1
|
Escalante J, Artaiz O, Diwakarla S, McQuade RM. Leaky gut in systemic inflammation: exploring the link between gastrointestinal disorders and age-related diseases. GeroScience 2024:10.1007/s11357-024-01451-2. [PMID: 39638978 DOI: 10.1007/s11357-024-01451-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
Global average life expectancy has steadily increased over the last several decades and is projected to reach ~ 77 years by 2050. As it stands, the number of people > 60 years currently outnumbers children younger than 5 years, and by 2050, it is anticipated that the global population of people aged > 60 years will double, surpassing 2.1 billion. This demographic shift in our population is expected to have substantial consequences on health services globally due to the disease burden associated with aging. Osteoarthritis, chronic obstructive pulmonary disease, diabetes, cardiovascular disease, and cognitive decline associated with dementia are among the most common age-related diseases and contribute significantly to morbidity and mortality in the aged population. Many of these age-related diseases have been linked to chronic low-grade systemic inflammation which often accompanies aging. Gastrointestinal barrier dysfunction, also known as "leaky gut," has been shown to contribute to systemic inflammation in several diseases including inflammatory bowel disease and irritable bowel syndrome, but its role in the development and/or progression of chronic low-grade systemic inflammation during aging is unclear. This review outlines current literature on the leaky gut in aging, how leaky gut might contribute to systemic inflammation, and the links between gastrointestinal inflammatory diseases and common age-related diseases to provide insight into a potential relationship between the intestinal barrier and inflammation.
Collapse
Affiliation(s)
- Jonathan Escalante
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
| | - Olivia Artaiz
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
| | - Shanti Diwakarla
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia
| | - Rachel M McQuade
- Gut-Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3021, Australia.
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3010, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), The Melbourne University and Western Health, Melbourne, VIC, 3021, Australia.
| |
Collapse
|
2
|
Zhou HM, Yang XY, Yue SJ, Wang WX, Zhang Q, Xu DQ, Li JJ, Tang YP. The identification of metabolites from gut microbiota in coronary heart disease via network pharmacology. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:145-155. [PMID: 38412071 DOI: 10.1080/21691401.2024.2319827] [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: 08/30/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Although the gut microbial metabolites exhibit potential effects on coronary heart disease (CHD), the underlying mechanism remains unclear. In this study, the active gut microbial metabolites acting on CHD and their potential mechanisms of action were explored through a network pharmacological approach. We collected a total of 208 metabolites from the gutMgene database and 726 overlapping targets from the similarity ensemble approach (SEA) and SwissTargetPrediction (STP) database, and ultimately identified 610 targets relevant to CHD. In conjunction with the gutMGene database, we identified 12 key targets. The targets of exogenous substances were removed, and 10 core targets involved in CHD were eventually retained. The microbiota-metabolites-targets-signalling pathways network analysis revealed that C-type lectin receptor signalling pathway, Lachnospiraceae, Escherichia, mitogen-activated protein kinase 1, prostaglandin-endoperoxidase synthase 2, phenylacetylglutamine and alcoholic acid are notable components of CHD and play important roles in the development of CHD. The results of molecular docking experiments demonstrated that AKT1-glycocholic acid and PTGS2-phenylacetylglutamine complexes may act on C-type lectin receptor signalling pathways. In this study, the key substances and potential mechanisms of gut microbial metabolites were analysed via network pharmacological methods, and a scientific basis and comprehensive idea were provided for the effects of gut microbial metabolites on CHD.
Collapse
Affiliation(s)
- Hao-Ming Zhou
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Xin-Yu Yang
- Department of Pharmacy, Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Wen-Xiao Wang
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Jia-Jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| |
Collapse
|
3
|
Xue J, Allaband C, Zuffa S, Poulsen O, Meadows J, Zhou D, Dorrestein PC, Knight R, Haddad GG. Gut Microbiota and Derived Metabolites Mediate Obstructive Sleep Apnea Induced Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624205. [PMID: 39605650 PMCID: PMC11601605 DOI: 10.1101/2024.11.18.624205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia/hypercapnia (IHC), affects predominantly obese individuals, and increases atherosclerosis risk. Since we and others have implicated gut microbiota and metabolites in atherogenesis, we dissected their contributions to OSA-induced atherosclerosis. Results Atherosclerotic lesions were compared between conventionally-reared specific pathogen free (SPF) and germ-free (GF) ApoE -/- mice following a high fat high cholesterol diet (HFHC), with and without IHC conditions. The fecal microbiota and metabolome were profiled using 16S rRNA gene amplicon sequencing and untargeted tandem mass spectrometry (LC-MS/MS) respectively. Phenotypic data showed that HFHC significantly increased atherosclerosis as compared to regular chow (RC) in both aorta and pulmonary artery (PA) of SPF mice. IHC exacerbated lesions in addition to HFHC. Differential abundance analysis of gut microbiota identified an enrichment of Akkermansiaceae and a depletion of Muribaculaceae (formerly S24-7) family members in the HFHC-IHC group. LC-MS/MS showed a dysregulation of bile acid profiles with taurocholic acid, taurodeoxycholic acid, and 12-ketodeoxycholic acid enriched in the HFHC-IHC group, long-chain N-acyl amides, and phosphatidylcholines. Interestingly, GF ApoE -/- mice markedly reduced atherosclerotic formation relative to SPF ApoE -/- mice in the aorta under HFHC/IHC conditions. In contrast, microbial colonization did not show a significant impact on the atherosclerotic progression in PA. Conclusions In summary, this research demonstrated that (1) IHC acts cooperatively with HFHC to induce atherosclerosis; (2) gut microbiota modulate atherogenesis, induced by HFHC/IHC, in the aorta not in PA; (3) different analytical methods suggest that a specific imbalance between Akkermansiaceae and Muribaculaceae bacterial families mediate OSA-induced atherosclerosis; and (4) derived bile acids, such as deoxycholic acid and lithocholic acid, regulate atherosclerosis in OSA. The knowledge obtained provides novel insights into the potential therapeutic approaches to prevent and treat OSA-induced atherosclerosis.
