Targeting of AKT1 enhances radiation toxicity of human tumor cells by inhibiting DNA-PKcs-dependent DNA double-strand break repair

M Toulany, R Kehlbach, U Florczak, A Sak… - Molecular cancer …, 2008 - AACR
M Toulany, R Kehlbach, U Florczak, A Sak, S Wang, J Chen, M Lobrich, HP Rodemann
Molecular cancer therapeutics, 2008AACR
We have already reported that epidermal growth factor receptor/phosphatidylinositol 3-
kinase/AKT signaling is an important pathway in regulating radiation sensitivity and DNA
double-strand break (DNA-dsb) repair of human tumor cells. In the present study, we
investigated the effect of AKT1 on DNA-dependent protein kinase catalytic subunit (DNA-
PKcs) activity and DNA-dsb repair in irradiated non-small cell lung cancer cell lines A549
and H460. Treatment of cells with the specific AKT pathway inhibitor API-59CJ-OH (API; 1-5 …
Abstract
We have already reported that epidermal growth factor receptor/phosphatidylinositol 3-kinase/AKT signaling is an important pathway in regulating radiation sensitivity and DNA double-strand break (DNA-dsb) repair of human tumor cells. In the present study, we investigated the effect of AKT1 on DNA-dependent protein kinase catalytic subunit (DNA-PKcs) activity and DNA-dsb repair in irradiated non-small cell lung cancer cell lines A549 and H460. Treatment of cells with the specific AKT pathway inhibitor API-59CJ-OH (API; 1-5 μmol/L) reduced clonogenic survival between 40% and 85% and enhanced radiation sensitivity of both cell lines significantly. As indicated by fluorescence-activated cell sorting analysis (sub-G1 cells) and poly(ADP-ribose) polymerase cleavage, API treatment or transfection with AKT1-small interfering RNA (siRNA) induced apoptosis of H460 but not of A549 cells. However, in either apoptosis-resistant A549 or apoptosis-sensitive H460 cells, API and/or AKT1-siRNA did not enhance poly(ADP-ribose) polymerase cleavage and apoptosis following irradiation. Pretreatment of cells with API or transfection with AKT1-siRNA strongly inhibited radiation-induced phosphorylation of DNA-PKcs at T2609 and S2056 as well as repair of DNA-dsb as measured by the γ-H2AX foci assay. Coimmunoprecipitation experiments showed a complex formation of activated AKT and DNA-PKcs, supporting the assumption that AKT plays an important regulatory role in the activation of DNA-PKcs in irradiated cells. Thus, targeting of AKT enhances radiation sensitivity of lung cancer cell lines A549 and H460 most likely through specific inhibition of DNA-PKcs-dependent DNA-dsb repair but not through enhancement of radiation-induced apoptosis. [Mol Cancer Ther 2008;7(7):1772–81]
AACR