431135 increased UC risk through elevated mannitol/sorbitol levels, mediating 28.38% of the effect. Sensitivity analyses confirmed the robustness of these findings, with minimal heterogeneity and pleiotropy detected. This study highlights the significant role of gut microbiota and their associated plasma metabolites in the pathogenesis of UC. Specific microbial species influence UC through metabolites, suggesting potential therapeutic targets. Modulating carnitine, tryptophan metabolism, or sugar alcohols could offer promising avenues for UC management.
Collapse
Affiliation(s)
- XuWen Zheng
- Emergency Department, Wujin Hospital Affiliated with Jiangsu University and Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - JinNuo Fan
- Emergency Department, Wujin Hospital Affiliated with Jiangsu University and Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - JinNan Yin
- Emergency Department, Wujin Hospital Affiliated with Jiangsu University and Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Ying Chu
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| |
Collapse
|
7
|
Marhuenda-Egea FC, Sanchez-Jerez P. Metabolomic Insights into Wild and Farmed Gilthead Seabream ( Sparus aurata): Lipid Composition, Freshness Indicators, and Environmental Adaptations. Molecules 2025; 30:770. [PMID: 40005081 PMCID: PMC11857973 DOI: 10.3390/molecules30040770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study explores the metabolic adaptations and quality differences between wild and farmed gilthead seabream (Sparus aurata), with a particular focus on lipid composition and metabolite profiles. These differences are examined in the context of varying environmental conditions, feeding regimes, and post-harvest processes. High-resolution magic-angle-spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy was employed to perform the metabolomic analysis. RESULTS Farmed seabream exhibited higher lipid content and PUFA levels (e.g., DHA and EPA) due to aquaculture diets, while wild seabream showed lower lipid concentrations and elevated levels of polar metabolites. Metabolic trade-offs in wild seabream reflected greater physical activity and environmental adaptation. The K-value indicated faster spoilage in farmed seabream, particularly from Greece, linked to handling conditions. HR-MAS provided precise, reproducible results, allowing direct quantification of key metabolites without altering sample integrity. METHODS HR-MAS NMR was employed to analyze muscle tissue from wild and farmed seabream (produced in Spain and imported from Greece), providing high-resolution spectra without requiring sample extraction. Metabolite quantification included polyunsaturated fatty acids (PUFAs), creatine, taurine, lactate, and trimethylamine N-oxide (TMAO). Freshness was monitored using the K-value index, calculated from ATP derivative levels in samples stored at 4 °C. CONCLUSIONS The study highlights the influence of diet and environment on the metabolic profiles of seabream. HR-MAS NMR emerges as a robust method for metabolomic studies and freshness assessment. Findings emphasize the potential for dietary adjustments to optimize aquaculture practices and fish quality while underscoring the importance of sustainable production strategies. Further research into lipid metabolism genes and environmental factors is recommended to deepen understanding of these adaptations.
Collapse
Affiliation(s)
- Frutos C. Marhuenda-Egea
- Department of Biochemistry and Molecular Biology and Agricultural Chemistry and Edafology, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690 Alicante, Spain
| | - Pablo Sanchez-Jerez
- Department of Marine Sciences and Applied Biology, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690 Alicante, Spain;
| |
Collapse
|
8
|
Liao JC, Xiang J, Gui WY, Luo HZ, You Q, He QR, Lu MX, Yang SY, Wang Q, Zou JD, Li CY. Broad range lipidomics and metabolomics coupled with 16S rRNA sequencing to reveal the mechanisms of Huangkui Capsule against cisplatin-induced nephrotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119197. [PMID: 39631718 DOI: 10.1016/j.jep.2024.119197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/22/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangkui Capsule (HKC) is a traditional Chinese medicinal preparation. Numerous clinical studies have reported that HKC has a good nephroprotection effect. The clinical application of cisplatin is greatly limited by its nephrotoxicity, and HKC shows promise in preventing cisplatin-induced nephrotoxicity (CIN). AIM OF THE STUDY To evaluate the effectiveness of HKC in alleviating CIN and explore its underlying action mechanisms. MATERIALS AND METHODS A rat model of CIN was established via single-dose injection of cisplatin. The effectiveness of HKC was evaluated by biochemical indices and pathological sections. Then, serum, kidney, and cecal endogenous metabolic profiles as well as the gut microbiota were characterized using lipidomics, metabolomics, and 16S rRNA high-throughput sequencing technique. Spearman's correlation analysis was carried out between gut microbiota, biomarkers, and biochemical indices. Finally, antibiotic treatment was performed to establish a pseudo-sterile rat model and validate the nephroprotection of HKC in a gut microbiota-dependent manner. RESULTS HKC could significantly attenuate the abnormal elevation of serum creatinine and urea nitrogen, kidney index, and kidney injury score in CIN rats, remarkably alleviate the disturbance of metabolic profiles of serum, kidney, and cecal contents, corresponding to the endogenous metabolites such as fatty acids, phosphatidylcholines, amino acids, acylcarnitines, and short-chain fatty acids, and enrich the diversity of gut microbiota. Spearman's correlation analysis revealed that Clostridium_sensu_stricto_1 was positively correlated with the altered short-chain fatty acids in serum and negatively correlated with the altered acylcarnitine in the kidney. In the pseudo-sterile rat model, the attenuation effect of HKC on the abnormal elevation of serum creatinine and urea nitrogen, along with the alleviation of metabolic profile disorders, was greatly diminished or even abolished, demonstrating the nephroprotective effect of HKC in a gut microbiota-dependent manner. CONCLUSIONS HKC exerted the nephroprotective effect on CIN in a gut microbiota-dependent manner, mainly by regulating Clostridium_sensu_stricto_1 mediated metabolisms of phosphatidylcholines, acylcarnitines, fatty acids, tryptophan, and short-chain fatty acids, thereby reducing the inflammatory response. The present study could provide reliable scientific evidence for gut microbiota-dependent mechanisms of HKC in the treatment of kidney injury and may widen the clinical application of HKC in cisplatin-containing cancer therapy.
Collapse
Affiliation(s)
- Jian-Cheng Liao
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Jie Xiang
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Wan-Yu Gui
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Hui-Zhi Luo
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Qing You
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Qi-Rui He
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Ming-Xia Lu
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Shu-Yun Yang
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China
| | - Qiong Wang
- Department of Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China.
| | - Jian-Dong Zou
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China.
| | - Chang-Yin Li
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, 210029, China.
| |
Collapse
|
9
|
Ou Z, Fu X, Norbäck D, Lin R, Wen J, Sun Y. MiMeJF: Application of Coupled Matrix and Tensor Factorization (CMTF) for Enhanced Microbiome-Metabolome Multi-Omic Analysis. Metabolites 2025; 15:51. [PMID: 39852393 PMCID: PMC11767930 DOI: 10.3390/metabo15010051] [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: 10/28/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: The integration of microbiome and metabolome data could unveil profound insights into biological processes. However, widely used multi-omic data analyses often employ a stepwise mining approach, failing to harness the full potential of multi-omic datasets and leading to reduced detection accuracy. Synergistic analysis incorporating microbiome/metabolome data are essential for deeper understanding. Method: This study introduces a Coupled Matrix and Tensor Factorization (CMTF) framework for the joint analysis of microbiome and metabolome data, overcoming these limitations. Two CMTF frameworks were developed to factorize microbial taxa, functional pathways, and metabolites into latent factors, facilitating dimension reduction and biomarker identification. Validation was conducted using three diverse microbiome/metabolome datasets, including built environments and human gut samples from inflammatory bowel disease (IBD) and COVID-19 studies. Results: Our results revealed biologically meaningful biomarkers, such as Bacteroides vulgatus and acylcarnitines associated with IBD and pyroglutamic acid and p-cresol associated with COVID-19 outcomes, which provide new avenues for research. The CMTF framework consistently outperformed traditional methods in both dimension reduction and biomarker detection, offering a robust tool for uncovering biologically relevant insights. Conclusions: Despite its stringent data requirements, including the reliance on stratified microbial-based pathway abundances and taxa-level contributions, this approach provides a significant step forward in multi-omics integration and analysis, with potential applications across biomedical, environmental, and agricultural research.
Collapse
Affiliation(s)
- Zheyuan Ou
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (Z.O.); (R.L.); (J.W.)
| | - Xi Fu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Dan Norbäck
- Occupational and Environmental Medicine, Department of Medical Science, University Hospital, Uppsala University, 75237 Uppsala, Sweden;
| | - Ruqin Lin
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (Z.O.); (R.L.); (J.W.)
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (Z.O.); (R.L.); (J.W.)
| | - Yu Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (Z.O.); (R.L.); (J.W.)
| |
Collapse
|
10
|
Wei Y, Mao H, Liu Q, Fang W, Zhang T, Xu Y, Zhang W, Chen B, Zheng Y, Hu X. Lipid metabolism and microbial regulation analyses provide insights into the energy-saving strategies of hibernating snakes. Commun Biol 2025; 8:45. [PMID: 39800781 PMCID: PMC11725596 DOI: 10.1038/s42003-025-07493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Hibernation is a necessary means for animals to maintain survival while coping with low temperatures and food shortages. While most studies have largely focused on mammalian hibernation, its reptilian equivalent has been less studied. In order to provide insights into the energy metabolism and potential microbial regulatory mechanisms in hibernating snakes, the serum, liver, gut content samples were measured by multi-omic methods. Here we show the active snakes have more vigorous lipid metabolism, whereas snakes in hibernation groups have higher sphingolipid metabolism. Furthermore, the results indicate that the potential energy supply pathway was gluconeogenesis. Microbial analysis reveals that Proteobacteria and Firmicutes showed dynamic changes with the transformation among active, pre-hibernation and hibernation periods. The correlation analysis reveals the potential role of Romboutsia, Providencia and Vagococcus in regulating above metabolism by producing certain metabolites. The results advance the understanding of the complex energy-saving strategy in hibernating poikilotherms.
Collapse
Affiliation(s)
- Yuting Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Huirong Mao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Qiuhong Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Wenjie Fang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Tianxiang Zhang
- Institute of Wildlife Conservation, Jiangxi Academy of Forestry, Nanchang, China
| | - Yongtao Xu
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China
- College of Forestry, Jiangxi Agricultural University, Nanchang, China
| | - Weiwei Zhang
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China
- College of Forestry, Jiangxi Agricultural University, Nanchang, China
| | - Biao Chen
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yunlin Zheng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xiaolong Hu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.