Collapse
|
4
|
Narum M, Seljeflot I, Bratseth V, Berg TJ, Sveen KA. Intestinal fatty acid binding protein is associated with coronary artery disease in long-term type 1 diabetes-the Dialong study. Cardiovasc Diabetol 2024; 23:419. [PMID: 39563343 PMCID: PMC11575117 DOI: 10.1186/s12933-024-02509-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Individuals with type 1 diabetes are at increased risk of accelerated atherosclerosis, causing coronary artery disease (CAD). The underlying mechanisms remain unclear, but new theories proposed are damage of gut mucosa causing leakage and translocation of gut microbiota products into the circulation, leading to inflammatory responses and atherosclerosis. We therefore aimed to study the associations between gut related inflammatory biomarkers and coronary atherosclerosis in individuals with long-term type 1 diabetes. METHODS In this cross-sectional, controlled study of 102 participants with type 1 diabetes and 63 control subjects, we measured circulating levels of intestinal fatty acid binding protein (I-FABP), soluble cluster of differentiation 14 (sCD14), lipopolysaccharide binding protein (LBP) and interleukin 18 (IL-18) by enzyme-linked immunosorbent assay (ELISA), and further gene expression of CD14 and toll-like receptor 4 (TLR4) by real time PCR in circulating leukocytes and peripheral blood mononuclear cells (PBMCs). The participants had either established coronary heart disease (CHD) or underwent computed tomography coronary angiography (CTCA) to assess for coronary atherosclerosis, including total, calcified and soft/mixed plaque volumes. RESULTS In the diabetes group, the levels of I-FABP were significantly higher in participants with established CHD or significant stenosis on CTCA compared to the participants with normal arteries or non-significant stenosis, with median 1.67 ng/ml (interquartile range [IQR] 1.02-2.32) vs. median 1.09 ng/ml (IQR 0.82-1.58), p = 0.003. I-FABP was associated with significant coronary artery stenosis by CTCA (> 50%) or previously established CHD in the adjusted analysis (odds ratio [OR] = 2.32, 95% confidence interval [CI]: 1.09-4.95; p = 0.029). The levels of I-FABP correlated also to total coronary plaque volume (r = 0.22, p < 0.05). This association remained significant after adjusting for age, sex, persistent albuminuria, eGFR, statin treatment, diabetes duration and mean time-weighted variables; HbA1c, LDL-cholesterol and systolic blood pressure (OR = 1.97, 95% CI: 1.28-3.01; p = 0.002). CONCLUSIONS In this cohort of individuals with long-term type 1 diabetes I-FABP associated significantly with coronary artery stenosis, suggesting a potential role of gut mucosa damage in the process of atherosclerosis in type 1 diabetes.
Collapse
Affiliation(s)
- Marte Narum
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway
| | - Vibeke Bratseth
- Oslo Center for Clinical Heart Research, Department of Cardiology Ullevaal, Oslo University Hospital, Oslo, Norway
| | - Tore Julsrud Berg
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kari Anne Sveen
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Yafarova AA, Dementeva EV, Zlobovskaya OA, Sheptulina AF, Lopatukhina EV, Timofeev YS, Glazunova EV, Lyundup AV, Doludin YV, Kiselev AR, Shipulin GA, Makarov VV, Drapkina OM, Yudin SM. Gut Microbiota and Metabolic Alterations Associated with Heart Failure and Coronary Artery Disease. Int J Mol Sci 2024; 25:11295. [PMID: 39457077 PMCID: PMC11508380 DOI: 10.3390/ijms252011295] [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: 09/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the role of gut microbiota in cardiovascular diseases, with an additional focus on pro-atherogenic metabolites. We use advanced network analysis and machine learning techniques to identify key microbial features linked to coronary artery disease (CAD) and heart failure with reduced ejection fraction (HFrEF). This cross-sectional study included 189 participants divided into three groups: coronary artery disease (n = 93), heart failure with reduced ejection fraction (n = 43), and controls (n = 53). Assessments included physical exams, echocardiography, dietary surveys, blood analysis, and fecal analysis. Gut microbiota composition was analyzed using next-generation sequencing (NGS) and quantitative polymerase chain reaction (qPCR). Statistical analysis methods for testing hypotheses and correlations, alpha and beta-diversity analyses, co-occurrence networks, and machine learning were conducted using Python libraries or R packages with multiple comparisons corrected using the Benjamini-Hochberg procedure. Significant gut microbiota alterations were observed, with higher Bacillota/Bacteroidota ratios in CAD and HFrEF groups compared to controls (p < 0.001). Significant differences were observed in α-diversity indices (Pielou, Chao1, Faith) between disease groups and controls (p < 0.001). β-diversity analyses also revealed distinct microbial profiles (p = 0.0015). Interestingly, trimethylamine N-oxide (TMAO) levels were lower in CAD and HFrEF groups compared to controls (p < 0.05), while indoxyl sulfate (IS) levels were comparable between the study groups. Co-occurrence network analysis and machine learning identified key microbial features linked to these conditions, highlighting complex interactions within the gut microbiota associated with cardiovascular disease.
Collapse
Affiliation(s)
- Adel A. Yafarova
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Dementeva
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Olga A. Zlobovskaya
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Anna F. Sheptulina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Elena V. Lopatukhina
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Yuriy S. Timofeev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Evgeniya V. Glazunova
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Aleksey V. Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, 117036 Moscow, Russia
| | - Yuriy V. Doludin
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Anton R. Kiselev
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - German A. Shipulin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| | - Oxana M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, Petroverigskyj Lane 10, Bld. 3, 101990 Moscow, Russia (A.R.K.)
| | - Sergey M. Yudin
- Federal State Budgetary Institution «Centre for Strategic Planning and Management of Biomedical Health Risks» of the Federal Medical and Biological Agency, Pogodinskaya Str., 10/1, 119121 Moscow, Russia
| |
Collapse
|
6
|
Xie K, Xiao C, Lin L, Li F, Hu W, Yang Y, Chen D, Miao Z, Sun TY, Yan Y, Zheng JS, Chen YM. Erythrocyte Very Long-Chain Saturated Fatty Acids, Gut Microbiota-Bile Acid Axis, and Incident Coronary Artery Disease in Adults: A Prospective Cohort Study. J Nutr 2024; 154:3019-3030. [PMID: 39128547 DOI: 10.1016/j.tjnut.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/12/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Prior research has highlighted inverse associations between concentrations of circulating very long-chain saturated fatty acids (VLCSFAs) and coronary artery disease (CAD). However, the intricate links involving VLCSFAs, gut microbiota, and bile acids remain underexplored. OBJECTIVES This study examined the association of erythrocyte VLCSFAs with CHD incidence, focusing on the mediating role of gut microbiota and fecal bile acids. METHODS This 10-y prospective study included 2383 participants without CHD at baseline. Erythrocyte VLCSFAs [arachidic acid (C20:0), behenic acid (C22:0), and lignoceric acid (C24:0)] were measured using gas chromatography at baseline, and 274 CHD incidents were documented in triennial follow-ups. Gut microbiota in 1744 participants and fecal bile acid metabolites in 945 participants were analyzed using 16S ribosomal ribonucleic acid sequencing and ultra-performance liquid chromatography-tandem mass spectrometry at middle-term. RESULTS The multivariable-adjusted hazard ratios (95% confidence interval) for CHD incidence in highest compared with lowest quartiles were 0.87 (0.61, 1.25) for C20:0, 0.63 (0.42, 0.96) for C22:0, 0.59 (0.41, 0.85) for C24:0, and 0.57 (0.39, 0.83) for total VLCSFAs. Participants with higher total VLCSFA concentrations exhibited increased abundances of Holdemanella, Coriobacteriales Incertae Sedis spp., Ruminococcaceae UCG-005 and UCG-010, and Lachnospiraceae ND3007 group. These 5 genera generated overlapping differential microbial scores (ODMSs) that accounted for 11.52% of the total VLCSFAs-CHD association (Pmediation = 0.018). Bile acids tauro_α_ and tauro_β_muricholic acid were inversely associated with ODMS and positively associated with incident CHD. Opposite associations were found for glycolithocholic acid and glycodeoxycholic acid. Mediation analyses indicated that glycolithocholic acid, glycodeoxycholic acid, and tauro_α_ and tauro_β_muricholic acid explained 56.40%, 35.19%, and 26.17% of the ODMS-CHD association, respectively (Pmediation = 0.002, 0.008, and 0.020). CONCLUSIONS Elevated erythrocyte VLCSFAs are inversely associated with CHD risk in the Chinese population, with gut microbiota and fecal bile acid profiles potentially mediating this association. The identified microbiota and bile acid metabolites may serve as potential intervention targets in future studies. This trial was registered at www. CLINICALTRIALS gov as NCT03179657.
