4.7 Article

RSL3 enhances the antitumor effect of cisplatin on prostate cancer cells via causing glycolysis dysfunction

Journal

BIOCHEMICAL PHARMACOLOGY
Volume 192, Issue -, Pages -

Publisher

PERGAMON-ELSEVIER SCIENCE LTD
DOI: 10.1016/j.bcp.2021.114741

Keywords

RSL3; Cisplatin; Glycolysis; Apoptosis; Prostate carcinoma

Funding

  1. National Natural Science Foundation of China [81773217]
  2. Fundamental Research Funds for the Central Universities, JLU, Research Fund of Jilin Provincial Science and Technology Department [20190701065GH, 20200404120YY]
  3. Jilin Province Health Technology Innovation Project [2019J030]
  4. Research Foundation of the Education Depart-ment of Jilin province [JJKH20211182KJ]
  5. China Post-doctoral Science Foundation [2018M631885]
  6. Chunhui International Research Project of Ministry of Education

Ask authors/readers for more resources

The study demonstrated that RSL3 improved the sensitivity of PCa to DDP by causing glycolysis dysfunction, effectively inhibiting PCa cell proliferation both in vitro and in vivo. Additionally, cotreatment of RSL3 and DDP could alleviate apoptosis and cell cycle abnormalities in PCa cells, providing a potential therapeutic strategy for PCa.
The resistance to cisplatin (DDP) and dose-related toxicity are the two important obstacles in the chemotherapy of prostate cancer (PCa) patients. The present study demonstrated that cotreatment of DDP and RSL3, a type of small molecular compound which can inactivate glutathione peroxidase 4 (GPX4) and induce ferroptosis, syn-ergistically inhibited the viability and proliferation of PCa cells in vitro and in vivo at low dose. In vitro studies revealed that RSL3 improved that sensitivity of PCa cells to DDP by producing ROS and aggravating the cell cycle arrest and apoptosis caused by DDP. Mechanistically, RSL3 could decrease the ATP and pyruvate content as well as the protein levels of HKII, PFKP, PKM2, which indicated that RSL3 induced glycolysis dysfunction in prostate cancer cells. Rescuing RSL3-induced glycolysis dysfunction by supplement of exterior sodium pyruvate not only inhibited RSL3/DDP-induced changes of apoptosis-related proteins levels, but also mitigated the cell death caused by RSL3/DDP. In vivo studies further confirmed that cotreatment of RSL3 and DDP at low dose signifi-cantly inhibited the growth of PCa with no obvious side effects. Taken together, we demonstrated that RSL3 improved the sensitivity of PCa to DDP via causing glycolysis dysfunction. Our findings indicated that DDP-based chemotherapy combined with RSL3 might provide a promising therapy for PCa.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available