- Jiangxi Provincial Key Laboratory of Conservation Biology, Jiangxi Agricultural University, Nanchang, China.
| |
Collapse
|
11
|
Valdivia-Garcia MA, Bi Y, Abaakil K, V Li J. Derivatization to reduce background interferences for simultaneous quantitation of trimethylamine (TMA) and trimethylamine-N-oxide (TMAO) using liquid chromatography with tandem mass spectrometry. J Pharm Biomed Anal 2025; 252:116480. [PMID: 39326376 DOI: 10.1016/j.jpba.2024.116480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Trimethylamine (TMA) and trimethylamine-N-oxide (TMAO) play a crucial role in many biochemical processes within diverse organisms including animals, plants, fungi and bacteria. Studies have linked these metabolites with cardiovascular and kidney diseases; however, emerging evidence demonstrates their protective properties. Owing to these controversies and co-existence of these metabolites in biological samples, it is crucial to accurately quantify these metabolites to associate their concentrations with various physiological and pathophysiological conditions to elucidate their potential roles. We reported interferences on TMA quantification without derivatizing the analyte. A combined sample preparation method, including sample derivatization with ethyl bromoacetate and use of ion pairing reagent (sodium heptanesulfonate), minimized these interferences and provided improved accuracy and precision for simultaneous quantification of TMA and TMAO. The linearity for TMAO ranged from 0.01 µM to 300 µM and 0.1 µM - 300 µM for TMA. With the application of this method, we reported that the circulating concentrations of TMA was 4 times higher in male mice (33.1 ± 5.9 µmol/L) compared to females (8.3 ± 1.39 µmol/L), whereas TMAO levels were 6 times lower in male (7.2 ± 0.4 µmol/L) than female mice (42.1 ± 4.5 µmol/L). In contrast, concentrations of TMA and TMAO in the colonic tissue did not differ significantly between males and females. The robust analytical method for simultaneously quantifying TMA and TMAO presents a significant value in facilitating investigations on TMA and TMAO biology.
Collapse
Affiliation(s)
- Maria A Valdivia-Garcia
- Section of Nutrition, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Campus, Commonwealth Building, Du Cane Road, London W12 0NN, United Kingdom
| | - Yang Bi
- Section of Nutrition, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Campus, Commonwealth Building, Du Cane Road, London W12 0NN, United Kingdom
| | - Kaoutar Abaakil
- Section of Nutrition, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Campus, Commonwealth Building, Du Cane Road, London W12 0NN, United Kingdom
| | - Jia V Li
- Section of Nutrition, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Campus, Commonwealth Building, Du Cane Road, London W12 0NN, United Kingdom.
| |
Collapse
|
12
|
Zheng X, Zhang Z, Shan T, Zhao M, Lu H, Zhang L, Liang X. Study on the Mechanism of Bifidobacterium animalis subsp. lactis F1-3-2 Regulating Bile Acid Metabolism Through TMA-TMAO Pathway to Improve Atherosclerosis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10417-x. [PMID: 39708191 DOI: 10.1007/s12602-024-10417-x] [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] [Accepted: 12/01/2024] [Indexed: 12/23/2024]
Abstract
Atherosclerosis is a major cause of cardiovascular disease (CVD). The trimethylamine (TMA)-trimethylamine N-oxide (TMAO) pathway is a key crossover pathway highly associated with diet, gut microbiome, and atherosclerosis. The Bifidobacterium animalis subsp. lactis F1-3-2 (Bif. animalis F1-3-2, No. CCTCCM2020832) was screened through in vitro and in vivo experiments in the early stage of this study with excellent lipid-lowering and anti-inflammatory function. By building an atherosclerosis model and focusing on TMAO, the specific mechanism of Bif. animalis F1-3-2 to improve atherosclerosis was explored. The study found that Bif. animalis F1-3-2 effectively improved the accumulation of aortic plaque in atherosclerotic mice. The strain improved lipid metabolism in serum and liver. It decreased the serum TMA and TMAO, regulated bile acid composition, participated in the farnesoid X receptor (FXR) pathway to improve lipid metabolism, and further reduced the aortic macrophage foam cell accumulation. In addition, the strain could improve the structure of the intestinal microbiome and reduce the proportion of Firmicutes and Bacteroidetes. The abundance of Turicibacter, Clostridium sensu stricto_1, and Romboutsia was reduced at the genus level. The differential microbiota is highly correlated with bile acid metabolism, which is speculated to be involved in ameliorating atherosclerotic lipid metabolism disorders.
Collapse
Affiliation(s)
- Xiumei Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, 266100, Shandong, China
| | - Zhe Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Tianhu Shan
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, 266100, Shandong, China
| | - Maozhen Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Haiyan Lu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong, China.
| | - Xi Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao, 266100, Shandong, China.
| |
Collapse
|
13
|
Altrawy A, Khalifa MM, Abdelmaksoud A, Khaled Y, Saleh ZM, Sobhy H, Abdel-Ghany S, Alqosaibi A, Al-Muhanna A, Almulhim J, El-Hashash A, Sabit H, Arneth B. Metabolites in the Dance: Deciphering Gut-Microbiota-Mediated Metabolic Reprogramming of the Breast Tumor Microenvironment. Cancers (Basel) 2024; 16:4132. [PMID: 39766032 PMCID: PMC11674667 DOI: 10.3390/cancers16244132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 01/05/2025] Open
Abstract
Breast cancer (BC), a major cause of death among women worldwide, has traditionally been linked to genetic and environmental factors. However, emerging research highlights the gut microbiome's significant role in shaping BC development, progression, and treatment outcomes. This review explores the intricate relationship between the gut microbiota and the breast tumor microenvironment, emphasizing how these microbes influence immune responses, inflammation, and metabolic pathways. Certain bacterial species in the gut either contribute to or hinder BC progression by producing metabolites that affect hormone metabolism, immune system pathways, and cellular signaling. An imbalance in gut bacteria, known as dysbiosis, has been associated with a heightened risk of BC, with metabolites like short-chain fatty acids (SCFAs) and enzymes such as β-glucuronidase playing key roles in this process. Additionally, the gut microbiota can impact the effectiveness of chemotherapy, as certain bacteria can degrade drugs like gemcitabine and irinotecan, leading to reduced treatment efficacy. Understanding the complex interactions between gut bacteria and BC may pave the way for innovative treatment approaches, including personalized microbiome-targeted therapies, such as probiotics and fecal microbiota transplants, offering new hope for more effective prevention, diagnosis, and treatment of BC.
Collapse
Affiliation(s)
- Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Maye M. Khalifa
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Asmaa Abdelmaksoud
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Yomna Khaled
- Department of Bioinformatics and Functional Genomics, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Zeinab M. Saleh
- Department of Agriculture Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Hager Sobhy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt;
| | - Amany Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Afnan Al-Muhanna
- King Fahad Hospital of the University, Alkhobar, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Jawaher Almulhim
- Department of Biological Sciences, King Faisal University, Alahsa 31982, Saudi Arabia;
| | - Ahmed El-Hashash
- Department of Biomedicine, Texas A&M University, College Station, TX 77840, USA;
| | - Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P. O. Box 77, Egypt; (A.A.); (M.M.K.); (H.S.); (H.S.)
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University, Feulgen Str., 35392 Giessen, Germany
| |
Collapse
|
14
|
Gautam R, Maan P, Patel AK, Vasudevan S, Arora T. Unveiling the complex interplay between gut microbiota and polycystic ovary syndrome: A narrative review. Clin Nutr 2024; 43:199-208. [PMID: 39481287 DOI: 10.1016/j.clnu.2024.10.028] [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: 04/04/2024] [Revised: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND & AIM Polycystic Ovary Syndrome (PCOS) is a complex endocrine disorder that affects women throughout their reproductive age and characterised via polycystic ovaries, hyperandrogenism, and irregular menstruation. There is rising evidence that the pathophysiology of PCOS is significantly affected via the gut microbiota and its metabolic products. METHODS This narrative review synthesizes current literature exploring the relationship between gut microbiota and PCOS. A comprehensive search of electronic databases was conducted to identify relevant studies. Further this review also analysed therapeutic options of probiotics, prebiotics, Fecal Microbiota Transplant (FMT), high fiber and poly phenol rich diet and novel therapeutic agents in treatment of PCOS. RESULTS Emerging evidence suggests alterations in the composition and diversity of gut microbiota in women with PCOS. The current literature showed a complex relationship of gut microbiota, short chain fatty acids (SCFAs) metabolism, intestinal permeability and LPS (Lipid Polysaccharide) metabolism, gut-brain axis and bile acid (BA) pathway within etiology and pathophysiology of PCOS. Additionally, the factors such as diet, lifestyle, genetics, and environmental influences may all contribute to alterations in gut microbiota that could potentially exacerbate or mitigate PCOS symptoms. CONCLUSION The review provides valuable insights into the intricate interplay between the gut and female reproductive health. The present evidence suggested that alterations in diversity and function of the gut microbiota may lead to specific pathogenic changes that lead to development of PCOS. A comprehensive understanding of these microbial dynamics may lead to new therapeutic approaches that target the gut micro biome.
Collapse
Affiliation(s)
- Rohit Gautam
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Pratibha Maan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Arbind Kumar Patel
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Sudharsan Vasudevan
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India
| | - Taruna Arora
- Division of Reproductive Child Health and Nutrition, Indian Council of Medical Research, New Delhi, India.
| |
Collapse
|
15
|
Shayista H, Prasad MN, Raj SN, Ranjini H, Manju K, Baker S. Mechanistic overview of gut microbiota and mucosal pathogens with respect to cardiovascular diseases. THE MICROBE 2024; 5:100160. [DOI: 10.1016/j.microb.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
16
|
Ghajavand B, Avesani C, Stenvinkel P, Bruchfeld A. Unlocking the Potential of Brewers' Spent Grain: A Sustainable Model to Use Beer for Better Outcome in Chronic Kidney Disease. J Ren Nutr 2024; 34:482-492. [PMID: 38621435 DOI: 10.1053/j.jrn.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/17/2024] Open
Abstract
The rising global incidence of chronic inflammatory diseases calls for innovative and sustainable medical solutions. Brewers' spent grain (BSG), a byproduct of beer production, presents a unique opportunity in this regard. This review explores the multifaceted health benefits of BSG, with a focus on managing chronic kidney disease (CKD). BSG is identified as a potent prebiotic with potential as a therapeutic agent in CKD. We emphasize the role of gut dysbiosis in CKD and discuss how BSG could help mitigate metabolic derangements resulting from dysbiosis and CKD. Fermentation of BSG further enhances its positive impact on gut health. Incorporating fermented BSG as a key component in preventive health care could promote a more sustainable and healthier future. By optimizing the use of this typically discarded byproduct, we can align proactive health-care strategies with responsible resource management, benefiting both people and the environment.