Collapse
Affiliation(s)
- Keliang Xie
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Congmei Xiao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Lishan Lin
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fanqin Li
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wei Hu
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yingdi Yang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Danyu Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zelei Miao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Ting-Yu Sun
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yan Yan
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China; School of Medicine, Westlake University, Hangzhou, China.
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Dean YE, Shebl MA, Doma M, Elmezayen RW, Loayza Pintado JJ, Rouzan SS, Hassan NAIF, Yaqout YE, Tokunaga A, Anozie C, ElKoumi O, Elawady SS, Mady T, Nizam SN, Etman Y, Nizam R, Hazimeh Y, Alazmy M, Aiash H. Intestinal microbiome as a diagnostic marker of coronary artery disease: a systematic review and meta-analysis. Ann Med Surg (Lond) 2024; 86:6105-6120. [PMID: 39359774 PMCID: PMC11444608 DOI: 10.1097/ms9.0000000000002516] [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: 03/20/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
Background The intestinal microbiome has been recently linked to several metabolic and chronic disorders, one of which is coronary artery disease (CAD). Our study aimed to analyze the intestinal microbiome of CAD patients and assess the eligibility of dysbiosis as a diagnostic marker of CAD. Methods PubMed, Scopus, Embase, and Web of Science were searched using terms, such as 'CAD' and 'microbiome'. Only observational controlled studies were included. R version 4.2.2 was used for the analysis. Results A significant association was found between the CAD group and increased Simpson and Shannon Indices compared with the control group (MD=0.04, 95% CI=0.03-0.05, and MD=0.11, 95% CI=0.01-0.22, respectively). Our analysis yielded a statistically significant association between the CAD group and increased Prevotella genus (MD=13.27, 95% CI=4.12-22.42, P-value=0.004), Catenibacterium genus (MD=0.09, 95% CI=0.09-0.10), Pseudomonas genus (MD=0.54, 95% CI=0.29-0.78, P-value), and Subdoligranulum (MD=-0.06, 95% CI=-0.06 to -0.06) compared with the control group. Another significant association was detected between the CAD group and decreased Bacteroides vulgatus and Bacteroides dorei (MD=-10.31, 95% CI=-14.78 to -5.84, P-value <0.00001). Conclusion Dysbiosis is an acceptable diagnostic marker of CAD. Decreased B. dorei and B. vulgatus among CAD patients suggests a protective role of these bacteria. Future clinical trials are necessary to investigate the potential benefit of supplementation of these bacteria in treating or preventing CAD.
Collapse
Affiliation(s)
- Yomna E Dean
- Alexandria University, Faculty of Medicine, Alexandria
| | | | - Mohamed Doma
- Alexandria University, Faculty of Medicine, Alexandria
| | | | | | | | | | | | | | | | - Omar ElKoumi
- Suez Universtiy, Faculty of Medicine, Suez, Egypt
| | | | - Tamer Mady
- International American University, College of Medicine, Saint Lucia
| | - Sana N Nizam
- University of California Los Angeles, California
| | - Yasser Etman
- Texas Health Hospital Rockwall, Director of Intensive Care Unit, Rockwall, Texas, USA
| | | | - Yusef Hazimeh
- Lebanese University
- Zahraa Hospital, University Medical Center, Lebanon
| | | | - Hani Aiash
- Suez Universtiy, Faculty of Medicine, Suez, Egypt
- SUNY Upstate Medical University, Syracuse
| |
Collapse
|
8
|
Arvelaez Pascucci J, Ghattas PE, Olivas Lerma R, Villa Nogueyra S, Nogales Bernal MB, Milani V, Floridia Rietmann LM, Alvarez SM, Salaz Diaz J. The Role of Microbiome in Cardiovascular Health: Insights for Primary Care Interventions. Cureus 2024; 16:e70311. [PMID: 39463572 PMCID: PMC11512747 DOI: 10.7759/cureus.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, highlighting the urgent need for effective prevention strategies. Emerging research suggests that the gut microbiome is critical in cardiovascular health, influencing pathophysiological processes associated with CVDs. This narrative review explores the intricate relationship between the gut microbiome and cardiovascular health, mainly focusing on how microbial composition affects inflammation, lipid metabolism, and endothelial function. Additionally, we discuss the implications of gut microbiome modulation through dietary interventions, prebiotics, and probiotics as potential therapeutic strategies for primary care practitioners. By emphasizing the importance of the microbiome in cardiovascular risk management, this review aims to inform primary care interventions that leverage microbiome research to improve patient outcomes and prevent CVDs. Ultimately, understanding and integrating gut health into cardiovascular care may provide a novel approach to enhancing cardiovascular resilience and reducing disease burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jorge Salaz Diaz
- Internal Medicine, Universidad Nacional Autónoma de México, Mexico, MEX
| |
Collapse
|
9
|
Florek K, Kübler M, Górka M, Kübler P. New Modifiable Risk Factors Influencing Coronary Artery Disease Severity. Int J Mol Sci 2024; 25:7766. [PMID: 39063008 PMCID: PMC11276953 DOI: 10.3390/ijms25147766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide with coronary artery disease (CAD) being the first culprit in this group. In terms of CAD, not only its presence but also its severity plays a role in the patient's treatment and prognosis. CAD complexity can be assessed with the indicator named the SYNTAX score (SS). A higher SS is associated with major adverse cardiovascular event (MACE) occurrence in short- and long-term observations. Hence, the risk factors affecting CAD severity based on SS results may help lower the risk among patients with already developed CAD to reduce their impact on coronary atherosclerosis progression. The well-established risk factors of CAD are consistent with those associated with the coronary plaque burden. However, recently, it was shown that new indicators exist, which we present in this paper, that significantly contribute to CAD complexity such as inflammatory parameters, C-reactive protein (CRP), ratios based on blood smear results, and uric acid. Moreover, microbiota alteration, vitamin D deficiency, and obstructive sleep apnea (OSA) also predicted CAD severity. However, sometimes, certain indicators were revealed as significant only in terms of chronic coronary syndromes (CCSs) or specific acute coronary syndromes (ACSs). Importantly, there is a need to apply the interdisciplinary and translational approach to the novel CAD severity risk assessment to maximize the impact of secondary prevention among patients at risk of coronary atherosclerosis progression.
