Cell Death and Disease (Jan 2024)

CYLD induces high oxidative stress and DNA damage through class I HDACs to promote radiosensitivity in nasopharyngeal carcinoma

  • Yueshuo Li,
  • Chenxing Yang,
  • Longlong Xie,
  • Feng Shi,
  • Min Tang,
  • Xiangjian Luo,
  • Na Liu,
  • Xudong Hu,
  • Yongwei Zhu,
  • Ann M. Bode,
  • Qiang Gao,
  • Jian Zhou,
  • Jia Fan,
  • Xuejun Li,
  • Ya Cao

DOI
https://doi.org/10.1038/s41419-024-06419-w
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 15

Abstract

Read online

Abstract Abnormal expression of Cylindromatosis (CYLD), a tumor suppressor molecule, plays an important role in tumor development and treatment. In this work, we found that CYLD binds to class I histone deacetylases (HDAC1 and HDAC2) through its N-terminal domain and inhibits HDAC1 activity. RNA sequencing showed that CYLD-HDAC axis regulates cellular antioxidant response via Nrf2 and its target genes. Then we revealed a mechanism that class I HDACs mediate redox abnormalities in CYLD low-expressing tumors. HDACs are central players in the DNA damage signaling. We further confirmed that CYLD regulates radiation-induced DNA damage and repair response through inhibiting class I HDACs. Furthermore, CYLD mediates nasopharyngeal carcinoma cell radiosensitivity through class I HDACs. Thus, we identified the function of the CYLD-HDAC axis in radiotherapy and blocking HDACs by Chidamide can increase the sensitivity of cancer cells and tumors to radiation therapy both in vitro and in vivo. In addition, ChIP and luciferase reporter assays revealed that CYLD could be transcriptionally regulated by zinc finger protein 202 (ZNF202). Our findings offer novel insight into the function of CYLD in tumor and uncover important roles for CYLD-HDAC axis in radiosensitivity, which provide new molecular target and therapeutic strategy for tumor radiotherapy.