Collapse
Affiliation(s)
- Babak Ghajavand
- Department of Renal Medicine, Linköping University Hospital, Linköping, Sweden.
| | - Carla Avesani
- Department of Renal Medicine, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Renal Medicine, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Renal Medicine, CLINTEC, Karolinska Institutet, Stockholm, Sweden; Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
17
|
Cui X, Wei W, Hu Y, Zhang Z, Lu M, Li Y, Wu J, Li C. Dietary inflammation and vascular calcification: a comprehensive review of the associations, underlying mechanisms, and prevention strategies. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 39340196 DOI: 10.1080/10408398.2024.2408447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death globally, and vascular calcification (VC) has been recognized as an independent and strong predictor of global CVD and mortality. Chronic inflammation has been demonstrated to play a significant role in the progression of VC. This review aims to summarize the literature that aimed to elucidate the associations between dietary inflammation (DI) and VC as well as to explore the mechanisms underlying the association and discuss strategies (including dietary interventions) to prevent VC. Notably, diets rich in processed foods, carbohydrates with high glycemic index/load, saturated fatty acids, trans-fatty acids, cholesterol, and phosphorus were found to induce inflammatory responses and accelerate the progression of VC, indicating a close relationship between DI and VC. Moreover, we demonstrate that an imbalance in the composition of the gut microbiota caused by the intake of specific dietary choices favored the production of certain metabolites that may contribute to the progression of VC. The release of inflammatory and adhesion cytokines, activation of inflammatory pathways, oxidative stress, and metabolic disorders were noted to be the main mechanisms through which DI induced VC. To reduce and slow the progression of VC, emphasis should be placed on the intake of diets rich in omega-3 fatty acids, dietary fiber, Mg, Zn, and polyphenols, as well as the adjustment of dietary pattern to reduce the risk of VC. This review is expected to be useful for guiding future research on the interplay between DI and VC.
Collapse
Affiliation(s)
- Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wendi Wei
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
18
|
Tain YL, Hou CY, Tzeng HT, Lin SF, Chang-Chien GP, Lee WC, Wu KLH, Yu HR, Chan JYH, Hsu CN. Effect of Purified Resveratrol Butyrate Ester Monomers against Hypertension after Maternal High-Fructose Intake in Adult Offspring. Nutrients 2024; 16:3132. [PMID: 39339732 PMCID: PMC11435219 DOI: 10.3390/nu16183132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Offspring hypertension arising from adverse maternal conditions can be mitigated through dietary nutritional supplementation, including resveratrol. Previously, we identified derivatives of resveratrol butyrate ester (RBE), specifically 3,4'-di-O-butanoylresveratrol (ED2) and 3-O-butanoylresveratrol (ED4), demonstrating their superior antioxidant capabilities compared to RBE itself. This study sought to assess the protective impact of maternal supplementation with ED2 or ED4 on offspring hypertension in a rat model subjected to a high-fructose (HF) diet during pregnancy and lactation. METHODS Female Sprague-Dawley rats were distributed into distinct dietary groups throughout pregnancy and lactation: (1) standard chow; (2) HF diet (60%); (3) HF diet supplemented with ED2 (25 mg/L); and (4) HF diet supplemented with ED4 (25 mg/L). Male offspring were euthanized at the age of 12 weeks. RESULTS The maternal HF diet induced hypertension in the offspring, which was mitigated by perinatal supplementation with either ED2 or ED4. These protective effects were attributed to the antioxidant properties of ED2 and ED4, resulting in an increased availability of nitric oxide (NO). Additionally, supplementation with ED2 was connected to an increased abundance of Bifidobacterium and Clostridium genera, which was accompanied by a decrease in Angelakisella and Christensenella. On the other hand, ED4 supplementation shielded rat offspring from hypertension by elevating concentrations of short-chain fatty acids (SCFAs) and their receptors while reducing trimethylamine-N-oxide (TMAO) levels. CONCLUSIONS These findings highlight the potential of purified RBE monomers, ED2 and ED4, as preventive measures against hypertension resulting from a maternal high-fructose diet. Further research is warranted to explore their clinical applications based on these promising results.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (Y.-L.T.); (H.-R.Y.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (H.-T.T.); (K.L.H.W.); (J.Y.H.C.)
- College of Medicine, Chang Gung University, Taoyuan 330, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung City 811, Taiwan;
| | - Hong-Tai Tzeng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (H.-T.T.); (K.L.H.W.); (J.Y.H.C.)
| | - Shu-Fen Lin
- Super Micro Mass Research and Technology Center, Cheng Shiu University, Kaohsiung City 833, Taiwan; (S.-F.L.); (G.-P.C.-C.)
- Institute of Environmental Toxin and Emerging-Contaminant, Cheng Shiu University, Kaohsiung City 833, Taiwan
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung City 833, Taiwan
| | - Guo-Ping Chang-Chien
- Super Micro Mass Research and Technology Center, Cheng Shiu University, Kaohsiung City 833, Taiwan; (S.-F.L.); (G.-P.C.-C.)
- Institute of Environmental Toxin and Emerging-Contaminant, Cheng Shiu University, Kaohsiung City 833, Taiwan
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung City 833, Taiwan
| | - Wei-Chia Lee
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung City 833, Taiwan;
| | - Kay L. H. Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (H.-T.T.); (K.L.H.W.); (J.Y.H.C.)
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (Y.-L.T.); (H.-R.Y.)
| | - Julie Y. H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan; (H.-T.T.); (K.L.H.W.); (J.Y.H.C.)
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
| |
Collapse
|
19
|
Zhong J, Chang Y, Liang M, Zhou Y, Ai Y. Phosphorylation-amplified synchronized droplet microfluidics sensitizes bacterial growth kinetic real-time monitoring. Biosens Bioelectron 2024; 259:116397. [PMID: 38772249 DOI: 10.1016/j.bios.2024.116397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
The necessity for rapid and accurate bacterial growth monitoring is imperative across various domains, including healthcare and environmental safety. We introduce the self-synchronized droplet-amplified electrical screening cytometry (SYNC) system, a novel meld of droplet microfluidics and electrochemical amplification tailored for precise bacterial growth kinetic monitoring. SYNC encapsulates single bacteria in picolitre droplets, enabling real-time, fluorescence-free electrochemical monitoring. A specially devised phosphorylation-amplified culture medium translates bacterial metabolic activity into discernible electrical impedance changes. The dual-channel design and a rail-based structure in SYNC facilitate parallel screening and self-synchronization of droplets, addressing the limitations of conventional impedance cytometry. SYNC showcases a 5-fold enhancement in detection sensitivity and reduces 50% of the detection time compared to traditional approaches. Notably, SYNC is pioneering in providing exact initial bacterial concentrations, achieve to 104 bacteria/ml, a capability unmatched by existing real-time techniques measuring electrochemical variations. Along with its robust performance, this earmarks SYNC as a powerful tool for applications such as antibiotic susceptibility testing, food quality monitoring, and real-time water bacteria monitoring, paving the way for enhanced microbial process management and infection control.
Collapse
Affiliation(s)
- Jianwei Zhong
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore
| | - Yifu Chang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Minhui Liang
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Ye Ai
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore, 487372, Singapore.
| |
Collapse
|
20
|
Furuta S. Microbiome-Stealth Regulator of Breast Homeostasis and Cancer Metastasis. Cancers (Basel) 2024; 16:3040. [PMID: 39272898 PMCID: PMC11394247 DOI: 10.3390/cancers16173040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cumulative evidence attests to the essential roles of commensal microbes in the physiology of hosts. Although the microbiome has been a major research subject since the time of Luis Pasteur and William Russell over 140 years ago, recent findings that certain intracellular bacteria contribute to the pathophysiology of healthy vs. diseased tissues have brought the field of the microbiome to a new era of investigation. Particularly, in the field of breast cancer research, breast-tumor-resident bacteria are now deemed to be essential players in tumor initiation and progression. This is a resurrection of Russel's bacterial cause of cancer theory, which was in fact abandoned over 100 years ago. This review will introduce some of the recent findings that exemplify the roles of breast-tumor-resident microbes in breast carcinogenesis and metastasis and provide mechanistic explanations for these phenomena. Such information would be able to justify the utility of breast-tumor-resident microbes as biomarkers for disease progression and therapeutic targets.
Collapse
Affiliation(s)
- Saori Furuta
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Jung YH, Chae CW, Han HJ. The potential role of gut microbiota-derived metabolites as regulators of metabolic syndrome-associated mitochondrial and endolysosomal dysfunction in Alzheimer's disease. Exp Mol Med 2024; 56:1691-1702. [PMID: 39085351 PMCID: PMC11372123 DOI: 10.1038/s12276-024-01282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/20/2024] [Accepted: 05/10/2024] [Indexed: 08/02/2024] Open
Abstract
Although the role of gut microbiota (GMB)-derived metabolites in mitochondrial and endolysosomal dysfunction in Alzheimer's disease (AD) under metabolic syndrome remains unclear, deciphering these host-metabolite interactions represents a major public health challenge. Dysfunction of mitochondria and endolysosomal networks (ELNs) plays a crucial role in metabolic syndrome and can exacerbate AD progression, highlighting the need to study their reciprocal regulation for a better understanding of how AD is linked to metabolic syndrome. Concurrently, metabolic disorders are associated with alterations in the composition of the GMB. Recent evidence suggests that changes in the composition of the GMB and its metabolites may be involved in AD pathology. This review highlights the mechanisms of metabolic syndrome-mediated AD development, focusing on the interconnected roles of mitochondrial dysfunction, ELN abnormalities, and changes in the GMB and its metabolites. We also discuss the pathophysiological role of GMB-derived metabolites, including amino acids, fatty acids, other metabolites, and extracellular vesicles, in mediating their effects on mitochondrial and ELN dysfunction. Finally, this review proposes therapeutic strategies for AD by directly modulating mitochondrial and ELN functions through targeting GMB metabolites under metabolic syndrome.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea.
| |
Collapse
|
22
|
Zhou Y, Zhang Y, Jin S, Lv J, Li M, Feng N. The gut microbiota derived metabolite trimethylamine N-oxide: Its important role in cancer and other diseases. Biomed Pharmacother 2024; 177:117031. [PMID: 38925016 DOI: 10.1016/j.biopha.2024.117031] [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: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
An expanding body of research indicates a correlation between the gut microbiota and various diseases. Metabolites produced by the gut microbiota act as mediators between the gut microbiota and the host, interacting with multiple systems in the human body to regulate physiological or pathological functions. However, further investigation is still required to elucidate the underlying mechanisms. One such metabolite involved in choline metabolism by gut microbes is trimethylamine (TMA), which can traverse the intestinal epithelial barrier and enter the bloodstream, ultimately reaching the liver where it undergoes oxidation catalyzed by flavin-containing monooxygenase 3 (FMO3) to form trimethylamine N-oxide (TMAO). While some TMAO is eliminated through renal excretion, remaining amounts circulate in the bloodstream, leading to systemic inflammation, endoplasmic reticulum (ER) stress, mitochondrial stress, and disruption of normal physiological functions in humans. As a representative microbial metabolite originating from the gut, TMAO has significant potential both as a biomarker for monitoring disease occurrence and progression and for tailoring personalized treatment strategies for patients. This review provides an extensive overview of TMAO sources and its metabolism in human blood, as well as its impact on several major human diseases. Additionally, we explore the latest research areas related to TMAO along with future directions.
