Login / Signup

p53N236S Activates Autophagy in Response to Hypoxic Stress Induced by DFO.

Kang GaoHuanhuan ZongKailong HouYanduo ZhangRuyi ZhangDan ZhaoXin GuoYing LuoShuting Jia
Published in: Genes (2022)
Hypoxia can lead to stabilization of the tumor suppressor gene p53 and cell death. However, p53 mutations could promote cell survival in a hypoxic environment. In this study, we found that p53N236S (p53N239S in humans, hereinafter referred to as p53S) mutant mouse embryonic fibroblasts (MEFs) resistant to deferoxamine (DFO) mimic a hypoxic environment. Further, Western blot and flow cytometry showed reduced apoptosis in p53 S/S cells compared to WT after DFO treatment, suggesting an antiapoptosis function of p53S mutation in response to hypoxia-mimetic DFO. Instead, p53 S/S cells underwent autophagy in response to hypoxia stress presumably through inhibition of the AKT/mTOR pathway, and this process was coupled with nuclear translocation of p53S protein. To understand the relationship between autophagy and apoptosis in p53 S/S cells in response to hypoxia, the autophagic inhibitor 3-MA was used to treat both WT and p53 S/S cells after DFO exposure. Both apoptotic signaling and cell death were enhanced by autophagy inhibition in p53 S/S cells. In addition, the mitochondrial membrane potential (MMP) and the ROS level results indicated that p53S might initiate mitophagy to clear up damaged mitochondria in response to hypoxic stress, thus increasing the proportion of intact mitochondria and maintaining cell survival. In conclusion, the p53S mutant activates autophagy instead of inducing an apoptotic process in response to hypoxia stress to protect cells from death.
Keyphrases