Abstract

Non-small cell lung cancer (NSCLC) cells often possess a hypermethylated Keap1 promoter, which decreases Keap1 mRNA and protein expression levels, thus impairing the Nrf2-Keap1 pathway and thereby leading to chemo- or radio-resistance. In this study, we showed that genistein selectively exhibited a radiosensitizing effect on NSCLC A549 cells but not on normal lung fibroblast MRC-5 cells. Genistein caused oxidative stress in A549 cells rather than MRC-5 cells, as determined by the oxidation of the ROS-sensitive probe DCFH-DA and oxidative damage marked by MDA, PCO or 8-OHdG content. In A549 instead of MRC-5 cells, genistein reduced the level of methylation in the Keap1 promoter region, leading to an increased mRNA expression, thus effectively inhibited the transcription of Nrf2 to the nucleus, which suppressed the Nrf2-dependent antioxidant and resulted in the upregulation of ROS. Importantly, when combined with radiation, genistein further increased the ROS levels in A549 cells whereas decreasing the radiation-induced oxidative stress in MRC-5 cells, possibly via increasing the expression levels of Nrf2, GSH and HO-1. Moreover, radiation combined with genistein significantly increased cell apoptosis in A549 but not MRC-5 cells. Together, the results herein show that the intrinsic difference in the redox status of A549 and MRC-5 cells could be the target for genistein to selectively sensitize A549 cells to radiation, thereby leading to an increase in radiosensitivity for A549 cells.

Highlights

  • Killing cancer cells without harming normal cells is a fundamental challenge in cancer therapy

  • The present study confirms these findings in A549 cells and further shows that the radiosensitizing effect of genistein was selective for A549 cells but not for MRC-5 cells (Figure 1)

  • Our results indicate that genistein protected the irradiated MRC-5 cells, because the cell growth rate (Figure 1D) and survival fractions (Figure 1E and 1F) after irradiation were restored to the untreated control level when combined with genistein treatment

Read more

Summary

Introduction

Killing cancer cells without harming normal cells is a fundamental challenge in cancer therapy. Elevated oxidative stress has been found in many types of cancer cells, due in part to their metabolic rates, when compared with their normal cell counterparts [1]. To adapt to this oxidative status, many tumor cells possess strong antioxidant defense mechanisms to counteract excessive reactive oxygen species (ROS), maintain redox status, suppress apoptosis and promote growth [2]. Selectively impairing the antioxidant defense system can make tumor cells have high constitutive oxidative stress levels, leading to cell death, whereas normal cells may still maintain redox homeostasis through adaptive responses. Numerous studies have shown that the Nrf2/Keap system can protect normal cells from exogenous ROS, but promotes the death of cancer cells under deleterious conditions [9, 10]

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