51
|
Huang WC, Tung CL, Yang YCSH, Lin IH, Ng XE, Tung YT. Endurance exercise ameliorates Western diet-induced atherosclerosis through modulation of microbiota and its metabolites. Sci Rep 2022; 12:3612. [PMID: 35256637 PMCID: PMC8901804 DOI: 10.1038/s41598-022-07317-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/08/2022] [Indexed: 12/21/2022] Open
Abstract
The World Health Organization determined cardiovascular disease to be the leading cause of death globally; atherosclerosis is the primary cause of the high morbidity and mortality rates. Regular physical activity is an effective strategy for maintaining endothelial health and function to prevent the development of atherosclerosis. Obesity is also a crucial risk factor for atherosclerotic progression in combination with various complications and systemic inflammation. Physiological homeostasis is modulated by the intestinal microbiota, but the mechanisms through which exercise attenuates atherosclerosis through the microbiota have not been elucidated. Therefore, we investigated the effects of endurance exercise on atherosclerosis induced by a Western diet (WD) and apolipoprotein E (ApoE) knockout in terms of microbiota parameters and metabolites. Genetically modified ApoE knockout mice (C57BL/6-Apoeem1Narl/Narl, ApoEKO) and wild-type mice (C57BL6/J) were divided into the following four groups (n = 6), namely, wild-type mice fed a chow diet (WT CD), ApoEKO mice fed a chow diet (ApoE CD), ApoEKO mice fed a WD (ApoE WD), and ApoEKO mice fed a WD and performing endurance exercise (ApoE WD EX), for a 12-week intervention. The WD significantly induced obesity and atherosclerotic syndrome in the ApoE WD group. Severe atherosclerotic lesions and arterial thickness were significantly elevated and accompanied by increases in VCAM-1, MCP-1, TNF-α, and IL-1β for immune cell chemotaxis and inflammation during atherosclerotic pathogenesis in the ApoE WD group. In addition, dysbiosis in the ApoE WD group resulted in the lowest short-chain fatty acid (SCFA) production. Endurance exercise intervention (ApoE WD EX) significantly alleviated atherosclerotic syndrome by reducing obesity, significantly inhibiting VCAM-1, MCP-1, TNF-α, and IL-1β expression, and increasing the production of SCFAs. Modulation of the microbiota associated with inflammation, such as Desulfovibrio, Tyzzerella, and Lachnospiraceae_ge, and increased SCFA production, particularly through an abundance of Rikenellaceae and Dubosiella, were also observed after exercise intervention. Endurance exercise can alleviate WD-induced atherosclerosis through the amelioration of obesity, inflammation, and chemotaxis signaling, which are modulated by the microbiota and derived SCFAs.
Collapse
Affiliation(s)
- Wen-Ching Huang
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, 112, Taiwan, ROC
| | - Chun-Liang Tung
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 600, Taiwan, ROC.,Department of Food Nutrition and Healthy Biotechnology, Asia University, Taichung, 413, Taiwan, ROC
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 110, Taiwan, ROC
| | - I-Hsuan Lin
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 110, Taiwan, ROC.,Bioinformatics Core Facility, University of Manchester, Manchester, M13 9PT, UK
| | - Xin Er Ng
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, 110, Taiwan, ROC
| | - Yu-Tang Tung
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, 110, Taiwan, ROC. .,Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 402, Taiwan, ROC.
| |
Collapse
|
52
|
Anto L, Blesso CN. Interplay Between Diet, the Gut Microbiome, and Atherosclerosis: Role of Dysbiosis and Microbial Metabolites on Inflammation and Disordered Lipid Metabolism. J Nutr Biochem 2022; 105:108991. [DOI: 10.1016/j.jnutbio.2022.108991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
|
53
|
Li X, Fan Z, Cui J, Li D, Lu J, Cui X, Xie L, Wu Y, Lin Q, Li Y. Trimethylamine N-Oxide in Heart Failure: A Meta-Analysis of Prognostic Value. Front Cardiovasc Med 2022; 9:817396. [PMID: 35252396 PMCID: PMC8888860 DOI: 10.3389/fcvm.2022.817396] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022] Open
Abstract
Objective The present study aimed to explore the prognostic value of trimethylamine N-oxide (TMAO) in heart failure (HF). Methods PubMed, Excerpta Medica Database (EMBASE), Cochrane Library, Web of Science, Wanfang Database, SINOMED, China Science and Technology Journal Database (VIP), and China National Knowledge Infrastructure (CNKI) were searched up to June 1, 2021. Studies recording the major adverse cardiovascular events (MACEs) or all-cause mortality in HF patients and their circulating TMAO concentrations were included. Meta-analysis was performed using Stata 13.0. Results Ten articles (12 studies) involving 13,425 participants from 2014 to 2021 were considered. Compared to low-level TMAO, elevated TMAO was correlated with MACEs and all-cause mortality in HF (RR: 1.28, 95% CI: 1.17, 1.39, P < 0.0001, random-effects model and RR: 1.35, 95% CI: 1.28, 1.42, P < 0.0001, random-effects model, respectively). Consistent results were obtained in all examined subgroups as well as in the sensitivity analysis. Conclusion Elevated TMAO may be an adverse prognostic indicator in patients with HF. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=267208
Collapse
Affiliation(s)
- Xingxing Li
- Second Clinical School of Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zongjing Fan
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Cui
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinjin Lu
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyun Cui
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liandi Xie
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Wu
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yan Li
| |
Collapse
|
54
|
Georgescu D, Caraba A, Ionita I, Lascu A, Hut EF, Dragan S, Ancusa OE, Suceava I, Lighezan D. Dyspepsia and Gut Microbiota in Female Patients with Postcholecystectomy Syndrome. Int J Womens Health 2022; 14:41-56. [PMID: 35136356 PMCID: PMC8816732 DOI: 10.2147/ijwh.s342882] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/06/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gallstone disease (GSD) represents one of the most frequent digestive disorders, highly reported in female gender. The purpose of the study was to explore the clinical and gut microbiota particularities of female patients with postcholecystectomy syndrome (PCS) and the possible relationship between gut dysbiosis (DB) and abdominal complaints. PATIENTS AND METHODS In total, 129 female participants: 104 outpatients divided into two equal groups, 52 PCS (+), 52 PCS (-) and 25 healthy controls were consecutively enrolled in this observational study. Patients underwent clinical examination with assessment of pain, bloating, transit disturbances, abdominal ultrasound/computer tomography/magnetic resonance imaging/endoscopic retrograde cholangiopancreatography, upper and lower digestive endoscopies. Laboratory work-ups and stool microbiology assessments were performed for all study participants (patients and controls). Stool microorganisms were identified by matrix-assisted laser desorption ionization - time-of-flight- mass spectrometry and in patients with DB also by next-generation sequencing. RESULTS Older age, complicated gallstones disease, associated conditions like diabetes mellitus/impaired glucose tolerance and irritable bowel syndrome were significantly present in PCS (+) group, as well as sedentary lifestyle and diets characterized by a low fiber intake (p<0.0001). PCS (+) patients displayed significant differences related to the incidence and severity of overall gut microbiota DB, decreased H index of biodiversity and the unbalanced Firmicutes/Bacteroidetes (F/B) ratios by comparison to the PCS (-) group (p<0.0001). Strong positive correlations of the severity of overall DB with bloating and the intestinal habit disorders, as well as of F/B ratios to all abdominal symptoms were noted. CONCLUSION PCS in female patients was associated with older age, sedentary lifestyle, specific dietary habits, history of complicated gallstone disease, diabetes mellitus/impaired glucose tolerance and irritable bowel syndrome, as well as gut microbiota particularities. Overall DB and unbalanced F/B ratios were strongly correlated to abdominal complaints.
Collapse
Affiliation(s)
- Doina Georgescu
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Alexandru Caraba
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Ionita
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ana Lascu
- Department of Functional Sciences, Discipline of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Emil Florin Hut
- Department IX of Surgery I/Compartment of Hepato-Bilio-Pancreatic Surgery, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Simona Dragan
- Department of Cardiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Oana Elena Ancusa
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Ioana Suceava
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Daniel Lighezan
- Department of Internal Medicine I, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
55
|
Abstract
The increasing prevalence of metabolic diseases has become a severe public health problem. Gut microbiota play important roles in maintaining human health by modulating the host's metabolism. Recent evidences demonstrate that Akkermansia muciniphila is effective in improving metabolic disorders and is thus considered as a promising "next-generation beneficial microbe". In addition to the live A. muciniphila, similar or even stronger beneficial effects have been observed in pasteurized A. muciniphila and its components, including the outer membrane protein Amuc_1100, A. muciniphila-derived extracellular vesicles (AmEVs), and secreted protein P9. Hence, this paper presents a systemic review of recent progress in the effects and mechanisms of A. muciniphila and its components in the treatment of metabolic diseases, including obesity, type 2 diabetes mellitus, cardiovascular disease, and nonalcoholic fatty liver disease, as well as perspectives on its future study.
Collapse
Affiliation(s)
- Juan Yan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Sheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Lili Sheng
| | - Houkai Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China,CONTACT Houkai Li Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai201203, China
| |
Collapse
|
56
|
Bandopadhyay P, Ganguly D. Gut dysbiosis and metabolic diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:153-174. [DOI: 10.1016/bs.pmbts.2022.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
57
|
Li Y, Zhao D, Qian M, Liu J, Pan C, Zhang X, Duan X, Zhang Y, Jia W, Wang L. Amlodipine, an anti-hypertensive drug, alleviates non-alcoholic fatty liver disease by modulating gut microbiota. Br J Pharmacol 2021; 179:2054-2077. [PMID: 34862599 DOI: 10.1111/bph.15768] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/08/2021] [Accepted: 11/21/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-alcoholic fatty liver disease (NAFLD) represents a severe public health problem. It often coexists with hypertension in the context of metabolic syndrome. Here, we investigated the effects of amlodipine on non-alcoholic fatty liver disease combined with hypertension and the underlying mechanism. EXPERIMENTAL APPROACH mice were fed with high-fat diet and 0.05% N-Nitro-L-arginine methylester sterile water to induce NAFLD with hypertension. Gut microbiota composition and function were assessed by 16S ribosomal DNA and metagenomic sequencing. Untargeted metabolome profiles were applied to identify differential metabolites in mice cecum. KEY RESULTS Amlodipine besylate (AB) and amlodipine aspartate (AA) significantly decreased liver injury, hepatic steatosis and improved lipid metabolism with a concomitant reduction in the expression of lipogenic genes in mice with NAFLD and hypertension. Mechanistically, AA and AB have potential in restoring intestinal barrier integrity and improving antimicrobial defense along with the elevated abundances of Akkermansia, Bacteroides and Lactobacillus. Noteworthily, the gut microbiota in AB and AA-treated mice had higher abundance of functional genes involved in taurine and hypotaurine metabolism. Consistently, the strengthened taurine and hypotaurine metabolism was confirmed by the untargeted metabolome analysis. Based on the correlation and causal analysis, the altered gut microbiota composition and the enhancement of taurine and hypotaurine metabolism may synergistically decreased ALT, liver triglycerides, lipogenic genes and plasma cholesterol in HFD-fed hypertensive mice. CONCLUSION AND IMPLICATIONS Collectively, AA and AB exert multi-factorial improvements in NAFLD and hypertension by modulating gut microbiota, and may serve as a promising therapeutic agent for treating these diseases.
