Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer

Mol Cell Proteomics. 2023 Jun;22(6):100551. doi: 10.1016/j.mcpro.2023.100551. Epub 2023 Apr 17.

Abstract

Esophageal cancer is the seventh most common cancer in the world. Although traditional treatment methods such as radiotherapy and chemotherapy have good effects, their side effects and drug resistance remain problematic. The repositioning of drug function provides new ideas for the research and development of anticancer drugs. We previously showed that the Food and Drug Administration-approved drug sulconazole can effectively inhibit the growth of esophageal cancer cells, but its molecular mechanism is not clear. Here, our study demonstrated that sulconazole had a broad spectrum of anticancer effects. It can not only inhibit the proliferation but also inhibit the migration of esophageal cancer cells. Both transcriptomic sequencing and proteomic sequencing showed that sulconazole could promote various types of programmed cell death and inhibit glycolysis and its related pathways. Experimentally, we found that sulconazole induced apoptosis, pyroptosis, necroptosis, and ferroptosis. Mechanistically, sulconazole triggered mitochondrial oxidative stress and inhibited glycolysis. Finally, we showed that low-dose sulconazole can increase radiosensitivity of esophageal cancer cells. Taken together, these new findings provide strong laboratory evidence for the clinical application of sulconazole in esophageal cancer.

Keywords: PANoptosis; esophageal cancer; glycolysis; oxidative stress; radiosensitivity; sulconazole.

MeSH terms

  • Apoptosis
  • Cell Line, Tumor
  • Esophageal Neoplasms* / metabolism
  • Glycolysis
  • Humans
  • Oxidative Stress
  • Proteomics*
  • Radiation Tolerance

Substances

  • sulconazole