Collapse
Affiliation(s)
- Kamila Florek
- Student Scientific Group of Invasive Cardiology, Institute of Heart Diseases, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Maja Kübler
- Student Scientific Group of Invasive Cardiology, Institute of Heart Diseases, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Magdalena Górka
- Student Scientific Group of Invasive Cardiology, Institute of Heart Diseases, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Piotr Kübler
- Institute of Heart Diseases, University Hospital, 50-556 Wroclaw, Poland
- Department of Cardiology, Faculty of Medicine, Institute of Heart Diseases, Wroclaw Medical University, 50-367 Wroclaw, Poland
| |
Collapse
|
10
|
Prins FM, Collij V, Groot HE, Björk JR, Swarte JC, Andreu-Sánchez S, Jansen BH, Fu J, Harmsen HJM, Zhernakova A, Lipsic E, van der Harst P, Weersma RK, Gacesa R. The gut microbiome across the cardiovascular risk spectrum. Eur J Prev Cardiol 2024; 31:935-944. [PMID: 38060843 DOI: 10.1093/eurjpc/zwad377] [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: 07/12/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 06/04/2024]
Abstract
AIMS Despite treatment advancements, cardiovascular disease remains a leading cause of death worldwide. Identifying new targets is crucial for enhancing preventive and therapeutic strategies. The gut microbiome has been associated with coronary artery disease (CAD), however our understanding of specific changes during CAD development remains limited. We aimed to investigate microbiome changes in participants without clinically manifest CAD with different cardiovascular risk levels and in patients with ST-elevation myocardial infarction (STEMI). METHODS AND RESULTS In this cross-sectional study, we characterized the gut microbiome using metagenomics of 411 faecal samples from individuals with low (n = 130), intermediate (n = 130), and high (n = 125) cardiovascular risk based on the Framingham score, and STEMI patients (n = 26). We analysed diversity, and differential abundance of species and functional pathways while accounting for confounders including medication and technical covariates. Collinsella stercoris, Flavonifractor plautii, and Ruthenibacterium lactatiformans showed increased abundances with cardiovascular risk, while Streptococcus thermophilus was negatively associated. Differential abundance analysis revealed eight species and 49 predicted metabolic pathways that were differently abundant among the groups. In the gut microbiome of STEMI patients, there was a depletion of pathways linked to vitamin, lipid, and amino acid biosynthesis. CONCLUSION We identified four microbial species showing a gradual trend in abundance from low-risk individuals to those with STEMI, and observed differential abundant species and pathways in STEMI patients compared to those without clinically manifest CAD. Further investigation is warranted to gain deeper understanding of their precise role in CAD progression and potential implications, with the ultimate goal of identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Femke M Prins
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Valerie Collij
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Hilde E Groot
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Johannes R Björk
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - J Casper Swarte
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Bernadien H Jansen
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection prevention, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Erik Lipsic
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| |
Collapse
|
11
|
Bridge LA, Hernández Vargas JA, Trujillo-Cáceres SJ, Beigrezaei S, Chatelan A, Salehi-Abargouei A, Muka T, Uriza-Pinzón JP, Raeisi-Dehkordi H, Franco OH, Grompone G, Artola Arita V. Two cosmoses, one universe: a narrative review exploring the gut microbiome's role in the effect of urban risk factors on vascular ageing. Maturitas 2024; 184:107951. [PMID: 38471294 DOI: 10.1016/j.maturitas.2024.107951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024]
Abstract
In the face of rising global urbanisation, understanding how the associated environment and lifestyle impact public health is a cornerstone for prevention, research, and clinical practice. Cardiovascular disease is the leading cause of morbidity and mortality worldwide, with urban risk factors contributing greatly to its burden. The current narrative review adopts an exposome approach to explore the effect of urban-associated physical-chemical factors (such as air pollution) and lifestyle on cardiovascular health and ageing. In addition, we provide new insights into how these urban-related factors alter the gut microbiome, which has been associated with an increased risk of cardiovascular disease. We focus on vascular ageing, before disease onset, to promote preventative research and practice. We also discuss how urban ecosystems and social factors may interact with these pathways and provide suggestions for future research, precision prevention and management of vascular ageing. Most importantly, future research and decision-making would benefit from adopting an exposome approach and acknowledging the diverse and boundless universe of the microbiome.
Collapse
Affiliation(s)
- Lara Anne Bridge
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Juliana Alexandra Hernández Vargas
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Silvia Juliana Trujillo-Cáceres
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sara Beigrezaei
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Angeline Chatelan
- Geneva School of Health Sciences, HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Amin Salehi-Abargouei
- Research Center for Food Hygiene and Safety, Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Julieth Pilar Uriza-Pinzón
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hamidreza Raeisi-Dehkordi
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Oscar H Franco
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Vicente Artola Arita
- Department of Global Public Health and Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
12
|
Dinetz E, Zeballos-Palacios C, Martinez CA. Addressing the Missing Links in Cardiovascular Aging. Clin Interv Aging 2024; 19:873-882. [PMID: 38774249 PMCID: PMC11107914 DOI: 10.2147/cia.s457180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/27/2024] [Indexed: 05/24/2024] Open
Abstract
The aim of this manuscript is to provide a review of available options to enhance cardiovascular health and prevent cardiovascular disease (CVD) in the aging population using a systems-biology approach. These include the role of the gut microbiome, the early identification and removal of environmental toxins, and finally age related sex hormones and supplement replacement which all influence aging. Implementing such a comprehensive approach has the potential to facilitate earlier risk assessment, disease prevention, and even improve mortality. Further study in these areas will continue to advance our understanding and refine therapeutic interventions for a healthier cardiovascular aging process.
Collapse
Affiliation(s)
- Elliot Dinetz
- Department of Integrative and Family Medicine, University of Miami Miller School of Medicine Miami, Miami, FL, USA
| | | | - Claudia A Martinez
- Department of Medicine, Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
13
|
McCoubrey LE, Ferraro F, Seegobin N, Verin J, Alfassam HA, Awad A, Marzorati M, Verstrepen L, Ghyselinck J, De Munck J, De Medts J, Steppe E, De Vleeschhauwer V, De Rocker G, Droesbeke A, De Rijck M, Vanthoor S, Moens F, Siepmann J, Siepmann F, Gaisford S, Orlu M, Basit AW. Poly(D,l-lactide-co-glycolide) particles are metabolised by the gut microbiome and elevate short chain fatty acids. J Control Release 2024; 369:163-178. [PMID: 38521168 DOI: 10.1016/j.jconrel.2024.03.039] [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: 01/04/2024] [Revised: 02/17/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
The production of short chain fatty acids (SCFAs) by the colonic microbiome has numerous benefits for human health, including maintenance of epithelial barrier function, suppression of colitis, and protection against carcinogenesis. Despite the therapeutic potential, there is currently no optimal approach for elevating the colonic microbiome's synthesis of SCFAs. In this study, poly(D,l-lactide-co-glycolide) (PLGA) was investigated for this application, as it was hypothesised that the colonic microbiota would metabolise PLGA to its lactate monomers, which would promote the resident microbiota's synthesis of SCFAs. Two grades of spray dried PLGA, alongside a lactate bolus control, were screened in an advanced model of the human colon, known as the M-SHIME® system. Whilst the high molecular weight (Mw) grade of PLGA was stable in the presence of the microbiota sourced from three healthy humans, the low Mw PLGA (PLGA 2) was found to be metabolised. This microbial degradation led to sustained release of lactate over 48 h and increased concentrations of the SCFAs propionate and butyrate. Further, microbial synthesis of harmful ammonium was significantly reduced compared to untreated controls. Interestingly, both types of PLGA were found to influence the composition of the luminal and mucosal microbiota in a donor-specific manner. An in vitro model of an inflamed colonic epithelium also showed the polymer to affect the expression of pro- and anti-inflammatory markers, such as interleukins 8 and 10. The findings of this study reveal PLGA's sensitivity to enzymatic metabolism in the gut, which could be harnessed for therapeutic elevation of colonic SCFAs.