Collapse
Affiliation(s)
- Yang Li
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Danyang Zhao
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Minyi Qian
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chuyue Pan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinxin Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xubin Duan
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yufei Zhang
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wenxin Jia
- School of Basic Medicine and Clinical Pharmacy, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lirui Wang
- Institute of Modern Biology, Nanjing University, Nanjing, China
| |
Collapse
|
58
|
Li L, Zhang Y, Speakman JR, Hu S, Song Y, Qin S. The gut microbiota and its products: Establishing causal relationships with obesity related outcomes. Obes Rev 2021; 22:e13341. [PMID: 34490704 DOI: 10.1111/obr.13341] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Gut microorganisms not only participate in the metabolism of carbohydrate, lipids, protein, and polypeptides in the intestine but also directly affect the metabolic phenotypes of the host. Although many studies have described the apparent effects of gut microbiota on human health, the development of metagenomics and culturomics in the past decade has generated a large amount of evidence suggesting a causal relationship between gut microbiota and obesity. The interaction between the gut microbiota and host is realized by microbial metabolites with multiple biological functions. We concentrated here on several representative beneficial species connected with obesity as well as the mechanisms, with particular emphasis on microbiota-dependent metabolites. Finally, we consider the potential clinical significance of these relationships to fuel the conception and realization of novel therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Yubing Zhang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,College of Life Sciences, Yantai University, Yantai, China
| | - John Roger Speakman
- Shenzhen Key Laboratory for Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shanliang Hu
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Yipeng Song
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
59
|
Probiotics Interactions and the Modulation of Major Signalling Pathways in Host Model Organism Caenorhabditis elegans. Indian J Microbiol 2021; 61:404-416. [PMID: 34744196 DOI: 10.1007/s12088-021-00961-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022] Open
Abstract
Microorganisms live in the human digestive system and the gut microbiome constitutes part of our prime determining component for healthy aging and wellness. Gut microbiota has broad influences on its host, beginning from the digestion of food and nutrients absorption to protective roles against invading pathogens and host immune system regulation. Dysbiosis of the gut microbial composition has been linked to numerous diseases and there is a need to have a better grasp on what makes a 'good' gut microbiome. Caenorhabditis elegans (C. elegans) model organism is considered as a well-suited in-vivo model system and, is at the frontline of probiotic research because of its well-defined characteristics and prolific nature. Most importantly, C. elegans feeds on bacteria, which speeds up manipulations and investigations in probiotics research tremendously. With its unique salient features of short lifespan, and ease of propagation, different unknown probiotics biological roles can be measured at an organism level with precision in the form of worm's stress responses, survivability, and lifespan. In this review, new insights on the different mechanisms underlying the establishment of probiotics regulations of conserved signalling pathways such as p38 MAPK/SKN-1, DAF-2/DAF-16, and JNK-1/DAF-16 is highlighted based on information obtained from C. elegans studies. Along with the current state of knowledge and the uniqueness of C. elegans as a model organism, explorations of its future contribution and scope in synthetic biology and probiotics engineering strains are also addressed. This is expected to strengthen our understanding of probiotics roles and to facilitate novel discovery and applications, for specific therapeutics against age-related disorders and various pathophysiological conditions.
Collapse
|
60
|
Animal Models of Neointimal Hyperplasia and Restenosis: Species-Specific Differences and Implications for Translational Research. JACC Basic Transl Sci 2021; 6:900-917. [PMID: 34869956 PMCID: PMC8617545 DOI: 10.1016/j.jacbts.2021.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/29/2022]
Abstract
Neointimal hyperplasia is the major factor contributing to restenosis after angioplasty procedures. Multiple animal models exist to study basic and translational aspects of restenosis formation. Animal models differ substantially, and species-specific differences have major impact on the pathophysiology of the model. Genetic, dietary, and mechanical interventions determine the translational potential of the animal model used and have to be considered when choosing the model.
The process of restenosis is based on the interplay of various mechanical and biological processes triggered by angioplasty-induced vascular trauma. Early arterial recoil, negative vascular remodeling, and neointimal formation therefore limit the long-term patency of interventional recanalization procedures. The most serious of these processes is neointimal hyperplasia, which can be traced back to 4 main mechanisms: endothelial damage and activation; monocyte accumulation in the subintimal space; fibroblast migration; and the transformation of vascular smooth muscle cells. A wide variety of animal models exists to investigate the underlying pathophysiology. Although mouse models, with their ease of genetic manipulation, enable cell- and molecular-focused fundamental research, and rats provide the opportunity to use stent and balloon models with high throughput, both rodents lack a lipid metabolism comparable to humans. Rabbits instead build a bridge to close the gap between basic and clinical research due to their human-like lipid metabolism, as well as their size being accessible for clinical angioplasty procedures. Every different combination of animal, dietary, and injury model has various advantages and disadvantages, and the decision for a proper model requires awareness of species-specific biological properties reaching from vessel morphology to distinct cellular and molecular features.
Collapse
Key Words
- Apo, apolipoprotein
- CETP, cholesteryl ester transferase protein
- ECM, extracellular matrix
- FGF, fibroblast growth factor
- HDL, high-density lipoprotein
- LDL, low-density lipoprotein
- LDLr, LDL receptor
- PDGF, platelet-derived growth factor
- TGF, transforming growth factor
- VLDL, very low-density lipoprotein
- VSMC, vascular smooth muscle cell
- angioplasty
- animal model
- neointimal hyperplasia
- restenosis
Collapse
|
61
|
Wu Q, Luo F, Wang XL, Lin Q, Liu GQ. Angiotensin I-converting enzyme inhibitory peptide: an emerging candidate for vascular dysfunction therapy. Crit Rev Biotechnol 2021; 42:736-755. [PMID: 34634988 DOI: 10.1080/07388551.2021.1948816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abnormal vasoconstriction, inflammation, and vascular remodeling can be promoted by angiotensin II (Ang II) in the renin-angiotensin system (RAS), leading to vascular dysfunction diseases such as hypertension and atherosclerosis. Researchers have recently focused on angiotensin I-converting enzyme inhibitory peptides (ACEIPs), that have desirable efficacy in vascular dysfunction therapy due to Ang II reduction by inhibiting ACE activity. Promising methods for the large-scale preparation of ACEIPs include selective enzymatic hydrolysis and microbial fermentation. Thus far, ACEIPs have been widely reported to be hydrolyzed from protein-rich sources, including animals, plants, and marine organisms, while many emerging microorganism-derived ACEIPs are theoretically biosynthesized through the nonribosomal peptide synthase (NRPS) pathway. Notably, vasodilatation, anti-inflammation, and vascular reconstruction reversal of ACEIPs are strongly correlated. However, the related molecular mechanisms underlying signal transduction regulation in vivo remain unclear. We provide a comprehensive update of the ACE-Ang II-G protein-coupled type 1 angiotensin receptor (AT1R) axis signaling and its functional significance for potential translation into therapeutic strategies, particularly targeting AT1R by ACEIPs, as well as specific related signaling pathways. Future studies are expected to verify the biosynthetic regulatory mechanism of ACEIPs via the NRPS pathway, the effect of gut microbiota metabolism on vascular dysfunction and rigorous studies of ACE-Ang II-AT1R signaling pathways mediated by ACEIPs in large animals and humans.
Collapse
Affiliation(s)
- Qiang Wu
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food and Chemical Engineering, Shaoyang University, Shaoyang, China
| | - Feijun Luo
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Xiao-Ling Wang
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China
| | - Qinlu Lin
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Gao-Qiang Liu
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
62
|
Zhao C, Qu Q, Yang F, Li Z, Yang P, Han L, Shi X. Monascus ruber fermented Panax ginseng ameliorates lipid metabolism disorders and modulate gut microbiota in rats fed a high-fat diet. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114300. [PMID: 34098018 DOI: 10.1016/j.jep.2021.114300] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng (Panax ginseng Meyer) is rich in a variety of biologically active ingredients, which shows good effect in the treatment of metabolic diseases. Monascus has lipid-lowering activity and one of its metabolites, lovastatin, is widely used in clinical practice. AIM OF THE STUDY The main purpose of this study was to clarify the effects of fermented Panax ginseng by Monascus ruber (PM) on lipid metabolism and gut microbiota in rats fed a high-fat diet. MATERIALS AND METHODS SPF Sprague-Dawley rats were randomly divided into 5 groups, the therapeutic effect of PM on HFD-induced obesity, hyperlipidemia, hepatic steatosis, and disordered gut microbiota were determined in rats. RESULTS PM could attenuate features of obesity in rats, decrease serum TC, LDL-C and IgA levels, increase excretion of bile acids in feces. Hepatic histopathologic analysis revealed that PM decrease lipid accumulation in hepatocytes. Consistently, mRNA expression levels of cholesterol metabolism-related genes were regulated in the livers of HFD-fed rats administered with PM. In addition, PM could enhance the diversity and relative abundance of gut microbiota, reduce the Firmicutes/Bacteroidetes (F/B) ratio, increase significantly the relative abundance of Prevotella_9, and decrease these of Muribaculaceae. CONCLUSIONS PM could regulate lipid metabolism and the structure of the gut microbiota in the HFD rats. Our findings provide valuable experience for the development of ginseng. PM could be a potentially effective strategy to prevent and treat metabolic diseases and alleviate the gut microbiota disturbance caused by it.
Collapse
Affiliation(s)
- Chongyan Zhao
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Qingsong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Fang Yang
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Zhixun Li
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Pengshuo Yang
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Lu Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China.
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, 102488, Beijing, China; Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing, 100029, China.
| |
Collapse
|
63
|
Cheng CK, Huang Y. The gut-cardiovascular connection: new era for cardiovascular therapy. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:23-46. [PMID: 37724079 PMCID: PMC10388818 DOI: 10.1515/mr-2021-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
Our gut microbiome is constituted by trillions of microorganisms including bacteria, archaea and eukaryotic microbes. Nowadays, gut microbiome has been gradually recognized as a new organ system that systemically and biochemically interact with the host. Accumulating evidence suggests that the imbalanced gut microbiome contributes to the dysregulation of immune system and the disruption of cardiovascular homeostasis. Specific microbiome profiles and altered intestinal permeability are often observed in the pathophysiology of cardiovascular diseases. Gut-derived metabolites, toxins, peptides and immune cell-derived cytokines play pivotal roles in the induction of inflammation and the pathogenesis of dysfunction of heart and vasculature. Impaired crosstalk between gut microbiome and multiple organ systems, such as gut-vascular, heart-gut, gut-liver and brain-gut axes, are associated with higher cardiovascular risks. Medications and strategies that restore healthy gut microbiome might therefore represent novel therapeutic options to lower the incidence of cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science; The Chinese University of Hong Kong, Hong Kong SAR999077, China
- Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR999077, China
| | - Yu Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science; The Chinese University of Hong Kong, Hong Kong SAR999077, China
- Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR999077, China
| |
Collapse
|
64
|
Ahrens AP, Culpepper T, Saldivar B, Anton S, Stoll S, Handberg EM, Xu K, Pepine C, Triplett EW, Aggarwal M. A Six-Day, Lifestyle-Based Immersion Program Mitigates Cardiovascular Risk Factors and Induces Shifts in Gut Microbiota, Specifically Lachnospiraceae, Ruminococcaceae, Faecalibacterium prausnitzii: A Pilot Study. Nutrients 2021; 13:3459. [PMID: 34684459 PMCID: PMC8539164 DOI: 10.3390/nu13103459] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) prevalence remains elevated globally. We have previously shown that a one-week lifestyle "immersion program" leads to clinical improvements and sustained improvements in quality of life in moderate to high atherosclerotic CVD (ASCVD) risk individuals. In a subsequent year of this similarly modeled immersion program, we again collected markers of cardiovascular health and, additionally, evaluated intestinal microbiome composition. ASCVD risk volunteers (n = 73) completed the one-week "immersion program" involving nutrition (100% plant-based foods), stress management education, and exercise. Anthropometric measurements and CVD risk factors were compared at baseline and post intervention. A subgroup (n = 22) provided stool, which we analyzed with 16S rRNA sequencing. We assessed abundance changes within-person, correlated the abundance shifts with clinical changes, and inferred functional pathways using PICRUSt. Reductions in blood pressure, total cholesterol, and triglycerides, were observed without reduction in weight. Significant increases in butyrate producers were detected, including Lachnospiraceae and Oscillospirales. Within-person, significant shifts in relative abundance (RA) occurred, e.g., increased Lachnospiraceae (+58.8% RA, p = 0.0002), Ruminococcaceae (+82.1%, p = 0.0003), Faecalibacterium prausnitzii (+54.5%, p = 0.002), and diversification and richness. Microbiota changes significantly correlated with body mass index (BMI), blood pressure (BP), cholesterol, high-sensitivity C-reactive protein (hsCRP), glucose, and trimethylamine N-oxide (TMAO) changes. Pairwise decreases were inferred in microbial genes corresponding to cancer, metabolic disease, and amino acid metabolism. This brief lifestyle-based intervention improved lipids and BP and enhanced known butyrate producers, without significant weight loss. These results demonstrate a promising non-pharmacological preventative strategy for improving cardiovascular health.
