1. Academic Validation
  2. Tissue-specific regulation of p53 by PKM2 is redox dependent and provides a therapeutic target for anthracycline-induced cardiotoxicity

Tissue-specific regulation of p53 by PKM2 is redox dependent and provides a therapeutic target for anthracycline-induced cardiotoxicity

  • Sci Transl Med. 2019 Feb 6;11(478):eaau8866. doi: 10.1126/scitranslmed.aau8866.
Bruno Saleme 1 2 Vikram Gurtu 1 2 Yongneng Zhang 1 2 Adam Kinnaird 1 3 Aristeidis E Boukouris 1 2 Keshav Gopal 2 4 John R Ussher 2 4 Gopinath Sutendra 5 2 6
Affiliations

Affiliations

  • 1 Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2J7, Canada.
  • 2 Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta T6G 2J7, Canada.
  • 3 Department of Surgery, University of Alberta, Edmonton, Alberta T6G 1Z1, Canada.
  • 4 Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2H1, Canada.
  • 5 Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2J7, Canada. [email protected].
  • 6 Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta T6G 2J7, Canada.
Abstract

Chemotherapy-induced cardiotoxicity (CIC) is a common clinical problem that compromises effective Anticancer therapies. Many chemotherapeutics (including anthracyclines, such as doxorubicin) induce the proapoptotic transcription factor p53 in the tumor and nonspecifically in the heart, promoting heart failure. Although inhibition of p53 shows benefit in preclinical heart failure models, it would not be an attractive adjuvant therapy for CIC, because it would prevent tumor regression. A p53-targeting therapy that would decrease chemotherapy-induced Apoptosis in the myocardium and, at the same time, enhance Apoptosis in the tumor would be ideal. Here, we propose that differences in oxygen tension between the myocardium and the tumor could provide a platform for redox-dependent tissue-specific therapies. We show by coimmunoprecipitation and mass spectrometry that the redox-regulated Pyruvate Kinase muscle 2 (PKM2) directly binds with p53 and that the redox status of cysteine-423 of tetrameric (but not monomeric) PKM2 is critical for the differential regulation of p53 transcriptional activity. Tetrameric PKM2 suppresses p53 transcriptional activity and Apoptosis in a high oxidation state but enhances them in a low oxidation one. We show that the oxidation state (along with cysteine-423 oxidation) is higher in the heart compared to the tumor of the same animal. Treatment with TEPP-46 (a compound that stabilizes tetrameric PKM2) suppressed doxorubicin-induced cardiomyocyte Apoptosis, preventing cardiac dysfunction, but enhanced Cancer cell Apoptosis and tumor regression in the same Animals in lung Cancer models. Thus, our work suggests that redox-dependent differences in common proteins expressed in the myocardium and tumor can be exploited therapeutically for tissue selectivity in CIC.

Figures
Products