Collapse
Affiliation(s)
- Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Fabiana Ferraro
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - Nidhi Seegobin
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Jérémy Verin
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - Haya A Alfassam
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 114422 Riyadh, Saudi Arabia
| | - Atheer Awad
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, United Kingdom
| | | | | | | | | | | | - Evi Steppe
- ProDigest BVB, Technologiepark 73, 9052 Ghent, Belgium
| | | | | | | | | | - Sara Vanthoor
- ProDigest BVB, Technologiepark 73, 9052 Ghent, Belgium
| | | | | | | | - Simon Gaisford
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Mine Orlu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom.
| |
Collapse
|
14
|
Wallin N, Bergman L, Smith GN. Pregnancy-associated cardiovascular risks and postpartum care; an opportunity for interventions aiming at health preservation and disease prevention. Best Pract Res Clin Obstet Gynaecol 2024; 92:102435. [PMID: 38048671 DOI: 10.1016/j.bpobgyn.2023.102435] [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: 04/26/2023] [Revised: 07/20/2023] [Accepted: 11/18/2023] [Indexed: 12/06/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of premature death and disability for female individuals around the world and the rates are increasing in those aged 35-44 years. Certain pregnancy complications (Pregnancy-associated Cardiovascular Risks (P-CVR))are linked to an increased risk of future CVD making pregnancy and the postpartum period as an ideal time to screen individuals for underlying, often unrecognized, cardiovascular risk factors. Pregnancy complications associated with an increased risk of future CVD including the hypertensive disorders of pregnancy, gestational diabetes, idiopathic preterm birth, delivery of a growth restricted baby and a placental abruption that leads to delivery. A number of guidelines and research groups recommend postpartum CVR screening, counseling and lifestyle intervention for all those who have had one or more of P-CVRs starting within the first six months postpartum. An individualized plan for postpartum screening should be created with the individual and lifestyle interventions discussed.
Collapse
Affiliation(s)
- Nicole Wallin
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lina Bergman
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Stellenbosch University, Cape Town, South Africa; Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Graeme N Smith
- Department of Obstetrics and Gynecology, Queen's University, Ontario, Canada.
| |
Collapse
|
15
|
Wang Y, Xie Y, Mahara G, Xiong Y, Xiong Y, Zheng Q, Chen J, Zhang W, Zhou H, Li Q. Intestinal microbiota and metabolome perturbations in ischemic and idiopathic dilated cardiomyopathy. J Transl Med 2024; 22:89. [PMID: 38254195 PMCID: PMC10804607 DOI: 10.1186/s12967-023-04605-6] [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: 06/17/2023] [Accepted: 10/06/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Various clinical similarities are present in ischemic (ICM) and idiopathic dilated cardiomyopathy (IDCM), leading to ambiguity on some occasions. Previous studies have reported that intestinal microbiota appeared dysbiosis in ICM, whether implicating in the IDCM remains unclear. The aim of this study was to assess the alterations in intestinal microbiota and fecal metabolites in ICM and IDCM. METHODS ICM (n = 20), IDCM (n = 22), and healthy controls (HC, n = 20) were enrolled in this study. Stool samples were collected for 16S rRNA gene sequencing and gas chromatography-mass spectrometry (GC-MS) analysis. RESULTS Both ICM and IDCM exhibited reduced alpha diversity and altered microbial community structure compared to HC. At the genus level, nine taxa including Blautia, [Ruminococcus]_torques_group, Christensenellaceae_R-7_group, UCG-002, Corynebacterium, Oceanobacillus, Gracilibacillus, Klebsiella and Citrobacter was specific to ICM, whereas one taxa Alistipes uniquely altered in IDCM. Likewise, these changes were accompanied by significant metabolic differences. Further differential analysis displayed that 18 and 14 specific metabolites uniquely changed in ICM and IDCM, respectively. The heatmap was generated to display the association between genera and metabolites. Receiver operating characteristic curve (ROC) analysis confirmed the predictive value of the distinct microbial-metabolite features in disease status. The results showed that microbial (area under curve, AUC = 0.95) and metabolic signatures (AUC = 0.84) were effective in discriminating ICM from HC. Based on the specific microbial and metabolic features, the patients with IDCM could be separated from HC with an AUC of 0.80 and 0.87, respectively. Furthermore, the gut microbial genus (AUC = 0.88) and metabolite model (AUC = 0.89) were comparable in predicting IDCM from ICM. Especially, the combination of fecal microbial-metabolic features improved the ability to differentiate IDCM from ICM with an AUC of 0.96. CONCLUSION Our findings highlighted the alterations of gut microbiota and metabolites in different types of cardiomyopathies, providing insights into the pathophysiological mechanisms of myocardial diseases. Moreover, multi-omics analysis of fecal samples holds promise as a non-invasive tool for distinguishing disease status.
Collapse
Affiliation(s)
- Yusheng Wang
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yandan Xie
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Gehendra Mahara
- Clinical Research Center, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qifang Zheng
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jianqin Chen
- Department of Cardiovascular Internal Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
16
|
Du SB, Zhou HH, Xue ZP, Gao S, Li J, Meng Y, Zhao YJ, Wang PF, Li N, Bai JX, Bai JQ, Wang XP. Metagenomic sequencing revealed the regulative effect of Danshen and Honghua herb pair on the gut microbiota in rats with myocardial ischemia injury. FEMS Microbiol Lett 2024; 371:fnad133. [PMID: 38100390 DOI: 10.1093/femsle/fnad133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/21/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023] Open
Abstract
In recent years, more and more evidence has shown that the disorder of gut microbiota (GM) is closely correlated with myocardial ischemia (MI). Even though the Danshen and Honghua herb pair (DHHP) is widely used in treating cardiovascular disease in China and exhibits obvious clinical efficacy on MI, the anti-MI mechanism of DHHP remains and needs to be explored in depth. Thus, in this study, we investigated whether the amelioration effect and molecular mechanism of DHHP on MI were related to regulating GM through pharmacodynamics evaluation and metagenomic sequencing. Histopathological testing results showed that DHHP treatment could alleviate the pathological changes of myocardial tissue in the acute MI (AMI) rats induced by isoproterenol (ISO), especially structural disorder, irregular distribution, and enlargement of the myocardial space. These pathological changes were all alleviated to some extent by DHHP treatment. Biochemical analysis results suggested that compared with the control group, the serum levels of AST, CTn-I, CK-MB, and TNF-α in model group rats were notably decreased, and the CAT and SOD levels in serum were markedly increased. These abnormal trends were significantly reversed by DHHP treatment. Furthermore, metagenomic sequencing analysis results indicated that DHHP could improve disorders in the composition and function of GM in AMI rats, mainly reflected in increasing diversity and richness, and obviously enhancing the abundance of Bacteroides fluxus, B. uniformis, B. stercoris, Roseburia hominis, Schaedlerella arabinosiphila, and R. intestinalis, and reducing the abundance of Enterococcus avium and E. canintestini, which were associated with purine metabolism, tyrosine metabolism, cyanoamino acid metabolism, and glutathione metabolism. In conclusion, DHHP may attenuate ISO-induced MI by regulating the structure, composition, and function of GM, thus contributing to further our understanding of the anti-MI mechanisms of DHHP and providing new therapeutic ideas and diagnostic targets for the clinical studies of MI.