Collapse
Affiliation(s)
- Angelica P. Ahrens
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, 1355 Museum Dr, Gainesville, FL 32603, USA; (A.P.A.); (E.W.T.)
| | - Tyler Culpepper
- Department of Medicine, College of Medicine, University of Florida, P.O. Box 100277, Gainesville, FL 32610, USA; (T.C.); (B.S.)
| | - Brittany Saldivar
- Department of Medicine, College of Medicine, University of Florida, P.O. Box 100277, Gainesville, FL 32610, USA; (T.C.); (B.S.)
| | - Stephen Anton
- Department of Aging and Geriatric Research, University of Florida, 210 E. Mowry Rd, Gainesville, FL 32611, USA;
| | - Scott Stoll
- Total Health Immersions, P.O. Box 741596, Boynton Beach, FL 33474, USA;
| | - Eileen M. Handberg
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; (E.M.H.); (C.P.)
| | - Ke Xu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, 2004 Mowry Rd, Gainesville, FL 32610, USA;
| | - Carl Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; (E.M.H.); (C.P.)
| | - Eric W. Triplett
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, 1355 Museum Dr, Gainesville, FL 32603, USA; (A.P.A.); (E.W.T.)
| | - Monica Aggarwal
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32610, USA; (E.M.H.); (C.P.)
| |
Collapse
|
65
|
Huang M, Zhu L, Jin Y, Fang Z, Chen Y, Yao Y. Association between Helicobacter Pylori Infection and Systemic Arterial Hypertension: A Meta-Analysis. Arq Bras Cardiol 2021; 117:626-636. [PMID: 34550168 PMCID: PMC8528373 DOI: 10.36660/abc.20200186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023] Open
Abstract
Fundamento: Estudos epidemiológicos recentes demonstraram que alterações na microbiota e seus metabólitos estão associadas à hipertensão arterial sistêmica. A Helicobacter pylori (H. pylori) é um dos patógenos bacterianos mais comuns, e a possível associação entre a infecção por H. pylori e a hipertensão é controversa. Objetivos: Este estudo teve o objetivo de esclarecer a associação entre eles e proporcionar uma nova base teórica para detectar a patogênese da hipertensão. Métodos: Foram selecionados estudos caso-controle e transversais sobre a associação entre H. pylori e hipertensão, publicados de 1996 a 2019 indexados nos bancos de dados PubMed, Google Scholar, Chinese Wan Fang Data, e Chinese National Knowledge Infrastructure (CNKI). As razões de chance (RC) combinadas e o intervalo de confiança (IC) 95% foram estimados. O I² foi realizado para avaliar a heterogeneidade estatística. O viés de publicação foi avaliado utilizando-se os testes de Beggs e de Egger. Os dados extraídos foram analisados no software Stata 12.0. A significância estatística foi definida com um p-valor < 0,05. Resultados: Foram cadastrados 17 estudos envolvendo 6376 casos de hipertensão e 10850 controles. A taxa de infecção por H. pylori em pacientes hipertensos e em controles foi de 64,9% e 56,3%, respectivamente. Foi demonstrada uma associação significativamente positiva entre a infecção por H. pylori e a hipertensão, com uma RC global de 2,07 (IC 95%: 1,46–2,94; p < 0,05). A análise de subgrupos revelou que a prevalência de infecção por H. pylori foi associada à hipertensão na região da Ásia e no grupo de caso-controle, as RC (IC 95%) foram 2,26 (1,51-3,38) e 2,53 (1,72-3,72), respectivamente. Depois de estratificar por métodos de detecção, ainda existiam diferenças entre os subgrupos (todos p < 0,05). Conclusão: Esta metanálise indicou que a infecção por H. pylori está associada positivamente à hipertensão.
Collapse
Affiliation(s)
- Mengyun Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China
| | - Lijun Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China
| | - Yuelong Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China
| | - Zhengmei Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China
| | - Yingshui Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College/ Institute of Chronic Disease Prevention and Control, Wuhu - China.,Department of Medicine, Anhui College of Traditional Chinese Medicine, Wuhu - China
| |
Collapse
|
66
|
Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications-Is There a Missing Link? Microorganisms 2021; 9:microorganisms9091913. [PMID: 34576810 PMCID: PMC8472650 DOI: 10.3390/microorganisms9091913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/21/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome is represented by the genome of all microorganisms (symbiotic, potential pathogens, or pathogens) residing in the intestine. These ecological communities are involved in almost all metabolic diseases and cardiovascular diseases are not excluded. Atherosclerosis, with a continuously increasing incidence in recent years, is the leading cause of coronary heart disease and stroke by plaque rupture and intraplaque hemorrhage. Vascular calcification, a process very much alike with osteogenesis, is considered to be a marker of advanced atherosclerosis. New evidence, suggesting the role of dietary intake influence on the diversity of the gut microbiome in the development of vascular calcifications, is highly debated. Gut microbiota can metabolize choline, phosphatidylcholine, and L-carnitine and produce vasculotoxic metabolites, such as trimethylamine-N-oxide (TMAO), a proatherogenic metabolite. This review article aims to discuss the latest research about how probiotics and the correction of diet is impacting the gut microbiota and its metabolites in the atherosclerotic process and vascular calcification. Further studies could create the premises for interventions in the microbiome as future primary tools in the prevention of atherosclerotic plaque and vascular calcifications.
Collapse
|
67
|
AbdelMassih A, Hassan AA, Abou-Zeid AS, Hassan A, Hussein E, Gadalla M, Hussein M, Eid MA, Elahmady M, El Nahhas N, Emad N, Zahra N, Aboushadi N, Ibrahim N, Mokhtar S, Ismail HA, El-Husseiny N, Moharam RK, Menshawey E, Menshawey R. Salivary markers and coronavirus disease 2019: insights from cross-talk between the oral microbiome and pulmonary and systemic low-grade inflammation and implications for vascular complications. Cardiovasc Endocrinol Metab 2021; 10:162-167. [PMID: 34386717 PMCID: PMC8352626 DOI: 10.1097/xce.0000000000000242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022]
Abstract
To date, coronavirus disease 2019 (COVID-19) has affected over 6.2 million individuals worldwide, including 1.46 million deaths. COVID-19 complications are mainly induced by low-grade inflammation-causing vascular degeneration. There is an increasing body of evidence that suggests that oral dysbiotic taxa are associated with worse prognosis in COVID-19 patients, especially the Prevotella genus, which was retrieved from nasopharyngeal and bronchoalveolar lavage samples in affected patients. Oral dysbiosis may act by increasing the likelihood of vascular complications through low-grade inflammation, as well as impairing respiratory mucosal barrier mechanisms against SARS-CoV-2. Salivary markers can be used to reflect this oral dysbiosis and its subsequent damaging effects on and the lungs and vasculature. Salivary sampling can be self-collected, and is less costly and less invasive, and thus may be a superior option to serum markers in risk stratification of COVID-19 patients. Prospective studies are needed to confirm such hypothesis. Video Abstract: http://links.lww.com/CAEN/A28.
Collapse
Affiliation(s)
- Antoine AbdelMassih
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Giza
- Pediatric Cardio-Oncology Department, Children Cancer Hospital of Egypt
| | - Alaa A Hassan
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Aya S Abou-Zeid
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Aya Hassan
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Engy Hussein
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Mahenar Gadalla
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Mahinour Hussein
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Maryam A Eid
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Maryam Elahmady
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Nadine El Nahhas
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Nadine Emad
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Nihal Zahra
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Nour Aboushadi
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Nourhan Ibrahim
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Sherouk Mokhtar
- Research Department, School of Oral and Dental Medicine, New Giza University, New Giza
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | | | - Nadine El-Husseiny
- Research Department, Faculty of Medicine, Cairo University, Cairo
- Department of Oral and Maxillo-facial Surgery, Faculty of Dentistry, Cairo University, Giza
- Department of Scientific Design, Pixagon Graphic Design Agency, Cairo
| | - Reham Khaled Moharam
- Residency Training Program of Plastic Surgery Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Esraa Menshawey
- Research Department, Faculty of Medicine, Cairo University, Cairo
| | - Rahma Menshawey
- Research Department, Faculty of Medicine, Cairo University, Cairo
| |
Collapse
|
68
|
He S, Jiang H, Zhuo C, Jiang W. Trimethylamine/Trimethylamine-N-Oxide as a Key Between Diet and Cardiovascular Diseases. Cardiovasc Toxicol 2021; 21:593-604. [PMID: 34003426 DOI: 10.1007/s12012-021-09656-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/27/2021] [Indexed: 02/08/2023]
Abstract
Trimethylamine (TMA) is a gut microbiota-derived metabolite which comes from diets rich of choline, betaine or L-carnitine and could be further converted to Trimethylamine-N-oxide (TMAO) in the liver. As the function of gut microbiota and its metabolites being explored so far, studies suggest that TMAO may be a potential risk factor of cardiovascular diseases independent of other traditional risk factors. However, the precise role of TMAO is controversial as some converse results were discovered. In recent studies, it is hypothesized that TMA may also participate in the progression of cardiovascular diseases and some cytotoxic effect of TMA has been discovered. Thus, exploring the relationship between TMA, TMAO and CVD may bring a novel insight into the diagnosis and therapy of cardiovascular diseases. In this review, we discussed the factors which influence the TMA/TMAO's process of metabolism in the human body. We have also summarized the pathogenic effect of TMA/TMAO in cardiovascular diseases, as well as the limitation of some controversial discoveries.
Collapse
Affiliation(s)
- Siyu He
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Hong Jiang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Caili Zhuo
- The Laboratory of Cardiovascular Diseases, Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wei Jiang
- The Laboratory of Cardiovascular Diseases, Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
69
|
The Role of Gut Microbiota on Cholesterol Metabolism in Atherosclerosis. Int J Mol Sci 2021; 22:ijms22158074. [PMID: 34360839 PMCID: PMC8347163 DOI: 10.3390/ijms22158074] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Hypercholesterolemia plays a causal role in the development of atherosclerosis and is one of the main risk factors for cardiovascular disease (CVD), the leading cause of death worldwide especially in developed countries. Current data show that the role of microbiota extends beyond digestion by being implicated in several metabolic and inflammatory processes linked to several diseases including CVD. Studies have reported associations between bacterial metabolites and hypercholesterolemia. However, such associations remain poorly investigated and characterized. In this review, the mechanisms of microbial derived metabolites such as primary and secondary bile acids (BAs), trimethylamine N-oxide (TMAO), and short-chain fatty acids (SCFAs) will be explored in the context of cholesterol metabolism. These metabolites play critical roles in maintaining cardiovascular health and if dysregulated can potentially contribute to CVD. They can be modulated via nutritional and pharmacological interventions such as statins, prebiotics, and probiotics. However, the mechanisms behind these interactions also remain unclear, and mechanistic insights into their impact will be provided. Therefore, the objectives of this paper are to present current knowledge on potential mechanisms whereby microbial metabolites regulate cholesterol homeostasis and to discuss the feasibility of modulating intestinal microbes and metabolites as a novel therapeutic for hypercholesterolemia.
