1. Academic Validation
  2. PRMT4 promotes ferroptosis to aggravate doxorubicin-induced cardiomyopathy via inhibition of the Nrf2/GPX4 pathway

PRMT4 promotes ferroptosis to aggravate doxorubicin-induced cardiomyopathy via inhibition of the Nrf2/GPX4 pathway

  • Cell Death Differ. 2022 Oct;29(10):1982-1995. doi: 10.1038/s41418-022-00990-5.
Yilong Wang  # 1 Shu Yan  # 2 3 Xuemei Liu 4 Fei Deng 5 Pengchao Wang 6 Liuye Yang 6 Lizhi Hu 6 Kai Huang 7 8 Jiangui He 9
Affiliations

Affiliations

  • 1 Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
  • 2 Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
  • 3 Heart Center and Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
  • 4 Department of functional medicine, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China.
  • 5 Department of Urology, Second Xiangya Hospital, Central South University, Changsha, China.
  • 6 Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 7 Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. [email protected].
  • 8 Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. [email protected].
  • 9 Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China. [email protected].
  • # Contributed equally.
Abstract

Doxorubicin (DOX), a commonly used antitumor agent, is often accompanied by its dosage-dependent cardiotoxicity, which incorporates Ferroptosis in its pathogenesis. Protein arginine methyltransferase 4 (PRMT4) is a transcription regulator involved in the modulation of oxidative stress and Autophagy, but its role in DOX-induced cardiomyopathy (DIC) and Ferroptosis remains elusive. Herein, we aimed to investigate the involvement and the underlying mechanisms of PRMT4 in the pathogenesis of DIC. Our present study revealed that the expression level of PRMT4 was markedly decreased in DOX-treated cardiomyocytes. Interestingly, it is noted that PRMT4 overexpression accelerated Ferroptosis to aggravate DIC, while its gene disruption or pharmaceutical inhibition exhibited the opposite effect. Mechanistically, our observation demonstrated that PRMT4 interacted with the nuclear factor erythroid 2-related factor 2 (Nrf2) to promote its enzymatic methylation, which restricted the nuclear translocation of Nrf2 and subsequently suppressed the transcription of Glutathione Peroxidase 4 (GPX4). Importantly, the detrimental role of PRMT4 in DOX-induced cardiomyocyte Ferroptosis was abolished by Nrf2 activation or Fer-1 administration. Collectively, our data reveal that PRMT4 inhibits Nrf2/GPX4 signaling to accelerate Ferroptosis in DIC, suggesting that targeting PRMT4 may present as a potential preventive strategy against the development of DIC.

Figures
Products