Collapse
Affiliation(s)
- Shao-Bing Du
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Hui-Hui Zhou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Zhi-Peng Xue
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Su Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Jing Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yi Meng
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yi-Jun Zhao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Peng-Fei Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Na Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Jia-Xin Bai
- Second Clinical College of Medicine, Heilongjiang University of Chinese Medicine, Harbin 150041, China
| | - Ji-Qing Bai
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiao-Ping Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| |
Collapse
|
17
|
Shi H, Yuan X, Wu F, Li X, Fan W, Yang X, Liu G. Genetic support of the causal association between gut microbiota and peripheral artery disease: a bidirectional Mendelian randomization study. Aging (Albany NY) 2024; 16:762-778. [PMID: 38198148 PMCID: PMC10817407 DOI: 10.18632/aging.205417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/04/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND The causal relationship between gut microbiota and peripheral artery disease (PAD) is still not clear. In this research, we employed the Mendelian randomization (MR) technique to explore the potential causal connection between 211 gut microbiota species and PAD. We also investigated whether the causal effects operate in both directions. METHODS We used Genome-wide Association Studies (GWAS) summary statistics data from the MiBioGen and FinnGen consortia to conduct a two-sample MR analysis to explore the causal link between gut microbiota and PAD. Sensitivity analysis is conducted to assess the robustness of the MR results. In addition to that, reverse MR analysis was performed to examine the inverse causal relationship. RESULTS The inverse variance weighted (IVW) method provided evidence supporting a causal relationship between 9 specific gut microbiota taxa and PAD. The study findings indicated that family Family XI (OR=1.11, CI 1.00-1.24, P=0.048), genus Lachnoclostridium (OR=1.24, 1.02-1.50, P=0.033), and genus Lachnospiraceae UCG001 (OR=1.17, 1.01-1.35, P=0.031) are risk factors associated with PAD. class Actinobacteria (OR=0.84, 0.72-0.99, P=0.034), family Acidaminococcaceae (OR=0.80, 0.66-0.98, P=0.029), genus Coprococcus2 (OR=0.79, 0.64-0.98, P=0.029), genus Ruminococcaceae UCG004 (OR=0.84, 0.72-0.99, P=0.032), genus Ruminococcaceae UCG010 (OR=0.74, 0.58-0.96, P=0.022), and order NB1n (OR=0.88, 0.79-0.98, P=0.02) may be associated with the risk factors of PAD. Moreover, our analysis did not uncover any evidence of a reverse causal relationship between PAD and the nine specific gut microbiota taxa investigated. CONCLUSIONS Our MR research has confirmed the potential causal relationship between gut microbiota and PAD while also identifying specific gut bacterial communities associated with PAD.
Collapse
Affiliation(s)
- Hongshuo Shi
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Yuan
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangfang Wu
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyu Li
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weijing Fan
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao Yang
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guobin Liu
- Department of Peripheral Vascular Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guangming Traditional Chinese Medicine Hospital Pudong New Area, Shanghai, China
| |
Collapse
|
18
|
Wang L, Yang L, Cheng XL, Qin XM, Chai Z, Li ZY. The Beneficial Effects of Dietary Astragali Radix Are Related to the Regulation of Gut Microbiota and Its Metabolites. J Med Food 2024; 27:22-34. [PMID: 38236693 DOI: 10.1089/jmf.2023.k.0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024] Open
Abstract
Astragali Radix (AR) or its extract has been used as an herbal medicine and dietary supplement in China, Europe, and the United States. The gut microbiota could provide new insights for exploring dietary supplements' underlying mechanism on organisms. However, no reports have focused on the regulatory effect of AR on the gut microbiota as a dietary supplement. In this study, healthy ICR mice of either sex were divided into AR and control (CON) groups and given AR water extract (4.55 mg/kg·day-1) or saline by gavage for 14 days, respectively. Then 16S rRNA gene sequencing and ultra-high-performance liquid chromatography quadrupole time-of-flight tandem mass spectrometry-based fecal metabolomics were integrated to investigate the benefits of dietary AR. Weighted gene coexpression network analysis was also introduced to investigate the metabolites with highly synergistic changes. AR supplementation influenced the structure of intestinal microflora, especially enriching short-chain fatty acid-producing bacteria g_Coprobacillus, g_Prevotella, and g_Parabacteroides. AR also significantly altered the fecal metabolome, mainly related to amino acid metabolism, nucleotide metabolism, and bile acid (BA) metabolism. Moreover, the increased secondary BAs and BA-sulfates might closely relate to intestinal microflora. These findings provide valuable insights for future research of dietary AR as a functional food.
Collapse
Affiliation(s)
- Ling Wang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, Shanxi, China
| | - Lan Yang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, Shanxi, China
| | - Xiao-Ling Cheng
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, Shanxi, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, Shanxi, China
| | - Zhi Chai
- College of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zhen-Yu Li
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Cai M, Lin L, Jiang F, Peng Y, Li S, Chen L, Lin Y. Gut microbiota changes in patients with hypertension: A systematic review and meta-analysis. J Clin Hypertens (Greenwich) 2023; 25:1053-1068. [PMID: 37853925 PMCID: PMC10710550 DOI: 10.1111/jch.14722] [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: 04/26/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 10/20/2023]
Abstract
Hypertension is a major public health issue worldwide. The imbalance of gut microbiota is thought to play an important role in the pathogenesis of hypertension. The authors conducted the systematic review and meta-analysis to clarify the relationship between gut microbiota and hypertension through conducting an electronic search in six databases. Our meta-analysis included 19 studies and the results showed that compared with healthy controls, Shannon significantly decreased in hypertension [SMD = -0.13, 95%CI (-0.22, -0.04), p = .007]; however, Simpson [SMD = -0.01, 95%CI (-0.14, 0.12), p = .87], ACE [SMD = 0.18, 95%CI (-0.06, 0.43), p = .14], and Chao1 [SMD = 0.11, 95%CI (-0.01, 0.23), p = .08] did not differ significantly between hypertension and healthy controls. The F/B ratio significantly increased in hypertension [SMD = 0.84, 95%CI (0.10, 1.58), p = .03]. In addition, Shannon index was negatively correlated with hypertension [r = -0.12, 95%CI (-0.19, -0.05)], but had no significant correlation with SBP [r = 0.10, 95%CI (-0.19, 0.37)] and DBP [r = -0.39, 95%CI (-0.73, 0.12)]. At the phylum level, the relative abundance of Firmicutes [SMD = -0.01, 95%CI (-0.37, 0.34), p = .94], Bacteroidetes [SMD = -0.15, 95%CI (-0.44, 0.14), p = .30], Proteobacteria [SMD = 0.25, 95%CI (-0.01, 0.51), p = .06], and Actinobacteria [SMD = 0.21, 95%CI (-0.11, 0.53), p = .21] did not differ significantly between hypertension and healthy controls. At the genus level, compared with healthy controls, the relative abundance of Faecalibacterium decreased significantly [SMD = -0.16, 95%CI (-0.28, -0.04), p = .01], while the Streptococcus [SMD = 0.20, 95%CI (0.08, 0.32), p = .001] and Enterococcus [SMD = 0.20, 95%CI (0.08, 0.33), p = .002] significantly increased in hypertension. Available evidence suggests that hypertensive patients may have an imbalance of gut microbiota. However, it still needs further validation by large sample size studies of high quality.