Collapse
|
70
|
Yang S, Li D, Yu Z, Li Y, Wu M. Multi-Pharmacology of Berberine in Atherosclerosis and Metabolic Diseases: Potential Contribution of Gut Microbiota. Front Pharmacol 2021; 12:709629. [PMID: 34305616 PMCID: PMC8299362 DOI: 10.3389/fphar.2021.709629] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis (AS), especially atherosclerotic cardiovascular diseases (ASCVDs), and metabolic diseases (such as diabetes, obesity, dyslipidemia, and nonalcoholic fatty liver disease) are major public health issues worldwide that seriously threaten human health. Exploring effective natural product-based drugs is a promising strategy for the treatment of AS and metabolic diseases. Berberine (BBR), an important isoquinoline alkaloid found in various medicinal plants, has been shown to have multiple pharmacological effects and therapeutic applications. In view of its low bioavailability, increasing evidence indicates that the gut microbiota may serve as a target for the multifunctional effects of BBR. Under the pathological conditions of AS and metabolic diseases, BBR improves intestinal barrier function and reduces inflammation induced by gut microbiota-derived lipopolysaccharide (LPS). Moreover, BBR reverses or induces structural and compositional alterations in the gut microbiota and regulates gut microbe-dependent metabolites as well as related downstream pathways; this improves glucose and lipid metabolism and energy homeostasis. These findings at least partly explain the effect of BBR on AS and metabolic diseases. In this review, we elaborate on the research progress of BBR and its mechanisms of action in the treatment of AS and metabolic diseases from the perspective of gut microbiota, to reveal the potential contribution of gut microbiota to the multifunctional biological effects of BBR.
Collapse
Affiliation(s)
- Shengjie Yang
- Guang’an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujuan Li
- Guang’an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
71
|
Silva CBP, Elias-Oliveira J, McCarthy CG, Wenceslau CF, Carlos D, Tostes RC. Ethanol: striking the cardiovascular system by harming the gut microbiota. Am J Physiol Heart Circ Physiol 2021; 321:H275-H291. [PMID: 34142885 DOI: 10.1152/ajpheart.00225.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ethanol consumption represents a significant public health problem, and excessive ethanol intake is a risk factor for cardiovascular disease (CVD), one of the leading causes of death and disability worldwide. The mechanisms underlying the effects of ethanol on the cardiovascular system are complex and not fully comprehended. The gut microbiota and their metabolites are indispensable symbionts essential for health and homeostasis and therefore, have emerged as potential contributors to ethanol-induced cardiovascular system dysfunction. By mechanisms that are not completely understood, the gut microbiota modulates the immune system and activates several signaling pathways that stimulate inflammatory responses, which in turn, contribute to the development and progression of CVD. This review summarizes preclinical and clinical evidence on the effects of ethanol in the gut microbiota and discusses the mechanisms by which ethanol-induced gut dysbiosis leads to the activation of the immune system and cardiovascular dysfunction. The cross talk between ethanol consumption and the gut microbiota and its implications are detailed. In summary, an imbalance in the symbiotic relationship between the host and the commensal microbiota in a holobiont, as seen with ethanol consumption, may contribute to CVD. Therefore, manipulating the gut microbiota, by using antibiotics, probiotics, prebiotics, and fecal microbiota transplantation might prove a valuable opportunity to prevent/mitigate the deleterious effects of ethanol and improve cardiovascular health and risk prevention.
Collapse
Affiliation(s)
- Carla B P Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jefferson Elias-Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Camilla F Wenceslau
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
72
|
Stepanova N. Role of Impaired Oxalate Homeostasis in Cardiovascular Disease in Patients With End-Stage Renal Disease: An Opinion Article. Front Pharmacol 2021; 12:692429. [PMID: 34122117 PMCID: PMC8193726 DOI: 10.3389/fphar.2021.692429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Natalia Stepanova
- State Institution “Institute of Nephrology National Academy of Medical Science of Ukraine”, Kyiv, Ukraine
| |
Collapse
|
73
|
Lakshmanan AP, Shatat IF, Zaidan S, Jacob S, Bangarusamy DK, Al-Abduljabbar S, Al-Khalaf F, Petroviski G, Terranegra A. Bifidobacterium reduction is associated with high blood pressure in children with type 1 diabetes mellitus. Biomed Pharmacother 2021; 140:111736. [PMID: 34034069 DOI: 10.1016/j.biopha.2021.111736] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Children with Type 1 diabetes mellitus (T1DM) have an elevated risk of abnormal blood pressure (BP) measurements and patterns. Both hypertension and T1DM are well-known risk factors for cardiovascular disease and kidney failure. The human microbiome has been linked to both diabetes and hypertension, but the relationship between the gut microbiome and BP in children with T1DM is not well-understood. In this cross-sectional study, we examined the relationship between resting office BP and gut microbiota composition, diversity, and richness in children with T1DM and healthy controls. We recruited 29 pediatric subjects and divided them into three groups: healthy controls (HC, n = 5), T1DM with normal BP (T1DM-Normo, n = 17), and T1DM with elevated BP (T1DM-HBP, n = 7). We measured the BP, dietary and clinical parameters for each subject. We collected fecal samples to perform the 16s rDNA sequencing and to measure the short-chain fatty acids (SCFAs) level. The microbiome downstream analysis included the relative abundance of microbiota, alpha and beta diversity, microbial markers using Linear Discriminant effect size analysis (LEfSe), potential gut microbial metabolic pathways using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) and metabolic pathways validation using Statistical Inference of Associations between Microbial Communities And host phenotype (SIAMCAT) machine learning toolbox. Our study results showed that T1DM-HBP group had distinct gut microbial composition (at multiple taxonomic levels) and reduced diversity (richness and abundance) compared with T1DM-Normo and HC groups. Children with T1DM-HBP showed a significant reduction of Bifidobacterium levels (especially B. adolescentis, B. bifidum, and B. longum) compared to the T1DM-Normo group. We also observed unique gut-microbial metabolic pathways, such as elevated lipopolysaccharide synthesis and glutathione metabolism in children with T1DM-HBP compared to T1DM-Normo children. We can conclude that the reduction in the abundance of genus Bifidobacterium could play a significant role in elevating the BP in pediatric T1DM subjects. More studies are needed to corroborate our findings and further explore the potential contributing mechanisms we describe.
Collapse
Affiliation(s)
| | - Ibrahim F Shatat
- Pediatric Nephrology and Hypertension, Sidra Medicine, HB. 7A. 106A, P.O. Box 26999, Doha, Qatar; Weill Cornell College of Medicine-Qatar, Ar-Rayyan, Doha, Qatar; Medical University of South Carolina, Charleston, SC, USA
| | - Sara Zaidan
- Research Department, Sidra Medicine, OPC, P.O. Box 26999, Doha, Qatar
| | - Shana Jacob
- Research Department, Sidra Medicine, OPC, P.O. Box 26999, Doha, Qatar
| | | | | | - Fawziya Al-Khalaf
- Pediatric Endocrinology, Sidra Medicine, OPC, P.O. Box 26999, Doha, Qatar
| | - Goran Petroviski
- Pediatric Endocrinology, Sidra Medicine, OPC, P.O. Box 26999, Doha, Qatar
| | | |
Collapse
|
74
|
Künstner A, Aherrahrou R, Hirose M, Bruse P, Ibrahim SM, Busch H, Erdmann J, Aherrahrou Z. Effect of Differences in the Microbiome of Cyp17a1-Deficient Mice on Atherosclerotic Background. Cells 2021; 10:1292. [PMID: 34070975 PMCID: PMC8224745 DOI: 10.3390/cells10061292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
CYP17A1 is a cytochrome P450 enzyme that has 17-alpha-hydroxylase and C17,20-lyase activities. Cyp17a11 deficiency is associated with high body mass and visceral fat deposition in atherosclerotic female ApoE knockout (KO, d/d or -/-) mice. In the present study, we aimed to investigate the effects of diet and Cyp17a1 genotype on the gut microbiome. Female Cyp17a1 (d/d) × ApoE (d/d) (DKO) and ApoE (d/d) (controls) were fed either standard chow or a Western-type diet (WTD), and we demonstrated the effects of genetics and diet on the body mass of the mice and composition of their gut microbiome. We found a significantly lower alpha diversity after accounting for the ecological network structure in DKO mice and WTD-fed mice compared with chow-fed ApoE(d/d). Furthermore, we found a strong significant positive association of the Firmicutes vs. Bacteroidota ratio with body mass and the circulating total cholesterol and triglyceride concentrations of the mice when feeding the WTD, independent of the Cyp17a1 genotype. Further pathway enrichment and network analyses revealed a substantial effect of Cyp17a1 genotype on associated cardiovascular and obesity-related pathways involving aspartate and L-arginine. Future studies are required to validate these findings and further investigate the role of aspartate/L-arginine pathways in the obesity and body fat distribution in our mouse model.
Collapse
Affiliation(s)
- Axel Künstner
- Medical Systems Biology Group, Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.K.); (H.B.)
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Redouane Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- Centre for Public Health Genomics, Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908-0717, USA
| | - Misa Hirose
- Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (S.M.I.)
| | - Petra Bruse
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Saleh Mohamed Ibrahim
- Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (M.H.); (S.M.I.)
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute for Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.K.); (H.B.)
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- DZHK (German Centre for Cardiovascular Research), University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany; (R.A.); (P.B.); (J.E.)
- DZHK (German Centre for Cardiovascular Research), University Heart Centre Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
75
|
Dubinski P, Czarzasta K, Cudnoch-Jedrzejewska A. The Influence of Gut Microbiota on the Cardiovascular System Under Conditions of Obesity and Chronic Stress. Curr Hypertens Rep 2021; 23:31. [PMID: 34014393 PMCID: PMC8137478 DOI: 10.1007/s11906-021-01144-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Based on the available data, it can be assumed that microbiota is an integral part of the human body. The most heavily colonized area of the human body is the gut, with bacterial accumulation ranging from 101-103 cells/g in the upper intestine to 1011-1012 cells/g in the colon. However, colonization of the gut is not the same throughout, as it was shown that there are differences between the composition of the microbiota in the intestine lumen and in the proximity of the mucus layer. RECENT FINDINGS Gut microbiota gradient can be differentially regulated by factors such as obesity and chronic stress. In particular, a high fat diet influences the gut microbial composition. It was also found that chronic stress may cause the development of obesity and thus change the organization of the intestinal barrier. Recent research has shown the significant effect of intestinal microflora on cardiovascular function. Enhanced absorption of bacterial fragments, such as lipopolysaccharide (LPS), promotes the onset of "metabolic endotoxemia," which could activate toll-like receptors, which mediates an inflammatory response and in severe cases could cause cardiovascular diseases. It is presumed that the intestinal microbiota, and especially its metabolites (LPS and trimethylamine N-oxide (TMAO)), may play an important role in the pathogenesis of arterial hypertension, atherosclerosis, and heart failure. This review focuses on how gut microbiota can change the morphological and functional activity of the cardiovascular system in the course of obesity and in conditions of chronic stress.
