Oncotarget

Research Papers:

The oxido-metabolic driver ATF4 enhances temozolamide chemo-resistance in human gliomas

Daishi Chen, Manfred Rauh, Michael Buchfelder, Ilker Y. Eyupoglu and Nicolai Savaskan _

PDF  |  HTML  |  How to cite

Oncotarget. 2017; 8:51164-51176. https://doi.org/10.18632/oncotarget.17737

Metrics: PDF 4662 views  |   HTML 5746 views  |   ?  


Abstract

Daishi Chen1,2, Manfred Rauh3, Michael Buchfelder1, Ilker Y. Eyupoglu1 and Nicolai Savaskan1,4

1Translational Cell Biology and Neurooncology laboratory at the Department of Neurosurgery, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander University of Erlangen–Nürnberg (FAU), Erlangen, Germany

2Department of Otolaryngology–Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China

3Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen (UKER), Friedrich-Alexander University of Erlangen–Nürnberg (FAU), Erlangen, Germany

4BiMECON Ent., Berlin, Germany

Correspondence to:

Nicolai Savaskan, email: [email protected], [email protected]

Keywords: autophagy, ferroptosis, apoptosis, cell death, glutamate

Received: August 25, 2016     Accepted: February 20, 2017     Published: May 09, 2017

ABSTRACT

Malignant gliomas are devastating neoplasia with limited curative treatment options. Temozolomide (TMZ, Temcat®, Temodal® or Temodar®) is a first-line treatment for malignant gliomas but the development of drug resistance remains a major concern. Activating transcription factor 4 (ATF4) is a critical oxido-metabolic regulator in gliomas, and its role in the pathogenesis of TMZ-resistance remains elusive. We investigated the effect of TMZ on human glioma cells under conditions of enhanced ATF4 expression (ATF4OE) and ATF4 knock down (ATF4KD). We monitored cell survival, ATF4 mRNA expression of ATF4 and xCT (SLC7a11) regulation within human gliomas. TMZ treatment induces a transcriptional response with elevated expression of ATF4, xCT and Nrf2, as a sign of ER stress and toxic cell damage response. ATF4 overexpression (ATF4OE) fosters TMZ resistance in human gliomas and inhibits TMZ-induced autophagy. Conversely, ATF4 suppression by small interfering RNAs (ATF4KD) leads to increased TMZ susceptibility and autophagy in comparison to wild type gliomas. ATF4OE gliomas show reduced cell cycle shift and apoptotic cell death, whereas ATF4KD gliomas reveal higher susceptibility towards cell cycle rearrangements. Hence, the migration capacity of ATF4OE glioma cells is almost not affected by TMZ treatment. In contrast, ATF4KD gliomas show a migratory stop following TMZ application. Mechanistically, xCT elevation is a consequence of ATF4 activation and increased levels of xCT amplifies ATF4-induced TMZ resistance. Our data show that ATF4 operates as a chemo-resistance gene in gliomas, and the tumor promoting function of ATF4 is mainly determined by its transcriptional target xCT. Therefore, therapeutic inactivation of ATF4 can be a promising strategy to overcome chemo-resistance and promote drug efficacy in human gliomas.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 17737