1. Academic Validation
  2. Autophagy activation is required for N6-methyladenosine modification to regulate ferroptosis in hepatocellular carcinoma

Autophagy activation is required for N6-methyladenosine modification to regulate ferroptosis in hepatocellular carcinoma

  • Redox Biol. 2023 Dec 1:69:102971. doi: 10.1016/j.redox.2023.102971.
Yujia Li 1 Mei Guo 2 Yangling Qiu 1 Mengran Li 1 Yang Wu 3 Min Shen 4 Yingqian Wang 1 Feng Zhang 1 Jiangjuan Shao 1 Xuefen Xu 5 Zili Zhang 6 Shizhong Zheng 7
Affiliations

Affiliations

  • 1 Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
  • 2 School of Nursing, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
  • 3 Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210023, China.
  • 4 Department of Biochemistry and Molecular Biology, Medical College, Yangzhou University, Yangzhou, 225009, China.
  • 5 Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine. Electronic address: 460103@njucm.edu.cn.
  • 6 Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China. Electronic address: zilizhang@njucm.edu.cn.
  • 7 Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China. Electronic address: nytws@njucm.edu.cn.
Abstract

Background & aims: Although Ferroptosis holds promise as a new strategy for treating hepatocellular carcinoma (HCC), there are several obstacles that need to be overcome. One major challenge is the lack of understanding about the mechanisms underlying Ferroptosis. Additionally, while the m6A modification has been shown to regulate various forms of cell death, its role in regulating Ferroptosis in HCC has been largely overlooked. Bridging this knowledge gap, our study aimed to elucidate the regulatory influence of m6A modification on HCC Ferroptosis.

Materials: Dot blot and EpiQuik m6A RNA Methylation Quantitative kit detected changes in overall m6A modification level during Ferroptosis in HCC. MeRIP-qPCR and RIP-qPCR identified that the m6A modification of ATG5 mRNA was significant changed. BALB/c nude mice were used to construct xenograft tumor models to verify the phenotypes upon YTHDC2 silencing. In addition, patient-derived organoid models were used to demonstrate that induction of Ferroptosis was an effective strategy against HCC.

Results: Our study has shown that inducing Ferroptosis is a promising strategy for combatting HCC. Specifically, we have found a significant correlation between Ferroptosis and high levels of m6A modification in HCC. Notably, we discovered that the elevation of ATG5 mRNA m6A modification mediated by WTAP is dependent on the reading protein YTHDC2. Importantly, inhibition of either WTAP or YTHDC2 effectively prevented Ferroptosis and suppressed HCC development in both in vitro and in vivo models.

Conclusion: Our study revealed that WTAP upregulates ATG5 expression post-transcriptionally in an m6A-YTHDC2-dependent manner, thereby promoting the translation of ATG5 mRNA during Ferroptosis in HCC. These findings have significant implications for the development of innovative and effective therapeutic approaches for HCC treatment.

Keywords

Autophagy; Ferroptosis; Hepatocellular carcinoma; Translation; m(6)A.

Figures
Products