Collapse
Affiliation(s)
- Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuwei Zhang
- Nantong University Medical School, Nantong, China
| | - Shengkai Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jing Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Menglu Li
- Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China; Nantong University Medical School, Nantong, China; Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| |
Collapse
|
23
|
Martin-Grau M, Monleón D. The Role of Microbiota-Related Co-Metabolites in MASLD Progression: A Narrative Review. Curr Issues Mol Biol 2024; 46:6377-6389. [PMID: 39057023 PMCID: PMC11276081 DOI: 10.3390/cimb46070381] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a growing health concern due to its increasing prevalence worldwide. Metabolic homeostasis encompasses the stable internal conditions vital for efficient metabolism. This equilibrium extends to the intestinal microbiota, whose metabolic activities profoundly influence overall metabolic balance and organ health. The metabolites derived from the gut microbiota metabolism can be defined as microbiota-related co-metabolites. They serve as mediators between the gut microbiota and the host, influencing various physiological processes. The recent redefinition of the term MASLD has highlighted the metabolic dysfunction that characterize the disease. Metabolic dysfunction encompasses a spectrum of abnormalities, including impaired glucose regulation, dyslipidemia, mitochondrial dysfunction, inflammation, and accumulation of toxic byproducts. In addition, MASLD progression has been linked to dysregulation in the gut microbiota and associated co-metabolites. Short-chain fatty acids (SCFAs), hippurate, indole derivatives, branched-chain amino acids (BCAAs), and bile acids (BAs) are among the key co-metabolites implicated in MASLD progression. In this review, we will unravel the relationship between the microbiota-related metabolites which have been associated with MASLD and that could play an important role for developing effective therapeutic interventions for MASLD and related metabolic disorders.
Collapse
Affiliation(s)
- Maria Martin-Grau
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
- University Clinical Hospital of Valencia Research Foundation (INCLIVA), 46010 Valencia, Spain
| | - Daniel Monleón
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
- University Clinical Hospital of Valencia Research Foundation (INCLIVA), 46010 Valencia, Spain
| |
Collapse
|
24
|
Martins D, Silva C, Ferreira AC, Dourado S, Albuquerque A, Saraiva F, Batista AB, Castro P, Leite-Moreira A, Barros AS, Miranda IM. Unravelling the Gut Microbiome Role in Cardiovascular Disease: A Systematic Review and a Meta-Analysis. Biomolecules 2024; 14:731. [PMID: 38927134 PMCID: PMC11201797 DOI: 10.3390/biom14060731] [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: 04/26/2024] [Revised: 06/04/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
A notable shift in understanding the human microbiome's influence on cardiovascular disease (CVD) is underway, although the causal association remains elusive. A systematic review and meta-analysis were conducted to synthesise current knowledge on microbial taxonomy and metabolite variations between healthy controls (HCs) and those with CVD. An extensive search encompassing three databases identified 67 relevant studies (2012-2023) covering CVD pathologies from 4707 reports. Metagenomic and metabolomic data, both qualitative and quantitative, were obtained. Analysis revealed substantial variability in microbial alpha and beta diversities. Moreover, specific changes in bacterial populations were shown, including increased Streptococcus and Proteobacteria and decreased Faecalibacterium in patients with CVD compared with HC. Additionally, elevated trimethylamine N-oxide levels were reported in CVD cases. Biochemical parameter analysis indicated increased fasting glucose and triglycerides and decreased total cholesterol and low- and high-density lipoprotein cholesterol levels in diseased individuals. This study revealed a significant relationship between certain bacterial species and CVD. Additionally, it has become clear that there are substantial inconsistencies in the methodologies employed and the reporting standards adhered to in various studies. Undoubtedly, standardising research methodologies and developing extensive guidelines for microbiome studies are crucial for advancing the field.
Collapse
Affiliation(s)
- Diana Martins
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Cláudia Silva
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - António Carlos Ferreira
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Sara Dourado
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ana Albuquerque
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Francisca Saraiva
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ana Beatriz Batista
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Pedro Castro
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Neurology, São João Hospital Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Adelino Leite-Moreira
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - António S. Barros
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Isabel M. Miranda
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
25
|
Vallianou NG, Kounatidis D, Psallida S, Panagopoulos F, Stratigou T, Geladari E, Karampela I, Tsilingiris D, Dalamaga M. The Interplay Between Dietary Choline and Cardiometabolic Disorders: A Review of Current Evidence. Curr Nutr Rep 2024; 13:152-165. [PMID: 38427291 PMCID: PMC11133147 DOI: 10.1007/s13668-024-00521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE OF REVIEW Choline is an essential nutrient for human health and cellular homeostasis as it is necessary for the synthesis of lipid cell membranes, lipoproteins, and the synthesis of the neurotransmitter acetylcholine. The aim of this review is to analyze the beneficial effects of choline and its significance in cellular metabolism and various inflammatory pathways, such as the inflammasome. We will discuss the significance of dietary choline in cardiometabolic disorders, such as non-alcoholic fatty liver disease (NAFLD), cardiovascular disease (CVD), and chronic kidney disease (CKD) as well as in cognitive function and associated neuropsychiatric disorders. RECENT FINDINGS Choline deficiency has been related to the development of NAFLD and cognitive disability in the offspring as well as in adulthood. In sharp contrast, excess dietary intake of choline mediated via the increased production of trimethylamine by the gut microbiota and increased trimethylamine-N-oxide (TMAO) levels has been related to atherosclerosis in most studies. In this context, CVD and CKD through the accumulation of TMAO, p-Cresyl-sulfate (pCS), and indoxyl-sulfate (IS) in serum may be the result of the interplay between excess dietary choline, the increased production of TMAO by the gut microbiota, and the resulting activation of inflammatory responses and fibrosis. A balanced diet, with no excess nor any deficiency in dietary choline, is of outmost importance regarding the prevention of cardiometabolic disorders as well as cognitive function. Large-scale studies with the use of next-generation probiotics, especially Akkermansia muciniphila and Faecalibacterium prausnitzii, should further examine their therapeutic potential in this context.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece.
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece.
| | - Dimitris Kounatidis
- Department of Internal Medicine, Hippokration General Hospital, 114 Vassilissis Sofias str, Athens, Greece
| | - Sotiria Psallida
- Department of Microbiology, KAT General Hospital of Attica, 2 Nikis str, Athens, Greece
| | - Fotis Panagopoulos
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini str, Athens, Greece
| | - Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100, Alexandroupoli, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece.
| |
Collapse
|
26
|
Lee J, Lee J, Kim K, Lee J, Jung Y, Hyeon JS, Seo A, Jin W, Weon B, Shin N, Kim S, Lim CS, Kim YS, Lee JP, Hwang GS, Yang SH. Antibiotic-induced intestinal microbiota depletion can attenuate the acute kidney injury to chronic kidney disease transition via NADPH oxidase 2 and trimethylamine-N-oxide inhibition. Kidney Int 2024; 105:1239-1253. [PMID: 38431216 DOI: 10.1016/j.kint.2024.01.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
Intestinal microbiota and their metabolites affect systemic inflammation and kidney disease outcomes. Here, we investigated the key metabolites associated with the acute kidney injury (AKI)-to chronic kidney disease (CKD) transition and the effect of antibiotic-induced microbiota depletion (AIMD) on this transition. In 61 patients with AKI, 59 plasma metabolites were assessed to determine the risk of AKI-to-CKD transition. An AKI-to-CKD transition murine model was established four weeks after unilateral ischemia-reperfusion injury (IRI) to determine the effects of AIMD on the gut microbiome, metabolites, and pathological responses related to CKD transition. Human proximal tubular epithelial cells were challenged with CKD transition-related metabolites, and inhibitory effects of NADPH oxidase 2 (NOX2) signals were tested. Based on clinical metabolomics, plasma trimethylamine N-oxide (TMAO) was associated with a significantly increased risk for AKI-to-CKD transition [adjusted odds ratio 4.389 (95% confidence interval 1.106-17.416)]. In vivo, AIMD inhibited a unilateral IRI-induced increase in TMAO, along with a decrease in apoptosis, inflammation, and fibrosis. The expression of NOX2 and oxidative stress decreased after AIMD. In vitro, TMAO induced fibrosis with NOX2 activation and oxidative stress. NOX2 inhibition successfully attenuated apoptosis, inflammation, and fibrosis with suppression of G2/M arrest. NOX2 inhibition (in vivo) showed improvement in pathological changes with a decrease in oxidative stress without changes in TMAO levels. Thus, TMAO is a key metabolite associated with the AKI-to-CKD transition, and NOX2 activation was identified as a key regulator of TMAO-related AKI-to-CKD transition both in vivo and in vitro.
Collapse
Affiliation(s)
- Jeonghwan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jinhaeng Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Kyuhong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jiwon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jin Seong Hyeon
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Areum Seo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Wencheng Jin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Boram Weon
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Nayeon Shin
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea.
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Kidney Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Liu S, Li W, Chen J, Li M, Geng Y, Liu Y, Wu W. The footprint of gut microbiota in gallbladder cancer: a mechanistic review. Front Cell Infect Microbiol 2024; 14:1374238. [PMID: 38774627 PMCID: PMC11106419 DOI: 10.3389/fcimb.2024.1374238] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Gallbladder cancer (GBC) is the most common malignant tumor of the biliary system with the worst prognosis. Even after radical surgery, the majority of patients with GBC have difficulty achieving a clinical cure. The risk of tumor recurrence remains more than 65%, and the overall 5-year survival rate is less than 5%. The gut microbiota refers to a variety of microorganisms living in the human intestine, including bacteria, viruses and fungi, which profoundly affect the host state of general health, disease and even cancer. Over the past few decades, substantial evidence has supported that gut microbiota plays a critical role in promoting the progression of GBC. In this review, we summarize the functions, molecular mechanisms and recent advances of the intestinal microbiota in GBC. We focus on the driving role of bacteria in pivotal pathways, such as virulence factors, metabolites derived from intestinal bacteria, chronic inflammatory responses and ecological niche remodeling. Additionally, we emphasize the high level of correlation between viruses and fungi, especially EBV and Candida spp., with GBC. In general, this review not only provides a solid theoretical basis for the close relationship between gut microbiota and GBC but also highlights more potential research directions for further research in the future.
