1. Academic Validation
  2. Mitochondrial ROS driven by NOX4 upregulation promotes hepatocellular carcinoma cell survival after incomplete radiofrequency ablation by inducing of mitophagy via Nrf2/PINK1

Mitochondrial ROS driven by NOX4 upregulation promotes hepatocellular carcinoma cell survival after incomplete radiofrequency ablation by inducing of mitophagy via Nrf2/PINK1

  • J Transl Med. 2023 Mar 25;21(1):218. doi: 10.1186/s12967-023-04067-w.
Chao Peng 1 Xi Li 1 Feng Ao 1 Ting Li 2 Jingpei Guo 1 Junfeng Liu 1 Xiaoting Zhang 1 Jinyan Gu 3 Junjie Mao 4 Bin Zhou 5 6
Affiliations

Affiliations

  • 1 Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
  • 2 Department of Anesthesiology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China.
  • 3 Library Department, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China. Gujy9@mail.sysu.edu.cn.
  • 4 Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China. herodoc@sina.com.
  • 5 Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China. zhoub2@mail.sysu.edu.cn.
  • 6 Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China. zhoub2@mail.sysu.edu.cn.
Abstract

Background: The recurrence of hepatocellular carcinoma (HCC) after radiofrequency ablation (RFA) remains a major clinical problem. Cells that survive the sublethal heat stress that is induced by incomplete RFA are the main source of HCC relapse. Heat stress has long been reported to increase intracellular Reactive Oxygen Species (ROS) generation. Although ROS can induce Apoptosis, a pro-survival effect of ROS has also been demonstrated. However, the role of ROS in HCC cells exposed to sublethal heat stress remains unclear.

Methods: HepG2 and HuH7 cells were used for this experiment. Insufficient RFA was performed in cells and in a xenograft model. ROS and antioxidant levels were measured. Apoptosis was analyed by Annexin-V/PI staining and flow cytometry. Protein expression was measured using western blotting. Colocalization of lysosomes and mitochondria was analyzed to assess Mitophagy. Corresponding activators or inhibitors were applied to verify the function of specific objectives.

Results: Here,we showed that sublethal heat stress induced a ROS burst, which caused acute oxidative stress. This ROS burst was generated by mitochondria, and it was initiated by upregulated NOX4 expression in the mitochondria. N-acetylcysteine (NAC) decreased HCC cell survival under sublethal heat stress conditions in vivo and in vitro. NOX4 triggers the production of mitochondrial ROS (mtROS), and NOX4 inhibitors or siNOX4 also decreased HCC cell survival under sublethal heat stress conditions in vitro. Increased mtROS trigger PINK1-dependent Mitophagy to eliminate the mitochondria that are damaged by sublethal heat stress and to protect cells from Apoptosis. Nrf2 expression was elevated in response to this ROS burst and mediated the ROS burst-induced increase in PINK1 expression after sublethal heat stress.

Conclusion: These data confirmed that the ROS burst that occurs after iRFA exerted a pro-survival effect. NOX4 increased the generation of ROS by mitochondria. This short-term ROS burst induced PINK1-dependent Mitophagy to eliminate damaged mitochondria by increasing Nrf2 expression.

Keywords

HCC; Mitochondria; Mitophagy; NOX4; Nrf2; ROS.

Figures
Products