Collapse
Affiliation(s)
- Meiling Cai
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Lingyu Lin
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Fei Jiang
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Yanchun Peng
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Sailan Li
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
| | - Liangwan Chen
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| | - Yanjuan Lin
- Department of NursingFujian Medical University Union HospitalFuzhouChina
- Department of Cardiovascular SurgeryFujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Special Reserve Talents LaboratoryFujian Medical University Union HospitalFuzhouChina
| |
Collapse
|
20
|
Gautam N, Mueller J, Alqaisi O, Gandhi T, Malkawi A, Tarun T, Alturkmani HJ, Zulqarnain MA, Pontone G, Al'Aref SJ. Machine Learning in Cardiovascular Risk Prediction and Precision Preventive Approaches. Curr Atheroscler Rep 2023; 25:1069-1081. [PMID: 38008807 DOI: 10.1007/s11883-023-01174-3] [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] [Accepted: 11/15/2023] [Indexed: 11/28/2023]
Abstract
PURPOSE OF REVIEW In this review, we sought to provide an overview of ML and focus on the contemporary applications of ML in cardiovascular risk prediction and precision preventive approaches. We end the review by highlighting the limitations of ML while projecting on the potential of ML in assimilating these multifaceted aspects of CAD in order to improve patient-level outcomes and further population health. RECENT FINDINGS Coronary artery disease (CAD) is estimated to affect 20.5 million adults across the USA, while also impacting a significant burden at the socio-economic level. While the knowledge of the mechanistic pathways that govern the onset and progression of clinical CAD has improved over the past decade, contemporary patient-level risk models lag in accuracy and utility. Recently, there has been renewed interest in combining advanced analytic techniques that utilize artificial intelligence (AI) with a big data approach in order to improve risk prediction within the realm of CAD. By virtue of being able to combine diverse amounts of multidimensional horizontal data, machine learning has been employed to build models for improved risk prediction and personalized patient care approaches. The use of ML-based algorithms has been used to leverage individualized patient-specific data and the associated metabolic/genomic profile to improve CAD risk assessment. While the tool can be visualized to shift the paradigm toward a patient-specific care, it is crucial to acknowledge and address several challenges inherent to ML and its integration into healthcare before it can be significantly incorporated in the daily clinical practice.
Collapse
Affiliation(s)
- Nitesh Gautam
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA
| | - Joshua Mueller
- Department of Internal Medicine, University of Arkansas for Medical Sciences Northwest Regional Campus, Fayetteville, AR, USA
| | - Omar Alqaisi
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tanmay Gandhi
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Abdallah Malkawi
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA
| | - Tushar Tarun
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA
| | - Hani J Alturkmani
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA
| | - Muhammed Ali Zulqarnain
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA
| | | | - Subhi J Al'Aref
- Division of Cardiology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St, Little Rock, AR, 72223, USA.
| |
Collapse
|
21
|
Jing J, Guo J, Dai R, Zhu C, Zhang Z. Targeting gut microbiota and immune crosstalk: potential mechanisms of natural products in the treatment of atherosclerosis. Front Pharmacol 2023; 14:1252907. [PMID: 37719851 PMCID: PMC10504665 DOI: 10.3389/fphar.2023.1252907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory reaction that primarily affects large and medium-sized arteries. It is a major cause of cardiovascular disease and peripheral arterial occlusive disease. The pathogenesis of AS involves specific structural and functional alterations in various populations of vascular cells at different stages of the disease. The immune response is involved throughout the entire developmental stage of AS, and targeting immune cells presents a promising avenue for its treatment. Over the past 2 decades, studies have shown that gut microbiota (GM) and its metabolites, such as trimethylamine-N-oxide, have a significant impact on the progression of AS. Interestingly, it has also been reported that there are complex mechanisms of action between GM and their metabolites, immune responses, and natural products that can have an impact on AS. GM and its metabolites regulate the functional expression of immune cells and have potential impacts on AS. Natural products have a wide range of health properties, and researchers are increasingly focusing on their role in AS. Now, there is compelling evidence that natural products provide an alternative approach to improving immune function in the AS microenvironment by modulating the GM. Natural product metabolites such as resveratrol, berberine, curcumin, and quercetin may improve the intestinal microenvironment by modulating the relative abundance of GM, which in turn influences the accumulation of GM metabolites. Natural products can delay the progression of AS by regulating the metabolism of GM, inhibiting the migration of monocytes and macrophages, promoting the polarization of the M2 phenotype of macrophages, down-regulating the level of inflammatory factors, regulating the balance of Treg/Th17, and inhibiting the formation of foam cells. Based on the above, we describe recent advances in the use of natural products that target GM and immune cells crosstalk to treat AS, which may bring some insights to guide the treatment of AS.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Dai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chaojun Zhu
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
22
|
Cimmino G, Natale F, Alfieri R, Cante L, Covino S, Franzese R, Limatola M, Marotta L, Molinari R, Mollo N, Loffredo FS, Golino P. Non-Conventional Risk Factors: "Fact" or "Fake" in Cardiovascular Disease Prevention? Biomedicines 2023; 11:2353. [PMID: 37760794 PMCID: PMC10525401 DOI: 10.3390/biomedicines11092353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases (CVDs), such as arterial hypertension, myocardial infarction, stroke, heart failure, atrial fibrillation, etc., still represent the main cause of morbidity and mortality worldwide. They significantly modify the patients' quality of life with a tremendous economic impact. It is well established that cardiovascular risk factors increase the probability of fatal and non-fatal cardiac events. These risk factors are classified into modifiable (smoking, arterial hypertension, hypercholesterolemia, low HDL cholesterol, diabetes, excessive alcohol consumption, high-fat and high-calorie diet, reduced physical activity) and non-modifiable (sex, age, family history, of previous cardiovascular disease). Hence, CVD prevention is based on early identification and management of modifiable risk factors whose impact on the CV outcome is now performed by the use of CV risk assessment models, such as the Framingham Risk Score, Pooled Cohort Equations, or the SCORE2. However, in recent years, emerging, non-traditional factors (metabolic and non-metabolic) seem to significantly affect this assessment. In this article, we aim at defining these emerging factors and describe the potential mechanisms by which they might contribute to the development of CVD.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Cardiology Unit, Azienda Ospedaliera Universitaria Luigi Vanvitelli, 80138 Naples, Italy
| | - Francesco Natale
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Roberta Alfieri
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Luigi Cante
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Simona Covino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Rosa Franzese
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Mirella Limatola
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Luigi Marotta
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Riccardo Molinari
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Noemi Mollo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Francesco S Loffredo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy (F.S.L.)
