Oncotarget

Research Papers:

This article has been corrected. Correction in: Oncotarget. 2016; 7(19):28761-62.

5-Fluorouracil-induced RNA stress engages a TRAIL-DISC-dependent apoptosis axis facilitated by p53

Birce Akpinar, Ethiene V. Bracht, Dorin Reijnders, Barbora Safarikova, Iva Jelinkova, Alf Grandien, Alena Hyrslova Vaculova, Boris Zhivotovsky _ and Magnus Olsson

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:43679-43697. https://doi.org/10.18632/oncotarget.6030

Metrics: PDF 2106 views  |   HTML 2339 views  |   ?  


Abstract

Birce Akpinar1, Ethiene V. Bracht1, Dorin Reijnders1, Barbora Safarikova2, Iva Jelinkova2, Alf Grandien3, Alena Hyrslova Vaculova1,2, Boris Zhivotovsky1, Magnus Olsson1

1Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden

2Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Brno, Czech Republic

3Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital–Huddinge, Stockholm, Sweden

Correspondence to:

Boris Zhivotovsky, e-mail: [email protected]

Keywords: colon cancer, apoptosis, 5-fluorouracil, p53, necrosis

Received: July 22, 2015     Accepted: October 12, 2015     Published: October 24, 2015

ABSTRACT

Despite recent advances in targeted therapeutics, administration of 5-fluorouracil (5-FU) remains a common clinical strategy for post-surgical treatment of solid tumors. Although it has been proposed that RNA metabolism is disturbed by 5-FU treatment, the key cytotoxic response is believed to be enzymatic inhibition of thymidylate synthase resulting in nucleotide pool disproportions. An operating p53 tumor suppressor signaling network is in many cases essential for the efficiency of chemotherapy, and malfunctions within this system remain a clinical obstacle. Since the fate of chemotherapy-insensitive tumor cells is rarely described, we performed a comparative analysis of 5-FU toxicity in p53-deficient cells and conclude that p53 acts as a facilitator rather than a gatekeeper of cell death. Although p53 can act as a regulator of several cellular stress responses, no rerouting of cell death mode was observed in absence of the tumor suppressor. Thus, the final death outcome of 5-FU-treated p53-/- cells is demonstrated to be caspase-dependent, but due to a slow pace, accumulation of mitochondrial reactive oxygen species contributes to necrotic characteristics. The oligomerization status of the p53 target gene DR5 is determined as a significant limiting factor for the initiation of caspase activity in an intracellular TRAIL-dependent manner. Using several experimental approaches, we further conclude that RNA-rather than DNA-related stress follows by caspase activation irrespectively of p53 status. A distinct 5-FU-induced stress mechanism is thereby functionally connected to a successive and discrete cell death signaling pathway. Finally, we provide evidence that silencing of PARP-1 function may be an approach to specifically target p53-deficient cells in 5-FU combinatorial treatment strategies. Together, our results disclose details of impaired cell death signaling engaged as a consequence of 5-FU chemotherapy. Obtained data will contribute to the comprehension of factors restraining 5-FU efficiency, and by excluding DNA as the main stress target in some cell types they propose alternatives to currently used and suggested synergistic treatment regimens.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 6030