4.7 Article

Rosuvastatin protects against coronary microembolization-induced cardiac injury via inhibiting NLRP3 inflammasome activation

Journal

CELL DEATH & DISEASE
Volume 12, Issue 1, Pages -

Publisher

SPRINGERNATURE
DOI: 10.1038/s41419-021-03389-1

Keywords

-

Categories

Funding

  1. Shanghai Municipal Commission of Health and Family Planning [2017YQ057]
  2. National Natural Science Foundation of China [81870267, 81970295, 81900307, 81521001, 81670318, 81570314]
  3. National Program on Key Basic Research Project of China [2019YFC0840601, 2014CBA02003]
  4. Shanghai Science and Technology Committee [19MC1910300, 17411962300]
  5. Shanghai Shenkang Development Project [SHDC12019104]
  6. Zhongshan Hospital [Zhupei-7, 2018ZSLC01, 2015ZSYXGG07, 2017ZSYQ08]
  7. VG Funding of Clinical Trials [2017-CCA-VG-036]
  8. Chinese Cardiovascular Association CMVD [2018-CCA-CMVD-03]
  9. Merck [Xinxin-merck-fund-051]
  10. Program for Outstanding Medical Academic Leader [2015-Weijiwei-24]

Ask authors/readers for more resources

In this study, it was found that the NLRP3 inflammasome plays an important role in CME-induced cardiac injury, and treatment with rosuvastatin (RVS) can improve cardiac function, reduce damage, and lower ROS levels.
Coronary microembolization (CME), a common reason for periprocedural myocardial infarction (PMI), bears very important prognostic implications. However, the molecular mechanisms related to CME remain largely elusive. Statins have been shown to prevent PMI, but the underlying mechanism has not been identified. Here, we examine whether the NLRP3 inflammasome contributes to CME-induced cardiac injury and investigate the effects of statin therapy on CME. In vivo study, mice with CME were treated with 40 mg/kg/d rosuvastatin (RVS) orally or a selective NLRP3 inflammasome inhibitor MCC950 intraperitoneally (20 mg/kg/d). Mice treated with MCC950 and RVS showed improved cardiac contractile function and morphological changes, diminished fibrosis and microinfarct size, and reduced serum lactate dehydrogenase (LDH) level. Mechanistically, RVS decreased the expression of NLRP3, caspase-1, interleukin-1 beta, and Gasdermin D N-terminal domains. Proteomics analysis revealed that RVS restored the energy metabolism and oxidative phosphorylation in CME. Furthermore, reduced reactive oxygen species (ROS) level and alleviated mitochondrial damage were observed in RVS-treated mice. In vitro study, RVS inhibited the activation of NLRP3 inflammasome induced by tumor necrosis factor alpha plus hypoxia in H9c2 cells. Meanwhile, the pyroptosis was also suppressed by RVS, indicated by the increased cell viability, decreased LDH and propidium iodide uptake in H9c2 cells. RVS also reduced the level of mitochondrial ROS generation in vitro. Our results indicate the NLRP3 inflammasome-dependent cardiac pyroptosis plays an important role in CME-induced cardiac injury and its inhibitor exerts cardioprotective effect following CME. We also uncover the anti-pyroptosis role of RVS in CME, which is associated with regulating mitochondrial ROS.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available