Collapse
Affiliation(s)
- Piotr Dubinski
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Katarzyna Czarzasta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
76
|
Yang Y, Zhao M, He X, Wu Q, Li DL, Zang WJ. Pyridostigmine Protects Against Diabetic Cardiomyopathy by Regulating Vagal Activity, Gut Microbiota, and Branched-Chain Amino Acid Catabolism in Diabetic Mice. Front Pharmacol 2021; 12:647481. [PMID: 34084135 PMCID: PMC8167056 DOI: 10.3389/fphar.2021.647481] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The disruption of gut microbes is associated with diabetic cardiomyopathy, but the mechanism by which gut microbes affect cardiac damage remains unclear. We explored gut microbes and branched-chain amino acid (BCAA) metabolite catabolism in diabetic cardiomyopathy mice and investigated the cardioprotective effect of pyridostigmine. The experiments were conducted using a model of diabetic cardiomyopathy induced by a high-fat diet + streptozotocin in C57BL/6 mice. The results of high-throughput sequencing showed that diabetic cardiomyopathy mice exhibited decreased gut microbial diversity, altered abundance of the diabetes-related microbes, and increased abundance of the BCAA-producing microbes Clostridiales and Lachnospiraceae. In addition, diabetes downregulated tight junction proteins (ZO-1, occludin, and claudin-1) and increased intestinal permeability to impair the intestinal barrier. These impairments were accompanied by reduction in vagal activity that manifested as increased acetylcholinesterase levels, decreased acetylcholine levels, and heart rate variability, which eventually led to cardiac damage. Pyridostigmine enhanced vagal activity, restored gut microbiota homeostasis, decreased BCAA-producing microbe abundance, and improved the intestinal barrier to reduce circulating BCAA levels. Pyridostigmine also upregulated BCAT2 and PP2Cm and downregulated p-BCKDHA/BCKDHA and BCKDK to improve cardiac BCAA catabolism. Moreover, pyridostigmine alleviated abnormal mitochondrial structure; increased ATP production; decreased reactive oxygen species and mitochondria-related apoptosis; and attenuated cardiac dysfunction, hypertrophy, and fibrosis in diabetic cardiomyopathy mice. In conclusion, the gut microbiota, BCAA catabolism, and vagal activity were impaired in diabetic cardiomyopathy mice but were improved by pyridostigmine. These results provide novel insights for the development of a therapeutic strategy for diabetes-induced cardiac damage that targets gut microbes and BCAA catabolism.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qing Wu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Dong-Ling Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
77
|
Guimarães KSDL, Braga VDA, Noronha SISRD, Costa WKAD, Makki K, Cruz JDC, Brandão LR, Chianca Junior DA, Meugnier E, Leulier F, Vidal H, Magnani M, de Brito Alves JL. Lactiplantibacillus plantarum WJL administration during pregnancy and lactation improves lipid profile, insulin sensitivity and gut microbiota diversity in dyslipidemic dams and protects male offspring against cardiovascular dysfunction in later life. Food Funct 2021; 11:8939-8950. [PMID: 33000822 DOI: 10.1039/d0fo01718c] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM Maternal dyslipidemia is recognized as a risk factor for the development of arterial hypertension (AH) and cardiovascular dysfunction in offspring. Here we evaluated the effects of probiotic administration of a specific strain of Lactiplantibacillus plantarum (WJL) during pregnancy and lactation on gut microbiota and metabolic profile in dams fed with a high-fat and high-cholesterol (HFHC) diet and its long-term effects on the cardiovascular function in male rat offspring. METHODS AND RESULTS Pregnant Wistar rats were allocated into three groups: dams fed a control diet (CTL = 5), dams fed a HFHC diet (DLP = 5) and dams fed a HFHC diet and receiving L. plantarum WJL during pregnancy and lactation (DLP-LpWJL). L. plantarum WJL (1 × 109 CFU) or vehicle (NaCl, 0.9%) was administered daily by oral gavage for 6 weeks, covering the pregnancy and lactation periods. After weaning, male offspring received a standard diet up to 90 days of life. Biochemical measurements and gut microbiota were evaluated in dams. In male offspring, blood pressure (BP), heart rate (HR) and vascular reactivity were evaluated at 90 days of age. Dams fed with a HFHC diet during pregnancy and lactation had increased lipid profile and insulin resistance and showed dysbiotic gut microbiota. Administration of L. plantarum WJL to dams having maternal dyslipidemia improved gut microbiota composition, lipid profile and insulin resistance in them. Blood pressure was augmented and vascular reactivity was impaired with a higher contractile response and a lower response to endothelium-dependent vasorelaxation in DLP male offspring. In contrast, male offspring of DLP-LpWJL dams had reduced blood pressure and recovered vascular function in later life. CONCLUSION Administration of L. plantarum WJL during pregnancy and lactation in dams improved gut microbiota diversity, reduced maternal dyslipidemia and prevented cardiovascular dysfunction in male rat offspring.
Collapse
Affiliation(s)
| | - Valdir de Andrade Braga
- Department of Biotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Sylvana I S Rendeiro de Noronha
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | | | - Kassem Makki
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Josiane de Campos Cruz
- Department of Biotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Larissa Ramalho Brandão
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, Brazil.
| | - Deoclecio Alves Chianca Junior
- Department of Biological Science, Laboratory of Cardiovascular Physiology, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Emmanuelle Meugnier
- Univ-Lyon, CarMeN (Cardio, Metabolism, Diabetes and Nutrition) Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - François Leulier
- Univ-Lyon, Institut de Génomique Fonctionnelle de Lyon (IGFL), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard Lyon 1, Lyon, France
| | - Hubert Vidal
- Univ-Lyon, CarMeN (Cardio, Metabolism, Diabetes and Nutrition) Laboratory, INSERM, INRAE, Université Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Marciane Magnani
- Department of Food Engineering, Technology Center, Federal University of Paraiba, Joao Pessoa, Brazil
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, Brazil.
| |
Collapse
|
78
|
Suslov AV, Chairkina E, Shepetovskaya MD, Suslova IS, Khotina VA, Kirichenko TV, Postnov AY. The Neuroimmune Role of Intestinal Microbiota in the Pathogenesis of Cardiovascular Disease. J Clin Med 2021; 10:1995. [PMID: 34066528 PMCID: PMC8124579 DOI: 10.3390/jcm10091995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Currently, a bidirectional relationship between the gut microbiota and the nervous system, which is considered as microbiota-gut-brain axis, is being actively studied. This axis is believed to be a key mechanism in the formation of somatovisceral functions in the human body. The gut microbiota determines the level of activation of the hypothalamic-pituitary system. In particular, the intestinal microbiota is an important source of neuroimmune mediators in the pathogenesis of cardiovascular disease. This review reflects the current state of publications in PubMed and Scopus databases until December 2020 on the mechanisms of formation and participation of neuroimmune mediators associated with gut microbiota in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Andrey V. Suslov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Elizaveta Chairkina
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Maria D. Shepetovskaya
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Irina S. Suslova
- Central State Medical Academy of the Administrative Department of the President of the Russian Federation, 19-1A Marshal Timoshenko Str., 121359 Moscow, Russia;
| | - Victoria A. Khotina
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315 Moscow, Russia
| | - Tatiana V. Kirichenko
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| | - Anton Y. Postnov
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| |
Collapse
|
79
|
Yang Y, Cai Y, Zhang Y, Yi X, Xu Z. Identification of Molecular Subtypes and Key Genes of Atherosclerosis Through Gene Expression Profiles. Front Mol Biosci 2021; 8:628546. [PMID: 33996893 PMCID: PMC8113832 DOI: 10.3389/fmolb.2021.628546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/02/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) caused by atherosclerosis (AS) is one of the highest causes of mortality worldwide. Although there have been many studies on AS, its etiology remains unclear. In order to carry out molecular characterization of different types of AS, we retrieved two datasets composed of 151 AS samples and 32 normal samples from the Gene Expression Omnibus database. Using the non-negative matrix factorization (NMF) algorithm, we successfully divided the 151 AS samples into two subgroups. We then compared the molecular characteristics between the two groups using weighted gene co-expression analysis (WGCNA) and identified six key modules associated with the two subgroups. Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analysis were used to identify the potential functions and pathways associated with the modules. In addition, we used the cytoscape software to construct and visualize protein-protein networks so as to identify key genes in the modules of interest. Three hub genes including PTGER3, GNAI1, and IGFBP5 were further screened using the least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) algorithms. Since the modules were associated with immune pathways, we performed immune cell infiltration analysis. We discovered a significant difference in the level of immune cell infiltration by naïve B cells, CD8 T cells, T regulatory cells (Tregs), resting NK cells, Monocytes, Macrophages M0, Macrophages M1, and Macrophages M2 between the two subgroups. In addition, we observed the three hub genes were positively correlated with Tregs but negatively correlated with Macrophages M0. We also found that the three key genes are differentially expressed between normal and diseased tissue, as well as in the different subgroups. Receiver operating characteristic (ROC) results showed a good performance in the validation dataset. These results may provide novel insight into cellular and molecular characteristics of AS and potential markers for diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Yujia Yang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yue Cai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Zhang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xu Yi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiqiang Xu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
80
|
Miryala SK, Anbarasu A, Ramaiah S. Gene interaction network to unravel the role of gut bacterial species in cardiovascular diseases: E. coli O157:H7 host-bacterial interaction study. Comput Biol Med 2021; 133:104417. [PMID: 33901711 DOI: 10.1016/j.compbiomed.2021.104417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/17/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cardiovascular Disease (CVD) is one of the most common causes of mortality in humans. Presently, the role of pathogens in the initiation and progression of the CVDs is not clearly understood. Hence, it is essential to understand the molecular-level interactions between the human proteins and the microbial proteins to deduce their functional roles in the CVDs. METHOD The host-pathogen interactions (HPI) related to CVDs in the case of E. coli str. O157:H7 colonization were curated, and also the protein-protein interactions (PPI) between humans and E. coli were collected. Gene interaction network (GIN) and functional enrichment analyses (FEA) were utilized for this. RESULTS The GIN revealed dense interactions between the functional partners. The FEA indicated that the essential pathways played a significant role in humans as well as in E. coli. The primary responses against most of the bacterial pathogens in humans are different from that of E. coli; Terpenoid biosynthesis and production of secondary metabolite pathways aid the survival of the E. coli inside the host. Interestingly, network analysis divulged that the E. coli genes ksgA, rpsT, ispE, rpsI, ispH, and the human genes TP53, CASP3, CYCS, EP300, RHOA communicated by significant numbers in direct interactions. CONCLUSIONS The results obtained from the present study will help researchers understand the molecular-level interactions in the CVDs between the human and the E. coli genes. The important genes with vital interactions can be considered as hub molecules and can be exploited for new drug discovery.
