4.7 Article

Vascular endothelial tissue factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis

期刊

CARDIOVASCULAR RESEARCH
卷 118, 期 10, 页码 2367-2384

出版社

OXFORD UNIV PRESS
DOI: 10.1093/cvr/cvab263

关键词

Microbiome; Trimethylamine N-oxide; Cardiovascular disease; Thrombosis; Tissue factor

资金

  1. NIH and Office of Dietary Supplements [P01 HL147823, R01HL103866]
  2. Leducq Foundation
  3. Deutsche Forschungsgemeinschaft [WI 5229/1-1]
  4. Charite'-Universitatsmedizin
  5. Berlin institute of health
  6. Swiss National Research Foundation
  7. AstraZeneca
  8. Roche Diagnostics
  9. Medtronic
  10. both Switzerland
  11. Eli Lilly, Indianapolis, USA

向作者/读者索取更多资源

The study demonstrates that TMAO increases the risk of cardiovascular events and affects platelets and vascular tissue factor TF. Experimental results indicate that TMAO enhances arterial thrombosis potential by inducing the expression of TF and VCAM1.
Aims Gut microbiota and their generated metabolites impact the host vascular phenotype. The metaorganismal metabolite trimethylamine N-oxide (TMAO) is both associated with adverse clinical thromboembolic events, and enhances platelet responsiveness in subjects. The impact of TMAO on vascular Tissue Factor (TF) in vivo is unknown. Here, we explore whether TMAO-enhanced thrombosis potential extends beyond TMAO effects on platelets, and is linked to TF. We also further explore the links between gut microbiota and vascular endothelial TF expression in vivo. Methods and results In initial exploratory clinical studies, we observed that among sequential stable subjects (n = 2989) on anti-platelet therapy undergoing elective diagnostic cardiovascular evaluation at a single-site referral centre, TMAO levels were associated with an increased incident (3 years) risk for major adverse cardiovascular events (MACE) (myocardial infarction, stroke, or death) [4th quartile (Q4) vs. Q1 adjusted hazard ratio (HR) 95% confidence interval (95% CI), 1.73 (1.25-2.38)]. Similar results were observed within subjects on aspirin mono-therapy during follow-up [adjusted HR (95% CI) 1.75 (1.25-2.44), n = 2793]. Leveraging access to a second higher risk cohort with previously reported TMAO data and monitoring of anti-platelet medication use, we also observed a strong association between TMAO and incident (1 year) MACE risk in the multi-site Swiss Acute Coronary Syndromes Cohort, focusing on the subset (n = 1469) on chronic dual anti-platelet therapy during follow-up [adjusted HR (95% CI) 1.70 (1.08-2.69)]. These collective clinical data suggest that the thrombosis-associated effects of TMAO may be mediated by cells/factors that are not inhibited by anti-platelet therapy. To test this, we first observed in human microvascular endothelial cells that TMAO dose-dependently induced expression of TF and vascular cell adhesion molecule (VCAM)1. In mouse studies, we observed that TMAO-enhanced aortic TF and VCAM1 mRNA and protein expression, which upon immunolocalization studies, was shown to co-localize with vascular endothelial cells. Finally, in arterial injury mouse models, TMAO-dependent enhancement of in vivo TF expression and thrombogenicity were abrogated by either a TF-inhibitory antibody or a mechanism-based microbial choline TMA-lyase inhibitor (fluoromethylcholine). Conclusion Endothelial TF contributes to TMAO-related arterial thrombosis potential, and can be specifically blocked by targeted non-lethal inhibition of gut microbial choline TMA-lyase.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据