Collapse
Affiliation(s)
- Shujie Liu
- Joint Program of Nanchang University and Queen Mary University of London, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Weijian Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Jun Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Maolan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Yajun Geng
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| | - Wenguang Wu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Shanghai, China
| |
Collapse
|
28
|
Zhou F, Zhang Q, Zheng X, Shi F, Ma K, Ji F, Meng N, Li R, Lv J, Li Q. Antiaging Effects of Human Fecal Transplants with Different Combinations of Bifidobacterium bifidum LTBB21J1 and Lactobacillus casei LTL1361 in d-Galactose-Induced Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:9818-9827. [PMID: 38647087 DOI: 10.1021/acs.jafc.3c09815] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The feces of healthy middle-aged and old people were first transplanted into d-galactose-induced aging mice to construct humanized aging mice with gut microbiota (FMTC) to confirm the antiaging effect of probiotics produced from centenarians. The mouse model was then treated with centenarian-derived Bifidobacterium bifidum (FMTL), Lactobacillus casei (FMTB), and their mixtures (FMTM), and young mice were used as the control. Compared with the FMTC group, the results demonstrated that the probiotics and their combinations alleviated neuronal damage, increased antioxidant capacity, decreased inflammation, and enhanced cognitive and memory functions in aging mice. In the gut microbiota, the relative abundance of Lactobacillus, Ligilactobacillus, and Akkermansia increased and that of Desulfovibrio and Colidextribacter decreased in the FMTM group compared with that in the FMTC group. The three probiotic groups displayed significant changes in 15 metabolites compared with the FMTC group, with 4 metabolites showing increased expression and 11 metabolites showing decreased expression. The groups were graded as Control > FMTM > FMTB > FMTL > FMTC using a newly developed comprehensive quantitative scoring system that thoroughly analyzed the various indicators of this study. The beneficial antiaging effects of probiotics derived from centenarians were quantitatively described using a novel perspective in this study; it is confirmed that both probiotics and their combinations exert antiaging effects, with the probiotic complex group exhibiting a larger effect.
Collapse
Affiliation(s)
- Fan Zhou
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Qinren Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Xiaohua Zheng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Fengcui Shi
- School of Chemical and Biological Engineering, Qilu Institute of Technology, Shandong 250200, China
| | - Kai Ma
- Jiangsu New-Bio Biotechnology Co.,Ltd, Jiangsu 214400, China
| | - Feng Ji
- Jiangsu New-Bio Biotechnology Co.,Ltd, Jiangsu 214400, China
| | - Ning Meng
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Ruiding Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jingwen Lv
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Quanyang Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| |
Collapse
|
29
|
Cicchinelli S, Gemma S, Pignataro G, Piccioni A, Ojetti V, Gasbarrini A, Franceschi F, Candelli M. Intestinal Fibrogenesis in Inflammatory Bowel Diseases: Exploring the Potential Role of Gut Microbiota Metabolites as Modulators. Pharmaceuticals (Basel) 2024; 17:490. [PMID: 38675450 PMCID: PMC11053610 DOI: 10.3390/ph17040490] [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: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Fibrosis, sustained by the transformation of intestinal epithelial cells into fibroblasts (epithelial-to-mesenchymal transition, EMT), has been extensively studied in recent decades, with the molecular basis well-documented in various diseases, including inflammatory bowel diseases (IBDs). However, the factors influencing these pathways remain unclear. In recent years, the role of the gut microbiota in health and disease has garnered significant attention. Evidence suggests that an imbalanced or dysregulated microbiota, along with environmental and genetic factors, may contribute to the development of IBDs. Notably, microbes produce various metabolites that interact with host receptors and associated signaling pathways, influencing physiological and pathological changes. This review aims to present recent evidence highlighting the emerging role of the most studied metabolites as potential modulators of molecular pathways implicated in intestinal fibrosis and EMT in IBDs. These studies provide a deeper understanding of intestinal inflammation and fibrosis, elucidating the molecular basis of the microbiota role in IBDs, paving the way for future treatments.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
30
|
Khan S, Ahmad F, Khalid N. Applications of Strain-Specific Probiotics in the Management of Cardiovascular Diseases: A Systemic Review. Mol Nutr Food Res 2024; 68:e2300675. [PMID: 38549453 DOI: 10.1002/mnfr.202300675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/14/2024] [Indexed: 05/08/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of global mortality and novel approaches for prevention and management are needed. The human gastrointestinal tract hosts a diverse microbiota that is crucial in maintaining metabolic homeostasis. The formulation of effective probiotics, alone or in combination, has been under discussion due to their impact on cardiovascular and metabolic diseases. Probiotics have been shown to impact cardiovascular health positively. An imbalance in the presence of Firmicutes and Bacteroidetes has been linked to the progression of CVDs due to their impact on bile acid and cholesterol metabolism. The probiotics primarily help in the reduction of plasma low-density lipoprotein levels and attenuation of the proinflammatory markers. These beneficial microorganisms contribute to lowering cholesterol levels and produce essential short-chain fatty acids. The impact of lipid-regulating probiotic strains on human health is quite significant. However, only a few have been tested for potential beneficial efficacy, and ambiguity exists regarding strain dosages, interactions with confounding factors, and potential adverse effects. Hence, more comprehensive studies and randomized trials are needed to understand the mechanisms of probiotics on CVDs and to ensure human health. This review assesses the evidence and highlights the roles of strain-specific probiotics in the management of CVDs.
Collapse
Affiliation(s)
- Saleha Khan
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Nauman Khalid
- Department of Human Nutrition and Dietetics, School of Food and Agricultural Sciences, University of Management and Technology, Lahore, 54000, Pakistan
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, 59911, United Arab Emirates
| |
Collapse
|
31
|
Gao Y, Huang R, Qiu Y, Liu Y, Chen L. Characterization of the chemical composition of different parts of Dolichos lablab L. and revelation of its anti-ulcerative colitis effects by modulating the gut microbiota and host metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117629. [PMID: 38135234 DOI: 10.1016/j.jep.2023.117629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a non-specific inflammatory disease characterized by long duration and easy relapse. Dolichos lablab L. (DLL) belongs to the family Fabaceae, was listed in a famous Chinese medical classic, Compendium of Materia Medic, and described as possessing features that invigorate the spleen, alleviate dampness, provide diarrhea relief, and other effects. The DLL-dried white mature seeds (DS) and dried flower (DF), which hold significant medicinal value in China, were used in clinical prescriptions to prevent and treat UC. DS and DF have appeared in different editions of the Pharmacopoeia of the People's Republic of China from 1977 to 2020. However, their chemical composition, pharmacological effects, and mechanism of treating UC are unclear. AIM OF THE STUDY This study aimed to characterize the chemical composition of different parts of DLL (seeds and flowers), further explore their pharmacological effects, and elaborate its underlying mechanism of treating UC. METHODS The chemical composition of DS and DF crude polysaccharides (DSP and DFP) and ethanolic extracts (DSE and DFE) were characterized by high-performance anion-exchange chromatography (HPAEC), ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS), and gas chromatography-mass spectrometry (GC-MS). Then, based on the acute UC mice model, the pharmacodynamic effects were investigated by Western blotting, ELISA, and other methods. Finally, the 16S rRNA gene sequencing and metabonomic analysis were used to explore the regulatory effects of DS and DF on intestinal microbiota and host metabolism. RESULTS DSE and DFE inhibited the oxidative stress response, reducing proinflammatory factor production and maintaining intestinal barrier integrity in UC mice. The 16S rRNA gene sequencing and metabonomic analysis revealed that DS and DF treated UC by regulating the intestinal microbiota structure and reversing the abnormal metabolism of the host. CONCLUSION This study suggested that different parts of DLL (flowers and seeds) may be potential medicines for treating UC, which exert their therapeutic effects through various active ingredients and might contribute significantly to reducing the economic pressures and challenges of UC treatment worldwide.
Collapse
Affiliation(s)
- Yanping Gao
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ruiting Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yongyi Qiu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
32
|
Cardoso AM. Microbial influence on blood pressure: unraveling the complex relationship for health insights. MICROBIOME RESEARCH REPORTS 2024; 3:22. [PMID: 38841410 PMCID: PMC11149090 DOI: 10.20517/mrr.2023.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 06/07/2024]
Abstract
Hypertension, a critical global health concern, is characterized by persistent high blood pressure and is a major cause of cardiovascular events. This perspective explores the multifaceted implications of hypertension, its association with cardiovascular diseases, and the emerging role of the gut microbiota. The gut microbiota, a dynamic community in the gastrointestinal tract, plays a pivotal role in hypertension by influencing blood pressure through the generation of antioxidant, anti-inflammatory, and short-chain fatty acids metabolites, and the conversion of nitrates into nitric oxide. Antihypertensive medications interact with the gut microbiota, impacting drug pharmacokinetics and efficacy. Prebiotics and probiotics present promising avenues for hypertension management, with prebiotics modulating blood pressure through lipid and cholesterol modulation, and probiotics exhibiting a general beneficial effect. Personalized choices based on individual factors are crucial for optimizing prebiotic and probiotic interventions. In conclusion, the gut microbiota's intricate influence on blood pressure regulation offers innovative perspectives in hypertension therapeutics, with targeted strategies proving valuable for holistic blood pressure management and health promotion.
Collapse
|
33
|
Satheesh Babu AK, Petersen C, Iglesias-Carres L, Paz HA, Wankhade UD, Neilson AP, Anandh Babu PV. Blueberry intervention mitigates detrimental microbial metabolite trimethylamine N-oxide by modulating gut microbes. Biofactors 2024; 50:392-404. [PMID: 37921575 PMCID: PMC11014767 DOI: 10.1002/biof.2014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/15/2023] [Indexed: 11/04/2023]
Abstract
Gut microbes play a pivotal role in host physiology by producing beneficial or detrimental metabolites. Gut bacteria metabolize dietary choline and L-carnitine to trimethylamine (TMA) which is then converted to trimethylamine-N-oxide (TMAO). An elevated circulating TMAO is associated with diabetes, obesity, cardiovascular disease, and cancer in humans. In the present study, we investigated the effect of dietary blueberries and strawberries at a nutritional dosage on TMA/TMAO production and the possible role of gut microbes. Blueberry cohort mice received a control (C) or freeze-dried blueberry supplemented (CB) diet for 12 weeks and subgroups received an antibiotics cocktail (CA and CBA). Strawberry cohort mice received a control (N) or strawberry-supplemented (NS) diet and subgroups received antibiotics (NA and NSA). Metabolic parameters, choline, TMA, and TMAO were assessed in addition to microbial profiling and characterization of berry powders. Blueberry supplementation (equivalent to 1.5 human servings) reduced circulating TMAO in CB versus C mice (~48%) without changing choline or TMA. This effect was not mediated through alterations in metabolic parameters. Dietary strawberries did not reduce choline, TMA, or TMAO. Depleting gut microbes with antibiotics in these cohorts drastically reduced TMA and TMAO to not-quantified levels. Further, dietary blueberries increased the abundance of bacterial taxa that are negatively associated with circulating TMA/TMAO suggesting the role of gut microbes. Our phenolic profiling indicates that this effect could be due to chlorogenic acid and increased phenolic contents in blueberries. Our study provides evidence for considering dietary blueberries to reduce TMAO and prevent TMAO-induced complications.
