4.7 Article

Mfn2-mediated mitochondrial fusion alleviates doxorubicin-induced cardiotoxicity with enhancing its anticancer activity through metabolic switch

期刊

REDOX BIOLOGY
卷 52, 期 -, 页码 -

出版社

ELSEVIER
DOI: 10.1016/j.redox.2022.102311

关键词

Doxorubicin cardiotoxicity; Mitochondrial fusion; Mfn2; FoxO1; Metabolism

资金

  1. National Natural Sci-ence Foundation of China [81970316, 82070387, 82070051, 81670354, 82111530058]
  2. Key Research and Development Plan of Shaanxi [2021SF-141, 2022KWZ-18]
  3. Fundamental Research Funds for the Central Universities [xzy012019115]

向作者/读者索取更多资源

This study found that inhibition of Mfn2-mediated mitochondrial fusion is associated with cardiac dysfunction and tumorigenesis. Restoring Mfn2-mediated mitochondrial fusion can enhance mitochondrial oxidative metabolism, reduce cellular injury and oxidative stress, and provide a dual therapeutic advantage in Dox-based chemotherapy by defending against Dox-induced cardiotoxicity and boosting its antitumor potency.
Imbalanced mitochondrial dynamics including inhibited mitochondrial fusion is associated with cardiac dysfunction as well as tumorigenesis. This study sought to explore the effects of promoting mitochondrial fusion on doxorubicin(Dox)-induced cardiotoxicity and its antitumor efficacy, with a focus on the underlying metabolic mechanisms. Herein, the inhibition of Mfn2-mediated mitochondrial fusion was identified as a key phenotype in Dox-induced cardiotoxicity. Restoration of Mfn2-mediated mitochondrial fusion enhanced mitochondrial oxidative metabolism, reduced cellular injury/apoptosis and inhibited mitochondria-derived oxidative stress in the Dox-treated cardiomyocytes. Application of lentivirus expressing Drp1 (mitochondrial fusion inhibitor) or Rote/Anti A (mitochondrial complex I/III inhibitors) blunted the above protective effects of Mfn2. Cardiacspecific Mfn2 transgenic mice showed preserved mitochondrial fusion and attenuated myocardial injury upon Dox exposure in vivo. The suppression of Mfn2-mediated mitochondrial fusion was induced by Dox-elicited upregulation of FoxO1, which inhibited the transcription of Mfn2 by binding to its promoter sites. In the B16 melanoma, Mfn2 upregulation not only attenuated tumor growth alone but also further delayed tumor growth in the presence of Dox. Mechanistically, Mfn2 synergized with the inhibitory action of Dox on glycolysis metabolism in the tumor cells. One common feature in both cardiomyocytes and tumor cells was that Mfn2 increased the ratio of oxygen consumption rate to extracellular acidification rate, suggesting Mfn2 triggered a shift from aerobic glycolysis to mitochondrial oxidative metabolism. In conclusion, targeting Mfn2-mediated mitochondrial fusion may provide a dual therapeutic advantage in Dox-based chemotherapy by simultaneously defending against Dox-induced cardiotoxicity and boosting its antitumor potency via metabolic shift.

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