Back to Search Start Over

GLS-driven glutamine catabolism contributes to prostate cancer radiosensitivity by regulating the redox state, stemness and ATG5-mediated autophagy

Authors :
Claudia Peitzsch
Giulia Negro
Mechthild Krause
Mirko Peitzsch
Marieta Toma
Fabian Lohaus
Tobias Hölscher
Sebastian Zschaeck
A Franken
Gustavo Baretton
Annett Linge
Nikolas H. Stoecklein
Tiago C. Alves
Anna Mukha
Mahdi Rivandi
Peter Mirtschink
Amir Abdollahi
Steffen Löck
Vladyslav Telychko
Christian Schwager
Oleg Chen
Susan Richter
Anna Dubrovska
Leoni A. Kunz-Schughart
Michael Baumann
Ulrich Sommer
Bertram Aschenbrenner
Bianca Behrens
Vasyl Lukiyanchuk
Maximilian Rehm
Ira-Ida Skvortsova
Hans Neubauer
Christer Groeben
Sergej Skvortsov
Uğur Kahya
Graeme Eisenhofer
Publication Year :
2021
Publisher :
Cold Spring Harbor Laboratory, 2021.

Abstract

Radiotherapy is one of the curative treatment options for localized prostate cancer (PCa). The curative potential of radiotherapy is mediated by irradiation-induced oxidative stress and DNA damage in tumor cells. However, PCa radiocurability can be impeded by tumor resistance mechanisms and normal tissue toxicity. Metabolic reprogramming is one of the major hallmarks of tumor progression and therapy resistance. Here, we found that radioresistant PCa cells and prostate cancer stem cells (CSCs) have a high glutamine demand. Glutaminase (GLS)-driven catabolism of glutamine serves not only for energy production but also for the maintenance of the redox state. Consequently, glutamine depletion or inhibition of critical regulators of glutamine utilization, such as glutaminase (GLS) and the transcription factor MYC results in PCa radiosensitization. On the contrary, we found that a combination of glutamine metabolism inhibitors with irradiation does not cause toxic effects on nonmalignant prostate cells. Glutamine catabolism contributes to the maintenance of CSCs through regulation of the alpha-ketoglutarate (α-KG)-dependent chromatin-modifying dioxygenase. The lack of glutamine results in the inhibition of CSCs with a high aldehyde dehydrogenase (ALDH) activity, decreases the frequency of the CSC populations in vivo and reduces tumor formation in xenograft mouse models. Moreover, this study shows that activation of the ATG5-mediated autophagy in response to a lack of glutamine is a tumor survival strategy to withstand radiation-mediated cell damage. In combination with autophagy inhibition, the blockade of glutamine metabolism might be a promising strategy for PCa radiosensitization. High blood levels of glutamine in PCa patients significantly correlate with a shorter prostate-specific antigen (PSA) doubling time. Furthermore, high expression of critical regulators of glutamine metabolism, GLS1 and MYC, is significantly associated with a decreased progression-free survival in PCa patients treated with radiotherapy. Our findings demonstrate that GLS-driven glutaminolysis is a prognostic biomarker and therapeutic target for PCa radiosensitization.

Details

Database :
OpenAIRE
Accession number :
edsair.doi...........68217130424b3f27e3f03f81acfccc81