학술논문

Ciprofloxacin as a potential radio-sensitizer to tumor cells and a radio-protectant for normal cells: differential effects on γ-H2AX formation, p53 phosphorylation, Bcl-2 production, and cell death
Document Type
Academic Journal
Source
Molecular and Cellular Biochemistry. August 1, 2014, Vol. 393 Issue 1-2, p133, 11 p.
Subject
Health aspects
Mortality
Apoptosis -- Health aspects
Tumor proteins -- Health aspects
Ionizing radiation -- Health aspects
Biochemistry -- Health aspects
Tumors -- Health aspects
DNA -- Health aspects
Ciprofloxacin -- Health aspects
Language
English
ISSN
0300-8177
Abstract
Introduction Ionizing irradiation is used to eliminate cancer cells while normal cells next to them are also exposed to it resulting in cell damage. Ionizing irradiation increases cell mortality in [...]
Ionizing radiation increases cell mortality in a dose-dependent manner. Increases in DNA double strand breaks, γ-H2AX, p53 phophorylation, and protein levels of p53 and Bax also occur. We investigated the ability of ciprofloxacin (CIP), a widely prescribed antibiotic, to inhibit DNA damage induced by ionizing radiation. Human tumor TK6, NH32 ([p53.sup.-/-] of TK6) cells, and human normal peripheral blood mononuclear cells (PBMCs) were exposed to 2-8 Gy [sup.60]Co-γ-photon radiation. γ-H2AX (an indicator of DNA strand breaks), phosphorylated p53 (responsible for cell-cycle arrest), Bcl-2 (an apoptotic protein, and cell death were measured. Ionizing irradiation increased γ-H2AX amounts in TK6 cells ([p53.sup.+/+]) within 1 h in a radiation dose-dependent manner. CIP pretreatment and posttreatment effectively inhibited the increase in γ-H2AX. CIP pretreatment reduced Bcl-2 production but promoted p53 phosphorylation, caspase-3 activation and cell death. In NH32 cells, CIP failed to significantly inhibit the radiation-induced γ-H2AX increase, suggesting that CIP inhibition involves in p53-dependent mechanisms. In normal healthy human PBMCs, CIP failed to block the radiation-induced γ-H2AX increase but effectively increased Bcl-2 production, but blocked the phospho-p53 increase and subsequent cell death. CIP increased Gadd45a, and enhanced p21 protein 24 h postirradiation. Results suggest that CIP exerts its effect in TK6 cells by promoting p53 phosphorylation and inhibiting Bcl-2 production and in PBMCs by inhibiting p53 phosphorylation and increasing Bcl-2 production. Our data are the first to support the view that CIP may be effective to protect normal tissue cells from radiation injury, while enhancing cancer cell death in radiation therapy. Keywords Radiation * γ-H2AX * p53 * p21 * Gadd45α * Bax * Bcl-2 * Caspase-3 * Cell viability