4.8 Article

KLF2 regulates neutrophil activation and thrombosis in cardiac hypertrophy and heart failure progression

期刊

JOURNAL OF CLINICAL INVESTIGATION
卷 132, 期 3, 页码 -

出版社

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI147191

关键词

-

资金

  1. American Heart Association (AHA) Transformative Project Award [18TPA34230033]
  2. National Natural Science Foundation of China [82074049]
  3. NIH grant [1R35HL135789]
  4. AHA Paul G. Allen Frontiers Group [19PABHI34580006]
  5. Leducq Foundation [18CVD03]
  6. Case Comprehensive Cancer Center at Case Western Reserve University School of Medicine [P30CA043703]
  7. CWRU Genomics Core and Applied Functional Genomics Core (AFGC) at Case Western Reserve University School of Medicine
  8. Elisabeth Severance Prentiss Foundation grant

向作者/读者索取更多资源

The role of inflammation and inflammatory cells, particularly neutrophils, in cardiac hypertrophy and heart failure has been investigated in this study. It was found that neutrophils play a crucial role in the pathogenesis and progression of heart failure by triggering thrombosis in small myocardial vessels through the KLF2/NETosis pathway. Targeting neutrophils, NETosis, or thrombosis can alleviate pathological changes and preserve cardiac dysfunction.
It is widely recognized that inflammation plays a critical role in cardiac hypertrophy and heart failure. However, clinical trials targeting cytokines have shown equivocal effects, indicating the need for a deeper understanding of the precise role of inflammation and inflammatory cells in heart failure. Leukocytes from human subjects and a rodent model of heart failure were characterized by a marked reduction in expression of Klf2 mRNA. Using a mouse model of angiotensin II-induced nonischemic cardiac dysfunction, we showed that neutrophils played an essential role in the pathogenesis and progression of heart failure. Mechanistically, chronic angiotensin II infusion activated a neutrophil KLF2/NETosis pathway that triggered sporadic thrombosis in small myocardial vessels, leading to myocardial hypoxia, cell death, and hypertrophy. Conversely, targeting neutrophils, neutrophil extracellular traps (NETs), or thrombosis ameliorated these pathological changes and preserved cardiac dysfunction. KLF2 regulated neutrophil activation in response to angiotensin II at the molecular level, partly through crosstalk with HIF1 signaling. Taken together, our data implicate neutrophil-mediated immunothrombotic dysregulation as a critical pathogenic mechanism leading to cardiac hypertrophy and heart failure. This neutrophil KLF2NETosis-thrombosis mechanism underlying chronic heart failure can be exploited for therapeutic gain by therapies targeting neutrophils, NETosis, or thrombosis.

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

4.8
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