Collapse
Affiliation(s)
- Sravan Kumar Miryala
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
81
|
Guo W, Kim SH, Wu D, Li L, Ortega EF, Thomas M, Meydani SN, Meydani M. Dietary Fruit and Vegetable Supplementation Suppresses Diet-Induced Atherosclerosis in LDL Receptor Knockout Mice. J Nutr 2021; 151:902-910. [PMID: 33561256 DOI: 10.1093/jn/nxaa410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/17/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Epidemiologic studies suggest that fruit and vegetable (F&V) consumption is inversely associated with incidence of cardiovascular disease (CVD). However, evidence for causality is lacking, and the underlying mechanisms are not well understood. OBJECTIVES We aimed to determine whether there is a causal relation between consuming high levels of F&V and prevention of atherosclerosis, the hallmark of CVD pathogenesis. Furthermore, the underlying mechanisms were determined. METHODS Six-week-old male LDL receptor-knockout mice were randomly assigned to 3 diet groups (12 mice/group) for 20 wk: control (CON, 10% kcal fat, 0.20 g/kg cholesterol), atherogenic (Ath, 27% kcal fat, 0.55 g/kg cholesterol), and Ath supplemented with 15% F&V (Ath + FV) (equivalent to 8-9 servings/d in humans). F&V was added as a freeze-dried powder that was prepared from the 24 most commonly consumed F&Vs in the United States. Body weight, aortic atherosclerotic lesion area, hepatic steatosis area, serum lipid profile and proinflammatory cytokine TNF-α concentrations, gut microbiota, and liver TNF-α and fatty acid synthase (Fasn) mRNA concentrations were assessed. RESULTS F&V supplementation did not affect weight gain. Mice fed the Ath + FV diet had a smaller aortic atherosclerotic lesion area (71.7% less) and hepatic steatosis area (80.7% less) than those fed the Ath diet (both P < 0.001) independent of impact on weight, whereas no difference was found between Ath + FV and CON groups in these 2 pathologic markers. Furthermore, F&V supplementation prevented Ath diet-induced dyslipidemia (high concentrations of serum TG and VLDL cholesterol and lower concentrations of HDL cholesterol), reduced serum TNF-α concentration (by 21.5%), suppressed mRNA expression of liver TNF-α and Fasn, and ameliorated Ath-induced gut microbiota dysbiosis. CONCLUSIONS Our results indicate that consuming a large quantity and variety of F&Vs causally attenuates diet-induced atherosclerosis and hepatic steatosis in mice. These effects of F&Vs are associated with, and may be mediated through, improved atherogenic dyslipidemia, alleviated gut dysbiosis, and suppressed inflammation.
Collapse
Affiliation(s)
- Weimin Guo
- Nutritional Immunology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sharon H Kim
- Vascular Biology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Dayong Wu
- Nutritional Immunology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Lijun Li
- Nutritional Immunology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Edwin Frank Ortega
- Nutritional Immunology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Michael Thomas
- Vascular Biology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Simin Nikbin Meydani
- Nutritional Immunology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Mohsen Meydani
- Vascular Biology Laboratories, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
82
|
Cesario A, D’Oria M, Bove F, Privitera G, Boškoski I, Pedicino D, Boldrini L, Erra C, Loreti C, Liuzzo G, Crea F, Armuzzi A, Gasbarrini A, Calabresi P, Padua L, Costamagna G, Antonelli M, Valentini V, Auffray C, Scambia G. Personalized Clinical Phenotyping through Systems Medicine and Artificial Intelligence. J Pers Med 2021; 11:jpm11040265. [PMID: 33918214 PMCID: PMC8065854 DOI: 10.3390/jpm11040265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Personalized Medicine (PM) has shifted the traditional top-down approach to medicine based on the identification of single etiological factors to explain diseases, which was not suitable for explaining complex conditions. The concept of PM assumes several interpretations in the literature, with particular regards to Genetic and Genomic Medicine. Despite the fact that some disease-modifying genes affect disease expression and progression, many complex conditions cannot be understood through only this lens, especially when other lifestyle factors can play a crucial role (such as the environment, emotions, nutrition, etc.). Personalizing clinical phenotyping becomes a challenge when different pathophysiological mechanisms underlie the same manifestation. Brain disorders, cardiovascular and gastroenterological diseases can be paradigmatic examples. Experiences on the field of Fondazione Policlinico Gemelli in Rome (a research hospital recognized by the Italian Ministry of Health as national leader in "Personalized Medicine" and "Innovative Biomedical Technologies") could help understanding which techniques and tools are the most performing to develop potential clinical phenotypes personalization. The connection between practical experiences and scientific literature highlights how this potential can be reached towards Systems Medicine using Artificial Intelligence tools.
Collapse
Affiliation(s)
- Alfredo Cesario
- Open Innovation Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Marika D’Oria
- Open Innovation Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Correspondence:
| | - Francesco Bove
- Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.B.); (P.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giuseppe Privitera
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ivo Boškoski
- Surgical Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.B.); (G.C.)
| | - Daniela Pedicino
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Luca Boldrini
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.B.); (V.V.)
| | - Carmen Erra
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Claudia Loreti
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Giovanna Liuzzo
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Filippo Crea
- Cardiology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (D.P.); (G.L.); (F.C.)
| | - Alessandro Armuzzi
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- CEMAD—IBD Unit—Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (A.A.); (A.G.)
- Department of Medicine and Translational Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Paolo Calabresi
- Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.B.); (P.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luca Padua
- High Intensity Neurorehabilitation Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.E.); (C.L.); (L.P.)
| | - Guido Costamagna
- Surgical Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.B.); (G.C.)
| | - Massimo Antonelli
- Anesthesia, Resuscitation, Intensive Care and Clinical Toxicology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Vincenzo Valentini
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (L.B.); (V.V.)
| | - Charles Auffray
- European Institute for Systems Biology and Medicine (EISBM), 69390 Vourles, France;
| | - Giovanni Scambia
- Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Gynecological Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
83
|
TMA/TMAO in Hypertension: Novel Horizons and Potential Therapies. J Cardiovasc Transl Res 2021; 14:1117-1124. [PMID: 33709384 DOI: 10.1007/s12265-021-10115-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/24/2021] [Indexed: 12/25/2022]
Abstract
Hypertension is the most prevalent chronic disease and a risk factor for various diseases. Although its mechanisms and therapies are constantly being updated and developed, they are still not fully clarified. In recent years, novel gut microbiota and its metabolites have attracted widespread attention. It is strongly linked with physiological and pathological systems, especially TMA and TMAO. TMA is formed by intestinal microbial metabolism of choline and L-carnitine and converted into TMAO by FMO3. This paper collected and collated the latest researches and mainly discussed the following four parts. It introduced gut microbiota; provided a focus on TMA, TMA-producing bacteria, and TMAO; summarized the alternations in hypertensive patients and animals; discussed the mechanisms of TMAO with two respects; and summarized the regulatory factors may be as new interventions and therapies of hypertension. And, more relevant studies are still prospected to be accomplished between hypertension and TMA/TMAO for further clinical services.
Collapse
|
84
|
Association between Gut Microbial Diversity and Carotid Intima-Media Thickness. ACTA ACUST UNITED AC 2021; 57:medicina57030195. [PMID: 33668894 PMCID: PMC7996485 DOI: 10.3390/medicina57030195] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
Background and Objectives: There is an increasing focus on the effect of the gut microbiome on developing atherosclerosis, but there is still no unified standpoint. We aimed to find associations between intestinal microbiome diversity and a marker of subclinical atherosclerosis, the carotid intima-media thickness (IMT). Materials and Methods: Recruited from the Hungarian Twin Registry, 108 monozygotic (MZ) twins (mean age 52.4 ± 14.1 years, 58% female) underwent a comprehensive carotid ultrasound examination (Samsung RS85). Of the 108 MZ twins, 14 pairs (mean age 65 ± 6.4 years, 71% female) discordant for carotid IMT were selected to undergo a stool sample collection. A special stool sampling container was mailed and received from each participant. After DNA extraction, library construction was performed specifically for the V3–V4 hypervariable region of microbial 16S rRNA. Next, the microbiome composition of the samples was determined using Kraken software. Two hypotheses were tested with the exact permutation test: (1) in the group with normal IMT, the Shannon index of the phyla is higher; and (2) the Firmicutes/Bacteroidetes ratio is greater in the group with high IMT values. Furthermore, the abundance of different bacterial strains present at higher and normal IMT was also explored. Statistical analysis was carried out using R software. Results: Increased Firmicutes/Bacteroidetes ratio was associated with increased IMT (mean Firmicutes/Bacteroidetes ratio of IMT > 0.9 and IMT < 0.9 groups: 2.299 and 1.436, respectively; p = 0.031). In the group with normal IMT values, a substantially higher fraction of Prevotellaceae was observed in contrast with subjects having subclinical atherosclerosis. However, there was no significant difference in the alpha diversity between the two groups. Conclusions: The determining role of individual genera and their proportions in the development and progression of atherosclerosis can be assumed. Further studies are needed to clarify if these findings can be used as potential therapeutic targets.
Collapse
|
85
|
Hsu CN, Tain YL. Preventing Developmental Origins of Cardiovascular Disease: Hydrogen Sulfide as a Potential Target? Antioxidants (Basel) 2021; 10:antiox10020247. [PMID: 33562763 PMCID: PMC7914659 DOI: 10.3390/antiox10020247] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
The cardiovascular system can be programmed by a diversity of early-life insults, leading to cardiovascular disease (CVD) in adulthood. This notion is now termed developmental origins of health and disease (DOHaD). Emerging evidence indicates hydrogen sulfide (H2S), a crucial regulator of cardiovascular homeostasis, plays a pathogenetic role in CVD of developmental origins. Conversely, early H2S-based interventions have proved beneficial in preventing adult-onset CVD in animal studies via reversing programming processes by so-called reprogramming. The focus of this review will first summarize the current knowledge on H2S implicated in cardiovascular programming. This will be followed by supporting evidence for the links between H2S signaling and underlying mechanisms of cardiovascular programming, such as oxidative stress, nitric oxide deficiency, dysregulated nutrient-sensing signals, activation of the renin–angiotensin system, and gut microbiota dysbiosis. It will also provide an overview from animal models regarding how H2S-based reprogramming interventions, such as precursors of H2S and H2S donors, may prevent CVD of developmental origins. A better understanding of cardiovascular programming and recent advances in H2S-based interventions might provide the answers to bring down the global burden of CVD.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-975-056-995; Fax: +886-7733-8009
| |
Collapse
|
86
|
Grylls A, Seidler K, Neil J. Link between microbiota and hypertension: Focus on LPS/TLR4 pathway in endothelial dysfunction and vascular inflammation, and therapeutic implication of probiotics. Biomed Pharmacother 2021; 137:111334. [PMID: 33556874 DOI: 10.1016/j.biopha.2021.111334] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
High blood pressure (BP) presents a significant public health challenge. Recent findings suggest that altered microbiota can exert a hypertensive effect on the host. One of the possible mechanisms involved is the chronic translocation of its components, mainly lipopolysaccharides (LPS) into systemic circulation leading to metabolic endotoxemia. In animal models, LPS has been commonly used to induce endothelial dysfunction and vascular inflammation. In human studies, plasma LPS concentration has been positively correlated with hypertension, however, the mechanistic link has not been fully elucidated. It is hypothesised here that the LPS-induced direct alterations to the vascular endothelium and resulting hypertension are possible targets for probiotic intervention. The methodology of this review involved a systematic search of the literature with critical appraisal of papers. Three tranches of search were performed: 1) existing review papers; 2) primary mechanistic animal, in vitro and human studies; and 3) primary intervention studies. A total of 70 peer-reviewed papers were included across the three tranches and critically appraised using SIGN50 for human studies and the ARRIVE guidelines for animal studies. The extracted information was coded into key themes and summarized in a narrative analysis. Results highlight the role of LPS in the activation of endothelial toll-like receptor 4 (TLR4) initiating a cascade of interrelated signalling pathways including: 1) Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase/ Reactive oxygen species (ROS)/ Endothelial nitric oxide synthase (eNOS) pathway leading to endothelial dysfunction; and 2) Mitogen-Activated Protein Kinase (MAPK) and Nuclear factor kappa B (NF-κB) pathways leading to vascular inflammation. Findings from animal intervention studies suggest an improvement in vasorelaxation, vascular inflammation and hypertension following probiotic supplementation, which was mediated by downregulation of LPS-induced pathways. Randomised controlled trials (RCTs) and systematic reviews provided some evidence for the anti-inflammatory effect of probiotics with statistically significant antihypertensive effect in clinical samples and may offer a viable intervention for the management of hypertension.
