Abstract

Cisplatin is one of the most effective and widely used anticancer agents for the treatment of several types of tumors. The cytotoxic effect of cisplatin is thought to be mediated primarily by the generation of nuclear DNA adducts, which, if not repaired, cause cell death as a consequence of DNA replication and transcription blockage. However, the ability of cisplatin to induce nuclear DNA (nDNA) damage per se is not sufficient to explain its high degree of effectiveness nor the toxic effects exerted on normal, post-mitotic tissues. Oxidative damage has been observed in vivo following exposure to cisplatin in several tissues, suggesting a role for oxidative stress in the pathogenesis of cisplatin-induced dose-limiting toxicities. However, the mechanism of cisplatin-induced generation of ROS and their contribution to cisplatin cytotoxicity in normal and cancer cells is still poorly understood. By employing a panel of normal and cancer cell lines and the budding yeast Saccharomyces cerevisiae as model system, we show that exposure to cisplatin induces a mitochondrial-dependent ROS response that significantly enhances the cytotoxic effect caused by nDNA damage. ROS generation is independent of the amount of cisplatin-induced nDNA damage and occurs in mitochondria as a consequence of protein synthesis impairment. The contribution of cisplatin-induced mitochondrial dysfunction in determining its cytotoxic effect varies among cells and depends on mitochondrial redox status, mitochondrial DNA integrity and bioenergetic function. Thus, by manipulating these cellular parameters, we were able to enhance cisplatin cytotoxicity in cancer cells. This study provides a new mechanistic insight into cisplatin-induced cell killing and may lead to the design of novel therapeutic strategies to improve anticancer drug efficacy.

Highlights

  • Cisplatin is one of the most effective and widely used drugs for the treatment of adult and pediatric cancers

  • Such a cisplatin-induced increase in mitochondrial reactive oxygen species (ROS) is due to apoptosis, as we were able to detect a significant increase in mitochondrial ROS levels in the non-apoptotic subgroup of cells exposed to the drug (Figure 1C-D and Figure S2)

  • In the present study we demonstrate that cisplatin exposure induces a mitochondria-dependent ROS response that significantly contributes to cell killing by enhancing the cytotoxic effect exerted through the formation of nuclear DNA (nDNA) damage

Read more

Summary

Introduction

Cisplatin is one of the most effective and widely used drugs for the treatment of adult and pediatric cancers. Bulky nDNA adducts formed by cisplatin cause a block in nDNA replication and/or transcription, resulting in apoptosis [6,7,8,9,10]. This nDNA damage mediated mechanism of cell killing explains the high toxicity of cisplatin in dividing cells. Toxicity of cisplatin does not entirely depend on the amount of drug accumulation in normal tissues [14], suggesting that blockage of nDNA transcription may not be the only mechanism determining the toxic effect of cisplatin in non-replicating cells. Cisplatin-induced generation of nDNA damage per se is not sufficient to explain its high degree of effectiveness as an anticancer agent as well as the tissue specificity of its cytotoxic effects on normal, post-mitotic tissues

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call