Collapse
Affiliation(s)
| | - Chrissa Petersen
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| | - Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC 28081, USA
| | - Henry A. Paz
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Umesh D. Wankhade
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Andrew P. Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC 28081, USA
| | - Pon Velayutham Anandh Babu
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
34
|
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
|
35
|
Getachew B, Hauser SR, Bennani S, El Kouhen N, Sari Y, Tizabi Y. Adolescent alcohol drinking interaction with the gut microbiome: implications for adult alcohol use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2024; 4:11881. [PMID: 38322648 PMCID: PMC10846679 DOI: 10.3389/adar.2024.11881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024]
Abstract
Reciprocal communication between the gut microbiota and the brain, commonly referred to as the "gut-brain-axis" is crucial in maintaining overall physiological homeostasis. Gut microbiota development and brain maturation (neuronal connectivity and plasticity) appear to be synchronized and to follow the same timeline during childhood (immature), adolescence (expansion) and adulthood (completion). It is important to note that the mesolimbic reward circuitry develops early on, whereas the maturation of the inhibitory frontal cortical neurons is delayed. This imbalance can lead to increased acquirement of reward-seeking and risk-taking behaviors during adolescence, and consequently eventuate in heightened risk for substance abuse. Thus, there is high initiation of alcohol drinking in early adolescence that significantly increases the risk of alcohol use disorder (AUD) in adulthood. The underlying causes for heightened AUD risk are not well understood. It is suggested that alcohol-associated gut microbiota impairment during adolescence plays a key role in AUD neurodevelopment in adulthood. Furthermore, alcohol-induced dysregulation of microglia, either directly or indirectly through interaction with gut microbiota, may be a critical neuroinflammatory pathway leading to neurodevelopmental impairments and AUD. In this review article, we highlight the influence of adolescent alcohol drinking on gut microbiota, gut-brain axis and microglia, and eventual manifestation of AUD. Furthermore, novel therapeutic interventions via gut microbiota manipulations are discussed briefly.
Collapse
Affiliation(s)
- Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
36
|
Meng C, Wang X, Fan L, Fan Y, Yan Z, Wang Y, Li Y, Zhang J, Lv S. A new perspective in the prevention and treatment of antitumor therapy-related cardiotoxicity: Intestinal microecology. Biomed Pharmacother 2024; 170:115588. [PMID: 38039758 DOI: 10.1016/j.biopha.2023.115588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 12/03/2023] Open
Abstract
The continuous development of antitumor therapy has significantly reduced the mortality of patients with malignancies. However, the antitumor-related cardiotoxicity has become the leading cause of long-term mortality in patients with malignancies. Besides, the pathogenesis of antitumor-related cardiotoxicity is still unclear, and practical means of prevention and treatment are lacking in clinical practice. Therefore, the major challenge is how to combat the cardiotoxicity of antitumor therapy effectively. More and more studies have shown that antitumor therapy kills tumor cells while causing damage to sensitive tissues such as the intestinal mucosa, leading to the increased permeability of the intestine and the dysbiosis of intestinal microecology. In addition, the dysbiosis of intestinal microecology contributes to the development and progression of cardiovascular diseases through multiple pathways. Thus, the dysbiosis of intestinal microecology may be a potential mechanism and target for antitumor-related cardiotoxicity. We summarized the characteristics of intestinal microecology disorders induced by antitumor therapy and the association between intestinal microecological dysbiosis and CVD. And on this basis, we hypothesized the potential mechanisms of intestinal microecology mediating the occurrence of antitumor-related cardiotoxicity. Then we reviewed the previous studies targeting intestinal microecology against antitumor-associated cardiotoxicity, aiming to provide a reference for future studies on the occurrence and prevention of antitumor-related cardiotoxicity by intestinal microecology.
Collapse
Affiliation(s)
- Chenchen Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Xiaoming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Lu Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Yajie Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Zhipeng Yan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Yanyang Li
- Department of integrated Chinese and Western medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China.
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China.
| |
Collapse
|
37
|
Kim K, Jang H, Kim E, Kim H, Sung GY. Recent advances in understanding the role of the skin microbiome in the treatment of atopic dermatitis. Exp Dermatol 2023; 32:2048-2061. [PMID: 37767872 DOI: 10.1111/exd.14940] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023]
Abstract
The skin is the largest organ in the human body, and histologically consists of the epidermis, dermis and subcutaneous tissue. Humans maintain a cooperative symbiotic relationship with their skin microbiota, a complex community of bacteria, fungi and viruses that live on the surface of the skin, and which act as a barrier to protect the body from the inside and outside. The skin is a 'habitat' and vast 'ecosystem' inhabited by countless microbes; as such, relationships have been forged through millions of years of coevolution. It is not surprising then that microbes are key participants in shaping and maintaining essential physiological processes. In addition to maintaining barrier function, the unique symbiotic microbiota that colonizes the skin increases the immune response and provides protection against pathogenic microbes. This review examines our current understanding of skin microbes in shaping and enhancing the skin barrier, as well as skin microbiome-host interactions and their roles in skin diseases, such as atopic dermatitis (AD). We also report on the current status of AD therapeutic drugs that target the skin microbiome, related research on current therapeutic strategies, and the limitations and future considerations of skin microbiome research. In particular, as a future strategy, we discuss the need for a skin-on-a-chip-based microphysiological system research model amenable to biomimetic in vitro studies and human skin equivalent models, including skin appendages.
Collapse
Affiliation(s)
- Kyunghee Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeji Jang
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Eunyul Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeju Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
- Major in Materials Science and Engineering, Hallym University, Chuncheon, Korea
| |
Collapse
|
38
|
Zong Y, Wang X, Wang J. Research progress on the correlation between gut microbiota and preeclampsia: microbiome changes, mechanisms and treatments. Front Cell Infect Microbiol 2023; 13:1256940. [PMID: 38029244 PMCID: PMC10644267 DOI: 10.3389/fcimb.2023.1256940] [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: 07/11/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Preeclampsia is a specific disease during pregnancy and is a significant factor in the increased mortality in perinatal women. Gut microbiota, an intricate and abundant microbial community in the digestive tract, is crucial for host metabolism, immunity, and nutrient absorption. The onset and progression of preeclampsia are closely correlated with the changes in maternal gut microbiota. Research purpose was to compile the existing bits of present scientific data and to close the gap in the knowledge of changes in gut microbiota in preeclampsia and their association with preeclampsia. We searched studies from two electronic databases (PubMed and Web of Science) included from 2014 to 2023. This review is divided into three parts. In the first part, the author elaborates longitudinal differences of maternal gut microbiota during different gestation periods. In the second part, we discuss that gut microbiota can lead to the occurrence of preeclampsia by systemic immune response, influencing the release of active peptides, short-chain fatty acids, trimethylamine-N-oxide (TMAO) and other metabolites, vascular factors and Microorganism-immune axis. In the third part, we proposed that a high-fiber diet combined with drugs and microecological regulators may be therapeutic in enhancing or preventing the emergence and evolution of preeclampsia, which needs further exploration. Although the pathogenesis of preeclampsia is still nebulous and there is no clear and valid clinical treatment, our study provides new ideas for the pathogenesis, prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
- Yichi Zong
- Department of Obstetrics and Gynecology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuguang Wang
- Sun Yatsen University Cancer Center, Guangzhou, Guangdong, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Shengjing Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
39
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
40
|
Andrikopoulos P, Aron-Wisnewsky J, Chakaroun R, Myridakis A, Forslund SK, Nielsen T, Adriouch S, Holmes B, Chilloux J, Vieira-Silva S, Falony G, Salem JE, Andreelli F, Belda E, Kieswich J, Chechi K, Puig-Castellvi F, Chevalier M, Le Chatelier E, Olanipekun MT, Hoyles L, Alves R, Helft G, Isnard R, Køber L, Coelho LP, Rouault C, Gauguier D, Gøtze JP, Prifti E, Froguel P, Zucker JD, Bäckhed F, Vestergaard H, Hansen T, Oppert JM, Blüher M, Nielsen J, Raes J, Bork P, Yaqoob MM, Stumvoll M, Pedersen O, Ehrlich SD, Clément K, Dumas ME. Evidence of a causal and modifiable relationship between kidney function and circulating trimethylamine N-oxide. Nat Commun 2023; 14:5843. [PMID: 37730687 PMCID: PMC10511707 DOI: 10.1038/s41467-023-39824-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 06/30/2023] [Indexed: 09/22/2023] Open
Abstract
The host-microbiota co-metabolite trimethylamine N-oxide (TMAO) is linked to increased cardiovascular risk but how its circulating levels are regulated remains unclear. We applied "explainable" machine learning, univariate, multivariate and mediation analyses of fasting plasma TMAO concentration and a multitude of phenotypes in 1,741 adult Europeans of the MetaCardis study. Here we show that next to age, kidney function is the primary variable predicting circulating TMAO, with microbiota composition and diet playing minor, albeit significant, roles. Mediation analysis suggests a causal relationship between TMAO and kidney function that we corroborate in preclinical models where TMAO exposure increases kidney scarring. Consistent with our findings, patients receiving glucose-lowering drugs with reno-protective properties have significantly lower circulating TMAO when compared to propensity-score matched control individuals. Our analyses uncover a bidirectional relationship between kidney function and TMAO that can potentially be modified by reno-protective anti-diabetic drugs and suggest a clinically actionable intervention for decreasing TMAO-associated excess cardiovascular risk.
Collapse
Affiliation(s)
- Petros Andrikopoulos
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Section of Genomic & Environmental Medicine, National Heart & Lung Institute, Imperial College London, London, UK.
| | - Judith Aron-Wisnewsky
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Rima Chakaroun
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antonis Myridakis
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Environmental Research Group, MRC Centre for Environment and Health, School of Public Health, Imperial College London, 86 Wood Lane, London, W12 0BZ, UK
| | - Sofia K Forslund
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin and the Max-Delbrück Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité University Hospital, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Solia Adriouch
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
| | | | - Julien Chilloux
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Joe-Elie Salem
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Fabrizio Andreelli
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Eugeni Belda
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Sorbonne Université, IRD, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, F-93143, Bondy, France
| | - Julius Kieswich
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kanta Chechi
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Section of Genomic & Environmental Medicine, National Heart & Lung Institute, Imperial College London, London, UK
| | - Francesc Puig-Castellvi
- European Genomics Institute for Diabetes, EGENODIA, INSERM U1283, CNRS UMR8199, Institut Pasteur de Lille, Lille University Hospital, University of Lille, Lille, France
| | - Mickael Chevalier
- European Genomics Institute for Diabetes, EGENODIA, INSERM U1283, CNRS UMR8199, Institut Pasteur de Lille, Lille University Hospital, University of Lille, Lille, France
| | | | - Michael T Olanipekun
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Lesley Hoyles
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Renato Alves
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Gerard Helft
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
- Institute of Cardiometabolism and Nutrition, ICAN, INSERM, 1166, Paris, France
| | - Richard Isnard
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Luis Pedro Coelho
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christine Rouault
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
| | - Dominique Gauguier
- INSERM UMR 1124, Université de Paris, 45 rue des Saint-Pères, 75006, Paris, France
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Jens Peter Gøtze
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Edi Prifti
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Sorbonne Université, IRD, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, F-93143, Bondy, France
| | - Philippe Froguel
- European Genomics Institute for Diabetes, EGENODIA, INSERM U1283, CNRS UMR8199, Institut Pasteur de Lille, Lille University Hospital, University of Lille, Lille, France
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Jean-Daniel Zucker
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France
- Sorbonne Université, IRD, Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, F-93143, Bondy, France
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Vestergaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Bornholms Hospital, Rønne, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jean-Michel Oppert
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
| | - Peer Bork
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, 03722, South Korea
| | - Muhammad M Yaqoob
- Diabetic Kidney Disease Centre, Renal Unit, Barts Health National Health Service Trust, The Royal London Hospital, London, UK
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Stumvoll
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte University Hospital, Copenhagen, Denmark
| | - S Dusko Ehrlich
- Department of Clinical and Movement Neurosciences, University College London, London, NW3 2PF, UK
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and obesities; systemic approaches (NutriOmics), Paris, France.