Collapse
Affiliation(s)
- Alina Grylls
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom.
| | - Karin Seidler
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom
| | - James Neil
- Centre for Nutrition Education and Lifestyle Management, Chapel Gardens, 14 Rectory Road, Wokingham RG40 1DH, England, United Kingdom
| |
Collapse
|
87
|
The influence of gut microbiota in cardiovascular diseases-a brief review. Porto Biomed J 2021; 6:e106. [PMID: 33490701 PMCID: PMC7817281 DOI: 10.1097/j.pbj.0000000000000106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Lately, the gut microbiota has emerged as an important mediator of the development and the outcomes of certain diseases. It's well known that the gut microbiota plays an important role in maintaining human health. Still far from being completely understood and analyzed is the complexity of this ecosystem, although a close relationship between the gut microbiota and cardiovascular diseases (CVD) has been established. A loss of diversity in the microbiota will lead to physiological changes, which can improve inflammatory or infection states like atherosclerosis and hypertension, the basic pathological process of CVD. Targeting the gut microbiota and its metabolites are new and promising strategies for the treatment and prognosis of CVD.
Collapse
|
88
|
Chiriac S, Stanciu C, Girleanu I, Cojocariu C, Sfarti C, Singeap AM, Cuciureanu T, Huiban L, Muzica CM, Zenovia S, Nastasa R, Trifan A. Nonalcoholic Fatty Liver Disease and Cardiovascular Diseases: The Heart of the Matter. Can J Gastroenterol Hepatol 2021; 2021:6696857. [PMID: 33505944 PMCID: PMC7815392 DOI: 10.1155/2021/6696857] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most frequent cause of liver disease worldwide, comprising a plethora of conditions, ranging from steatosis to end-stage liver disease. Cardiovascular disease (CVD) has been associated with NAFLD and CVD-related events represent the main cause of death in patients with NAFLD, surpassing liver-related mortality. This association is not surprising as NAFLD has been considered a part of the metabolic syndrome and has been related to numerous CVD risk factors, namely, insulin resistance, abdominal obesity, dyslipidemia, hyperuricemia, chronic kidney disease, and type 2 diabetes. Moreover, both NAFLD and CVD present similar pathophysiological mechanisms, such as increased visceral adiposity, altered lipid metabolism, increased oxidative stress, and systemic inflammation that could explain their association. Whether NAFLD increases the risk for CVD or these diagnostic entities represent distinct manifestations of the metabolic syndrome has not yet been clarified. This review focuses on the relation between NAFLD and the spectrum of CVD, considering the pathophysiological mechanisms, risk factors, current evidence, and future directions.
Collapse
Affiliation(s)
- Stefan Chiriac
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Carol Stanciu
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Irina Girleanu
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Camelia Cojocariu
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Catalin Sfarti
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Ana-Maria Singeap
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| | - Tudor Cuciureanu
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Laura Huiban
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Cristina Maria Muzica
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Sebastian Zenovia
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Robert Nastasa
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Anca Trifan
- 1Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
- 2Institute of Gastroenterology and Hepatology, “St. Spiridon” Emergency Hospital, Iasi 700111, Romania
| |
Collapse
|
89
|
Chang E, Kobayashi R, Fujihashi K, Komiya M, Kurita-Ochiai T. Impaired salivary SIgA antibodies elicit oral dysbiosis and subsequent induction of alveolar bone loss. Inflamm Res 2021; 70:151-158. [PMID: 33165644 DOI: 10.1007/s00011-020-01418-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Secreted IgA (SIgA) plays a central role in preventing bacterial and viral infections on mucosal surfaces by neutralizing toxins and viruses and inhibiting bacterial attachment to epithelial cells. However, the role of salivary SIgA antibodies (Abs) in regulating oral flora is still unknown. This study aimed to evaluate the association among oral bacteria, their metabolites and periodontitis in IgA-deficient (IgA KO) and wild-type (WT) control mice. METHODS Microcomputed tomography (micro-CT) analysis was used to assess alveolar bone resorption as a development of periodontitis. The bacterial profiles of saliva were determined using the next-generation sequencing assays. Furthermore, the metabolites in saliva were measured and compared using CE-TOFMS. RESULTS Salivary microbiota of IgA KO mice revealed a remarkably decreased frequency of Streptococcus, and increased percentages of Aggregatibacer, Actinobacillus, and Prevotella at the genus level when compared with those of WT. Compared to WT control mice of the same age, the level of alveolar bone loss was significantly increased in IgA KO mice, and infiltration of osteoclasts was found on the surface of the alveolar bone. The metabolome profile indicated that the metabolites of IgA KO mice had greater variability in carbon metabolic, urea cycle, and lipid pathways than WT mice. CONCLUSION These results suggest that salivary SIgA plays an important role in regulating and maintaining normal oral microflora to prevent the development of periodontal disease.
Collapse
Affiliation(s)
- Emily Chang
- Departments of Oral Surgery, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Ryoki Kobayashi
- Infection and Immunology, Nihon University, School of Dentistry at Matsudo, Chiba, Japan
| | - Kohtaro Fujihashi
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masamichi Komiya
- Departments of Oral Surgery, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Tomoko Kurita-Ochiai
- Infection and Immunology, Nihon University, School of Dentistry at Matsudo, Chiba, Japan.
- Department of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-nishi, Matsudo-shi, Chiba, 271-8587, Japan.
| |
Collapse
|
90
|
Karabulut A. The Role of Microbiologic Agents in the Progression of the Atherosclerosis: a Comprehensive Review. J Saudi Heart Assoc 2020; 32:440-450. [PMID: 33299789 PMCID: PMC7721457 DOI: 10.37616/2212-5043.1198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/07/2022] Open
Abstract
Atherosclerosis is a leading cause of disability, morbidity and mortality in the world. Atherosclerosis is accepted as a chronic progressive inflammatory disease. The inflammatory cascade in the vascular wall is well-defined. However, the predictors and contributors of the inflammatory response in atherosclerosis are not completely understood. Systemic and local inflammation, which enhance the burden of inflammation in the vascular wall, have been proposed as risk factors for the progression of atherosclerosis. Infectious micro-organisms are one of the major triggering factors for local and systemic inflammation. In this review, we aimed to emphasize the linkage between micro-organisms and the progression of atherosclerosis. We briefly summarize the current medical literature and discuss the future perspectives of the linkage between microbial agents and atherosclerosis representing cause and effect.
Collapse
Affiliation(s)
- Ahmet Karabulut
- Department of Cardiology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey.,Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| |
Collapse
|
91
|
Zhao X, Oduro PK, Tong W, Wang Y, Gao X, Wang Q. Therapeutic potential of natural products against atherosclerosis: Targeting on gut microbiota. Pharmacol Res 2020; 163:105362. [PMID: 33285231 DOI: 10.1016/j.phrs.2020.105362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/08/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Gut microbiota (GM) has emerged as an essential and integral factor for maintaining human health and affecting pathological outcomes. Metagenomics and metabolomics characterization have furthered gut metagenome's understanding and unveiled that deviation of specific GM community members and GM-dependent metabolites imbalance orchestrate metabolic or cardiovascular diseases (CVDs). Restoring GM ecosystem with nutraceutical supplements keenly prebiotics and probiotics relatively decreases CVDs incidence and overall mortality. In Atherosclerosis, commensal and pathogenic gut microbes correlate with atherogenesis events. GM-dependent metabolites-trimethylamine N-oxide and short-chain fatty acids regulate atherosclerosis-related metabolic processes in opposite patterns to affect atherosclerosis outcomes. Therefore, GM might be a potential therapeutic target for atherosclerosis. In atherogenic animal models, natural products with cardioprotective properties could modulate the GM ecosystem by revitalizing healthier GM phylotypes and abrogating proatherogenic metabolites, paving future research paths for clinical therapeutics.
Collapse
Affiliation(s)
- Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China
| | - Patrick Kwabena Oduro
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanyu Tong
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuefei Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China
| | - Xiumei Gao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China.
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin, China.
| |
Collapse
|
92
|
Szulc-Dąbrowska L, Bossowska-Nowicka M, Struzik J, Toka FN. Cathepsins in Bacteria-Macrophage Interaction: Defenders or Victims of Circumstance? Front Cell Infect Microbiol 2020; 10:601072. [PMID: 33344265 PMCID: PMC7746538 DOI: 10.3389/fcimb.2020.601072] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages are the first encounters of invading bacteria and are responsible for engulfing and digesting pathogens through phagocytosis leading to initiation of the innate inflammatory response. Intracellular digestion occurs through a close relationship between phagocytic/endocytic and lysosomal pathways, in which proteolytic enzymes, such as cathepsins, are involved. The presence of cathepsins in the endo-lysosomal compartment permits direct interaction with and killing of bacteria, and may contribute to processing of bacterial antigens for presentation, an event necessary for the induction of antibacterial adaptive immune response. Therefore, it is not surprising that bacteria can control the expression and proteolytic activity of cathepsins, including their inhibitors – cystatins, to favor their own intracellular survival in macrophages. In this review, we summarize recent developments in defining the role of cathepsins in bacteria-macrophage interaction and describe important strategies engaged by bacteria to manipulate cathepsin expression and activity in macrophages. Particularly, we focus on specific bacterial species due to their clinical relevance to humans and animal health, i.e., Mycobacterium, Mycoplasma, Staphylococcus, Streptococcus, Salmonella, Shigella, Francisella, Chlamydia, Listeria, Brucella, Helicobacter, Neisseria, and other genera.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Magdalena Bossowska-Nowicka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Justyna Struzik
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Felix N Toka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland.,Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
93
|
Abbasalizad Farhangi M, Vajdi M. Gut microbiota–associated trimethylamine N-oxide and increased cardiometabolic risk in adults: a systematic review and dose-response meta-analysis. Nutr Rev 2020; 79:1022-1042. [DOI: 10.1093/nutrit/nuaa111] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Abstract
Context
Several studies suggest a relationship between trimethylamine N-oxide (TMAO) concentrations and increased cardiometabolic risk, but findings are controversial.
Objective
The aim of this systematic review and meta-analysis was to summarize evidence of the relationship between circulating TMAO levels and risk of hypertension and increased serum lipids in a dose-response and 2-class meta-analysis of discrete and continuous variables.
Data Sources
PubMed, Scopus, Cochrane, and ProQuest databases were searched.
Study Selection
Observational studies that reported disease status of participants (≥ 18 years), type of sample in which TMAO was measured (serum or plasma), and results based on at least 2 categories of TMAO concentrations, including relative risks, hazard ratios, or odds ratios with 95%CIs for cardiometabolic risk factors in association with circulating TMAO levels were selected. Papers were reviewed independently by both authors. The Newcastle-Ottawa Scale was used to assess the quality of included studies.
Data Extraction
The following data were extracted: first author’s name, publication year, study design, study location, demographic information of participants, and concentrations of circulating TMAO.
Results
Eighteen studies were included in the meta-analysis. There was a dose-response relationship between circulating TMAO and increased odds of hypertension in cohort studies (P for nonlinearity = 0.049), in plasma-derived TMAO samples (P for nonlinearity = 0.043), in patients with cardiovascular disease (P for nonlinearity = 0.048), and in apparently healthy individuals from community-based studies (P for nonlinearity = 0.005). Moreover, the highest category of TMAO concentrations was associated with a 2.36 mmHg increase in systolic blood pressure when compared with the lowest category. The dose-response meta-analysis of continuous variables revealed that an increase in TMAO is associated with reduced high-density lipoprotein cholesterol in apparently healthy individuals and reduced high-density lipoprotein cholesterol and increased total cholesterol in patients with cardiovascular disease.
