Frontiers in Cell and Developmental Biology (Dec 2021)

Hypoxia Stimulates SUMOylation-Dependent Stabilization of KDM5B

  • Bingluo Zhou,
  • Yiran Zhu,
  • Wenxia Xu,
  • Qiyin Zhou,
  • Linghui Tan,
  • Liyuan Zhu,
  • Hui Chen,
  • Lifeng Feng,
  • Tianlun Hou,
  • Xian Wang,
  • Dingwei Chen,
  • Hongchuan Jin

DOI
https://doi.org/10.3389/fcell.2021.741736
Journal volume & issue
Vol. 9

Abstract

Read online

Hypoxia is an important characteristic of the tumor microenvironment. Tumor cells can survive and propagate under the hypoxia stress by activating a series of adaption response. Herein, we found that lysine-specific demethylase 5B (KDM5B) was upregulated in gastric cancer (GC) under hypoxia conditions. The genetic knockdown or chemical inhibition of KDM5B impaired the growth of GC cell adapted to hypoxia. Interestingly, the upregulation of KDM5B in hypoxia response was associated with the SUMOylation of KDM5B. SUMOylation stabilized KDM5B protein by reducing the competitive modification of ubiquitination. Furthermore, the protein inhibitor of activated STAT 4 (PIAS4) was determined as the SUMO E3 ligase, showing increased interaction with KDM5B under hypoxia conditions. The inhibition of KDM5B caused significant downregulation of hypoxia-inducible factor-1α (HIF-1α) protein and target genes under hypoxia. As a result, co-targeting KDM5B significantly improved the antitumor efficacy of antiangiogenic therapy in vivo. Taken together, PIAS4-mediated SUMOylation stabilized KDM5B protein by disturbing ubiquitination-dependent proteasomal degradation to overcome hypoxia stress. Targeting SUMOylation-dependent KDM5B upregulation might be considered when the antiangiogenic therapy was applied in cancer treatment.

Keywords