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France.
| | - Marc-Emmanuel Dumas
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Section of Genomic & Environmental Medicine, National Heart & Lung Institute, Imperial College London, London, UK.
- European Genomics Institute for Diabetes, EGENODIA, INSERM U1283, CNRS UMR8199, Institut Pasteur de Lille, Lille University Hospital, University of Lille, Lille, France.
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC, H3A 0G1, Canada.
| |
Collapse
|
41
|
Volmer JG, McRae H, Morrison M. The evolving role of methanogenic archaea in mammalian microbiomes. Front Microbiol 2023; 14:1268451. [PMID: 37727289 PMCID: PMC10506414 DOI: 10.3389/fmicb.2023.1268451] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
Methanogenic archaea (methanogens) represent a diverse group of microorganisms that inhabit various environmental and host-associated microbiomes. These organisms play an essential role in global carbon cycling given their ability to produce methane, a potent greenhouse gas, as a by-product of their energy production. Recent advances in culture-independent and -dependent studies have highlighted an increased prevalence of methanogens in the host-associated microbiome of diverse animal species. Moreover, there is increasing evidence that methanogens, and/or the methane they produce, may play a substantial role in human health and disease. This review addresses the expanding host-range and the emerging view of host-specific adaptations in methanogen biology and ecology, and the implications for host health and disease.
Collapse
Affiliation(s)
- James G. Volmer
- Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba, QLD, Australia
| | - Harley McRae
- Faculty of Medicine, University of Queensland Frazer Institute, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Mark Morrison
- Faculty of Medicine, University of Queensland Frazer Institute, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
42
|
Basnet TB, GC S, Basnet R, Fatima S, Safdar M, Sehar B, Alsubaie ASR, Zeb F. Interaction between gut microbiota metabolites and dietary components in lipid metabolism and metabolic diseases. Access Microbiol 2023; 5:acmi000403. [PMID: 37424550 PMCID: PMC10323789 DOI: 10.1099/acmi.0.000403] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/04/2023] [Indexed: 07/11/2023] Open
Abstract
Gut microbiota composition has caused perplexity in developing precision therapy to cure metabolic disorders. However, recent research has focused on using daily diet and natural bioactive compounds to correct gut microbiota dysbiosis and regulate host metabolism. Complex interactions between the gut microbiota and dietary compounds disrupt or integrate the gut barrier and lipid metabolism. In this review, we investigate the role of diet and bioactive natural compounds in gut microbiota dysbiosis and also the modulation of lipid metabolism by their metabolites. Recent studies have revealed that diet, natural compounds and phytochemicals impact significantly on lipid metabolism in animals and humans. These findings suggest that dietary components or natural bioactive compounds have a significant impact on microbial dysbiosis linked to metabolic diseases. The interaction between dietary components or natural bioactive compounds and gut microbiota metabolites can regulate lipid metabolism. Additionally, natural products can shape the gut microbiota and improve barrier integrity by interacting with gut metabolites and their precursors, even in unfavourable conditions, potentially contributing to the alignment of host physiology.
Collapse
Affiliation(s)
- Til Bahadur Basnet
- Department of Epidemiology and Biostatistics, School of Public Health, Fujian Medical University, Fuzhou, PR China
| | - Srijana GC
- Kanti Children’s Hospital, Kathmandu, Nepal
| | - Rajesh Basnet
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, PR China
| | - Sadia Fatima
- Department of Biochemistry, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Mahpara Safdar
- Department of Environmental Design, Health and Nutritional Sciences, Allama Iqbal Open University, Islamabad, Pakistan
| | - Bismillah Sehar
- Department of Health and Social Sciences, University of Bedfordshire, Bedford, UK
| | - Ali Saad R. Alsubaie
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Falak Zeb
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, UAE
| |
Collapse
|
43
|
Wei Y, Zhou M, Fang W, Liu Q, Mao H, Chen B, Zhang T, Xu Y, Zhang W, Zheng Y, Hu X. Differences in the luminal and mucosal gut microbiomes and metabolomes of oriental rat snake (Ptyas mucosus). Appl Microbiol Biotechnol 2023; 107:3257-3271. [PMID: 37071138 DOI: 10.1007/s00253-023-12524-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Previous studies regarding the gastrointestinal biogeography of microbiomes generally focused on longitudinal comparisons, whereas few studies have compared luminal and mucosal microbiomes. Investigations of the snake gut microbiome have attracted interest because of the unique digestive physiology and hibernation behavior, but adequate sampling methods must be developed. Here, we used an omics approach combining 16S rRNA gene sequencing with untargeted metabolomics to profile the luminal and mucosal gut microbiomes and metabolomes in oriental rat snakes, with the goal of revealing the heterogeneity and co-occurrence at these sites. The α-diversity of the gut microbiome was significantly higher at mucosal sites than at luminal sites. Microbial composition also differed according to sampling site, with significant differences in the abundances of dominant phyla and genera, as well as β-diversity clustering and distribution. Metabolome profiling revealed differences that were mainly related to cholinergic substances and nucleic acids. Analysis of variations in Kyoto Encyclopedia of Genes and Genomes functions of microbes and metabolites showed that the mucosal microbiome was more frequently involved in genetic information processing and cellular processes, whereas the luminal microbiome generally participated in metabolic regulation. Notably, we found a greater abundance of the opportunistic pathogen genus Escherichia-Shigella at luminal sites and higher levels of the lipid-regulator metabolite fenfluramine at mucosal sites. Despite the extensive differences between the two sampling sites, the results revealed similarities in terms of amplicon sequence variant composition and dominant core microbes. This pilot exploration of luminal and mucosal microbiomes and metabolites provides key insights to guide future research. KEY POINTS: • Snake luminal and mucosal microbiota was distinct in composition and function. • Metabolome profiling revealed differences related to different metabolites. • The pathogenic microbes are more likely to colonize the gut lumina.
Collapse
Affiliation(s)
- Yuting Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Mingfang Zhou
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Wenjie Fang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qiuhong Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Huirong Mao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Biao Chen
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Tianxiang Zhang
- Institute of Wildlife Conservation, Jiangxi Academy of Forestry, Nanchang, 330045, China
| | - Yongtao Xu
- College of Forestry, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Weiwei Zhang
- College of Forestry, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yunlin Zheng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Xiaolong Hu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
44
|
Marhuenda-Egea FC, Narro-Serrano J, Shalabi-Benavent MJ, Álamo-Marzo JM, Amador-Prous C, Algado-Rabasa JT, Garijo-Saiz AM, Marco-Escoto M. A metabolic readout of the urine metabolome of COVID-19 patients. Metabolomics 2023; 19:7. [PMID: 36694097 PMCID: PMC9873393 DOI: 10.1007/s11306-023-01971-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Analysis of urine samples from COVID-19 patients by 1H NMR reveals important metabolic alterations due to SAR-CoV-2 infection. Previous studies have identified biomarkers in urine that reflect metabolic alterations in COVID-19 patients. We have used 1H NMR to better define these metabolic alterations since this technique allows us to obtain a broad profile of the metabolites present in urine. This technique offers the advantage that sample preparation is very simple and gives us very complete information on the metabolites present. To detect these alterations, we have compared urine samples from COVID-19 patients (n = 35) with healthy people (n = 18). We used unsupervised (Robust PCA) and supervised (PLS-LDA) multivariate analysis methods to evaluate the differences between the two groups: COVID-19 and healthy controls. The differences focus on a group of metabolites related to energy metabolism (glucose, ketone bodies, glycine, creatinine, and citrate) and other processes related to bacterial flora (TMAO and formic acid) and detoxification (hippuric acid). The alterations in the urinary metabolome shown in this work indicate that SARS-CoV-2 causes a metabolic change from a normal situation of glucose consumption towards a gluconeogenic situation and possible insulin resistance.
Collapse
Affiliation(s)
- F C Marhuenda-Egea
- Departamento de Agroquímica y Bioquímica, Universidad de Alicante, Alicante, Spain.
| | - J Narro-Serrano
- Departamento de Química Física, Universidad de Alicante, Alicante, Spain
| | | | - J M Álamo-Marzo
- Biochemical Laboratory, Hospital Marina Baixa, Villajoyosa, Spain
| | | | | | | | | |
Collapse
|
45
|
Jalandra R, Makharia GK, Sharma M, Kumar A. Inflammatory and deleterious role of gut microbiota-derived trimethylamine on colon cells. Front Immunol 2023; 13:1101429. [PMID: 36726978 PMCID: PMC9885123 DOI: 10.3389/fimmu.2022.1101429] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Trimethylamine (TMA) is produced by the intestinal microbiota as a by-product of metabolism of dietary precursors. TMA has been implicated in various chronic health conditions. However, the effect of TMA in the colon and the underlying mechanism was not clear. In this study, TMA exhibited toxic effects in vitro as well as in vivo. TMA-induced oxidative stress causes DNA damage, and compromised cell membrane integrity leading to the release of LDH outside the cells which ultimately leads to cell death. Besides, TMA also exhibited pronounced increase in cell cycle arrest at G2/M phase in both HCT116 and HT29 cell lines. TMA was found to be genotoxic and cytotoxic as the TMA concentration increased from 0.15 mM. A decreased ATP intracellular content was observed after 24 h, 48 h, and 72 h treatment in a time and dose-dependent manner. For in vivo research, TMA (100 mM, i.p. and intra-rectal) once a week for 12 weeks caused significant changes in cellular morphology of colon and rectum epithelium as assessed by H & E staining. TMA also significantly increased the infiltration of inflammatory cells in the colon and rectal epithelium indicating the severity of inflammation. In addition, TMA caused extensive mucosal damage and distortion in the epithelium, decrease in length of small intestine compared to control mice. In conclusion, these results highlight the detrimental effects of TMA in the colon and rectal epithelium.
