1. Academic Validation
  2. Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway

Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway

  • Cell Death Dis. 2021 Jan 18;12(1):88. doi: 10.1038/s41419-020-03357-1.
Shaohong Fang  # 1 2 Xin Wan  # 2 3 Xiaoyi Zou  # 2 3 Song Sun 2 3 Xinran Hao 2 3 Chenchen Liang 1 2 Zhenming Zhang 1 2 Fangni Zhang 2 3 Bo Sun 2 3 Hulun Li 4 5 Bo Yu 6 7
Affiliations

Affiliations

  • 1 Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
  • 2 The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.
  • 3 Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China.
  • 4 The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China. [email protected].
  • 5 Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China. [email protected].
  • 6 Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China. [email protected].
  • 7 The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China. [email protected].
  • # Contributed equally.
Abstract

Inducing Autophagy and inhibiting Apoptosis may provide a therapeutic treatment for atherosclerosis (AS). For the treatment of progressive AS, arsenic trioxide (ATO) has been used to coat vascular stents. However, the effect of ATO on Autophagy of macrophages is still unknown. Therefore, the aims of this study were to characterize the effects and the mechanism of actions of ATO on Autophagy in macrophages. Our results showed that ATO-induced activation of Autophagy was an earlier event than ATO-induced inhibition of the expression of Apoptosis markers in macrophages and foam cells. Nuclear transcription factor EB (TFEB) prevents atherosclerosis by activating macrophage Autophagy and promoting lysosomal biogenesis. Here, we report that ATO triggered the nuclear translocation of TFEB, which in turn promoted Autophagy and autophagosome-lysosome fusion. Both the latter events were prevented by TFEB knockdown. Moreover, ATO decreased the p-AKT and p-mTOR in the PI3K/Akt/mTOR signaling pathway, thus inducing Autophagy. Correspondingly, treatment with the Autophagy Inhibitor 3-methyladenine (3-MA) abolished the autophagy-inducing effects of ATO. Meanwhile, PI3K Inhibitor (LY294002) and mTOR Inhibitor (rapamycin) cooperated with ATO to induce Autophagy. Furthermore, Reactive Oxygen Species (ROS) were generated in macrophages after treatment with ATO. The ROS scavenger N-acetyl-1-cysteine (NAC) abolished ATO-induced nuclear translocation of TFEB, as well as changes in key molecules of the Akt/mTOR signaling pathway and downstream Autophagy. More importantly, ATO promoted Autophagy in the aorta of ApoE-/- mice and reduced atherosclerotic lesions in early AS, which were reversed by 3-MA treatment. In summary, our data indicated that ATO promoted ROS induction, which resulted in nuclear translocation of TFEB and inhibition of the PI3K/Akt/mTOR pathway. These actions ultimately promoted macrophage Autophagy and reduced atherosclerotic lesions at early stages. These findings may provide a new perspective for the clinical treatment of early-stage atherosclerosis and should be further studied.

Figures
Products
Inhibitors & Agonists
Other Products