Conclusions
Circulating TMAO is positively associated with an increased risk of hypertension and other cardiometabolic disorders in adults.
Systematic Review Registration
PROSPERO identification number CRD42019138296.
Collapse
Affiliation(s)
| | - Mahdi Vajdi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
94
|
He M, Tan CP, Xu YJ, Liu Y. Gut microbiota-derived trimethylamine-N-oxide: A bridge between dietary fatty acid and cardiovascular disease? Food Res Int 2020; 138:109812. [DOI: 10.1016/j.foodres.2020.109812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/14/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023]
|
95
|
Lewis CV, Taylor WR. Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol 2020; 319:H1227-H1233. [PMID: 32986965 PMCID: PMC7792706 DOI: 10.1152/ajpheart.00612.2020] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
The gut microbiome and intestinal dysfunction have emerged as potential contributors to the development of cardiovascular disease (CVD). Alterations in gut microbiome are well documented in hypertension, atherosclerosis, and heart failure and have been investigated as a therapeutic target. However, a perhaps underappreciated but related role for intestinal barrier function has become evident. Increased intestinal permeability is observed in patients and mouse models of CVD. This increased intestinal permeability can enhance systemic inflammation, alter gut immune function, and has been demonstrated as predictive of adverse cardiovascular outcomes. The goal of this review is to examine the evidence supporting a role for intestinal barrier function in cardiovascular disease and its prospect as a novel therapeutic target. We outline key studies that have investigated intestinal permeability in hypertension, coronary artery disease, atherosclerosis, heart failure, and myocardial infarction. We highlight the central mechanisms involved in the breakdown of barrier function and look at emerging evidence for restored barrier function as a contributor to promising treatment strategies such as short chain fatty acid, probiotic, and renin angiotensin system-targeted therapeutics. Recent studies of more selective targeting of the intestinal barrier to improve disease outcomes are also examined. We suggest that although current data supporting a contribution of intestinal permeability to CVD pathogenesis are largely associative, it appears to be a promising avenue for further investigation. Additional studies of the mechanisms of barrier restoration in CVD and testing of intestinal barrier-targeted compounds will be required to confirm their potential as a new class of CVD therapeutic.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Cardiology Division, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
96
|
Simonsen JR, Järvinen A, Harjutsalo V, Forsblom C, Groop PH, Lehto M. The association between bacterial infections and the risk of coronary heart disease in type 1 diabetes. J Intern Med 2020; 288:711-724. [PMID: 32754939 DOI: 10.1111/joim.13138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/07/2020] [Accepted: 05/27/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Diabetes increases the risk of infections and coronary heart disease (CHD). Whether infections increase the risk of CHD and how this applies to individuals with diabetes is unclear. OBJECTIVES To investigate the association between bacterial infections and the risk of CHD in type 1 diabetes. METHODS Individuals with type 1 diabetes (n = 3781) were recruited from the Finnish Diabetic Nephropathy Study (FinnDiane), a prospective follow-up study. CHD was defined as incident events: fatal or nonfatal myocardial infarction, coronary artery bypass surgery or percutaneous coronary intervention, identified through national hospital discharge register data. Infections were identified through national register data on all antibiotic purchases from outpatient care. Register data were available from 1 January 1995 to 31 December 2015. Bacterial lipopolysaccharide (LPS) activity was measured from serum samples at baseline. Data on traditional risk factors for CHD were collected during baseline and consecutive visits. RESULTS Individuals with an incident CHD event (n = 370) had a higher mean number of antibiotic purchases per follow-up year compared to those without incident CHD (1.34 [95% CI: 1.16-1.52], versus 0.79 [0.76-0.82], P < 0.001), as well as higher levels of LPS activity (0.64 [0.60-0.67], versus 0.58 EU mL-1 [0.57-0.59], P < 0.001). In multivariable-adjusted Cox proportional hazards models, the mean number of antibiotic purchases per follow-up year was an independent risk factor for incident CHD (HR 1.21, 95% CI: 1.14-1.29, P < 0.0001). High LPS activity was a risk factor for incident CHD (HR 1.93 [1.34-2.78], P < 0.001) after adjusting for static confounders. CONCLUSION Bacterial infections are associated with an increased risk of incident CHD in individuals with type 1 diabetes.
Collapse
Affiliation(s)
- J R Simonsen
- From the, Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - A Järvinen
- Division of Infectious Diseases, Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - V Harjutsalo
- From the, Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland
| | - C Forsblom
- From the, Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - P-H Groop
- From the, Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Vic., Australia
| | - M Lehto
- From the, Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Nephrology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
97
|
Mutual Interplay of Host Immune System and Gut Microbiota in the Immunopathology of Atherosclerosis. Int J Mol Sci 2020; 21:ijms21228729. [PMID: 33227973 PMCID: PMC7699263 DOI: 10.3390/ijms21228729] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is the key for the initiation and progression of atherosclerosis. Accumulating evidence has revealed that an altered gut microbiome (dysbiosis) triggers both local and systemic inflammation to cause chronic inflammatory diseases, including atherosclerosis. There have been some microbiome-relevant pro-inflammatory mechanisms proposed to link the relationships between dysbiosis and atherosclerosis such as gut permeability disruption, trigger of innate immunity from lipopolysaccharide (LPS), and generation of proatherogenic metabolites, such as trimethylamine N-oxide (TMAO). Meanwhile, immune responses, such as inflammasome activation and cytokine production, could reshape both composition and function of the microbiota. In fact, the immune system delicately modulates the interplay between microbiota and atherogenesis. Recent clinical trials have suggested the potential of immunomodulation as a treatment strategy of atherosclerosis. Here in this review, we present current knowledge regarding to the roles of microbiota in contributing atherosclerotic pathogenesis and highlight translational perspectives by discussing the mutual interplay between microbiota and immune system on atherogenesis.
Collapse
|
98
|
Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes 2020; 12:1802866. [PMID: 32835590 PMCID: PMC7524325 DOI: 10.1080/19490976.2020.1802866] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/10/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last two decades our understanding of the gut microbiota and its contribution to health and disease has been transformed. Among a new 'generation' of potentially beneficial microbes to have been recognized are members of the genus Eubacterium, who form a part of the core human gut microbiome. The genus consists of phylogenetically, and quite frequently phenotypically, diverse species, making Eubacterium a taxonomically unique and challenging genus. Several members of the genus produce butyrate, which plays a critical role in energy homeostasis, colonic motility, immunomodulation and suppression of inflammation in the gut. Eubacterium spp. also carry out bile acid and cholesterol transformations in the gut, thereby contributing to their homeostasis. Gut dysbiosis and a consequently modified representation of Eubacterium spp. in the gut, have been linked with various human disease states. This review provides an overview of Eubacterium species from a phylogenetic perspective, describes how they alter with diet and age and summarizes its association with the human gut and various health conditions.
Collapse
Affiliation(s)
- Arghya Mukherjee
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Cathy Lordan
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - R. Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
99
|
Oynotkinova OS, Nikonov EL, Demidova TY, Baranov AP, Kryukov EV, Dedov EI, Karavashkina EA. [Changes in the intestinal microbiota as a risk factor for dyslipidemia, atherosclerosis and the role of probiotics in their prevention]. TERAPEVT ARKH 2020; 92:94-101. [PMID: 33346437 DOI: 10.26442/00403660.2020.09.000784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
The review presents an analysis of studies on the role of the intestinal microbiota and microbiome in lipid metabolism and the development of dyslipidemia, atherosclerosis and cardiovascular diseases. The role of the intestine as a metabolic organ with a multifactorial strain evolution, involved in lipid metabolism, cholesterol homeostasis and enterohepatic circulation is shown. The influence of microbial imbalance on the development of dyslipidemia and atherosclerosis is considered. Special attention is paid to preventive therapy with hypolipidemic probiotics. It is shown that the use of probiotics with hypolipidemic properties and consisting of a mixture of such strains asLactobacillus plantarumCECT7527, CET7528 and CECT7529, mixtures ofLactobacillus acidophilusLa-5,Bifidobacterium lactisBB-12,Bifidobacterium animalis lactisBB-12 contribute to reducing the level of LDL-C, CCS, TG, are safe and well tolerated, can be used as an adjuvant non-drug therapy in combination with hypolipidemic drugs for dyslipidemia, multifocal atherosclerosis.
Collapse
Affiliation(s)
- O S Oynotkinova
- Research Institute of the Organization of Health Care and Medical Management.,Pirogov Russian National Research Medical University.,Lomonosov Moscow State University
| | - E L Nikonov
- Pirogov Russian National Research Medical University
| | - T Y Demidova
- Pirogov Russian National Research Medical University
| | - A P Baranov
- Pirogov Russian National Research Medical University.,Lomonosov Moscow State University
| | | | - E I Dedov
- Pirogov Russian National Research Medical University
| | | |
Collapse
|
100
|
Roth WH, Cai A, Zhang C, Chen ML, Merkler AE, Kamel H. Gastrointestinal Disorders and Risk of First-Ever Ischemic Stroke. Stroke 2020; 51:3577-3583. [PMID: 33040706 DOI: 10.1161/strokeaha.120.030643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE Recent studies suggest that alteration of the normal gut microbiome contributes to atherosclerotic burden and cardiovascular disease. While many gastrointestinal diseases are known to cause disruption of the normal gut microbiome in humans, the clinical impact of gastrointestinal diseases on subsequent cerebrovascular disease remains unknown. We conducted an exploratory analysis evaluating the relationship between gastrointestinal diseases and ischemic stroke. METHODS We performed a retrospective cohort study using claims between 2008 and 2015 from a nationally representative 5% sample of Medicare beneficiaries. We included only beneficiaries ≥66 years of age. We used previously validated diagnosis codes to ascertain our primary outcome of ischemic stroke. In an exploratory manner, we categorized gastrointestinal disorders by anatomic location, disease chronicity, and disease mechanism. We used Cox proportional hazards models to examine associations of gastrointestinal disorder categories and ischemic stroke with adjustment for demographics and established vascular risk factors. RESULTS Among a mean of 1 725 246 beneficiaries in each analysis, several categories of gastrointestinal disorders were associated with an increased risk of ischemic stroke after adjustment for established stroke risk factors. The most notable positive associations included disorders of the stomach (hazard ratio, 1.17 [95% CI, 1.15-1.19]) and functional (1.16 [95% CI, 1.15-1.17]), inflammatory (1.13 [95% CI, 1.12-1.15]), and infectious gastrointestinal disorders (1.13 [95% CI, 1.12-1.15]). In contrast, we found no associations with stroke for diseases of the anus and rectum (0.97 [95% CI, 0.94-1.00]) or neoplastic gastrointestinal disorders (0.97 [95% CI, 0.94-1.00]). CONCLUSIONS In exploratory analyses, several categories of gastrointestinal disorders were associated with an increased risk of future ischemic stroke after adjustment for demographics and established stroke risk factors.
Collapse
Affiliation(s)
- William H Roth
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.).,Division of Neurocritical Care, Department of Neurology, University of Florida Medicine, Gainesville (W.H.R.)
| | - Anna Cai
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.)
| | - Cenai Zhang
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.)
| | - Monica L Chen
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.)
| | - Alexander E Merkler
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.)
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute and Department of Neurology, Weill Cornell Medicine, NY (W.H.R., A.C., C.Z., M.L.C., A.E.M., H.K.)
| |
Collapse
|