- Vanvitelli Cardiology Unit, Monaldi Hospital, 80131 Naples, Italy
| |
Collapse
|
23
|
Abstract
Cardiometabolic disease comprises cardiovascular and metabolic dysfunction and underlies the leading causes of morbidity and mortality, both within the United States and worldwide. Commensal microbiota are implicated in the development of cardiometabolic disease. Evidence suggests that the microbiome is relatively variable during infancy and early childhood, becoming more fixed in later childhood and adulthood. Effects of microbiota, both during early development, and in later life, may induce changes in host metabolism that modulate risk mechanisms and predispose toward the development of cardiometabolic disease. In this review, we summarize the factors that influence gut microbiome composition and function during early life and explore how changes in microbiota and microbial metabolism influence host metabolism and cardiometabolic risk throughout life. We highlight limitations in current methodology and approaches and outline state-of-the-art advances, which are improving research and building toward refined diagnosis and treatment options in microbiome-targeted therapies.
Collapse
Affiliation(s)
- Curtis L Gabriel
- Division of Gastroenterology, Hepatology and Nutrition (C.L.G.), Vanderbilt University Medical Center, Nashville
- Tennessee Center for AIDS Research (C.L.G.), Vanderbilt University Medical Center, Nashville
| | - Jane F Ferguson
- Division of Cardiovascular Medicine (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Microbiome Innovation Center (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Institute for Infection, Immunology, and Inflammation (J.F.F.), Vanderbilt University Medical Center, Nashville
| |
Collapse
|
24
|
Zhuo X, Luo H, Lei R, Lou X, Bian J, Guo J, Luo H, Zhang X, Jiao Q, Gong W. Association between Intestinal Microecological Changes and Atherothrombosis. Microorganisms 2023; 11:1223. [PMID: 37317197 DOI: 10.3390/microorganisms11051223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of large- and medium-sized arteries that causes ischemic heart disease, strokes, and peripheral vascular disease, collectively called cardiovascular disease (CVD), and is the leading cause of CVD resulting in a high rate of mortality in the population. AS is pathological by plaque development, which is caused by lipid infiltration in the vessel wall, endothelial dysfunction, and chronic low-grade inflammation. Recently, more and more scholars have paid attention to the importance of intestinal microecological disorders in the occurrence and development of AS. Intestinal G-bacterial cell wall lipopolysaccharide (LPS) and bacterial metabolites, such as oxidized trimethylamine (TMAO) and short-chain fatty acids (SCFAs), are involved in the development of AS by affecting the inflammatory response, lipid metabolism, and blood pressure regulation of the body. Additionally, intestinal microecology promotes the progression of AS by interfering with the normal bile acid metabolism of the body. In this review, we summarize the research on the correlation between maintaining a dynamic balance of intestinal microecology and AS, which may be potentially helpful for the treatment of AS.
Collapse
Affiliation(s)
- Xinyu Zhuo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Hui Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Rumei Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Xiaokun Lou
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Jing Bian
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Junfeng Guo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Hao Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Xingwei Zhang
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Qibin Jiao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
| | - Wenyan Gong
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| |
Collapse
|
25
|
Liao XX, Hu K, Xie XH, Wen YL, Wang R, Hu ZW, Zhou YL, Li JJ, Wu MK, Yu JX, Chen JW, Ren P, Wu XY, Zhou JJ. Banxia Xiexin decoction alleviates AS co-depression disease by regulating the gut microbiome-lipid metabolic axis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116468. [PMID: 37044233 DOI: 10.1016/j.jep.2023.116468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin decoction (BXD) is a classic Chinese herbal formulation consisting of 7 herbs including Pinelliae Rhizoma, Scutellariae Radix, Zingiberis Rhizoma, Ginseng Radix, Glycyrrhizae Radix, Coptidis Rhizoma, and Jujubae Fructus, which can exert effects on lowering lipids and alleviating depressive mood disorders via affecting gastrointestinal tract. AIM OF THE STUDY The pathogenesis of atherosclerosis (AS) co-depression disease has not been well studied, and the current clinical treatment strategies are not satisfactory. As a result, it is critical to find novel methods of treatment. Based on the hypothesis that the gut microbiome may promote the development of AS co-depression disease by regulating host lipid metabolism, this study sought to evaluate the effectiveness and action mechanism of BXD in regulation of the gut microbiome via an intervention in AS co-depression mice. MATERIALS AND METHODS To determine the primary constituents of BXD, UPLC-Q/TOF-MS analysis was carried out. Sixteen C56BL/6 mice were fed normal chow as a control group; 64 ApoE-/- mice were randomized into four groups (model group and three treatment groups) and fed high-fat chow combined with daily bind stimulation for sixteen weeks to develop the AS co-depression mouse model and were administered saline or low, medium or high concentrations of BXD during the experimental modeling period. The antidepressant efficacy of BXD was examined by weighing, a sucrose preference test, an open field test, and a tail suspension experiment. The effectiveness of BXD as an anti-AS treatment was evaluated by means of biochemical indices, the HE staining method, and the Oil red O staining method. The impacts of BXD on the gut microbiome structure and brain (hippocampus and prefrontal cortex tissue) lipids in mice with the AS co-depression model were examined by 16S rDNA sequencing combined with lipidomics analysis. RESULTS The main components of BXD include baicalin, berberine, ginsenoside Rb1, and 18 other substances. BXD could improve depression-like behavioral characteristics and AS-related indices in AS co-depression mice; BXD could regulate the abundance of some flora (phylum level: reduced abundance of Proteobacteria and Deferribacteres; genus level: reduced abundance of Clostridium_IV, Helicobacter, and Pseudoflavonifractor, Acetatifactor, Oscillibacter, which were significantly different). The lipidomics analysis showed that the differential lipids between the model and gavaged high-dose BXD (BXH) groups were enriched in glycerophospholipid metabolism, and lysophosphatidylcholine (LPC(20:3)(rep)(rep)) in the hippocampus and LPC(20:4)(rep) in the prefrontal cortex both showed downregulation in BXH. The correlation analysis illustrated that the screened differential lipids were mainly linked to Deferribacteres and Actinobacteria. CONCLUSION BXD may exert an anti-AS co-depression therapeutic effect by modulating the abundance of some flora and thus intervening in peripheral lipid and brain lipid metabolism (via downregulation of LPC levels).
Collapse
Affiliation(s)
- Xing-Xing Liao
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Ke Hu
- Department of Rehabilitation Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xin-Hua Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - You-Liang Wen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Rui Wang
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zi-Wei Hu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yu-Long Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jia-Jun Li
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Ming-Kun Wu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jing-Xuan Yu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Jia-Wei Chen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Peng Ren
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Xiao-Yun Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Jun-Jie Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|