Collapse
Affiliation(s)
- Rekha Jalandra
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
- Department of Zoology, Maharshi Dayanand University, Rohtak, India
| | - Govind K. Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Minakshi Sharma
- Department of Zoology, Maharshi Dayanand University, Rohtak, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
46
|
Iodomethylcholine Inhibits Trimethylamine-N-Oxide Production and Averts Maternal Chronic Kidney Disease-Programmed Offspring Hypertension. Int J Mol Sci 2023; 24:ijms24021284. [PMID: 36674799 PMCID: PMC9866155 DOI: 10.3390/ijms24021284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Chronic kidney disease (CKD) affects 10% of the global population, including pregnant women. Adverse maternal conditions determine the developmental programming of many diseases later in life. We previously demonstrated that adult rat offspring born to dams with CKD developed hypertension and renal hypertrophy. Trimethylamine-N-oxide (TMAO), a uremic toxin derived from the gut microbiota, has been linked to hypertension. This study assesses the effects of TMAO inhibition by iodomethylcholine (IMC) treatment on offspring hypertension programmed by maternal CKD. Female rats were fed either a control or a 0.5% adenine diet before conception, with or without IMC treatment during pregnancy and lactation. Maternal IMC treatment averted maternal CKD-primed offspring hypertension and renal hypertrophy in 12-week-old offspring. Offspring hypertension is associated with increases in the plasma TMAO concentration and oxidative stress and shifts in gut microbiota. The beneficial effects of IMC are related to a reduction in TMAO; increases in genera Acetatifactor, Bifidobacterium, and Eubacterium; and decreases in genera Phocacecola and Bacteroides. Our findings afford insights into the targeting of the gut microbiota to deplete TMAO production, with therapeutic potential for the prevention of offspring hypertension programmed by maternal CKD, although these results still need further clinical translation.
Collapse
|
47
|
Bertram HC. NMR foodomics in the assessment of diet and effects beyond nutrients. Curr Opin Clin Nutr Metab Care 2023:00075197-990000000-00051. [PMID: 36942870 DOI: 10.1097/mco.0000000000000906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
PURPOSE OF REVIEW This review provides an overview of most recent research studies employing nuclear magnetic resonance (NMR)-based metabolomics in the assessment of effects of diet and food ingestion. RECENT FINDINGS NMR metabolomics is a useful tool in the elucidation of specific diets, for example, the Mediterranean diet, the New Nordic diet types, and also for comparing vegan, vegetarian and omnivore diets where specific diet-linked metabolite perturbations have been identified. Another core area where NMR metabolomics is employed involves research focused on examining specific food components or ingredients, including dietary fibers and other functional components. In several cases, NMR metabolomics has aided to document how specific food components exert effects on the metabolic activity of the gut microbiota. Research has also demonstrated the potential use of NMR metabolomics in assessing diet quality and interactions between specific food components such as meat and diet quality. The implications of these findings are important as they address that background diet can be decisive for if food items turn out to exert either harmful or health-promoting effects. SUMMARY NMR metabolomics can provide important mechanistic insight and aid to biomarker discovery with implications for compliance and food registration purposes.
Collapse
|
48
|
Rehman A, Tyree SM, Fehlbaum S, DunnGalvin G, Panagos CG, Guy B, Patel S, Dinan TG, Duttaroy AK, Duss R, Steinert RE. A water-soluble tomato extract rich in secondary plant metabolites lowers trimethylamine-n-oxide and modulates gut microbiota: a randomized, double-blind, placebo-controlled cross-over study in overweight and obese adults. J Nutr 2023; 153:96-105. [PMID: 36913483 DOI: 10.1016/j.tjnut.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/26/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Natural products rich in polyphenols have been shown to lower plasma trimethylamine-n-oxide (TMAO) known for its proatherogenic effects by modulating the intestinal microbiota. OBJECTIVES We aimed to determine the impact of Fruitflow, a water-soluble tomato extract, on TMAO, fecal microbiota, and plasma and fecal metabolites. METHODS Overweight and obese adults (n = 22, BMI 28-35 kg/m2) were included in a double-blind, placebo-controlled, cross-over study receiving 2×150 mg Fruitflow per day or placebo (maltodextrin) for 4 wk with a 6-week wash-out between interventions. Stool, blood, and urine samples were collected to assess changes in plasma TMAO (primary outcome) as well as fecal microbiota, fecal and plasma metabolites, and urine TMAO (secondary outcomes). In a subgroup (n = 9), postprandial TMAO was evaluated following a choline-rich breakfast (∼450 mg). Statistical methods included paired t-tests or Wilcoxon signed rank tests and permutational multivariate analysis of variance. RESULTS Fruitflow, but not placebo, reduced fasting levels of plasma (-1.5 μM, P ≤ 0.05) and urine (-19.1 μM, P ≤ 0.01) TMAO as well as plasma lipopolysaccharides (-5.3 ng/mL, P ≤ 0.05) from baseline to the end of intervention. However, these changes were significant only for urine TMAO levels when comparing between the groups (P ≤ 0.05). Changes in microbial beta, but not alpha, diversity paralleled this with a significant difference in Jaccard distance-based Principal Component (P ≤ 0.05) as well as decreases in Bacteroides, Ruminococccus, and Hungatella and increases in Alistipes when comparing between and within groups (P ≤ 0.05, respectively). There were no between-group differences in SCFAs and bile acids (BAs) in both faces and plasma but several changes within groups such as an increase in fecal cholic acid or plasma pyruvate with Fruitflow (P ≤ 0.05, respectively). An untargeted metabolomic analysis revealed TMAO as the most discriminant plasma metabolite between groups (P ≤ 0.05). CONCLUSIONS Our results support earlier findings that polyphenol-rich extracts can lower plasma TMAO in overweight and obese adults related to gut microbiota modulation. This trial was registered at clinicaltrials.gov as NCT04160481 (https://clinicaltrials.gov/ct2/show/NCT04160481?term= Fruitflow&draw= 2&rank= 2).
Collapse
Affiliation(s)
| | | | | | | | | | - Bertrand Guy
- DSM Nutritional Products, Kaiseraugst, Switzerland
| | | | - Timothy G Dinan
- Atlantia Clinical Trials, Cork, Ireland, APC Microbiome Ireland, Cork, Ireland, Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Nutrition, Faculty of Medicine, University of Oslo, Norway
| | - Ruedi Duss
- DSM Nutritional Products, Kaiseraugst, Switzerland
| | - Robert E Steinert
- DSM Nutritional Products, Kaiseraugst, Switzerland; Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
49
|
Choroszy M, Litwinowicz K, Bednarz R, Roleder T, Lerman A, Toya T, Kamiński K, Sawicka-Śmiarowska E, Niemira M, Sobieszczańska B. Human Gut Microbiota in Coronary Artery Disease: A Systematic Review and Meta-Analysis. Metabolites 2022; 12:1165. [PMID: 36557203 PMCID: PMC9788186 DOI: 10.3390/metabo12121165] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
In recent years, the importance of the gut microbiome in human health and disease has increased. Growing evidence suggests that gut dysbiosis might be a crucial risk factor for coronary artery disease (CAD). Therefore, we conducted a systematic review and meta-analysis to determine whether or not CAD is associated with specific changes in the gut microbiome. The V3-V4 regions of the 16S rDNA from fecal samples were analyzed to compare the gut microbiome composition between CAD patients and controls. Our search yielded 1181 articles, of which 21 met inclusion criteria for systematic review and 7 for meta-analysis. The alpha-diversity, including observed OTUs, Shannon and Simpson indices, was significantly decreased in CAD, indicating the reduced richness of the gut microbiome. The most consistent results in a systematic review and meta-analysis pointed out the reduced abundance of Bacteroidetes and Lachnospiraceae in CAD patients. Moreover, Enterobacteriaceae, Lactobacillus, and Streptococcus taxa demonstrated an increased trend in CAD patients. The alterations in the gut microbiota composition are associated with qualitative and quantitative changes in bacterial metabolites, many of which have pro-atherogenic effects on endothelial cells, increasing the risk of developing and progressing CAD.
Collapse
Affiliation(s)
- Marcin Choroszy
- Department of Microbiology, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Kamil Litwinowicz
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Robert Bednarz
- Ninewells Hospital and Medical School, James Arrott Drive, Dundee DD1 9SY, UK
| | - Tomasz Roleder
- Research and Development Centre, Regional Specialist Hospital, 51-124 Wroclaw, Poland
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Division of Cardiology, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Karol Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Emilia Sawicka-Śmiarowska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Cardiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | |
Collapse
|
50
|
Carneiro TJ, Vojtek M, Gonçalves-Monteiro S, Batista de Carvalho ALM, Marques MPM, Diniz C, Gil AM. Effect of Pd 2Spermine on Mice Brain-Liver Axis Metabolism Assessed by NMR Metabolomics. Int J Mol Sci 2022; 23:13773. [PMID: 36430252 PMCID: PMC9693583 DOI: 10.3390/ijms232213773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Cisplatin (cDDP)-based chemotherapy is often limited by severe deleterious effects (nephrotoxicity, hepatotoxicity and neurotoxicity). The polynuclear palladium(II) compound Pd2Spermine (Pd2Spm) has emerged as a potential alternative drug, with favorable pharmacokinetic/pharmacodynamic properties. This paper reports on a Nuclear Magnetic Resonance metabolomics study to (i) characterize the response of mice brain and liver to Pd2Spm, compared to cDDP, and (ii) correlate brain-liver metabolic variations. Multivariate and correlation analysis of the spectra of polar and lipophilic brain and liver extracts from an MDA-MB-231 cell-derived mouse model revealed a stronger impact of Pd2Spm on brain metabolome, compared to cDDP. This was expressed by changes in amino acids, inosine, cholate, pantothenate, fatty acids, phospholipids, among other compounds. Liver was less affected than brain, with cDDP inducing more metabolite changes. Results suggest that neither drug induces neuronal damage or inflammation, and that Pd2Spm seems to lead to enhanced brain anti-inflammatory and antioxidant mechanisms, regulation of brain bioactive metabolite pools and adaptability of cell membrane characteristics. The cDDP appears to induce higher extension of liver damage and an enhanced need for liver regeneration processes. This work demonstrates the usefulness of untargeted metabolomics in evaluating drug impact on multiple organs, while confirming Pd2Spm as a promising replacement of cDDP.
Collapse
Affiliation(s)
- Tatiana J. Carneiro
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Martin Vojtek
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | - Salomé Gonçalves-Monteiro
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | | | - Maria Paula M. Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Carmen Diniz
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | - Ana M. Gil
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|